CA2637175C - Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution - Google Patents

Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution Download PDF

Info

Publication number
CA2637175C
CA2637175C CA2637175A CA2637175A CA2637175C CA 2637175 C CA2637175 C CA 2637175C CA 2637175 A CA2637175 A CA 2637175A CA 2637175 A CA2637175 A CA 2637175A CA 2637175 C CA2637175 C CA 2637175C
Authority
CA
Canada
Prior art keywords
water solution
orp water
solution
orp
reductive potential
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA2637175A
Other languages
French (fr)
Other versions
CA2637175A1 (en
Inventor
Hojabr Alimi
Andres Gutierrez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oculus Innovative Sciences Inc
Original Assignee
Oculus Innovative Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38198117&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2637175(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Oculus Innovative Sciences Inc filed Critical Oculus Innovative Sciences Inc
Publication of CA2637175A1 publication Critical patent/CA2637175A1/en
Application granted granted Critical
Publication of CA2637175C publication Critical patent/CA2637175C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/20Elemental chlorine; Inorganic compounds releasing chlorine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/40Peroxides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Provided is a method for preventing or treating inflammation and associated states (e.g. infection, hypersensitivity, pain) by administering a therapeutically effective amount of an oxidative reduction potential (ORP) water solution that is stable for at least about twenty-four hours. The ORP water solution administered in accordance with the invention can be combined with one or more suitable carriers and can be administered in conjunction with one or more additional therapeutic agents.

Description

METHODS OF TREATING OR PREVENTING INFLAmivIATIoN. AND
HYPERSENSITIVITY WITH
OXIDATIVE REDUCTIVE POTENTIAL WATER SOLUTION
;
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] [BLANK]
BACKGROUND OF THE INVENTION
[0002] Inflammation is a biological response that can result from a noxious stimulus and is normally intended to remove that stimulus or ameliorate its effects.
Although normally intended to promote survival, inflammation can cause damage te the host, especially in mammals. The stimulus or insult initiating inflammation can be caused by endogenous factors (e.g., an auto-antigen or irritating body fluid) or exogenous factors (e,g., a foreign body or infectious agent).
[0003] Inflammation has been classified as "acute" and "chronic." Acute inflammation is typically of relatively short duration, lasting minutes to hours and, in some cases, a few days, Acute inflammation can be characterized by the exudation of fluid and plasma proteins and the accumulation of polymorphonaclear leukocytes (PMNLs) at the site of the insult. Acute inflammation usually includes an increase in blood flow to the area of the insult mediated by cellular molecules released in response to the insult.' Increased vascular permeability also results from cellular mediators and leads to an accumulation of protein-rich fluid. Important mediators of this increased blood flow and vascular permeability include histamine from mast cells, serotonin and bradykinin.
[0004] In acute inflammation, PMNL5 are also attracted to the area of insult and migrate out of the blood stream toward the insult. The PMNIds release toxic metabolites and proieinases that can cause tissue damage. These proteinases include proteins in the complement system, which can damage cell membranes and kallikrains which generate bradykinin. Acute inflammation can undergo complete resolution, lead to the formation of an abscess, result in scarring fibrosis or progress to chronic inflammation.
[00051 Chronic inflammation is of longer duration, lasting weeks to months, and possibly years, in which tissue destruction and biological processes that are intended to repair the injury are simultaneously ongoing. Chronic inflammation more typically involves lymphocytes and macrophages and may also include a proliferation of blood vessels, fibrosis and/or necrosis. Chronic inflammation can result from a number of conditions including persistent infections, prolonged exposure to toxic agents, and - - wpatrA 'WIMP) A.116,Ci 111I Flo- -L.
I

autoimmune reactions. Chronic inflammation is often maintained by the production of cytokines by lymphocytes and macrophages at the site of the persistent insult.
Chronic inflammation can result in permanent tissue damage or complete healing.
[0006] Hypersensitivity generally refers to inflammation that causes damage to the host, in which the damage outweighs the benefit to the host. Hypersensitivity can result in significant pathology including, e.g., anaphylaxis, transplant rejection, and autoimmune diseases. The most common type of hypersensititvity is allergy.
[0007] Independently of the inducing factor -and the length of the exposure-an inflammatory reaction is mediated by a varied number and type of cells and molecules, the later including cytokines, growth factors, clotting factors, enzymes, neurotransmitters and complement proteins, among others. These molecules are primarily secreted by fibroblasts, endothelial and infiltrating cells (e.g. macrophages, lymphocytes, mast cells, polymorphonuclear cells, etc), and local nerves in response to the insulting agent. The mixture and amount of cytokines therein released will depend on the type, concentration and exposure time of the inducing agent. Therefore, these proteins could mediate from an acute local inflammatory reaction to systemic life-threatening responses (e.g.
acute systemic inflammatory response syndrome, SIRS; multiple organ failure as in septic shock;
anaphylaxis, etc). In chronic inflammatory processes, the cytokines continuously recruit more and more infiltrating cells that generate, for example, granulomas, induration of the tissues, and encapsulated abscesses. In any case, proteins secreted during an inflammatory process are central players in the grade and persistence of the final reaction.
[0008] Stimulation of the aforementioned cells by the induction agent leads to a cascade of intracellular signaling events that ultimately result in production and secretion of cytokines and other inflammatory mediators that constitute the pro-inflammatory response.
While the pro-inflammatory response is crucial for effective clearance of the pathogen or allergen, the inflammatory mediators produced cause tissue damage and inflammation.
Hence, a balance needs to be maintained between the activation and down-regulation of this response in order to avoid severe tissue damage (Cohen, J. :The immunopathogenesis of sepsis. Nature 2002 420, 885-891). Dysregulation of this response could induce local damage (e.g. lung fibrosis) or could lead to potentially lethal conditions like septic shock and systemic inflammatory response syndrome (SIRS) as previously mentioned.
Thus, microbes allergens, endotoxins, and many other molecules induce the production of pro-inflammatory mediator proteins by different cells in the human body. The combined effects of all these molecules in living tissues could mediate changes in the clotting system, wound healing process, anti-microbial activity, antibody production and the perception of pain, among many other reactions.

[0009] The systemic inflammatory response syndrome (SIRS), a syndrome that encompasses the features of systemic inflammation without end-organ damage or identifiable bacteremia. SIRS is separate and distinct from sepsis, severe sepsis or septic shock. The key transition from SIRS to sepsis is the presence of an identified pathogen in the blood. The pathophysiology of SIRS includes, but is not limited to, complement activation, cytokine and arachidonic acid metabolites secretion, stimulated cell-mediated immunity, activation of the clotting cascades, and humoral immune mechanisms.
Clinically SIRS is characterized by tachycardia, tachypnea, hypotension, hypoperfusion, oliguria, leukocytosis or leukopenia, pyrexia or hypothermia, metabolic acidosis, and the need for volume support. SIRS may affect all organ systems and may lead to multiple organ dysfunction syndrome (MODS). Thus, even in early stages (i.e. SIRS), there is accumulation of pro-inflammatory cytokines at the primary site of inflammation and in the blood that can contribute to the establishment of multi-organ failure and death.
[0010] Typically, inflammation is treated with steroidal or non-steroidal anti-inflammatory drugs. However, conventional anti-inflammatory therapy suffers from several drawbacks, e.g., systemic toxicity, allergic reactions, insulin resistance, hypertension, cardiac toxicity, renal toxicity, various coagulopathies and gastric erosions.
Accordingly, there is a need for mild, yet safe and effective methods for treating or preventing inflammation.
[0011] The present invention provides such methods. These and other advantages of the invention, as well as additional inventive features, will be apparent from the description of the invention provided herein.
BRIEF SUMMARY OF THE INVENTION
[0012] The present invention provides a method of preventing or treating inflammation in a patient by administering to the patient a therapeutically effective amount of an oxidative reductive potential (ORP) water solution, wherein the solution is stable for at least twenty-four hours. The method of the present invention can be used in the treatment of inflammation resulting from a variety of causative factors, e.g., allergic reaction, autoimmtme reaction, infection, contact with one or more inflammation-causing substances, and combinations of such causative factors.
[0013] The method of the present invention can farther include administering the ORP
water solution in conjunction with one or more therapeutic agents, e.g., one or more compounds selected from the group consisting of antibiotics, anti-viral agents, anti-inflammatory agents, and combinations thereof. Administering such therapeutic agents in conjunction with the ORP water solution includes administering one or more of such agents, e.g., prior to, during (e.g., contemporaneously, by co-administration or in combination with), or following administration of the ORP water solution.
[0014] The ORP water solution can be administered by any suitable route in accordance with the present invention, e.g., by delivering the ORP water solution topically or parenterally, so as to contact a therapeutically effective amount of the ORP
water solution with one or more affected tissues, which may reside inside or outside of the body.
Accordingly, the invention provides a method wherein the ORP water solution is administered to one or more tissues, e.g., nasal, sinus, pharyngeal, tracheal, pulmonary, esophageal, gastric, intestinal, mesothelial, peritoneal, synovial, urinary bladder, urtheral, vaginal, uterine, fallopian, pancreatic, nervous, oral, cutaneous, and subcutaneous. The ORP water solution can be administered in any suitable faun in accordance with the present invention, e.g., as a liquid, spray, mist, aerosol or steam, and, if desired, can be combined with one or more suitable carriers, e.g., vehicles, adjuvants, excipients, diluents, and the like.
[0015] The ORP water solution administered in accordance with the present invention can be contained within a sealed container and is stable for at least twenty-four hours. The ORP water solution administered in accordance with the invention can be produced by electrolysis, and preferably comprises a mixture of anode water and cathode water, which contains one or more species, including, e.g., reactive species, ionic species, radical species, precursors thereof and combinations thereof. The ORP water solution administered in accordance with the invention exhibits potent anti-inflammatory activity, yet is virtually free of toxicity to noiinal tissues and noinial eukaryotic cells.
BRIEF DESCRIPTION OF THE DRAWINGS
[0016] FIG. 1 illustrates a three-chambered electrolysis cell for producing an exemplary ORP water solution.
[0017] FIG. 2 illustrates a three-chambered electrolysis cell and depicts ionic species that are believed to be generated during the production process.
[0018] FIG. 3 is a schematic flow diagram of a process for producing an exemplary ORP water solution.
[0019] FIG. 4A-4C depict a graphical comparison of cell viability, apoptosis and necrosis in human diploid fibroblasts (HDFs) treated with an exemplary ORP
water solution (MCN) versus hydrogen peroxide (HP).
[0020] FIG. 5 is a graphical comparison of the levels of 8-hydroxy-2'-deoxiguanosine (8-0HdG) adducts in HDFs treated with an exemplary ORP water solution (MCN) versus 500 p,M hydrogen peroxide (HP).

[0021] FIG. 6 illustrates cellular senescence demonstrated by 13-galactosidase expression in HDFs after chronic exposure to low concentrations of an exemplary ORP
water solution (MCN) versus hydrogen peroxide (HP).
[00221 FIG. 7 illustrates the effect on degranulation of antigen-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
[00231 FIG. 8 comparatively illustrates the effect on degranulation of antigen-activated mast cells treated with cromoglycate.
[00241 FIG. 9 illustrates the effect on degranulation of antigen-activated and calcium ionophore (A23187)-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
[0025] FIG. 10A-10B are RNAse protection assays illustrating cytokine mRNA
levels after antigen challenge in control versus ORP water solution-treated mast cells.
[00261 FIG. 11 is a graphical comparison of TNF-a secretion by antigen-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
[0027] FIG. 12 is a graphical comparison of MIP1-a secretion by antigen-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
[00281 FIG. 13 is a graphical comparison of IL-6 secretion by antigen-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
[0029] FIG. 14 is a graphical comparison of IL-13 secretion by antigen-activated mast cells treated with various concentrations of an exemplary ORP water solution (MCN).
DETAILED DESCRIPTION OF THE INVENTION
[0030] The present invention provides a method of preventing or treating inflammation in a patient, which method comprises administering to the patient a therapeutically effective amount of an oxidative reductive potential (ORP) water solution (also known as super-oxidized water (SOW)), wherein the solution is stable for at least about twenty-four hours.
The method of the present invention can be used for treating or preventing (e.g., inhibiting the onset of, inhibiting the escalation of, decreasing the likelihood of) acute inflammation and chronic inflammation, including hypersensitivity such as, e.g., in allergies. The inflammation and hypersensitivity treatable or preventable in accordance with the method of the present invention can include inflammation that results from, e.g., contact with a noxious stimulus, injury, infection, autoimmune reaction, hypersensitivity, and allergic reaction, including allergic reactions associated with cellular histamine and pro-inflammatory cytokine release.
[0031] Surprisingly, it has been found that the ORP water solution administered in accordance with the invention is a highly effective inhibitor of mast cell degranulation, one of the primary inflammation and allergy-causing biological cascades. The ORP
water solution administered in accordance with the invention inhibits degranulation of mast cells regardless of whether they are activated with an antigen or a calcium ionophore. Also surprisingly, it has been found that the ORP water solution administered in accordance with the present invention non-selectively inhibits the secretion of pro-inflammatory cytokines in mast cells. For example, the ORP water solution of the present invention can inhibit the secretion of, e.g., TNF-a, MIP1-a, IL-6, and IL-13 in mast cells. It is believed that the ORP
water solution administered in accordance with the invention also can inhibit the secretion of pro-inflammatory cytokines in other cytokine-secreting cells including, but not limited to, macrophages, monocytes, lymphocytes, macrophages, PMN, fibroblasts and endothelial cells. . These findings demonstrate that the ORP water administered in accordance with the present invention should exhibit broad anti-inflammatory efficacy.
[0032] The ORP water solution administered in accordance with the invention preferably inhibits mast cell degranulation by more than about 50% relative to untreated mast cells, more preferably by more than about 80% relative to untreated mast cells, still more preferably by more than about 90% relative to untreated mast cells, and even more preferably by more than about 95% relative to untreated mast cells, when contacted with the ORP water solution for up to about 30 minutes, more preferably up to about 15 minutes, and still more preferably up to about 5 minutes.
[0033] The ORP water solution administered in accordance with the invention also preferably inhibits the secretion of TNF-a by more than about 50%, more preferably by more than about 60%, still more preferably by more than about 70%, and even more preferably by more than about 85%. In addition, the ORP water solution administered in accordance with the invention also preferably inhibits the secretion of MLP1-a by more than 25%, more preferably by more than about 50%, and still more preferably by more than about 60%. Further, the ORP water solution administered in accordance with the invention also preferably inhibits the secretion of IL-6 and/or IL-13 by more than 25%, more preferably by more than about 50%, and still more preferably by more than about 60%. In accordance with the method of the invention, secretion of these and that of other cytokines, can be therapeutically inhibited down to certain % by the administration of the ORP water solution alone or in combination with a diluent (e.g., water), by increasing the concentration of the components of the ORP water solution, by utilizing special delivery systems and /or by increasing the exposure time. For instance, cytokine secretion can be therapeutically inhibited by administering compositions in which the ORP water solution is diluted, e.g., by a ratio of up to about 50% (vol./vol.) ORP water solution/diluent, by a ratio of up to about 25% (vol./vol.) ORP water solution/diluent, by a ratio of up to about 10%
(vol./vol.) ORP

water solution/diluent, by a ratio of up to about 5 % (vol./vol.) ORP water solution/diluent, or even by a ratio of up to about 1% (vol./vol.) ORP water solution/diluent.
[0034] The method of the present invention can be used for treating or preventing cell-mediated inflammation , which results from an autoimmune reaction, including, but not limited to, SLE, autoimmune thyroiditis, sarcoidosis, inflammatory bowel disease, rheumatoid arthritis, rheumatic fever, psoriasis, pemphigus, erythema multifonne, other bullous diseases of the skin, and atopias. The method of the invention can be used for treating or preventing inflammation, which results from infection, allergens, foreign bodies, and autoimmune processes. The method of the invention can also be used for treating or preventing inflammation, which results from infection, e.g., from an infection by one or more microorganisms selected from the group consisting of viruses, bacteria, and fungi, including hypersensitivity and autoimmune-mediated inflammation resulting from infection.
[0035] The method of the present invention can be used for treating or preventing inflammation associated with an upper respiratory condition. When the inflammation is associated with an upper respiratory condition, the ORP water solution is preferably administered to the upper airway, e.g., as a spray, mist, aerosol or steam, so as to contact one or more upper airway tissues affected by the condition. Any suitable method can be employed for delivering the ORP water solution to the upper airway so as to treat or prevent one or more upper respiratory conditions in accordance with the present invention, including one or more routes of administration described herein.
[0036] The method of the present invention can be used for preventing or treating inflammation affecting one or more upper respiratory airway tissues (e.g., nasal tissue, sinus tissue) or lung tissues. Such conditions can include, for example, sinusitis (e.g., rhinosinusitis, acute sinusitis, chronic sinusitis, and the like), pharyngitis, asthma, and the like, which are preventable or treatable with the ORP solution administered in accordance with the invention.
[0037] Chronic sinusitis typically refers to inflammation of the sinuses that continues for at least 3 weeks, but the inflammation can (and often does) continue for months or even years. Allergies are frequently associated with chronic sinusitis. In addition, patients with asthma have a particularly high frequency of chronic sinusitis. Inhalation of airborne allergens (substances that provoke an allergic reaction), such as dust, mold, and pollen, often set off allergic reactions (e.g., allergic rhinitis) that, in turn, may contribute to sinusitis (particularly rhinosinusitis or rhinitis). People who are allergic to fungi can develop a condition called "allergic fungal sinusitis." Damp weather or pollutants in the air and in buildings also can affect people subject to chronic sinusitis.

[0038] Like acute sinusitis, chronic sinusitis is more common in patients with immune deficiency or abnoimalities of mucus secretion or movement (e.g., immune deficiency, HIV
infection, cystic fibrosis, Kartagener's syndrome). In addition, some patients have severe asthma, nasal polyps, and severe asthmatic responses to aspirin and aspirin-like medications (so-called non-steroidal anti-inflammatory drugs, or NSAIlls). These latter patients have a high frequency of chronic sinusitis.
[0039] A doctor can diagnose sinusitis by medical history, physical examination, X-rays, and if necessary, MRIs or CT scans (magnetic resonance imaging and computed tomography). After diagnosing sinusitis and identifying a possible cause, a doctor can prescribe a course of treatment that will reduce the inflammation and relieve the symptoms. Treating acute sinusitis typically requires re-establishing drainage of the nasal passages, controlling or eliminating the source of the inflammation, and relieving the pain.
Doctors generally recommend decongestants to reduce the congestion, antibiotics to control a bacterial infection, if present, and pain relievers to reduce the pain.
[0040] When treatment with drugs fails, surgery may be the only alternative for treating chronic sinusitis, e.g., removal of adenoids, removal of nasal polyps, repair of a deviated septum, endoscopic sinus surgery, and the like. It is believed that the administration of ORP water in accordance with the method of the present invention can be used for treating chronic sinusitis and inflammation associated therewith as an alternative to potentially avoid more aggressive therapies, such as antibiotics and surgery.
[0041] With regard to pharyngitis, it is estimated that worldwide, 1 to 2%
of all visits to doctors' offices, clinics and emergency rooms are because of pharyngitis. In the United States and Mexico, pharyngitis and tonsillitis is believed to account for about 15 and 12 million consultations per year, respectively. These cases are typically caused by various bacteria and viruses. Also, pharyngitis and tonsillitis caused by group A 13-hemolytic Streptococcus can significantly raise the risk of rheumatic fever in poor populations;
however it is believed that only 5 to 15% of pharyngitis cases are caused by this bacterium, and that the rest of the acute cases are due to bacteria and viruses of little epidemiological relevance. The latter cases tend to be self-limiting in a few days and do not leave sequelae.
[0042] It has been verified that a great number of doctors worldwide prescribe antibiotics indiscriminately for acute pharyngitis. This occurs in a daily practice, often because patients tend to request powerful antibiotics. Unfortunately, it is difficult to establish an accurate diagnosis of streptococcal pharyngitis/tonsillitis clinically and the cost/benefit ratio of treating acute pharyngitis/tonsillitis with antibiotics is questionable.
[0043] It is believed that the method of the present invention provides a safe, efficacious and cost-effective adjuvant therapy for the treatment or prevention of acute pharyngitis andior tonsillitis due to bacteria and/or viruses. . The empirical treatment of acute pharyngitis/tonsillitis may begin with administering an ORP water solution in accordance with the present invention, and, depending on evolution or the result of the rapid test for Streptococcus, antibiotics may be initiated from 48-72 hours thereafter only if needed. The method of the present invention may thus allow the use of antibiotics to be deferred and, at the seine time, reduce the symptomatology of the patient and accelerate the patient's recovery if the pharyngitis/tonsillitis is not from group A Streptococcus. The adjuvant use of an ORP water solution of the present invention with antibiotics for the treatment of streptococcal pharyngitis/tonsillitis also .may shorten the period of clinical response and decrease the incidence of recurrences.
[0044] The method of the present invention also can be used for treating or preventing inflammation associated with hypersensitivity. Historically, hypersensitivity reactions have been classified as one of four types, from which significant disease can result, The ORP
water solution administered in accordance with the invention can be used to treat and/or prevent (e.g., inhibit the onset of, inhibit the escalation of or decrease the likelihood of) one or more of such reactions. Type I hypersensitivity typically results from the combination of an antigen with an antibody bound to a mast cell or basophil. Type I reactions occur within minutes of exposure to the antigen in individuals who have been previously sensitized to the antigen. In humans, Type I reactions are mediated by IgE which has high affinity Pc receptors on mast cells and basophils, [0045] Mast cells' role in Type I hypersensitivity is especially important because they reside in tissues under the epithelial surface near blood vessels and nerves.
Multiple clinical symptoms observed in atopic dermatitia, allergic rhinitis and atopic asthma are produced by IgE-antigen stimulation of mast cells located in distinct affected tissues.
The currently accepted view of the pathogenesis of atopic asthma is that allergens initiate the process by triggering IgE-bearing pulmonary mast cells (NICs) to release mediators such as histamine, leukotrienes, prostaglandins, kininis, platelet activating factor (PAF), etc.
in the so-called early phase of the reaction (see Kumar et al., Robbins 84 Caren Pathologic Basis of Disease, 2004, pp. 193-268. In turn, these mediators induce bronchoconstriction and enhance vascular permeability and mucus production. According to this model, following mast cell activation in the late phase, those cells secrete various cytokines, including tumor necrosis factor alpha (TNP-a), IL-4, IL-5 and 1L-6, which participate in the local recruitment and activation of other inflammatory cells such as eosinophils, basophils, T lymphocytes, platelets and mononuclear phagocytes.
These recruited cells, in turn, contribute to the development of an inflammatory response that may then become autonomous and aggravate the asthmatic symptoms, This late phase _ -. _ response constitutes a long term inflammatory process which will induce changes in surrounding tissues (Kumar et al., pp. 193-268). Clinically, Type I reactions can have local effects such as allergic rhinitis, or systemic effects as is found in anaphylaxis which manifests with itching, hives, respiratory distress, and circulatory collapse.
[00461 Type II hypersensitivity is mediated by antibodies directed to antigens on the surfaces of cells and in the extracellular space. These antibodies can direct cell lysis or result in opsonization of the target molecules (preparation for phagocytosis by other cells).
Alternatively, the antibodies can be directed to and activate cell surface receptors.
Conditions resulting from Type II reactions include transfusion reactions, Graves disease (thyrotoxicosis), drug reactions, pernicious anemia, and acute rheumatic fever. In rheumatic fever the antibodies are formed against Streptococcal antigens but, cross-react with human tissues such as heartyalves.
[0047] Type III hypersensitivity is caused by immune complexes, which are combinations of antibodies and other host immune system proteins, most typically complement proteins. It is the normal function of antibodies to bind and active complement. However, when the resulting macromolecular immune complexes are not adequately processed, they can lead to persistent tissue damage. Macrophages and PMNLs can be activated by immune complexes and lead to the release of toxic chemicals by these cells. Immune complex reactions can be local and may result in conditions such as, e.g., the arthus reaction or cause systemic disease such as serum sickness or some of the aspects of systemic lupus erythematous (SLE).
[0048] Type IV hypersensitivity is cell mediated and is sometimes called delayed-type hypersensitivity. Type IV hypersensitivity is mediated by T lymphocytes and often results in the formation of a granulomatous reaction. In a granulomatous reaction, a form of macrophage called an epitheloid cells attempts to, but fails, to digest an antigen. The antigen's persistence leads to the release of cytokines that attract additional lymphocytes resulting in chronic foci of inflammation. The foci have high concentrations of cyotoxic T-lymphocytes which release granzymes and perforins which are toxic to adjacent cells. Type IV hypersensitivity is a prominent component of autoimmune diseases such as, e.g., Sjogrren's Syndrome, Sarcoidosis, and contact dermatitis.
[0049] Pathologic states can combine different types of hypersensitivity reactions. In autoimmune diseases host antigens stimulate hypersensitivity with serious consequences for the host. For example, in SLE host antigens induce Type II reactions against blood cells while Type III reactions lead to blood vessel and renal glomerular damage. In addition, hypersensitivity reactions are also seen in iatragenic conditions such as drug reactions and transplant rejection. Transplant rejection includes components of Type II and Type IV

hypersensivity. Accordingly, ORP water solution used in accordance with the invention in transplantable organs or cells could greatly reduced the possibility of being rejected by the host.
[0050] It has been found that the ORP water solution administered in accordance with the invention is virtually free of toxicity to normal tissues and normal mammalian cells.
The ORP water solution administered in accordance with the invention causes no significant decrease in the viability of eukaryotic cells, no significant increase in apoptosis, no significant acceleration of cell aging and/or no significant oxidative DNA
damage in mammalian cells. The non-toxicity is particularly advantageous, and perhaps even surprising, given that the disinfecting power of the ORP water solution administered in accordance with the invention is roughly equivalent to that of hydrogen peroxide, yet is significantly less toxic than hydrogen peroxide is to normal tissues and normal mammalian cells. These findings demonstrate that the ORP water solution administered in accordance with the present invention is safe for use, e.g., in mammals, including humans.
[0051] For the ORP water solution administered in accordance with the invention, the cell viability rate is preferably at least about 65%, more preferably at least about 70%, and still more preferably at least about 75% after an about 30 minute exposure to the ORP water solution. In addition, the ORP water solution administered in accordance with the invention preferably causes only up to about 10% of cells, more preferably only up to about 5% of cells, and still more preferably only up to about 3% of cells to expose Annexin-V on their cellular surfaces when contacted with the ORP water solution for up to about thirty minutes or less (e.g., after about thirty minutes or after about five minutes of contact with the ORP
water solution).
[0052] Further, the ORP water solution administered in accordance with the invention preferably causes less than about 15% of cells, more preferably less than about 10% of cells, and still more preferably less than about 5% of cells to express the SA-13-galactosidase enzyme after chronic exposure to the OPR water solution. The ORP water solution administered in accordance with the invention preferably causes caused the same fraction of the oxidative DNA adduct formation caused by saline solution, e.g., less than about 20% of the oxidative DNA adduct formation, less than about 10% of the oxidative DNA
adduct formation, or about 5% or less of the oxidative DNA adduct formation normally caused by hydrogen peroxide in cells treated under equivalent conditions.
[0053] The ORP water solution administered in accordance with the invention produces no significant RNA degradation. Accordingly, RNA extracted from human cell cultures after an about 30 minutes exposure to the ORP water solution or r at about 3 hours after an about 30 minute-exposureõ and analyzed by denaturing gel electrophoresis, will typically show no significant RNA degradation and will typically exhibit two discreet bands corresponding to the ribosomal eukaryotic RNAs (i.e. 28S and 18S) indicating that the ORP
water solution administered in accordance with the invention leaves the RNA
substantially intact. Similarly, RNA extracted from human cell cultures after about 30 minutes of exposure to the ORP water solution or after about 3 hours of exposure, can be subjected reverse transcription and amplification (RT-PCR) of the constitutive human GAPDH
(Glyceraldehyde-3-phosphate dehydrogenase) gene and result in a strong GAPDH
band on gel electrophoresis of the RT-PCR products. By contrast, cells treated with HP
for a similar period show significant RNA degradation and little if any GAPDH RT-PCR
product.
[0054] The ORP water solution used in accordance with the present invention can be administered using any suitable method of administration known in the art. For instance, the ORP water solution can be administered parenterally, endoscopically or directly to the surface of any affected biological tissue, e.g., to the skin and/or one or more mucosal surfaces. Parenteral administration can include using, for example, administering the ORP
water solution intramuscularly, subcutaneously, intravenously, intra-arterially, intrathecally, intravesically or into a synovial space. Endoscopic administration of the ORP
water solution can include using, e.g., bronchoscopy, colonoscopy, sigmoidoscopy, hysterscopy, laproscopy, athroscopy, gastroscopy or a transurethral approach. Administering the ORP
water solution to a mucosal surface can include, e.g., administration to a nasal, oral, tracheal, bronchial, esophageal, gastric, intestinal, peritoneal, urethral, vesicular, urethral, vaginal, uterine, fallopian, and synovial mucosal surface.
[0055] Parenteral administration also can include administering the ORP
water solution used in accordance with the invention intravenously, subcutaneously, intramuscularly, or intraperitoneally. The ORP water solution of the present invention can be administered intravenously as described, e.g., in U.S. Patent Nos. 5,334,383 and 5,622,848 (hereby incorporated by reference), which describe methods of treating viral myocarditis, multiple sclerosis, and AIDS via intravenous administration of ORP water solutions.
Other applications include the treatment of any hypersensitivity and infectious processes, as mentioned above.
[0056] The ORP water solution used in accordance with the invention can be administered topically, e.g., as a liquid, spray, mist, aerosol or steam by any suitable process, e.g., by aerosolization, nebulization or atomization. The ORP
solution of the present invention can be administered to the upper airway as a steam or a spray. When the ORP water solution is administered by aerosolization, nebulization or atomization, it is preferably administered in the form of droplets having a diameter in the range of from about 0.1 micron to about 100 microns, preferably from about 1 micron to about 10 microns. In one embodiment, the method of the present invention includes administering the ORP water solution in the form of droplets having a diameter in the range of from about 1 micron to about 10 microns to one or more mucosal tissues, e g., one or more upper respiratory tissues and/or lung tissues.
[0057] Methods and devices, which are useful for aerosolization, nebulization and atomization, are well known in the art. Medical nebulizers, for example, have been used to deliver a metered dose of a physiologically active liquid into an inspiration gas stream for inhalation by a recipient. See, e.g., Patent No. 6,598,602.
Medical nebulizers can operate to generate liquid droplets, which form an aerosol with the inspiration gas. In other circumstances medical nebulizers may be used to inject water droplets into an inspiration gas stream to provide gas with a suitable moisture content to a recipient, which is particularly useful where the inspiration gas stream is provided by a mechanical breathing aid such as a respirator, ventilator or anaesthetic delivery system.
10058] An exemplary nebulizer is described, for example, in WO 95/01137, which describes a hand held device that operates to eject droplets of a medical liquid into a passing air stream (inspiration gas stream), which is generated by a recipient's inhalation through a mouthpiece. Another example can-be found in U.S. Patent No, 5,388,571.
which describes a positive-pressure ventilator system which provides control and augmentation of breathing for a patient with respiratory insufficiency and which includes a nebulizer for delivering particles of liquid medication into the airways and alveoli of the lungs of a patient. U.S. Patent No. 5,312,281 describes an ultrasonic wave nebulizer, which atomizes water or liquid at low temperature and reportedly can adjust the size of mist. In addition, U.S.
Patent No.
5,287,847 describes a pneumatic nebulizing apparatus with scalable flow rates and output volumes for delivering a medicinal aerosol to neonates, children and adults, Further, U.S. Patent No. 5,063,922 describes an ultrasonic atomizer. The ORE water solution also may be dispensed in aerosol form as part of an inhaler system for treatment of infections in the lungs and/or air passages or for the healing of wounds in such parts of the body.
[00591 For larger scale applications, a suitable device may be used to disperse the ORP
water solution into the air including, but not limited to, humidifiers, misters, foggers, vaporizers, atomizers, water sprays, and other spray deviees. Such devices permit the dispensing of the ORE water solution on a continuous basis. An ejector which directly mixes air and water in a nozzle may be employed. The ORE water solution may be converted to steam, such as low pressure steam, and released into the air stream, Various _ .
. - .

types of humidifiers may be used such as ultrasonic humidifiers, stream humidifiers or vaporizers, and evaporative humidifiers. The particular device used to disperse the ORP
water solution may be incorporated into a ventilation system to provide for widespread application of the ORP water solution throughout an entire house or healthcare facility (e.g., hospital, nursing home, etc.).
[0060] In accordance with the invention, the ORP water solution can be administered alone or in combination with one or more pharmaceutically acceptable carriers, e.g., vehicles, adjuvants, excipients, diluents, combinations thereof, and the like, which are preferably compatible with one or more of the species that exist in the ORP
water solution.
One skilled in the art can easily determine the appropriate formulation and method for administering the ORP water solution used in accordance with the present invention. Any necessary adjustments in dose can be readily made by a skilled practitioner to address the nature and/or severity of the condition being treated in view of one or more clinically relevant factors, such as, e.g., side effects, changes in the patient's overall condition, and the like.
[0061] For example, the ORP water solution can be formulated by combining or diluting the ORP water solution with up to about 25% (wt./wt. or vol./vol.) of a suitable carrier, up to about 50% (wt./wt. or vol./vol.) of a suitable carrier, up to about 75% (wt./wt.
or vol./vol.) of a suitable carrier, up to about 90% (wt./wt. or vol./vol.) of a suitable carrier, up to about 95% (wt./wt. or vol./vol.) of a suitable carrier, or even with up to about 99%
(wt./wt. or vol./vol.) or more of a suitable carrier. Suitable carriers can include, e.g., water (e.g., distilled water, sterile water, e.g., sterile water for injection, sterile saline and the like).
Suitable carriers also can include one or more carriers described in U.S.
Patent Application No. 10/916,278 (hereby incorporated by reference). Exemplary formulations can include, e.g., solutions in which the ORP water solution is diluted with sterile water or sterile saline, wherein the ORP water solution is diluted by up to about 25% (vol./vol.), by up to about 50% (vol./vol.), by up to about 75% (vol./vol.), by up to about 90%
(vol./vol.), by up to about 95% (vol./vol.), or by up to 99% (vol./vol.) or more of a suitable carrier.
[0062] The ORP water solution administered in accordance with the invention can further be combined with (or be administered in conjunction with) one or more additional therapeutic agents, e.g., one or more active compounds selected from the group consisting of antibacterial agents (e.g., antibiotics), anti-viral agents, anti-inflammatory agents, and combinations thereof.
[0063] The therapeutically effective amount administered to the patient, e.g., a mammal, particularly a human, in the context of the present invention should be sufficient to affect a therapeutic or prophylactic response in the patient over a reasonable time frame. The dose can be readily determined using methods that are well known in the art. One skilled in the art will recognize that the specific dosage level for any particular patient will depend upon a variety of potentially therapeutically relevant factors. For example, the dose can be determined based on the strength of the particular ORP water solution employed, the severity of the condition, the body weight of the patient, the age of the patient, the physical and mental condition of the patient, general health, sex, diet, the frequency of applications, and the like. The size of the dose also can be determined based on the existence, nature, and extent of any adverse side effects that might accompany the administration of a particular ORP water solution. It is desirable, whenever possible, to keep adverse side effects to a minimum.
[0064] Factors, which can be taken into account for a specific dosage can include, for example, bioavailability, metabolic profile, time of administration, route of administration, rate of excretion, the pharrnacodynamics associated with a particular ORP
water solution in a particular patient, and the like. Other factors can include, e.g., the potency or effectiveness of the ORP water solution with respect to the particular condition to be treated, the severity of the symptoms presented prior to or during the course of therapy, and the like.
In some instances, what constitutes a therapeutically effective amount also can be determined, in part, by the use of one or more of the assays, e.g., bioassays, which are reasonably clinically predictive of the efficacy of a particular ORP water solution for the treatment or prevention of a particular condition.
[0065] The ORP water solution used in accordance with the present invention can be administered, alone or in combination with one or more additional therapeutic agents, to a patient, e.g., a human, e.g., to treat an existing condition. The ORP water solution of the present invention also can be administered prophylactically, alone or in combination with one or more additional therapeutic agents, to a patient, e.g., a human, that has been exposed to one or more causative agents associated with the condition. For example, the ORP
water solution can be suitably administered prophylactically to a patient that has been exposed to one or more inflammation-causing microorganisms (e.g., infections, viruses, bacteria and/or frmgi) -or hypersensitivity epitope or allergen- to inhibit or decrease the likelihood of inflammation (and even infection) associated with the microorganism or epitope in a patient, or decrease the severity of an inflammation (and even infection or allergy) that develops as a result of such exposure.
[0066] One skilled in the art will appreciate that suitable methods of administering the ORP water solution used in accordance with the present invention are available, and, although more than one route of administration can be used, a particular route can provide a more immediate and more effective reaction than another route. The therapeutically effective amount can be the dose necessary to achieve an "effective level" of the ORP
water solution in an individual patient, independent of the number of applieadons a day. The therapeutically effective amount can be defined, for example, as the amount required to be administered to an individual patient to achieve a blood level, tissue level, and/or 1 intracellular level of the ORP water solution (or one or more active species contained therein) to prevent or treat the condition in. the patient.
(0067] 'When the effective level is used as a preferred endpoint for dosing, the actual dose and schedule can vary depending, for example, upon interindividual differences in phannacolcinetics, distribution, metabolism, and the like. The effective level also can vary when the ORP water solution is used in combination with one or more additional therapeutic agents, e.g., one or more anti-infective agents, one or more "moderating,"
"modulating" or 'neutralizing agents," e.g., as described in Patent Nos.
5,334,383 and 5,622,848, one or more anti-inflammatory agents, and the like, [0068] An appropriate indicator can be used for determining and/or monitoring the effective level. For example, the effective level can be determined by direct analysis (e.g.) analytical chemistry) or by indirect analysis (e.g., with clinical chemistry indicators) of appropriate patient samples (e.g., blood and/or tissues). The effective level also can be determined, for example, by direct or indirect observations such as, e.g., the concenrration of urinary metabolites, changes in markers associated with the condition (e.g., viral count in the Case of a viral infection), histopathology and immunochemistry analysis, positive changes in image analysis (e.g. X ray, CT scan, NIVfR, PET, etc), nuclear medicine studies, decrease in the symptoms associated with the conditions, and the like.
(0069] Cenventional ORP water solutions have an extremely limited shelf-life, usually only a few hours. As a result of this short lifeapan, using conventional ORP
water solutions requires the production to take place in close proximity to the point of use.
From a practical standpoint, this means that the facility, e.g., a healthcare facility such as a hospital, must purchase, house and maintain the equipment necessary to produce conventional ORP water solution. Additionally, conventional manufacturing techniques have not been able to produce sufficient commercial-scale quantities to permit widespread use, e.g., as a general disinfecting agent for healthcare facilities.
NOM Unlike conventional ORP water solutions, the ORP water solution administered in accordance with the invention is stable for at least about twenty-hours after its preparation. In addition, the ORP water solution administered in accordance with the invention is generally environmentally safe and, thus, avoids the need for costly disposal procedures. Preferably, the ORP water solution administered in accordance with the ----------------------------------------------------------------------- , invention is stable for at least about one week (e.g., one week, two weeks, three weeks, four weeks or more.), and more preferably at least about two months. Still more preferably, the ORP water solution administered in accordance with the invention is stable for at least about six months. Even more preferably, the ORP water solution administered in accordance with the invention is stable for at least about one year, and most preferably is stable for more than about one year, e.g., at least about two years or at least about three years.
[0071] Stability can be measured based on the ability of the ORP water solution to remain suitable for one or more uses, for example, inhibiting mast cell degranulation, inhibiting cytokine secretion, decontamination, disinfection, sterilization, anti-microbial cleansing, and wound cleansing, for a specified period of time after its preparation under normal storage conditions (e.g., room temperature). The stability of the ORP
water solution administered in accordance with the invention also can be measured by storage under accelerated conditions, e.g., from about 30 C to about 60 C, in which the ORP water solution preferably is stable for up to about 90 days, and more preferably for up to about 180 days.
[0072] Stability also can be measured based on the concentration over time of one or more species (or precursors thereof) present in solution during the shelf-life of the ORP
water solution. Preferably, the concentrations of one or more species, e.g., free chlorine, hypocholorous acid and one or more additional superoxidized water species and are maintained at about 70% or greater of their initial concentration for at least about two months after preparation of the ORP water solution. More preferably, the concentration of one of more of these species is maintained at about 80% or greater of their initial concentration for at least about two months after preparation of the ORP water solution.
Still more preferably, the concentration of one or more of such species is maintained at about 90% or greater, and most preferably is maintained at about 95% or greater, of their initial concentration for at least about two months after preparation of the ORP water solution.
[0073] Stability also can be determined based on the reduction in the amount of organisms present in a sample following exposure to the ORP water solution.
Measuring the reduction of organism concentration can be made on the basis of any suitable organism including, e.g., bacteria, fungi, yeasts, or viruses. Suitable organisms can include, e.g., Escherichia coli, Staphylococcus aureus, Candida albicans, and Bacillus athrophaeus (fofinerly B. subtilis).
[0074] The ORP water solution administered in accordance with the invention can function as a low-level disinfectant capable of a four log (104) reduction in the concentration of live microorganisms, and also can function as a high-level disinfectant capable of a six log (106) reduction in concentration of live microorganisms.
Preferably, the ORP water solution administered in accordance with the invention is capable of yielding at least about a four log (104) reduction in total organism concentration, following exposure for one minute when measured at least about two months after preparation of the solution.
More preferably, the ORP water solution is capable of a 104-106 reduction of organism concentration when measured at least about six months after preparation of the solution.
Still more preferably, the ORP water solution is capable of a 104-106 reduction of organism concentration when measured at least about one year after preparation of the ORP water solution, and most preferably when measured more than about one year, e.g., at least about two years or at least about three years, after preparation of the ORP water solution.
[0075] For instance, the ORP water solution administered in accordance with the present invention can be capable of at least about a five log (105) reduction in the concentration of a sample of live microorganisms from the group consisting of Pseudomonas aeruginosa, Escherichia coil, Enterococcus hirae, Acinetobacter baurnannii, Acinetobacter species, Bacteroides fragilis, Enterobacter aerogenes, Enterococcus faecalis, Vancomycin resistant- Enterococcus faecium (VRE, MDR), Haernophilus influenzae, Klebsiella oxytoca, Klebsiella pneumoniae, Micrococcus luteus, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus haemolyticus, Staphylococcus hominis, Staphylococcus saprophyticus, Streptococcus pneumoniae, Streptococcus pyogenes, Candida albicans within thirty seconds of exposure, when measured at least two months after preparation of the ORP water solution (BioSciences Labs, Montana, US). Preferably, the ORP water solution is capable of achieving a 105 reduction of all these organisms when measured at least about six months after preparation, and more preferably when measured at least about one year after preparation.
[0076] The invention also provides methods for killing bacteria in biofilms, e.g., Pseudomonas aeruginosa in biofilms. The invention further provides methods for killing of Moraexlla catarrhalis and antibotic resistant bacteria, e.g., penicillin resistant Streptococcus. The methods disclosed herein can be used in accordance with the invention for killing bacteria using ORP water solutions faster than with using Bacitracin.
[0077] In one embodiment, the ORP water solution administered in accordance with the invention can reduce a sample of live microorganisms including, but not limited to, Escherichia colt, Pseudomonas aeruginosa, Staphylococcus aureus and Candida albicans, from an initial concentration of between about 1 x 106 and about 1 x 108 organisms/ml to a final concentration of about zero organisms/ml within about one minute of exposure when measured at least about two months after preparation of the ORP water solution. This corresponds to from about a six log (106) to about an eight log (108) reduction in organism concentration. Preferably, the ORP water solution is capable of achieving a reduction of Escherichia coli, Pseudoinonas aeruginosa, Staphylococcus aureus or Candida albicans organisms when measured at least about six months after preparation, and more preferably when measured at least about one year after preparation.
[0078] Alternatively, the ORP water solution administered in accordance with the present invention can produce about a six log (106) reduction in the concentration of a spore suspension of Bacillus athrophaeus spores within about five minutes of exposure when measured at least about two months after preparation of the ORP water solution.
Preferably, the ORP water solution administered in accordance with the invention can achieve about a 106 reduction in the concentration of Bacillus athrophaeus spores when measured at least about six months after preparation, and more preferably when measured at least about one year after preparation.
[0079] The ORP water solution administered in accordance with the invention also can produce about a four log (104) reduction in the concentration of a spore suspension of Bacillus athrophaeus spores within about thirty (30) seconds of exposure when measured at least about two months after preparation of the ORP water solution.
Preferably, the ORP
water solution can achieve this reduction in the concentration of Bacillus athrophaeus spores when measured at least about six months after preparation, and more preferably when measured, at least about one year after preparation.
[0080] The ORP water solution administered in accordance with the invention further can produce about a six log (106) reduction in the concentration of fungal spores, such as Aspergillis niger spores, within about five to about ten minutes of exposure when measured at least about two months after preparation of the ORP water solution.
Preferably, the ORP
water solution can achieve a 106 reductionin the concentration of fungal spores when measured at least about six months after preparation, and more preferably when measured at least about one year after preparation.
[0081] The ORP water solution administered in accordance with the invention can be acidic, neutral or basic, and generally can have a pH of from about 1 to about 14. Within this pH range, the ORP water solution can be safely applied in suitable quantities, e.g., to surfaces without damaging the surfaces or harming objects, such as human skin, that comes into contact with the ORP water solution. Preferably, the pH of the ORP water solution administered in accordance with the invention is from about 3 to about 8. More preferably, the pH of the ORP water solution is from about 6.4 to about 7.8, and still more preferably, the pH is from about 7.4 to about 7.6.

[0082] The ORP water solution administered in accordance with the invention can have an oxidation-reduction potential of from about ¨1000 millivolts (mV) to about +1150 millivolts (mV). This potential is a measure of the tendency (i.e., the potential) of a solution to either accept or transfer electrons that are sensed by a metal electrode and compared with a reference electrode in the same solution. This potential may be measured by standard techniques including, for example, measuring the electrical potential in millivolts of the ORP water solution relative to standard reference such as, e.g., a silver/silver chloride electrode.
[0083] The ORP water solution administered in accordance with the invention preferably has a potential of from about ¨400 mV to about +1300 mV. More preferably, the ORP water solution has a potential of from about 0 mV to about +1250 mV, and still more preferably from about +500 mV to about +1250 mV. Even more preferably, the ORP
water solution administered in accordance with the present invention has a potential of from about +800 mV to about +1100 mV, and most preferably from about +800 mV to about +1000 mV.
[0084] Various ionic and other species may be present in the ORP water solution administered in accordance with the invention. For example, the ORP water solution may contain chlorine (e.g., free chlorine and bound chlorine), and dissolved oxygen and, optionally, ozone and peroxides (e.g., hydrogen peroxide). The presence of one or more of these species is believed to contribute to at least the disinfectant ability of the ORP water solution to kill a variety of microorganisms, such as bacteria and fungi, as well as viruses.
Although not wishing to be bound by any particular theory, it is believed that or more of such species also may contribute the anti-inflammatory efficacy of the ORP
water solution.
[0085] Free chlorine typically includes, but is not limited to, hypochlorous acid (HC10), hypochlorite ions (C10), sodium hypochlorite (Na0C1), and precursors thereof.
The ratio of hypochlorous acid to hypochlorite ion is dependent upon pH. At a pH of 7.4, hypochlorous acid levels are typically from about 25 ppm to about 75 ppm.
Temperature also impacts the ratio of the free chlorine component.
[0086] Bound chlorine typically includes chlorine in chemical combination with, e.g., ammonia or organic amines (e.g., chloramines). Bound chlorine is preferably present in an amount of up to about 20 ppm.
[0087] One or more chlorine species, one or more additional superoxidized water species (e.g., one or more additional oxidizing species such as, e.g., oxygen) can be present in the ORP water solution administered in accordance with the invention in any suitable amount. The levels of these components may be measured by any suitable method, including methods known in the art.

[0088] The total amount of free chlorine species is preferably from about 10 ppm to about 400 ppm, more preferably from about 50 ppm to about 200 ppm, and most preferably from about 50 ppm to about 80 ppm. The amount of hypochlorous acid is preferably from about 15 ppm to about 35 ppm. The amount of sodium hypochlorite is preferably in the range of from about 25 ppm to about 50 ppm. Optionally, Chlorine dioxide levels are preferably less than about 5 ppm.
[0089] The chlorine content may be measured by methods known in the art, such as the DPD colorimeter method (Lamotte Company, Chestertown, Maryland) or other known methods such as, e.g., methods established by the Environmental Protection Agency. In the DPD colorimeter method, a yellow color is foinied by the reaction of free chlorine with N,N-diethyl-p-phenylenediamine (DPD) and the intensity is measured with a calibrated calorimeter that provides the output in parts per million. Further addition of potassium iodide turns the solution a pink color to provide the total chlorine value.
The amount of bound chlorine present is then determined by subtracting free chlorine from the total chlorine.
I[0090] The total amount of oxidizing chemical species present in the ORP
water solution is preferably in the range of about 2 millimolar (mM), which includes the aforementioned chlorine species, oxygen species, and additional species, including those, which can be difficult to measure such as, e.g., a, C103, 02-, and C10x-[0091] In one embodiment, the ORP water solution administered in accordance with the invention comprises one or more chlorine species and one or more additional superoxidized water species (e.g., one or more additional oxidizing species such as, e.g., oxygen).
Preferably, the chlorine species present is a free chlorine species. The free chlorine species can include one or more species selected from the group consisting of hypochlorous acid (HOC1), hypochlorite ions (00"), and sodium hypochlorite (Na0C1), chloride ion (Cl), and optionally, chlorine dioxide (C102), dissolved chlorine gas (C12), precursors thereof and mixtures thereof.
[0092] In one embodiment, the ORP water solution includes one or more chlorine species or one or more precursors thereof, and one or more additional superoxidized water species or one or more precursors thereof, and, optionally, hydrogen peroxide, and is stable for at least about 24 hours, preferably for at least about one week, more preferably for at about least two months, and still more preferably for at least about six months after its preparation. Even more preferably, such ORP water solution is stable for at least about one year, and most preferably for more than about one year, e.g., at least about two years or at least about three years.

.1 [0093) It is also preferred that the ORP water solution includes one or more chlorine species (e.g., hypocholorous acid and sodium hypochlorite) or one or more precursors thereof and one or one or more additional auperoxiclized water species (e.g., one or more oxygen species, dissolved oxygen) or one or more precursors thereof and has a pH of from about 6 to about 8. More preferably from about 6.2 to about 7.8, and most preferably from = about 7.4 to about 7.6. An exemplary ORP water solution administered in accordance with the present invention can comprise, e.g., from about 15 ppm to about 35 ppm hypochlorous = acid, from about 25 ppm to about 50 ppm sodium hypochlorite, from about I
ppm to about 4 ppm of one or more additional superoxidized water species and a pH of from about 6.2 to about 7,8, and can be stable for at least about one week, e.g., at least about two months, at least about six months, at least about one year, or more than about one year, e.g,, at least about two years or at least about three years.
[0094J While in no way limiting the present invention, it is believed that the control of pH and other variables (e.g., salinity) can provide stable ORP water solutions, which contain one or more chlorine species or precursors thereof, such as, e.g., hypochlorous acid and hypoehlorite ions, and one or more additional superoxidized water species (e.g., oxygen) or one or more precursors thereof.
[00951 The ORP water solutions administered in accordance with the invention preferably comprises one or more oxidized water Species which can yield free radicals (such = aa,..e.g., hydroxyl radicals) on exposure to iron. The ORP water can optionally include one or more chemical compounds generated during the production thereof such as, e.g., sodium hydroxide (NaOH), chlorine dioxide (d02), peroxides (e,g., hydrogen peroxide (H202), and ozone (03) although, it has been reported that sodium hydroxide, chlorine dioxide, hydrogen peroxide, and ozone may rent with hypocholrite resulting in their consumption and the production of other chemical species.
[0096) The ORP water solution administered hi accordance with the present invention can be produced by an oxidation-reduction process, e.g., by an electrolytic process or redox reaction, in which electrical energy is used to produce one or more chemical changes in an aqueous solution. Exemplary processes for preparing suitable ORP water solutions are described, e.g., in U.S. Patent Application Publication Nos, US 2005/0139808 and US
2005/0142157, = [0097J, In the electrolytic process, electrical energy is introduced into and transported through water by the conduction of electrical charge from one point to another in the form of an electrical current, In order for the electrical current to arise and subsist there should be charge carriers in the water, and there should be a force that makes the carriers move.
The charge carriers can be electrons, as in the case of metal and semiconductors, or they can I

be positive and negative ions in the case of solutions. A reduction reaction occurs at the cathode while an oxidation reaction occurs at the anode. At least some of the reductive and oxidative reactions that are believed to occur are described in International Application WO
03/048421 Al.
[0098] As used herein, water produced at an anode is referred to as anode water and water produced at a cathode is referred to as cathode water. Anode water typically contains oxidized species produced from the electrolytic reaction while cathode water typically contains reduced species from the reaction. Anode water generally has a low pH, typically of from about 1 to about 6.8. The anode water preferably contains chlorine in various foi. ms including, for example, chlorine gas, chloride ions, hydrochloric acid and/or hypochlorous acid, or one or more precursors thereof. Oxygen in various forms is also preferably present including, for example, oxygen gas, and possibly one or more species foHned during production (e.g., peroxides, and/or ozone), or one or more precursors thereof.
Cathode water generally has a high pH, typically from about 7.2 to about 11. Cathode water can contain hydrogen gas, hydroxyl radicals, and/or sodium ions.
[0099] The ORP water solution administered in accordance with the invention can include a mixture of anode water (e.g., water produced in the anode chamber of an electrolytic cell) and cathode water (e.g., water produced in the cathode chamber of an electrolysis cell). Preferably, the ORP water solution administered in accordance with the present invention contains cathode water, e.g., in an amount of from about 10%
by volume to about 90% by volume of the solution. More preferably, cathode water is present in the ORP water solution in an amount of from about 10% by volume to about 50% by volume, and still more preferably of from about 20% by volume to about 40% by volume of the solution, e.g., from about 20% by volume to about 30% by volume of the solution.
Additionally, anode water can be present in the ORP water solution, e.g., in an amount of from about 50% by volume to about 90% by volume of the solution. Exemplary ORP
water solutions can contain from about 10% by volume to about 50% by volume of cathode water and from about 50% by volume to about 90% by volume of anode water. The anode and cathode water can be produced using the three-chambered electrolysis cell shown in FIG. 1.
[00100] The ORP water solution administered in accordance with the invention is preferably produced using at least one electrolysis cell comprising an anode chamber, a cathode chamber and a salt solution chamber located between the anode and cathode chambers, wherein at least some of the anode and cathode water are combined such that the ORP water solution comprises anode water and cathode water. A diagram of an exemplary three chamber electrolysis cell that can be used in preparing an exemplary ORP
water solution is shown in FIG. 2.

[0100] The electrolysis cell 100 has an anode chamber 102, cathode chamber 104 and salt solution chamber 106. The salt solution chamber is located between the anode chamber 102 and cathode chamber 104. The anode chamber 102 has an inlet 108 and outlet 110 to permit the flow of water through the anode chamber 100. The cathode chamber similarly has an inlet 112 and outlet 114 to pelinit the flow of water through the cathode chamber 104. The salt solution chamber 106 has an inlet 116 and outlet 118.
The electrolysis cell 100 preferably includes a housing to hold all of the components together.
[0101] The anode chamber 102 is separated from the salt solution chamber by an anode electrode 120 and an anion ion exchange membrane 122. The anode electrode 120 may be positioned adjacent to the anode chamber 102 with the membrane 122 located between the anode electrode 120 and the salt solution chamber 106. Alternatively, the membrane 122 may be positioned adjacent to the anode chamber 102 with the anode electrode 120 located between the membrane 122 and the salt solution chamber 106.
[0102] The cathode chamber 104 is separated from the salt solution chamber by a cathode electrode 124 and a cathode ion exchange membrane 126. The cathode electrode 124 may be positioned adjacent to the cathode chamber 104 with the membrane 126 located between the cathode electrode 124 and the salt solution chamber 106.
Alternatively, the membrane 126 may be positioned adjacent to the cathode chamber 104 with the cathode electrode 124 located between the membrane 126 and the salt solution chamber 106.
[0103] The electrodes preferably are constructed of metal to permit a voltage potential to be applied between the anode chamber and cathode chamber. The metal electrodes are generally planar and have similar dimensions and cross-sectional surface area to that of the ion exchange membranes. The electrodes are configured to expose a substantial portion of the surface of the ion exchange members to the water in their respective anode chamber and cathode chamber. This permits the migration of ionic species between the salt solution chamber, anode chamber and cathode chamber. Preferably, the electrodes have a plurality of passages or apertures evenly spaced across the surface of the electrodes.
[01041 A source of electrical potential is connected to the anode electrode 120 and cathode electrode 124 so as to induce an oxidation reaction in the anode chamber 102 and a reduction reaction in the cathode chamber 104.
[0105] The ion exchange membranes 122 and 126 used in the electrolysis cell 100 may be constructed of any suitable material to permit the exchange of ions between the salt solution chamber 106 and the anode chamber 102 such as, e.g., chloride ions (Cl) and between the salt solution salt solution chamber 106 and the cathode chamber 104 such as, e.g., sodium ions (Nat). The anode ion exchange membrane 122 and cathode ion exchange membrane 126 may be made of the same or different material of construction.
Preferably, the anode ion exchange membrane comprises a fluorinated polymer. Suitable fluorinated polymers include, for example, perfluorosulfonic acid polymers and copolymers such as perfluorosulfonic acid/PTFE copolymers and perfluorosulfonic acid/TFE
copolymers. The ion exchange membrane may be constructed of a single layer of material or multiple layers.
Suitable ion exchange membrane polymers can include one or more ion exchange membrane polymers marketed under the trademark Nafion .
[0106] The source of the water for the anode chamber 102 and cathode chamber 104 of the electrolysis cell 100 may be any suitable water supply. The water may be from a municipal water supply or alternatively pretreated prior to use in the electrolysis cell.
Preferably, the water is pretreated and is selected from the group consisting of softened water, purified water, distilled water, and deionized water. More preferably, the pretreated water source is ultrapure water obtained using reverse osmosis purification equipment.
[0107] The salt water solution for use in the salt water chamber 106 can include any aqueous salt solution that contains suitable ionic species to produce the ORP
water solution.
Preferably, the salt water solution is an aqueous sodium chloride (NaC1) salt solution, also commonly referred to as a saline solution. Other suitable salt solutions can include other chloride salts such as potassium chloride, ammonium chloride and magnesium chloride as well as other halogen salts such as potassium and bromine salts. The salt solution can contain a mixture of salts.
[0108] The salt solution can have any suitable concentration. For example, the salt solution can be saturated or concentrated. Preferably, the salt solution is a saturated sodium chloride solution.
[0109] FIG. 2 illustrates what are believed to be various ionic species produced in the three chambered electrolysis cell useful in connection with the invention. The three chambered electrolysis cell 200 includes an anode chamber 202, cathode chamber 204, and a salt solution chamber 206. Upon application of a suitable electrical current to the anode 208 and cathode 210, the ions present in the salt solution flowing through the salt solution chamber 206 migrate through the anode ion exchange membrane 212 and cathode ion exchange membrane 214 into the water flowing through the anode chamber 202 and cathode chamber 204, respectively.
[0110] Positive ions migrate from the salt solution 216 flowing through the salt solution chamber 206 to the cathode water 218 flowing through the cathode chamber 204.
Negative ions migrate from the salt solution 216 flowing through the salt solution chamber 206 to the anode water 220 flowing through the anode chamber 202.
[0111] Preferably, the salt solution 216 is aqueous sodium chloride (NaCl), which contains both sodium ions (Nat) and chloride ions (a) ions. Positive Na+ ions migrate from the salt solution 216 to the cathode water 218. Negative Cl ions migrate from the salt solution 216 to the anode water 220.
[0112] The sodium ions and chloride ions may undergo further reaction in the anode chamber 202 and cathode chamber 204. For example, chloride ions can react with various oxygen ions and other species (e.g., oxygen containing free radicals, 02, 03) present in the anode water 220 to produce CIOn- and C10-. Other reactions may also take place in the anode chamber 202 including the formation of oxygen free radicals, hydrogen ions (11+), oxygen (e.g., as 02), ozone (03), and peroxides. In the cathode chamber 204, hydrogen gas (H2), sodium hydroxide (NaOH), hydroxide ions (OH), and other radicals may be formed.
[01131 The apparatus for producing the ORP water solution also can be constructed to include at least two three chambered electrolysis cells. Each of the electrolytic cells includes an anode chamber, cathode chamber, and salt solution chamber separating the anode and cathode chambers. The apparatus includes a mixing tank for collecting the anode water produced by the electrolytic cells and a portion of the cathode water produced by one or more of the electrolytic cells. Preferably, the apparatus further includes a salt recirculation system to permit recycling of the salt solution supplied to the salt solution chambers of the electrolytic cells. A diagram of an exemplary process for producing an ORP water solution using two electrolysis cells is shown in FIG. 3.
[0114] The process 300 includes two three-chambered electrolytic cells, specifically a first electrolytic cell 302 and second electrolytic cell 304. Water is transferred, pumped or otherwise dispensed from the water source 305 to anode chamber 306 and cathode chamber 308 of the first electrolytic cell 302 and to anode chamber 310 and cathode chamber 312 of the second electrolytic cell 304. Advantageously, this process can produce from about 1 liter/minute to about 50 liters/minute of ORP water solution. The production capacity may be increased by using additional electrolytic cells. For example, three, four, five, six, seven, eight, nine, ten or more three-chambered electrolytic cells may be used to increase the output of the ORP water solution administered in accordance with the invention.
[0115] The anode water produced in the anode chamber 306 and anode chamber 310 are collected in the mixing tank 314. A portion of the cathode water produced in the cathode chamber 308 and cathode chamber 312 is collected in mixing tank 314 and combined with the anode water. The remaining portion of cathode water produced in the process is discarded. The cathode water may optionally be subjected to gas separator 316 and/or gas separator 318 prior to addition to the mixing tank 314. The gas separators remove gases such as hydrogen gas that are fomied in cathode water during the production process.
[0116] The mixing tank 314 may optionally be connected to a recirculation pump 315 to permit homogenous mixing of the anode water and portion of cathode water from electrolysis cells 302 and 304. Further, the mixing tank 314 may optionally include suitable devices for monitoring the level and pH of the ORP water solution. The ORP
water solution may be transferred from the mixing tank 314 via pump 317 for application in disinfection or sterilization at or near the location of the mixing tank.
Alternatively, the ORP water solution may be dispensed into one or more suitable containers for shipment to a remote site (e.g., warehouse, hospital, etc.).
[0117] The process 300 further includes a salt solution recirculation system to provide the salt solution to salt solution chamber 322 of the first electrolytic cell 302 and the salt solution chamber 324 of the second electrolytic cell 304. The salt solution is prepared in the salt tank 320. The salt is transferred via pump 321 to the salt solution chambers 322 and 324. Preferably, the salt solution flows in series through salt solution chamber 322 first followed by salt solution chamber 324. Alternatively, the salt solution may be pumped to both salt solution chambers simultaneously.
[0118] Before returning to the salt tank 320, the salt solution may flow through a heat exchanger 326 in the mixing tank 314 to control the temperature of the ORP
water solution as needed.
[0119] The ions present in the salt solution are depleted over time in the first electrolytic cell 302 and second electrolytic cell 304. An additional source of ions periodically can be added to the mixing tank 320 to replace the ions that are transferred to the anode water and cathode water. The additional source of ions may be used, e.g., to maintain a constant pH
of the salt solution, which can to drop (i.e., become acidic) over time. The source of additional ions may be any suitable compound including, for example, salts such as, e.g., sodium chloride. Preferably, sodium hydroxide is added to the mixing tank 320 to replace the sodium ions (Na) that are transferred to the anode water and cathode water.
[0120] Following its preparation, the ORP water solution can be transferred to one or more suitable containers, e.g., a sealed container for distribution and sale to end users such as, e.g., health care facilities including, e.g., hospitals, nursing homes, doctor offices, outpatient surgical centers, dental offices, and the like. Suitable containers can include, e.g., a sealed container that maintains the sterility and stability of the ORP water solution held by the container. The container can be constructed of any material that is compatible with the ORP water solution. Preferably, the container is generally non-reactive with one or more ions or other species present in the ORP water solution.
[0121] Preferably, the container is constructed of plastic or glass. The plastic can be rigid so that the container is capable of being stored on a shelf.
Alternatively, the container can be flexible, e.g., a container made of flexible plastic such as, e.g., a flexible bag.

[0122] Suitable plastics can include, e.g., polypropylene, polyester terephthalate (PET), polyolefin, cycloolefin, polycarbonate, ABS resin, polyethylene, polyvinyl chloride, and mixtures thereof. Preferably, the container comprises one or more polyethylenes selected from the group consisting of high-density polyethylene (HDPE), low-density polyethylene (LDPE), and linear low-density polyethylene (LLDPE). Most preferably, the container is constructed of high density polyethylene.
[0123] The container preferably has an opening to permit dispensing of the ORP water solution. The container opening can be sealed in any suitable manner. For example, the container can be sealed with a twist-off cap or stopper. Optionally, the opening can be further sealed with a foil layer.
[0124] The headspace gas of the sealed container can be air or any other suitable gas, which preferably does not react with one or more species in the ORP water solution.
Suitable headspace gases can include, e.g., nitrogen, oxygen, and mixtures thereof.
[0125] The ORP water solution administered in accordance with the invention also can be used for the prevention or treatment of an infection, e.g., by one or more infectious pathogens such as, for example, infectious microorganisms. Such microorganisms can include, for example, viruses, bacteria, and fungi. The viruses can include, e.g., one or more viruses selected from the group consisting of adenovintses, herpes viruses, coxsackie viruses, HIV, rhinoviruses, comavimses, and flu viruses. The bacteria can include, e.g., one or more bacteria selected from the group consisting of Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Mycobaterium tuberculosis. The fungi can include, e.g., one or more fungi selected from the group consisting of Candida albicans, Bacillus subtilis and Bacillus athrophaeus.
[0126] The ORP water solution administered in accordance with the invention also can be effective against adenovirus. Preferably, the ORP water solution administered in accordance with the invention preferably achieves a log-10 reduction in the adenoviral load of greater than about 2, more preferably greater than about 2.5, and still more preferably greater than about 3, after exposure to the ORP water solution for about 20 minutes, more preferably after exposure for about 15 minutes, and still more preferably after exposure for about 10 minutes. The ORP water solution administered in accordance with the invention also can be effective for reducing the viral load of HIV-1, preferably by a log reduction factor greater than about 2, more preferably by a log reduction factor of greater than about 2.5, and still more preferably by a log reduction factor of greater than about 3 after exposure to the ORP water solution for about five minutes.

[0127] In accordance with the method of the present invention, administering the ORP
water solution for the prevention or treatment of infection also can serve to prevent or treat inflammation associated with the infection (or the affected tissues) as described herein.
[0128] The ORP water solution administered in accordance with the invention also can be used for treating impaired or damaged tissue, e.g., by contacting one or more impaired or damaged tissues with a therapeutically effective amount of the ORP water solution. Any suitable method can be used for contacting the impaired or damaged tissue, so as to treat the impaired or damaged tissue. For example, the impaired or damaged tissue can be treated by irrigating the tissue with the ORP water solution, so as to contact the impaired or damaged tissue with a therapeutically effective amount of the ORP water solution. The ORP water solution can be administered as a steam or a spray, or by aerosolization, nebulization or atomization, as described herein, so as to contact the impaired or damaged tissue with a therapeutically effective amount of the ORP water solution.
[0129] The ORP water solution administered in accordance with the invention can be used for treating tissues, which have been impaired or damaged, e.g., by surgery. For instance, the ORP water solution can be used for treating tissues, which have been impaired or damaged by an incision. In addition, the ORP water solution can be used for treating tissues, which have been impaired or damaged by oral surgery, graft surgery, implant surgery, transplant surgery, cauterization, amputation, radiation, chemotherapy, and combinations thereof. The oral surgery can include, for example, dental surgery such as, e.g., root canal surgery, tooth extraction, gum surgery, and the like.
[0130] The ORP water solution administered in accordance with the invention can be used for treating tissues, which have been impaired or damaged by one or more burns, cuts, abrasions, scrapes, rashes, ulcers, puncture wounds, combinations thereof, and the like, which are not necessarily caused by surgery. The ORP water solution administered in accordance with the invention can be used for treating impaired or damaged tissue, which is infected, or tissue impaired or damaged due to infection. Such infection can be caused by one or more infectious pathogens, such as, e.g., one or more microorganisms selected from the group consisting of viruses, bacteria, and fungi, as described herein.
[0131] In accordance with the present invention, administering the ORP
water solution for treating impaired or damaged tissue also can serve to prevent or treat inflammation associated with the impairment or damage (or with the impaired or damaged tissue) .
[0132] The ORP water solution administered in accordance with the invention also can be used as a disinfectant to eradicate microorganisms, including bacteria, viruses and spores, in a variety of settings, e.g., in the healthcare and medical device fields, to disinfect surfaces and medical equipment, and also can be applied in wound care, medical device sterilization, food sterilization, hospitals, consumer households and anti-bioterrorism. The ORP water solution can be used for disinfecting a surface, e.g., by contacting the surface with an anti-infective amount of the ORP water solution. The surface can be contacted using any suitable method. For example, the surface can be contacted by irrigating the surface with the ORP water solution, so as to disinfect the surface.
Additionally, the surface can be contacted by applying the ORP water solution to the surface as a steam or a spray, or by aerosolization, nebulization or atomization, as described herein, so as to disinfect the surface. Further, the ORP water solution can be applied to the surface with a cleaning wipe, as described herein. By disinfecting a surface, the surface may be cleansed of infectious microorganisms. Alternatively (or additionally), the ORP water solution administered in accordance with the present invention can be applied to the surface to provide a barrier to infection, to thereby disinfect the surface.
[01331 The surface(s) can include one or more biological surfaces, one or more inanimate surfaces, and combinations thereof. Biological surfaces can include, for example, tissues within one or more body cavities such as, for example, the oral cavity, the sinus cavity, the cranial cavity, the abdominal cavity, and the thoracic cavity.
Tissues within the oral cavity include, e.g., mouth tissue, gum tissue, tongue tissue, and throat tissue. The biological tissue also can include muscle tissue, bone tissue, organ tissue, mucosal tissue, vascular tissue, neurological tissue, and combinations thereof. Biological surfaces also include any other cultured tissue in vitro, such as primary and established cell lines, stem cells of any nature, xenotransplants, tissue substitutes (e.g. made of collagen or any other organic material in addition or not of cellular elements), any other tissue-engineered substitutes and combinations thereof.
[0134] Inanimate surfaces include, for example, surgically implantable devices, prosthetic devices, and medical devices. In accordance with the method of the present invention, the surfaces of internal organs, viscera, muscle, and the like, which may be exposed during surgery, can be disinfected, e.g., to maintain sterility of the surgical environment. In accordance with the present invention, administering the ORP
water solution for disinfecting a surface also can serve to treat or prevent inflammation affecting one or more biological tissues associated with such surfaces.
[0135] The ORP water solution may also be applied to humans and/or animals to treat various conditions, including inflammation, hypersensitivity, and associated systemic effects associated with one or more of the following: surgical/open wound cleansing agent;
skin pathogen disinfection (e.g., for bacteria, mycoplasmas, virus, fungi, prions); battle wound disinfection; wound healing promotion; burn healing promotion; treatment of stomach ulcers; wound irrigation; skin fungi; psoriasis; athlete's foot;
pinkeye and other eye infections; ear infections (e.g., swimmer's ear); lung/nasal/sinus infections;
and other medical applications on or in the human or animal body, as well as environmental remediation, The use of ORP water solutions as a tissue cell growth promoter is further described in U.S. Patent Application Publication 2002/0160053.
= [0136) The ORP water solution may be used as a disinfectant, sterilization agent, decontaminant, antiseptic and/or cleanser. The ORP water solution administered in accordance with the invention is suitable for use in the following representative applications: medical, dental and/or veterinary equipment and devices; food industry (e.g., hard surfaces, fruits, vegetables, meats); hospitals/health care facilities (e.g., hard surfaces);
cosmetic industry (e.g., skin cleaner); households (e.g., floors, counters, hard surfaces);
electronics industry (e.g., cleaning circuitry, hard drives); and bio-terrorism (e.g., anthrax, infectious microbes).
[0137] Organisms that can be controlled, reduced, killed or eradicated by treatment with the ORP water solution include, but are not limited to, bacteria, fiangi, yeasts, and viruses.
Susceptible bacteria include, but are not limited to, .81scherichia coli, Staphylococcus mamas, Bacillus athrophaeus, Streptococcus ,pyagenes, Salpnonella choleraesuis, Pseudomonas aeruginosa, Shingella dysenteriae, and other susceptible bacteria, Fungi and yeasts that may be treated with the ORP water solution include, for example, Candida albicans and Trichophyton mentagrophytes. The ORP water solution may also be applied to viruses including, for example, adenovirus, human immunodeficiency virus (HIV), rbieovirus, influenza (e.g., influenza A), hepatitis (e.g., hepatitis A), coronavirus (responsible for Severe Acute Respiratory Syndrome (SARS)), rotavinis, respiratory syncytial virus, herpes simplex virile, varicella zoster virus, rubella virus, and other susceptible viruses.
101381 The ORP water solution may be applied to disinfect and sterilize in any suitable manner. For example, to disinfect and sterilize medical or dental equipment, the equipment can be maintained in contact with the ORP water solution for a sufficient period of time to reduce the level of organisms present on the equipment to a desired level.
Alternatively, the ' ORP water solution can be applied to medical or dental equipment by immersing the equipment in a container with or without the application of enhancing physical procedures, e.g. ultrasound, shakers, heaters, and the like.
101391 For disinfection and sterilization of hard surfaces, the ORP water solution can be = applied to the hard surface directly from a. container in which the ORP
water solution is stored. For example, the ORP water solution can be poured, sprayed or otherwise directly . _ _ applied to the hard surface. The ORP water solution can then be distributed over the hard surface using a suitable substrate such as, for example, cloth, fabric or paper towel. In hospital applications, the substrate is preferably sterile. Alternatively, the ORP water solution can first be applied to a substrate such as cloth, fabric or paper towel. The wetted substrate can then be contacted with the hard surface. Alternatively, the ORP
water solution can be applied to hard surfaces by dispersing the solution into the air as described herein.
The ORP water solution can be applied in a similar manner to humans and animals.
[0140] The ORP water solution also can be applied with a cleaning wipe comprising a water insoluble substrate and the ORP water solution as described herein, wherein the ORP
water solution is dispensed onto the substrate. The ORP water solution can be impregnated, coated, covered or otherwise applied to the substrate. Preferably, the substrate is pretreated with the ORP water solution before distribution of the cleaning wipes to end users.
[0141] The substrate for the cleaning wipe can be any suitable water-insoluble absorbent or adsorbent material. A wide variety of materials can be used as the substrate. It should have sufficient wet strength, abrasivity, loft and porosity. Further, the substrate should not adversely impact the stability of the ORP water solution. Examples include non woven substrates, woven substrates, hydroentangled substrates and sponges.
[0142] The substrate can have one or more layers. Each layer can have the same or different textures and abrasiveness. Differing textures can result from the use of different combinations of materials or from the use of different manufacturing processes or a combination thereof. The substrate should not dissolve or break apart in water. The substrate can thereby provide a vehicle for delivering the ORP water solution to the surface to be treated.
[0143] The substrate can be a single nonwoven sheet or multiple nonwoven sheets. The nonwoven sheet can be made of wood pulp, synthetic fibers, natural fibers, and blends thereof. Suitable synthetic fibers for use in the substrate can include, without limitation, polyester, rayon, nylon, polypropylene, polyethylene, other cellulose polymers, and mixtures of such fibers. The nonwovens can include nonwoven fibrous sheet materials which include meltblown, cofonn, air-laid, spun bond, wet laid, bonded-carded web materials, hydroentangled (also known as spunlaced) materials, and combinations thereof.
These materials can comprise synthetic or natural fibers or combinations thereof. A binder can optionally be present in the substrate.
[0144] Examples of suitable nonwoven, water insoluble substrates include 100%
cellulose Wadding Grade 1804 from Little Rapids Corporation, 100%
polypropylene needlepunch material NB 701-2.8-W/R from American Non-wovens Corporation, a blend of cellulosic and synthetic fibres-Hydraspun 8579 from Ahlstrom Fibre Composites, and 70% Viscose/30% PBS Code 9881 from POI Nonwovens Polymer Corp. Additional examples .of nonwoven substrates suitable for use in the cleaning wipes are described in U.S. Pat. Nos. 4,781,974, 4,615,937, 4,666,621, and 5,908,707, and International Patent Application Publications WO 98/03713, WO 97/40814, and WO 96/14835-[0145j The substrate also can be made of woven materials, such as cotton fibers, cotton/nylon blends, or other textiles. Regenerated cellulose, polyurethane foams, and the like, which are used in making sponges, also can be suitable for use.
[0146] The liquid loading capacity of the substrate should be at least about 50%4000%
of the dry weight thereof, most preferably at least about 200%-800%. This is expressed as loading 112 to 10 times the weight of the substrate. The weight of the substrate varies without limitation from about 0.01 to about 1,000 grams per square meter, most preferably 25 to 120 grams/m2 (referred to as "basis weight") and typically is produced as a sheet or web which is cut, die-cut, or otherwise sired into the appropriate shape and sin. The cleaning wipes will preferably have a certain wet tensile strength which is without limitation about 25 to about 250 Newtons/m, more preferably about 75470 Newtons/m.
101.47] The ORP water solution can be dispensed, impregnated, coated, covered or otherwise applied to.the substrate by any suitable method. For example, individual portions of substrate can be treated with a discrete amount of the ORP water solution.
Preferably, a mass treatment of a continuous web of substrate material with the ORP water solution is can-fed out. The entire web of substrate material can be soaked in the ORP
water solution.
Alternatively, as the substrate web is spooled, or even during creation of a nonwoven substrate, the ORP water solution can be sprayed or metered onto the web. A
stack of individually cut and sized portions of substrate can be impregnated or coated with the ORP
water solution in its container by the manufacturer, [0148] The cleaning wipes optionally can contain additional components to improve the properties of the wipes. Fr example, the cleaning wipes can further comprise polymers, surfactants, polysaccharides, polycarboxylates, polyvinyl aloohols, solvents, cnelating agents, buffers, thickeners, dyes, colorants, fragrances, and mixtures thereof to improve the properties of the wipes. These optional components should not adversely impact the stability of the ORP water solution, Examples of various components that may optionally be included in the cleaning wipes are described in U.S. Patents 6,340,663,
6,649,584 and 6,624,135.
[0149] The cleaning wipes can be individually sealed with a heat-sealable or glueable thermoplastic overwrap (such as polyethylene, Mylar, and the like). The wipes can also be packaged as numerous, individual sheets for more economical dispensing. The cleaning .............

wipes can be prepared by first placing multiple sheets of the substrate in a dispenser and then contacting the substrate sheets with the ORP water solution administered in accordance with the invention. Alternatively, the cleaning wipes can be fanned as a continuous web by applying the ORP water solution to the substrate during the manufacturing process and then loading the wetted substrate into a dispenser.
[0150] The dispenser includes, but is not limited to, a canister with a closure, or a tub with closure. The closure on the dispenser is to seal the moist wipes from the external environment and to prevent premature volatilization of the liquid ingredients.
[0151] The dispenser can be made of any suitable material that is compatible with both the substrate and the ORP water solution. For example, the dispenser can be made of plastic, such as high density polyethylene, polypropylene, polycarbonate, polyethylene terephthalate (PET), polyvinyl chloride (PVC), or other rigid plastics.
[0152] The continuous web of wipes can be threaded through a thin opening in the top of the dispenser, most preferably, through the closure. A means of sizing the desired length or size of the wipe from the web can then be desirable. A knife blade, serrated edge, or other means of cutting the web to desired size can be provided on the top of the dispenser, for non-limiting example, with the thin opening actually doubling in duty as a cutting edge.
Alternatively, the continuous web of wipes can be scored, folded, segmented, perforated or partially cut into uniform or non-unifoun sizes or lengths, which would then obviate the need for a sharp cutting edge. Further, the wipes can be interleaved, so that the removal of one wipe advances the next.
[0153] The ORP water solution alternatively can be dispersed into the environment through a gaseous medium, such as air. The ORP water solution can be dispersed into the air by any suitable means. For example, the ORP water solution can be fowled into droplets of any suitable size and dispersed into a room.
For small scale applications, the ORP water solution can be dispensed through a spray bottle that includes a standpipe and pump. Alternatively, the ORP water solution can be packaged in aerosol containers. Aerosol containers can include the product to be dispensed, propellant, container, and valve. The valve can include both an actuator and dip tube. The contents of the container can be dispensed by pressing down on the actuator.
The various components of the aerosol container should be compatible with the ORP water solution.
Suitable propellants can include a liquefied halocarbon, hydrocarbon, or halocarbon-hydrocarbon blend, or a compressed gas such as carbon dioxide, nitrogen, or nitrous oxide.
Aerosol systems preferably yield droplets that range in size from about 0.15 pm to about 5 pm.

[0154] Applications can also be conducted by using various hydrosurgery equipments for debriding and cleaning (e.g VersaJet devices sold in the United States by Smith and Nephew, Debritom in Europe by Medaxis, JetOx in the United States and Europe by DeRoyal or PulsaVac in Italy), irrigation systems with negative pressure (e.g., VAC
Instill), and the like.
[0155] Optionally, several adjuvant therapies can also be utilized in accordance with the invention including bioengineered skin (Apligraf, Organogenesis, Inc., Canton), acellular skin substitutes (Oasis Wound Matrix, Healthpoint), ultrasonic application of ORP water solutions, and local oxygen replacement or hyperbaric oxygen treatment (such as, e.g., hyperbaric boots, the Vent-Ox System).
[0156] For some applications, the ORP water solution optionally can contain a bleaching agent. The bleaching agent can include, e.g., any suitable compound that lightens or whitens a substrate. The ORP water solution containing a bleaching agent can be used in home laundering to disinfect and sterilize bacteria and geinis as well as brighten clothing.
Suitable bleaching agents include, but are not limited to, chlorine-containing bleaching agents and peroxide-containing bleaching agents. Mixtures of bleaching agents also can be added to the ORP water solution. Preferably, the bleaching agent is added in the foini of an aqueous solution to the ORP water solution.
[0157] Suitable chlorine-containing bleaching agents can include, e.g., chlorine, hypochlorites, N-chloro compounds, and chlorine dioxide. Preferably, the chlorine-containing bleaching agent added to the ORP water solution is sodium hypochlorite or hypochlorous acid. Other suitable chlorine-containing bleaching agents include, e.g., chlorine, calcium hypochlorite, bleach liquor (e.g., aqueous solution of calcium hypochlorite and calcium chloride), bleaching powder (e.g., mixture of calcium hypochlorite, calcium hydroxide, calcium chloride, and hydrates thereof), dibasic magnesium hypochlorite, lithium hypochlorite, chlorinated trisodium phosphate and mixtures thereof.
[0158] The addition of a bleaching agent to the ORP water solution can be carried out in any suitable manner. Preferably, an aqueous solution containing the bleaching agent is first prepared. The aqueous solution containing the bleaching agent can be prepared using household bleach (e.g., Clorox() bleach) or other suitable source of chlorine-containing bleaching agent or other bleaching agent. The bleaching agent solution can then be combined with the ORP water solution.
[0159] The bleaching agent can be added to the ORP water solution in any suitable amount. Preferably, the ORP water solution containing a bleaching agent is non-irritating to human or animal skin. Preferably, the total chloride ion content of the ORP
water solution containing a chlorine-containing bleaching agent is from about 1000 ppm to about 5000 ppm, and preferably from about 1000 ppm to about 3000 ppm. The pH of the ORP
water solution containing a chlorine-containing bleaching agent is preferably from about 8 to about 10, and the oxidative-reductive potential is preferably from about +700 mV to about +800 mV.
[0160] The following examples further illustrate the invention but, of course, should not be construed as in any way limiting in its scope.

[0161] These examples demonstrate the unique features of the ORP water solution used in accordance with the invention. The samples of the ORP water solution in Examples 1-3 were analyzed in accordance with the methods described herein to determine the physical properties and levels of ionic and other chemical species present in each sample. The results obtained for chlorine dioxide, ozone and hydrogen peroxide are based on standard tests used to measure such species but may be indicative of different species, which can also generate positive test results. Further, it has been reported that chlorine dioxide, ozone and hydrogen peroxide react with hypocholrite resulting in their consumption and the production of other compounds (e.g., HC1 and 02.) The pH, oxidative-reductive potential (ORP) and ionic species present are set forth in Table 1 for each sample of the ORP water solution.
Table 1: Physical characteristics and ion species present for the ORP water solution samples pH 7.45 7.44 7.45 ORP (mV) +879 +881 +874 Total a- (ppm) 110 110 120 Bound a (ppm) 5 6 6 [0162] The ORP water solution has suitable physical characteristics for use in, e.g., disinfection, sterilization, cleaning, and/or the prevention and/or treatment of inflammation, sinusitis, peritonitis, or infection.

[0163] These examples demonstrate the addition of a bleaching agent to the ORP water solution according to the invention in various amounts. In particular, these examples demonstrate the antimicrobial activity and fabric bleaching ability of the compositions.
[0164] A 10% Clorox bleach solution was prepared using distilled water.
The following solutions were then prepared using the 10% bleach solution: 80% ORP
water solution/20% bleach (Example 4); 60% ORP water solution/40% bleach (Example 5); 40%
ORP water solution/60% bleach (Example 6); 20% ORP water solution/80% bleach (Example 7); and 0% ORP water solution/I 00% bleach (Example 8). Two control solutions were also used for comparison including 100% ORP water solution/0% bleach (Example 9) and an ORP water solution with 0.01% Tween 20 detergent (Example 10). The physical characteristics of these samples were determined, specifically pH, oxidative-reductive potential (ORP), total chlorine (Cl) content, hypochlorous acid (HC10") content, and were tested for chlorine dioxide content and peroxide content, the results are set forth in Table 2.

Table 2: Physical characteristics of ORP water solution/bleach compositions Total cr HC10-pH ORP
(10Prn) (10Pm) Ex. 4 8.92 +789 1248 62 Ex. 5 9.20 +782 2610 104 Ex. 6 9.69 +743 4006 80 Ex. 7 9.86 +730 4800 48 Ex. 8 9.80 +737 5000 50 Ex. 9 7.06 +901 64 32 Ex. 10 6.86 +914 51 26 [0165] The large bolus of chlorine ions added as part of the bleaching agent prevented the accurate measurement of the chlorine dioxide and peroxide levels as indicated with the n.d. designations. Also, the results obtained for chlorine dioxide and peroxide are based on standard tests used to measure such species but may be indicative of different species, which can also generate positive test results. Further, it has been reported that chlorine dioxide, ozone and hydrogen peroxide react with hypocholrite resulting in their consumption and the production of other compounds (e.g., HC1 and 02.) As these examples demonstrate, the hypochlorous acid levels of the ORP water solution with and without the addition of a bleaching agent are similar.
[0166] The samples of Examples 4-10 were subjected to a high spore count test using Bacillus subtilis var. niger spores (ATCC #9372 obtained from SPS Medical of Rush, New York). Spore suspensions were concentrated (by evaporation in a sterile hood) to 4 x 106 spores per 100 microliters. A 100 microliter sample of the spore suspension were mixed with 900 microliters of each of the samples in Examples 4-10. The samples were incubated at room temperature for periods of 1 to 5 minutes as set forth in Table 3. At the indicated times, 100 microliters of the incubated samples were plated onto individual TSA plates and incubated for 24 hours at 35 C 2 C, after which the number of resulting colonies on each plate was determined. The control plates demonstrated that the starting spore concentrations were > 1 x 106 spores/100 microliters. The concentration of Bacillus spores for the various samples at the various incubation times (as the average of two detemnnations) is set forth in Table 3.

Table 3: Bacillus spore concentrations 1 minute 2 minutes 3 minutes 4 minutes 5 minutes Ex. 4 >>1000 411 1 0 2 Ex. 5 >> 1000 1000 1 0 0 Ex. 6 >> 1000 >> 1000 > 1000 22 0 Ex. 7 >> 1000 >> 1000 > 1000 15 0 Ex. 8 >> 1000 >> 1000 > 1000 3 1 Ex. 9 >>1000 74 0 0 0 Ex 10 >> 1000 239 3 0 0 [0167] As these results demonstrate, as the concentration of bleach (as 10%
aqueous bleach solution) increases, the amount of Bacillus spores killed is reduced for the samples incubated for 2-3 minutes. However, for samples incubated for 5 minutes, the bleach concentration does not impact Bacillus spore kill. Further, the results demonstrate that the addition of 0.01% detergent to the ORP water solution does not reduce spore kill.
[0168] The samples of Examples 4-10 were subjected to a fabric bleaching test. The fabric upon which the samples were tested was a 100% rayon children's t-shirt with dark blue dye patches. Two inch square pieces of dyed fabric were placed into 50 mL
plastic tubes. Each fabric piece was covered by a sample of the solution in Examples 4-10. The elapsed time until complete bleaching was obtained, as detemiined by the whitening of the fabric, is set forth in Table 4.
Table 4: Time until complete bleaching of fabric sample Example Time Ex. 4 39 minutes Ex. 5 23 minutes Ex. 6 18 minutes Ex. 7 19 minutes Ex. 8 10 minutes Ex. 9 > 6 hours Ex. 10 > 6 hours [01691 As demonstrated by these examples, as the concentration of the ORP water solution increases in the composition, the time until complete bleaching is achieved increases.

[0170) The purpose of this study was to assess the safety of the test an exemplary ORP
water solution, Microcyrr, when administered as drops into the nasal cavity of rabbits.
Thirty-three rabbits were randomly assigned to two groups, Groups I and II.
Group 1 (18 animals) served as the control group and Group II (15 animals) was dosed with the test article. On Day -1 or Day 0, body weights were recorded and blood samples were, collected for analysis of selected parameters. On Day 0, 500121, of sterile saline was administered to the Group I animals and 500A of the test article (at a 50% concentration) was administered to Group n annuals. Both the control and the test articles were administered twice daily as drops into the right nostril. The animals were dosed in the same manner on Days 1-6. Animals were observed daily for signs of pharmacologic and/or toxicologic effects with special attention paid to the nose. Body weights were recorded weekly through study termination. On Day 7, one-third of the animals from each group were selected for blood collection, sacrifice and necropsy. The remaining animals continued to be dosed through Day 14, when half of the animals from each group were selected for blood collection, sacrifice and necropsy. On Day 21, after a 7-day recovery period), the remaining , animals had blood collected and were sacrificed and necropsied. Samples of the nasal mucosa from both nostrils were collected from each animal for histopathological analysis.
[0171J The necropsy consisted of gross observations of the respiratory tract, The entire nasal passage and associated bone were taken and fixed in buffered forrnalin.
Samples of any visible abnormalities in the respiratory tract were also collected for histopathology.
Three biopsy samples (anterior, middle and posterior nasal cavity) per nostril (treated right and untreated left) Were examined. The microscopic histopathology of the nasal mucosa included: integrity of epithelium, presence or loss of epithelial cilia, inflammatory cell iniatration, edema, presence of goblet cells, hyperplaaia of glands, changes in number or characteristics of blood vessels arid any other changes or observations.
[01721 The results (in-life observations including nasal observations, body weights, blood analysis, gross necropsy and histopathology results) from the test gaup were compared to the control group. The test group was not significantly different from animals treated with saline in terms of mild irritation.
= F r"
- = -= [01731 This example illustrates the lack of toxicity from the use of an exemplary OR?
= water solution.
[01741 The characterization of local and systemic toxicity from topically applied MicrocynTm 60 to a deep wound was evaluated in rats. No abnormalities, significant differences in the parameters of the blood chemistry or hematic cytology were observed, nor anomalies in the autopsies. The skin irritation gradings and the histopathology of the wounds and the tissues around the place of application did not reveal any difference between the wounds treated with Microcynm60 and those of the control group treated with saline solution.
101751 The systemic toxicity of Microcyn 60 was also evaluated by means of an intraperitoneal injection in mice. For this, five mice were injected with a single dose (50 ml/kg) of Microcy60 by the intraperitoneal route. In the same way, five control mice were injected with a single dose (50 milicg) of saline solution (sodium chloride at 0,9%). In this investigation, neither mortality nor any evidence of systemic toxicity was observed in any of the animals that received the single intraperitoneal dose of Microeyn 60, indicating that the LD50 is above 50 m1,/lcg.
[0176] Microcy-A0 was administered by the oral route to rats to allow its absorption and to characterize any inherent toxic effect of the product. In this study, a single dose (4.9g ml/kg) was administered by esophageal tube to three albino rats of the Sprague-Dawley strain. There was no mortality, nor were there clinical signs or abnormalities in the autopsies of any of the animals exposed to the single oral dose of Microcy160.
[0177] The potential of topically applied Microcyn"60 for ocular irritation was also evaluated in rabbits, Ocular irritation was not observed nor any other clinical sip in any animal exposed to Microcyti60 by topical administration through the ocular route.
[01781 Microcyri 60 was applied by the inhalatory route to rats to determine potential acute toxicity by inhalation. All the animals showed a very slight or slight reduction in activity and piloerection after the exposure, but they were all asymptomatic on the following day. Mortality or abnormalities were not observed at autopsy of the animals exposed to Microcyn 60 by inhalation, TOI
101791 Bvaluation of the potential for sensitization of the skin with Microcyn 60 was carried out in guinea pigs using a modiAed occlusion patch method (Buehler).
Irritation was not observed in the animals of the control group after a simple treatment challenge, nor in the animals evaluated (treated by induction) after challenge with the treatment. These studies demonstrate that Microcyrirm60 does not provoke a sensitizing reaction. =

[01801 Thus, when it has been applied to the intact skin, deep open dermal wounds, in the conjunctival sac, by oral and inhalation routes or by means of intraperitoneal injection, Microcyn1M 60 has not shown adverse effects related to the product. There is also experience in having treated more than thousands of patients with wounds of very diverse nature in the skin and mucosae, with excellent antiseptic and cosmetic results. Accordingly, topically applied Microcyn'60 should be effective and well-tolerated in this clinical trial.
[01811 Microcyris60 is packaged in transparent 240 mL PET sealed bottles.
This product is stored at ambient temperature and remains stable for up to 2 years in such bottles.
From its profile of high biological safety, IVIicrocyr760 can be safely disposed of, e.g., emptied into the sink without risk of contamination or corrosion.

10182) This example illustrates a clinical study, which can be used to determine the effectiveness of an exemplary ORP water solution for treating pharyngitis.
[0183] Multiple microbial trials have been run with Microcyri60, both in the United States and in Mexico. Eradication of more than 90% of the bacteria occurs in the first few TM
seconds of exposure. The antibacterial and antimycotic activity that Microoyn 60 exhibits in accordance with this standard is summarized in Table 5.
Table 5, Kill Times, Bacterium Catalog Time of action (reduction below 99.999%) Ps. aeruginosa ATCC 256191 min . .
St. aureus ATCC 6538 1 min E. colt ATCC 11229 1 ruin S. typhi CDC 99 1 min C. albicans ATCC 1 min . .
B. subtilis 9372 Low spore (104) 10 min High spore (106) _ _ 15 min [0184] The sporicidal activity trial was carried out in accordance with the PA.H0 [Pan-American Health Organization]fWHO protocol_ T.
[0185] The virucidal activity of Microcyn 60 has recently been confirmed in studies carried out in the United States against HIV and its activity against Listerta rnonocytogenes, IVIRSA and Mycobacterium tuberculasis has also been demonstrated. Thus, it has been = demonstrated that Microcyn 60, when it is administered as recommended, can eradicate bacteria, fungi, viruses and spores from one to fifteen minutes of exposure.
[01861 Additionally, the following is a clinical study that can be used to assess the efficacy of Microcyn 60 for the treatment of pharyngitishonsilitis. In this study, 40 patients with acute pharyngitis/tonsillitis caused by group A13-hemolytic Streptococcus and who = have not received treatment are recruited. The inclusion criteria are as follows: age 12 to 40 years and two or more of the following symptoms: oropharyngea] burning;
pain on swallowing; pharyngeal erythema or of the tonsils (with or without exudate);
cervical lymphadenopathy; and positive immunoassay for group A Streptococcus' antigen (StrepA
Test-Abbott Labs). The exclusion criteria are as follows: fever >38 C;
bronchospaam (excluded by the clinic); severe cough; sinusitis-rhinitis (excluded by the clinic); esophageal reflux (excluded by the clinic); use of antibiotics in the two weeks prior to the study;
patients who have taken part in another clinical study in the last 8 weeks;
rheumatic fever;
postatreptococcal glomerulonephritis; severe chronic cardiopathy; severe renal, hepatic or pulmonary insufficiencies; and pregnancy or lactation.
101871 At the beginning of the study, patients may use such concomitant medicines as antipyretics and analgesics, including paracetamol and acetylsalicylies but not TM
anti-inflammatories such as ibuprofen, Mesulid, COX-2 inhibitors, or steroids.
Written informed consent must be obtained before the patient submits to any specific procedure of the study.
[0188] The patients are evaluated in three visits. In the first visit, the patient clinically presents acute pharyngitis/tonsillitis, and the clinical history is taken, and a medical examination, rapid immunoassay for Streptococcus, and taking of a pharyngeal exudate is carried out. After being declared eligible and after having signed the letter of informed consent, the patient is prescribed two =pharyngeal cleansings of 30 sec and 5 mL
Microcern 60 each, These rinsings are done every 3 h for a total of four times a day for 3 days.
[0189] The second is made 72 h after having been treated with Microcyn 60. In the fM
second visit, the clinical evolution and side effects of Microeyn 60 are evaluated. A new pharyngeal exudate is taken, and it will be decided, in accordance with the clinical evolution, if the continuing treatment will be with antibiotics or a palliative. A third visit is done after 10 days to discharge the patient.
[0190] To be eligible and clinically evaluated in this study, each patient must present A
13-hemolytic Streptococcus pharyngitis/tonsillitis confirmed by culture. All the patients must Tt4 comply with 18 rinsings of 30 see and 5 rnL of Microcyn 60 each, or a maximum of 24 rinsings in the space of 72 h.
[0191] The primary parameter of efficacy is a reduction by 3 orders of magnitude in the bacterial load of the initial culture compared to the culture taken after the administration of TM TM
Microcyn 60. This bacteriological evaluation is realized 72 h after treatment with Microcyn 60. Secondary parameters of efficacy are the improvement reported clinically, with particular emphasis on the reduction of pharyngeal pain and dysphagia.
Clinical symptoms are reported in visits 1, 2 and 3.
[01921 Tolerance is evaluated by reports of adverse events. An adverse event is defined TM
as any symptomatic declaration of the patient who submits to the treatment with Microcyn 60, related or not to the antiseptic, that appears in the course of the treatment.
[0193] The results of bacteriological efficacy (the principal criterion of efficacy) are issued by a bacteriologist independently of the clinical symptoms. The testa for the group A
Streptococcus antigen and the initial pharyngeal exudate culture are done in the first visit (Visit 1), in accordance with the Schedule of Evainations and before the administration of TM
Microcyn 60. The second taking and culture of pliaryngeal exudate is carried out 72 h after the administration of IVIicrocylr60 (Visit 2). An antibiogram is done on all the cultures to determine the bacterial resistance to penicillin, erythrom.ycin, claiithromycin and lincomycin by means of the standard diffusion disc test. Bacteriological efficacy is defined as the reduction by three orders of magnitude of the bacterial count between the initial culture and the culture taken 72 h after administering MicrocynTM 60.
[01941 Bacteriological failure is indicated by a reduction of less than three orders of magnitude of the bacterial count in the culture at 72 h posttreatment.
Indeterminate responses are documented in those cases in which the transport of the sample has been delayed for more than 48 h, in those cases in which the swab has not been immersed in the transport medium, or in those cases in which the sample has been lost. These cases are outside the analysis of the study and are replaced by new cases until those of forty eligible patients have been completed.
[0195] The follow-up and reporting phase begins when the patient finishes the administration of IVIicrocyri 60, and from the second visit, In this evaluation, according to the clinical evolution and the presence of possible adverse effects, the patients are categorized as follows:
[0196) Therapeutic failurea if their initial signs and symptoms have not been eliminated or if there is worsening of their general condition with systemic symptoms, In these cases an oral antibiotic is prescribed, such as procaine penicillin, clarithromycin or azithromycin t at the dose and for the time that the treating doctor indicates, and they are evaluated in one week.
=
10191 Clinically cured if the symptoms and signs that were present in Visit I have been eliminated. In these cases in which the acute process is resolved, the patient is discharged and reported as clinically cured. In any case, the patient is asked to return for a third = check-up visit in one week.
[0198] Indeterminate evolution. The evolution of any patient who could not have been evaluated clinically for any good reason; for example, a coinfeetion, or if the evaluation was done very late, later than 72 h. In these cases, the patients is still able to be included in the analysis of the study provided it is possible to document the result of the pharyngeal exudate and culture at 72 h.
[0199] The statistical analysis used in this clinical study takes into account all the patients who have received at least 18 rinsinga of IVIierocynT"60 of 30 sec each in a period of 72 h. This same criterion is considered to include any patient in the analysis of tolerance.
The principal criterion for analysis of efficacy is the reduction of the bacterial count of P-hemolytic Streptococcus by three orders of magnitude in the culture carried out at 72 h TM
. posttreatment with IvEcrocyn 60. The statistical analysis is realized by means of a Wilcoxon paired samples teat. Statistical analysis of the elinioal variables is realized using the ANOVA test for quantitative variables. The minimal evaluable number of patients is 30 patients.
=
[0200] An adverse event is any contrary medical occurrence in a patient or subject of clinical investigation to whom a pharmaceutical product is administered and that does not necessarily have a causal relationship with that medicine. An adverse event can, therefore, be any unfavorable and unintended sign (including an abnormal laboratory finding), symptom or illness temporarily associated with the use of a medical product, whether it is considered to be related to this use or not. ,Preexisting conditions that deteriorate during a study are reported as adverse events, [0201] The treatment is suspended at any time during the 72 h of duration in case of adverse events that are moderate to severe in intensity. Subsequent treatment is determined by the treating doctor. In accordance with this example, the effectiveness of an MP water solution of the present invention for treating sinusitis is thus demonstrated.

[0202] This example demonstrates the virieidal activity of an exemplary ORP water solution against Adenovims-serotype 5. For this example Adenoviral (Ad) vectors based on human adenovirus type 5 which are Ela-, partially El-b, and partially E3-deleted were /-used. A shuttle plasmid containing the Green Fluorescent Protein (GFP) reporter gene under the transcriptional control of pCMV was prepared (pAd-Track ).
Homologous recombination of this pShuttle plasmid with AdEasy 1 plasmid was caiiied out in electrocompetent bacteria. Clones that had inserts were tested by restriction endonuclease digestions. Once confirmed, supercoiled plasmid DMA was transformed into DH1OB
cells for large scale amplification. Subsequently, 293 cells (ATCC 1573) were cultured in serum-free medium (OptiMEM-GIBCO) and transfected with recombinant plasmid digested with Pad. Infected cells were monitored for cytopathic effect, collected and lysed with three cycles of freezing and thawing. The resultant viruses (AdGFP) were purified with AdenoPure columns (BD Clontech) according to the manufacturer's instructions.
Viruses were quantitated by OD 260/280. Final yield was 1.52 X1011 pfil/mL.
[0203] The efficacy of the ORP water solution for inactivating adenovirus encoding the green fluorescence protein gene (AdGFP), was evaluated using a test based on the detection of fluorescence emission from HeLa cells infected with either, control AdGFP
viruses or ORP water solution-treated AdGFP, using fluorescence-activated flow cytometry.
Infection of HeLa cells was always carried out with 7.5 X 107 pfii/mL (i.e. 150 m.o.i.).
In all test conditions, cells appeared normal under light microscopy. The background fluorescence measured in control HeLa cells was 0.06%. After infection with control AdGFP, 88.51% of HeLa cells expressed GFP. Following exposure to the ORP water solution, adenovirus infectivity decreased inversely proportionally to the exposure period.
Accordingly, ORP
water solution-treated virus for 1, 5, and 10 min could only express GFP in 2.8%, 0.13%, and 0.09% of HeLa cell cultures, respectively. Considering the autofluorescence and the initial viral load for all tested conditions (i.e. 7.5 X 107 pfit), the infectious titer was 6.6 X
107 pfu in the control AdGFP-HeLa group. In the groups where the virus had been treated with the ORP water solution, the infectious titers were 2.0 X 106, 5.2 X 104 and 2.2 X 104 at one, five and ten minutes of virus exposure to the ORP water solution, respectively.
Therefore, the log-10 reduction factor was 1.5, 3.1, and 3.5 at one, five and ten minutes of viral exposure to the ORP water solution. Taken together, these results demonstrate that the virus exposure to the ORP water solution for 5 minutes achieves a log-10 reduction in the viral load of > 3.

[0204] This example demonstrates the viricidal effectiveness of an exemplary ORP
water solution against HIV using the United States Environmental Protection Agency protocol for disinfection of inanimate environmental surfaces.

[0205] The SF33 strain of HIV-1 used for this study. Peripheral blood mononuclear cells from healthy donors were activated with PHA (3 p.g/mL, Sigma) and human IL-2 (20 U/mL, Roche) in HUT media for three days. Cells were washed and infected with strain. Supernatant was collected on days 4 and 6, and tested for the p24 HIV-1 antigen by ELISA (Beckman Coulter). Superantant was centrifuged to remove cell and debris at 3000 RPM for 20 min at room temperature. Supernatant was removed, aliquoted, and the virus was stored at -80 C until the day of use.
[0206] Frozen aliquots were thawed at 37 C for two minutes immediately prior to its use. Serial logarithmic dilutions (-1 to -5) in HUT medium were used. Films of virus were prepared by spreading 0.2 ml of virus inoculum uniformly over the bottoms of 55 cm2 sterile polystyrene Petri dishes. The virus films were air-dried at room temperature (21 C) in a biological safety cabinet until they looked visibly dry (20 minutes). (To assure that the virus strain (SF33) was capable of replicating and causing cytopathic effects, the procedure was repeated with a viral suspension that had remained in HUT medium without being dried.) [0207] The control film was exposed to 2 ml HUT media for five minutes. The virus was then scraped and diluted. Separate dried films were exposed to 2 ml each of the ORP
water solution for five minutes at room temperature. Following the exposure time, the plates were scraped and their contents were resuspended. The virus-ORP water solution mixture was immediately diluted (10:1) in HUT medium. Serial log dilutions of this resulting suspension were assayed for infectivity. (To control for a possible direct cytotoxic effect of ORP water solution on MT-2 cells, a 2 ml aliquot of ORP water solution was diluted serially (10:1 to 10:5) in medium and inoculated into MT-2 cell cultures.) [0208] The MT-2 cell line was used as the indicator cell line in the infectivity assays.
This line shows a cytopathic effect consisting of sincitia formation when infected with HIV-1. Four microwells were inoculated with 0.2 ml of each dilution of the reconstituted virus suspension from test (reconstituted in ORP water) and control (reconsituted with control medium) groups. Uninfected cell controls were inoculated with test medium only. Cultures were incubated at 37 C and 5% CO2.
[0209] The cultures were scored periodically every two days for the presence or absence of cytopathic effect as well as presence of p24-HIV-1 antigen by ELISA.
Experimental infection with control HIV-1 exerted a cytopathic effect and Ag p24 protein release into the supernatant in infected MT-2 cultures. In contrast, treatment of HIV-1 with the ORP water solution for five minutes, achieved a log reduction factor > 3 in the viral load as measured in MT-2 cultures by both assays. These results thus demonstrate the level of efficacy that is in conformity with the EPA requirements for HIV-1 virucidal activity on inanimate surfaces.

[0210] This example demonstrates the effect of an exemplary ORP water solution versus hydrogen peroxide (HP) on the viability of human diploid fibroblasts (HDFs). To study this potential toxicity, HDFs were exposed in vitro to ORP water solution and hydrogen peroxide (HP). HP is known to be toxic to eukaryotic cells, increasing apoptosis and necrosis and reducing cellular viability. In this example, cell viability, apoptosis and necrosis were measured in HDFs exposed to pure ORP water solution and 880 mM
HP (a concentration employed for antiseptic uses of HP) for 5 and 30 minutes.
[02111 HDF cultures were obtained from three different foreskins, which were pooled and cryopreserved together for the purpose of this study. Only diploid cells were used for all experiments. On cell cycle analysis, DNA diploidy was defined as the presence of a single GO-G1 peak with a CV <1= 7% and a corresponding G2/M peak collected from at least 20,000 total events. FIG. 4A-4C disclose the results where exposure times of 5 and 30 minutes are depicted in white and black bars, respectively. Simultaneous analyses of these parameters were perfatnied in the same cell populations by flow cytometry using: A) 7-aminoactinomycin D (7AAD); B) Annexin V-FITC; and C) Propidiurn iodide. FIG.

disclose percentage values expressed as mean SD (ri=3).
[0212] Cell viability was 75% and 55% after a 5 minute exposure to antiseptic concentrations of full strength-ORP water solution and 880mM HP , respectively (FIG. 4A).
The effect of full strength ORP water solution on cell viability was comparable to a very diluted HP solution considered sublethal but not disinfectant (i.e. 500 p.M).
If the exposure was prolonged to 30 min, cell viability further decreased to 70% and 5%, respectively.
Apparently, the ORP water solution induced cell death through necrosis because 15% of the cells incorporated propidium iodide in the flow cytometry analysis at both times (FIG. 4C).
Apoptosis does not seem to be the mechanism by which the ORP water solution induces cell death because only 3 % of ORP water solution-treated cells exposed Annexin -V in the cellular surface (a marker of apoptosis) (FIG. 4B). This percentage was actually similar to the one measured in the control group. On the contrary, HP induced necrosis in 20% and 75% of treated cells and apoptosis in 15% and 20% after 5 and 30 min of exposure, respectively. Altogether these results show that the (undiluted) ORP water solution is far less toxic for HDFs than an antiseptic concentration of HP.

[0213] This example demonstrates the effect of an exemplary ORP water solution relative to hydrogen peroxide (HP) on oxidative DNA damage and formation of the DNA
adduct 8-hydroxy-2'-deoxiguanosine (8-0HdG) in HDFs. It is known that the production of 8-0HdG adducts in a cell is a marker of oxidative damage at specific residues of DNA. In addition, high cellular levels of this adduct correlate with mutagenesis, carcinogenesis and cellular aging.
[0214] FIG. 5 shows the levels of 8-0HdG adducts present in DNA samples from HDFs after control treatments, ORP water solution treatments and HP-treatments for 30 minutes.
DNA was extracted right after the exposure (TO, white bars) or three hours after the challenge period (T3, black bars). DNA was digested and the 8-0HdG adducts were measured by ELISA kit as per the manufacturer's instructions. Values are shown (ng/mL) as mean SD (n=3). The exposure to ORP water solution for 30 minutes did not increase the formation of adducts in the treated cells in comparison to control cells after incubation for 30 minutes. In contrast, the treatment with 500 tiM HP for 30 minutes increased the number of 8-0HdG adducts by about 25 fold relative to the control-treated or ORP water solution-treated cells.
[0215] The ORP water solution-treated cells were able to decrease the levels of 8-OHdG adducts if left in supplemented DMEM for 3 hours after exposure to the ORP water solution. Despite being allowed the same 3 hour recovery period, HP-treated cells still presented about 5 times more adducts than control-treated or ORP water solution treated cells. Altogether, these results demonstrate that acute exposure to the ORP
water solution does not induce significant DNA oxidative damage. These results also indicate that the ORP water solution will not likely induce mutagenesis or carcinogenesis in vitro or in vivo.

[0216] This example demonstrates the effects on HDFs of chronic exposure to low concentrations of an exemplary ORP water solution versus HP. It is known that chronic oxidative stress induces premature aging of cells. In order to mimick a prolonged oxidative stress, primary HDF cultures were chronically exposed to low concentrations of the ORP
water solution (10%) or HP (511M) during 20 population doublings. The expression and activity of the SA- p-galactosidase enzyme has previously been associated with the senescence process in vivo and in vitro. In this example the expression of the galactosidase enzyme was analyzed after one month of continuous exposure of HDF to the ORP water solution or HP. The results are depicted in FIG. 6. The expression of the enzyme SA-P-galactosidase was analyzed by counting the number of blue cells in microscopic fields. FIG. 6 shows that only HP treatment accelerated the aging of cells as indicated by the number of cells over-expressing SA-13-galactosidase (n= 3).
Chronic treatment with a low dose of HP increased the SA- J3-Gal expression in 86% of cells while the treatment with the ORP water solution did not induce the overexpression of this protein.
It can be concluded from this example that ORP water solution is not an inducer of premature cellular aging.

[0217] This example demonstrates the effect of an exemplary ORP water solution on the reduction of peritoneal bacterial load and on the reduction in the length of hospital stay in patients with peritonitis. All patients admitted to the Hospital Ruben Lefiero in Mexico City from June 2004 to January 2005, and with a diagnosis of acute generalized,secondary peritonitis, were included in the ORP water solution-treated group. Secondary peritonitis was defined as the result of the loss of integrity of the gastrointestinal or genito-urinary tract leading to contamination of the peritoneal space. Retrospective analysis of paired-cases presenting similar peritoneal infections between 2003 and 2004 at the same Institution was undertaken for the control group. Twenty consecutive patients were prospectively included in the ORP water solution-treated group (i.e. study group).
[0218] Upon admission, all patients underwent open surgery and intra-operative peritoneal lavage ("IOPL") of all quadrants of the abdomen. Intraoperative peritoneal-culture samples were taken in both groups. IOPL was perfoimed with 10 L of saline solution in both groups and followed by 5 L of the ORP water solution in the study group only. The excess ORP water solution was removed and no further rinsing was conducted.
The abdominal cavity was covered with a plastic mesh in both groups. However, in the study group, a dressing soaked in ORP water solution was left on top of the mesh. The dressing was changed Li. d. Emperic antimicrobial therapy was started in all patients with two antibiotics including clindamycin and cefotaxime or amikacin. Post-operative management in the study group included daily irrigation of the mesh with 100 mL of the ORP water solution t.i. d., without further rinsing or lavage. Severe cases of peritonitis required re-laparotomy and IOPL every 72 hours. Cultures of the peritoneal fluid for aerobic bacteria and fungi were taken every 72 hours in both groups for up to one week.
The duration of length of stay in the hospital was recorded.
[0219] Twenty control cases were selected from the medical records of the Institution and paired to the study group by age, sex and etiology of peritonitis. The control and study populations were comparable in age, sex and prognostic factors at entry. The anatomic origin and etiology of peritonitis was also similar for both groups (Table 6).

Table 6. Diagnoses.
DIAGNOSIS CONTROL STUDY TOTAL
Appendicitis 3 6 9 23.0 Post-trauma 1 3 4 10.0 Pancreatitis 6 3 9 23.0 Cholecystitis 1 2 3 7.5 Colon cancer 0 1 1 2.5 Small bowel 4 1 5 12.5 fistula Diverticulitis 1 1 2 5.0 Gastric 4 0 4 10.0 perforation Other Organ 0 2 2 5.0 perforation Other 0 1 1 2.5 TOTAL 20 20 40 100.0 [0220] Post-operative peritonitis was present in 19 and 17 patients of the control and study groups, respectively. All cases underwent surgical treatment followed by IOPL. The types of surgeries perfoimed in control /study groups, were: appendicectomy (3/6), gastric resection (4/0), cholecystectomy (1/2), pancreatic necrosectomy (6/3), small bowel suture/
resection with anastomosis (4/3), Hartman's operation (1/1); colonic resection (0/1) and miscellaneous (1/4). The use of antibiotics was very similar in both groups.
For control and study groups, three antibiotics were administered in 16 and 15 patients and more than 3 antibiotics in 4 and 5 cases, respectively. Patients were kept at the ICU and were mechanically ventilated post-operatively. Pen-operative intra-abdominal samples were taken in all 40 patients (Table 7).

Table 7. Microorganisms isolated from intraperitoneal samples and length of hospital stay in patients with peritonitis.
CONTROL GROUP STUDY GROUP
Isolated strains (n) Isolated strains (n) Hospital Days Hospital Days Organism Pen-op Post-op Pen-op Post-op C. albicans 10 7 19.4 7 0 6.3 E. coil 3 2 17.6 6 1 10.2 S. aureus 10 9 22.3 8 1 14.1 coagulase 0 0 0 2 0 17.8 neg. Staph.
A. baumanii 0 0 0 1 0 22.4 E. faecalis 3 3 23.7 1 0 28.6 A.
0 0 0 1 0 28.6 xilosoxidans P.
2 2 24.0 3 0 33.9 aeruginosa E. coacae 1 1 13.0 1 0 37.0 TOTAL 29 24 31.9 30 2 22.4 [0221] Samples were obtained in the pen-operative period and in the following week after intra-operative lavage with saline solution only (control group) or saline solution and ORP water solution (study group). The average hospital stay was then analyzed for each microorganism isolated at entry and for the whole group.
[0222] Pen-operative samples were taken in all 40 patients (Table 7). The average numbers of microorganisms grown from these samples were 29 in the control and 30 in the study group. The microorganisms isolated are shown in Table 8. Escherichia coli, Enterococcus, Staphylococcus aureus, Pseudomonas aeruginosa and fungi were isolated from these groups in 3/6, 4/2, 10/8, 2/3 and 10/7 occasions, respectively.
Positive cultures for A. xilosoxidans (1), coagulase negative Staphylococci (2) and A. baumanii (1) were only found in the study group.
[0223] A second intra-abdominal culture was taken during the first week after surgery (Table 7). At this time, the average number of organisms isolated in the control group (24) was almost the same as in the peri-operative sample (29). Importantly, there was a strong reduction in the number of positive samples in the study group. From 30 positive cultures in the peri-operative samples, only one remained positive for 3, aureus nd another one for B. con. In the analysis of hospital days, the control group had a longer stay (31.9 days) in comparison to the study group (22.4 days). Thus, the ORP water solution effectively reduced the peritoneal bacterial load and length of hospital stay in patients with peritonitis.
[0224] The mortality rates were also analyzed. There were six deaths in the control group and 3 in the study one. All deaths occurred in the first 30 days after the Drat surgery and the calculated relative risk was higher for the control group (i.e. 3.3 versus 0).
However, the sample size was too small to achieve statistical significance. No local side effects were recorded with the use of ORP water in the IOPL. Surviving patients in the study group were followed for 6 to 12 months. None of the 20 patients in the ORP water-treated group presented intestinal occlusion or data suggesting sclerosing peritonitis in the follow-up period.

[0225] This example demonstrates the effectiveness of an exemplary ORP water solution (Mycrocyn) in inhibiting mast cell degranulation. Mast cells have been recognized as principal players in type 1 hypersensitivity disorders, Multiple clinical symptoms observed in atopic dermatitis, allergic rhinitis, and.atopic asthma are produced by IgE-antigen stimulation of mast cells located in distinct affected tissues. The ciurently accepted view of the pathogenesis of atopic asthma is that allergens initiate the process by triggering IgE-bearing pulmonary mast cells (hies) to release mediators such as histamine, leukotri.enes, prostaglandins, kininie, platelet activating factor (PAY), etc.
in the so-called early phase of the reaction. In turn, these mediators induce bronchoconstriction and = enhance vascular permeability and mucus production. According to this model, following mast cell activation, those cells secrete various pro-inflammatory cytolcines in a late phase, including tumor necrosis factor alpha (TNFee), IL-4, IL-5 and 1L-6, which participate in the local recruitment and activation of other inflammatory cells such as eosinophils, basophile, T lymphocytes, platelets and. mononuclear phagocytes. These recruited ceps, in turn, contribute to the development of an inflammatory response that may then become autonomous and aggravate the asthmatic symptoms. This late phase response constitutes a long term inflammation process which can induce plastic changes in surrounding tissues (see Kumar et al., pp. 193-268).
[02261 Antigenic stimulation of mast cells occurs via the activation of the high affinity receptor for Iga (the FceRI receptor), which is a multimeric protein that binds IgE and subsequently can be aggregated by the interaction of the receptor-bound Igr, with a specific ¨

antigen, Its structure comprises four polypeptides, an IgE binding a chain, a p chain that serves to amplify its signaling capacity, and two disulfide-linked chains, which are the principal signal transducers via the encoded immunoreceptor tyrosine-based (ITAM) activation motif. Signaling pathways activated by the cross-linking of this receptor have been characterized using bone marrow-derived mast cells (EMMC), the rat leukemia cell line RBI. 2H3, mouse and rat peritoneal mast cells, and other mast cell lines, such as MC-9, In all of them, the presence of antigen bound to IgE causes mast cell degranulation, calcium mobilization, cytoskeletal re-arrangements and activation of different transcription factors (NFAT, NFKB, AP-1, PU,1, SP1, Ets, etc.) which activate cytoldne gene transcription that culminate with cytokine production.
[0227] Mature marine bone-derived mast cells (BMMC) were loaded with a monoclonal anti-Dinitrophenol IgE (300 ng/million cell) during 4 hours at 37 C. Culture media was removed and cells were resuspended in physiological buffer (Tyrode's Buffer/BSA). Cells were then treated 15 minutes at 37 C with distinct concentrations of the ORP water solution (in its Micrecyrrembodiment). Buffer was removed and cells resuspended in fresh Tyrode's/BSA and stimulated with different concentrations of antigen (Human Albumin coupled to Dinitroph.enol) during a 30 minute incubation at 37 C.
Degrannlation was measured by P-hexosaminidase activity determination in supernatants and pellets of the stimulated cells, using a colorimetrl.c reaction based on the capacity of this enzyme to hydrolize distinct carbohydrates. (1-hexosaminidase has been shown to be located in the same granules that contain histamine in mast cells.) The results (FIG. 7) demonstrate that degranulation is significantly reduced with increasing concentrations of the ORP water solution.
TM
= [0228] Surprisingly, the inhibitory effect of the ORP water solution (Microcyn) on mast cell degranulation at least is similar to that observed with the clinically effective "mast cell stabilizer" and established anti-allergic compound sodium cromoglycate (IntelTm).
Degranulation was again measured by 0-hexosaminidase enzymatic activity in the pellet and supernatant of stimulated cells, using a colorimetric reaction based on the capacity of this enzyme to hydrolize distinct carbohydrates. Cells loaded with anti-DNP
monoclonal IgE were stimulated with or without a 15 minute pre-incubation with sodium cromoglycate = (Interrm). Cromoglycate was no more effective than the ORP water solution in reducing = deiganulations (Compare FI(3.7 with PIG, g; both achieving at least about 50% reduction in degranulation.) Br "

.1 [0229j This example demonstrates the inhibitory activity of an exemplary OR? water solution on mast cell activation by a calcium ionophore.
(02301 Mast cells can be stimulated via the activation of calcium fluxes induced by a calcium ionophore. Signaling pathways activated by calcium ionophores have been characterized using bone marrow-derived mast cells (BMMC), the rat leukemia cell line RBL 2H3, mouse and rat peritoneal mast cells, and other mast cell lines, such as MC-9. In all of these systems the calcium mobilization causes mast cell degranulation (e.g.
histamine release), cytoskeletal re-arrangements, and activation of different transcription factors (e.g., NFAT, NFKB, AP-1, PU.1, SP1, Ets,) which activate cytokine gene transcription that culminate with cytokine production and secretion.
[0231) Mature murine BMMC were loaded with a monoclonal anti-Dinitrophenol IgE
(300 ng/million cell) during 4 hours at 37 C. Culture media was removed and cells were resuspended in physiological buffer (Tyrode's13uffer/BSA). Cells were then treated for 15 TM
minutes at 37 C with distinct concentrations of the ORP water solution (Microcyn). Buffer was removed and cells were resuspended in fresh Tyrode's/13SA and stimulated with calcium ionophore (100 rriM .A23187) during a 30 minute incubation at 37 C.
Degranulation was measured by ii-hexosaminidase activity determination in supernatants and pellets of the stimulated celled using a calorimetric reaction based on the capacity of this enzyme to hydrolyze distinct carbohydrates. (I3-hexosaminidase has been shown to be located in the same granules that contain histamine in mast cells.) The results (FIG. 8) demonstrate that degranulation is significantly reduced with increasing concentrations of the ORP water solution.
[0232] These results suggest that ORP water solution is a non-specific inhibitor of histamine release. Thus, ORP water solution ¨even at different concentrations-will inhibit the degranulation of mast cells independently of the stimulus (e.g. antigen or ionophqre), While not desiring to be bound by any theory, ORP water solution probably modifies the secretory pathway system at the level of the plasma membrane and/or cytoskeleton.
Because the mechanism of action of ORP water solution is believed to be non-specific, it is believed that ORP water solution can have broad potential clinical applications.

10233]. This example demonstrates the effect of an exemplary ORP water solution on the activation of mast cell cytokine gene transcription.
[0234) FIG.s 10A and 10B are RNAase protection assays from mast cells treated with ORP water solution at different concentration for 15 minutes and further stimulated by ....................................................................... "f''' antigen as described in Example 20. After stimulation, rnRNA was extracted using affinity chromatography columns (RNAeasy kit, Qiagene) and the RNAse Protection Assay VVEM
performed using standard kit conditions (Clontech, Becton & Dickinson) in order to detect mRNA production of distinct cytokines after antigen challenge, The cytokines included TNF-a, LIF, ILI 3, M-CSF, 11,6, MIT and 02.
[0235] FIG.s 10A and 10B show that the ORP solution water (Microcyn) did not modify cytokine m,RNA levels after antigen challenge in mast cells irrespective of the concentrations of ORP water solution or antigen used for the experiment.
[0236] In this study, the level of transcripts (i.e., the RNA content of stimulated mast cells) of proinflammatory genes was not changed in OR? water solution-treated mast cells after being stimulated with various concentrations of antigen. Thus, the ORP
water solution inhibited the secretory pathway of these cytokines without affecting their transcription.

[02371 This example demonstrates the inhibitory activity of an exemplary ORP water solution on mast cell secretion of TNF-11.
[0238] Mast cells were treated with different concentrations of ORP water solution for 15 minutes and further stimulated by antigen as described in Example 20, Thereafter, the tissue culture medium was replaced and samples of the fresh medium were collected at various periods of time (2-8 hours), for measuring TI\IF-a, levels. Samples were frozen and further analyzed with a commercial ELISA kit (Biosource) according to the manufacturer's instructions.
[0239] FIG. 11 shows that the level of secreted TNF-a to the medium from OR? water solution-treated cells after antigen stimulation is significantly decreased in comparison to the untreated cells.
[0240] Since the release of TNF-a and that of' various other pro-inflammatory molecules depends on a separate secretory pathway than that of histamine, it is possible that the ORP
solution can stop the secretion of those other cytokines leading the late inflammatory phase.
[0241] Thus, the ORP water solution inhibited TNF-a secretion of antigen-stimulated mast cells. These results are in agreement with clinical observations that the use of ORP
water solutions can decrease the inflammatory reaction in various wounds after surgical procedures.

[0242] This example demonstrates the inhibitory activity of an exemplary ORP water solution on mast cell secretion of Ml? 1-a.

[0243] Mast cells were treated with different concentrations of an exemplary ORP water solution (Microceei5 for 15 minutes and further stimulated by antigen as described in Example 70, Thereafter, the tissue culture medium was replaced and samples of the fresh medium were collected at various periods of time (2-8 hours) for measuring MI?
1-a. levels, Samples were frozen and further analyzed with a commercial ELISA kit (Eiiosource) according to the manufacturer% instructions.
[0244] FIG, 12 shows that the level of secreted MT 1-a to the medium from ORP water =
solution-treated oells after antigen stimulation was significantly decreased in comparison to the untreated cells.
[0245] This, the ORP water solution inhibited MIP 1-a secretion of antigen-stimulated mast cells. These results are in agreement with clinical observations that the use of ORP
water solutions can decrease the inflammatory reaction in various wounds after surgical procedures.
[0746] Since the release of MI? 1.-a and that of various other pro-inflammatory molecules depends on a separate secretory pathway than that of histamine, it is possible that the ORP solution can stop the secretion of those other cytokines leading the late = inflammatory phase, [02471 The results of analogous studies measuring IL-6 and 114-13 secretion are depicted in FIGS. 13 and 14.
[0248] Examples 20-23 and this example further demonstrate that the ORP water solution is able to inhibit early and late phase allergic responses initiated by IgE receptor crosslinking.
EX.AMPLE 25 [0249] This example demonstrates the results of a toxicity study using an exemplary ORP water solution.
[0250] An acute systemic toxicity study was performed in mice to determine the potential systemic toxicity of Miorocyn 60, an exemplary ORP water solution. A
single TM
dose (50 mlikg) of Microeyn 60 was injected intraperitone,ally in five mice.
Five control mice were injected with a single dose (50 mIlleg) of saline (0.9% sodium chloride). All animals were observed for mortality and adverse reactions immediately following the injection, at 4 hours after injection, end then once daily for 7 days. All animals were also weighed prior to the injection and again on Day 7. There was no mortality during the study.
All animals appeared clinically normal throughout the study. All animals gained weight.
The estimated Microcyn"I60 acute intraperitoneal LD50 from this study is greater than 50 , = ..................................................................... .
vvi.A.V1-1) Id111111livil k "

( mL/kg This example demonstrates that Microcyn 60 lacks significant toxicity and should be safe for therapeutic use accordance with the invention.

102511 This example illustrates a study conducted to determine the potential cytogenetic toxicity of an exemplary ORP water solution.
[02521 A micronucleus test was performed using an exemplary ORP
water solution (10% MicrooynTM ) to evaluate the inutagenic potential of intraperitoneal injection of an ORP water solution into mice. The mammalian in vivo micronucleus test is used for the identification of substances which cause damage to chromosomes or the mitotic apparatus of =rine polychromatic erythrocytes. This damage results in the formation of "micronuclei,- intracellular structures containing lagging chromosome fragments Or isolated whole chromosomes.The OR? water solution study included 3 groups of 10 mice each (5 males / 5 females): a test group, dosed with the OR? water solution; a negative control group, dosed with a 0.9% NaC1 solution; and a positive control group, dosed with a mutagenic cyclophosphamide solution. The test and the negative control groups received an intraperitoneal injection (12.5 ml/kg) of the ORP water solution or 0.9% NaC1 solution, respectively, for two consecutive days (days 1 and 2). The positive control mice received a single intraperitoneal injection of cyclophosphamide (8 mg/mL, 12.5 ml/kg) on day 2. All mice were observed immediately after injection for any adverse reactions. Al]
animals appeared clinically normal throughout the study and no sign of toxicity was noted in any group. On day 3, all mice were weighed and terminated.
102531 The femurs were excised from the terminated mice, the bone marrow was extracted, and duplicate smear preparations were performed for each mouse. The bone marrow slides for each animal were read at 40X magnification. The ratio of polychromatic erythrocytes (PCE) to normochromatic erythrocytes (NCE), an index of bone marrow toxicity, was determined for each mouse by counting a total of at least 200 erythrocytes.
Then a minimum of 2000 scoreable PCE per mouse were evaluated for the incidence of micronucleated polychromatic erythrocytes. Statistical analysis of the data were done using the Mann and Whitney test (at 5% risk threshold) from a statistical software package (Statview 5.0, SAS institute Inc., USA).
[02541 The positive control mice had statistically significant lower PCE/NC ratios when compared to their respective negative controls (males : 0.77 vs. 0.90 and females:
033 vs. 1.02), showing the toxicity of the cyclophosphamide on treated bone marrow However, there was no statistically significant difference between the PCE/NCE
ratios for the ORP water solution-treated mice and negative controls. Similarly, positive control mice 1,¨. &wry,' n had a statistically significant higher number of polychromatic erythrocytes bearing micronuclei as compared to both the OR? water solution-treated mice (males:
11.0 vs. 1.4 /
females: 12,6 vs. 0.8) and the negative controls (males: 11.0 vs. 0.6 /females: 12.6 vs. 1.0).
There was no statistically significant difference between the number of polychromatic erythrocytes bearing micronculei in ORP water solution-treated and negative control mice.
[0255] This example demonstrates that MicrocyriTM 10% did not induce toxicity or mutagenic effects after intraperitoneal injections into mice.

[0256] This study demonstrates the lack of toxicity of an exemplary OR?
water TM
solution, Dermacyn, [02571 This study was done in accordance with ISO 10993-51999 standard to determine the potential of an exemplary ORP water solution, Dermacyn:to cause cytotoxicity. A filter disc.with 0.1 mL of Dermacyri was placed onto an agarose surface, directly overlaying a monolayer of mouse fibroblast cells (L-929). The prepared samples were observed for crotoxic damage after 24 hours of incubation at 37 C in the presence of 5% COB. Observations were compared to positive and negative control samples.
The Dennacyricontaining samples did not reveal any evidence of cell lysis or toxicity, while positive and negative control performed as anticipated.
10258] Based on this study Dernaacyrt was concluded not to generate cytotoxic effects on murine fibroblasts.

[02591 This study was conducted with 16 rats to evaluate the local tolerability of an TM
exemplary ORP water solution, Dermacyn, and its effects on the histopathology of wound beds in a model of full-thickness dermal wound healing. Wounds were made on both sides of the subject rat. During the healing process skin sections were taken on either the left or TM
the right sides (e.g., Dermacyn-treated and saline-treated, respectively).
102601 1Vlasson's trichrome-stained sections and Collagen Type II stained sections of the Dermacynand saline-treated surgical wound sites were evaluated by a board-certified veterinary pathologist. The sections were assessed for the amount of Collagen Type 2 expression as a metnifestiation of connective tissue proliferation, fibroblast morphology and collagen formation, presence of neoepiderrnis in cross section, inflammation and extent of dermal ulceration.
[0261] The findings indicate that Derrnacyn was well tolerated in rats.
There were no treatment-related histopathologic lesions in the akin sections from either sides' wounds = a =al v/ V 41, I 1/141Y.,,JVV ,¨rl vvvg.vwv 1,01,511,1411 µ111.11 (DermacyrT-treated and saline-treated, respectively). There were no relevant histopathologie differences between the saline-treated and the Dermacyn-treated wound sites, indicating that the Dennaoyn-treatement was well tolerated. There were no significant differences between Collagen Type 2 expression between the saline-treated and the DermacynTm-treated wound Trvi sites indicating that the Dermacyn does not have an adverse effect on fibroblasts or on collagen elaboration during wound healing.

10262) This example demonstrates the use of an exemplary oxidative reductive potential water, Microcyn, in accordance with the invention as an effective antimicrobial solution.
[0263] An In-Vitro Time-Kill evaluation was performed using Microcyn oxidative reductive potential water. lvlieroeyn was evaluated versus challenge suspensions of fifty different microorganism strains ¨ twenty-five American Type Culture Collection (ATCC) strains and twenty-five Clinical Isolates of those same species -- as described in the Tentative Final Monograph, 'Federal Register, 17 June 1994, vol. 59:116, pg.
31444. The percent reductions and the Logic, reductions from the initial population of each challenge strain were determined following exposures to Microcyn for thirty (30) seconds, one (1) minute, three (3) minutes, five (5) minutes, seven (7) minutes, nine (9) minutes, eleven (11) minutes, thirteen (13) minutes, fifteen (15) minutes, and twenty (20) minutes.
All agar-plating was perforrned in duplicate and Microcyn was evaluated at a 99 % (v/v) concentration. All testing was performed in accordance with Good Laboratory Practices, as specified in 21 C.F.R. Part 58.
[0264] The following table surrnnarizes the results of the abovementioned In-Vitro Time-Kill evaluation at the thirty second exposure mark for all populations tested which were reduced by more than 5.0 Logi 0:
Table 8. 30-Second In-Vitro Kill.
Initial Post-Exposure Logo Percent No. Microorganism Species Population Population (CFU/mL) (CFU/mL) Reduction Reduction Acinetobacter baumannii 1 2340x 109 < 1.00 x 103 6.3692 99.9999 (ATCC #19003) Acinetobacter baumanaii Clinical Isolate . 1,8150 x 109 <1.0Q x 103 6.2589 99.9999 BSLI #061901Ab3 Bacteroides 3 4,40 x 101 <1.00 x 103 7.6435 99.9999 (ATCC #43858) , Bacteroides fragilis 4 Clinical Isolate 2.70 x 1010 <1.00 x 103 7.4314 99.9999 BSLI #061901Bf6 Candida albicans 2.70 x 1010 <1.00 x 103 6.3345 99.9999 (ATCC #10231) Candida albicans 6 Clinical Isolate 5.650 x 109 <1.00 x 103 6.7520 99.9999 BSLI #042905Ca Enterobacter aerogenes
7 1.2250 x 109 <1.00 x 103 6.0881 99.9999 (ATCC #29007) Enterobacter aerogenes
8 Clinical Isolate 1.0150 x 109 <1.00 x 103 6.0065 99.9999 BSLI #042905Ea Enterococcus faecalis
9 2.610 x 109 <1.00 x 103 6.4166 99.9999 (ATCC #29212) Enterococcus faecalis Clinical Isolate 1.2850 x 109 <1.00 x 103 6.1089 99.9999 BSLI #061901Efs2 Enterococcus faecium 11 VRE, MDR 3.250x 109 < 1.00 x 103 6.5119 99.9999 (ATCC #51559) Enterococcus faecium 12 Clinical Isolate 1.130 x 109 <1.00 x 103 6.0531 99.9999 BSLI #061901Efm1 Escherichia colt 13 5.00 x 108 <1.00 x 103 5.6990 99.9998 (ATCC #11229) Escherichia coli 14 Clinical Isolate 3.950 x 108 <1.00 x 103 5.5966 99.9997 BSLI #042905Ec1.
Escherichia coli 6.650x 108 < 1.00 x 103 5.8228 99.9998 (ATCC #25922) Escherichia coil 16 Clinical Isolate 7.40 x 108 <1.00 x 103 5.8692 99.9998 BSLI #042905Ec2 Haemophilus influenzae 17 1.5050 x 109 <1.00 x 104 5.1775 99.9993 (ATCC #8149) Haernophilus influenzae 18 Clinical Isolate 1.90 x 109 <1.00 x 104 5.2788 99.9995 BSLI #072605Hi Klebsiella oxytoca 19 MDR 1.120x 109 < 1.00 x 103 6.0492 99.9999 (ATCC #15764) Klebsiella oxytoca Clinical Isolate 1.810 x 109 <1.00 x 103 6.2577 99.9999 BSLI #061901Ko1 Klebsiella pneumoniae 21 subsp. ozaenae 1.390x 109 < 1.00 x 103 6.1430 99.9999 (ATCC #29019) Klebsiella pneumoniae 22 Clinical Isolate 9.950 x 108 <1.00 x 103 5.9978 99.9999 BSLI #061901Kpn2 Micrococcus luteus 23 6.950 x 108 <1.00 x 103 5.8420 99.9999 (ATCC #7468) Micrococcus luteus 24 Clinical Isolate 1.5150 x 109 <1.00 x 103 6.1804 99.9999 BSLI #061901M12 Proteus mirabilis 25 1.5950x 109 < 1.00 x 103 6.2028 99.9999 (ATCC #7002) Proteus mirabilis 26 Clinical Isolate 2.0950 x 109 <1.00 x 103 6.3212 99.9999 BSLI #061901Pm2 Pseudomonas aeruginosa 27 6.450 x 108 <1.00 x 103 5.8096 99.9999 (ATCC #15442) Pseudomonas aeruginosa 28 Clinical Isolate 1.3850 x 109 <1.00 x 103 6.1414 99.9999 BSLI #072605Pa Pseudomonas aeruginosa 29 5.550 x 108 <1.00 x 103 5.7443 99.9999 (ATCC #27853) Pseudomonas aeruginosa 30 Clinical Isolate 1.1650 x 109 <1.00 x 103 6.0663 99.9999 BSLI #061901Pa2 Serratia marcescens 31 9.950 x 108 <1.00 x 103 5.9978 99.9999 (ATCC #14756) Serratia marcescens 32 Clinical Isolate 3.6650 x 109 <1.00 x 103 6.5641 99.9999 BSLI #042905Sm Staphylococcus aureus 33 1.5050 x 109 <1.00 x 103 6.1775 99.9999 (ATCC #6538) Staphylococcus aureus 34 Clinical Isolate 1.250 x 109 <1.00 x 103 6.0969 99.9999 BSLI #061901Sa1 Staphylococcus aureus 35 1.740 x 109 <1.00 x 103 6.2405 99.9999 (ATCC #29213) Staphylococcus aureus 36 Clinical Isolate 1.1050 x 109 <1.00 x 103 6.0434 99.9999 BSLI #061901Sa2 Staphylococcus epidermidis 37 1.0550 x 109 <1.00 x 103 6.0233 99.9999 (ATCC #12228) Staphylococcus epidermidis 38 Clinical Isolate 4.350 x 108 <1.00 x 103 5.6385 99.9998 BSLI #072605Se Staphylococcus haemolyticus 39 8.150 x 10 < 1.00 x 103 5.9112 999999 (ATCC #29970) _ _ Staphylococcus haemolyticus 40 Clinical Isolate 8.350x 108 <
1.00 x 103 5.9217 99.9999 BSLI #042905Sha _ Staphylococcus hominis 41 2.790x 102 < 1.00 x 103 5.4456 99.9996 v(ATCC #27844) Staphylococcus hominis 42 Clinical Isolate 5,20 x 108 < 1.00 x 103 5.7160 99.999B
BSLI#042905Sho 43 Staphylococcus saprophyticus 9.10 x 108 <1.00 x 103 5.9590 99.9999 (ATCC #35552) - -Staphylococcus sap rophyticus 44 Clinical Isolate 1.4150x 10 <l.00x 103 6A508 99.9999 BSLI #042905Ss . _ 45 Streptococcus pneumonia 2,1450x 109 <1.00 x 104 5.3314 99.9995 = (ATCC #33400) _ Streptococcus pyogenes 5.20 x 109 < 1.00 x 103 6.7160 99.9999 _ , (ATCC #19615) Streptococcus pyogenes 47 Clinical Isolate 2.5920 x 109 <1.00 x 103 6.4141 99_9999 BSLI #061901Spy7 (0265) While their microbial reductions were measured at less than 5.0 Logi , Microcyn'm also demonstrated antimicrobial activity against the remaining three species not included in Ti Table 8. More specifically, a thirty second exposure to Microcyn reduced the population of Strepwcoccus pneunzonieze (Clinical Isolate; BSLI #072605Spn1) by more than 4,5 Logic), which was the limit of deteotion versus this species. Further, when challenged with TPI
Canclicla tropicalts (ATCC #750), Microcyn demonstrated a microbial reduction in excess of 3.0 Logo following a thirty second exposure. TAdditionally, when challenged with =
Candida tropica tis (BSLI #042905Ct), Microcynmdemonstrated a microbial reduction in excess of 3.0 Logo following a twenty minute exposure, 10266j The exemplary results of this In-Vitro Time-Kill evaluation demonstrate that MiCrOCylTMl oxidative reductive potential water exhibits rapid (i.e., less than 30 seconds in =
most cases) antimicrobial activity versus a broad spectrum of challenging microorganisms.
Microbial populations of forty-seven out of the fifty Gram-positive, Gram-negative, and yeast species evaluated were reduced by more than 5.0 Logi within thirty seconds of exposure to the product.
. .
_ LW ni ri illILVIVI,LJ,JV I .

[02671 This example demonstrates a comparison of the antimicrobial activity of an TM
exemplary oxidative reductive potential water, Mieroeyn, used in accordance with the invention versus HIBICLENSID chlorhexidine gluconate solution 4.0 % (w/v) and 0.9 %
sodium chloride irrigation (USP).
[0268j An In-Vitro Time-Kill evaluation was performed as described in Example 29 using HIBICLENS6 chlorhexidine gluconate solution 4.0 % (w/v) and a sterile 0.9 %
sodium chloride irrigation solution (T.ISP) as reference products. Each reference product was evaluated versus suspensions of the ten American Type Culture Collection (ATCC) strains specifically denoted in the Tentative Final Monograph. The data collected was then analyzed against the Microcyn microbial reduction activity recorded in Example 29.
102691 IVIicroc-yiroxidative reductive potential water reduced microbial populations of five of the challenge strains to a level comparable to that observed for the HIBICLENS41) chlorhexidine gluconate solution. Both Microcyrimand HIBICLENS* provided a microbial reduction of more than 5.0 Logio following a thirty second exposure to the following species: .kscherichia colt (ATCC #11229 and ATCC #25922), Pseudotnonas aeruginosa 1 (ATCC #15442 and ATCC #27853), and Serratia marcescens (ATCCI#14756). rurther, as TM
shown above in Table 9, Microcyn demonstrated excellent antimicrobial activity against Micr000ccus luteus (ATCC #7468) by providing a 5.8420 Logic, reduction after a thirty .second exposure. However, a direct Microcaccus luteus (ATCC #7468) activity comparison to H1BICLENS was not possible because after a thirty second exposure.
HIBICLENe reduced the population by the detection limit of the test (in this specific case, by more than 4.8 Logio). It is noted that the sterile 0,9 % sodium chloride irrigation solution reduced microbial populations of each of the six challenge strains discussed above by less than 0.3 Logi following a full twenty minute exposure.
102101 Microcyloxidative reductive potential water provided greater antimicrobial activity than both 14IBICLENSe and the sodium chloride irrigation for four of the challenge strains tested; Enter000ceus faecalis (ATCC #29212), Staphylococcus aureu.s.
(ATCC
#6538 and ATCC #29213), and Staphylococcus epidermidis (ATCC #12228). The following table summarizes the microbial reduction results of the In-Vitro Time-Kill evaluation for these four species:
Table 9. Comparative Results Microorganism Lo.glo Reduction Exposure Time Species Microcyn HIBICLENV NaC1 Irrigation 30 seconds 6.4166 1.6004 0,3180 30 seconds 6.4166 1.6004 0.3180 1 minute 6.4166 2.4648 0.2478 3 minutes 6.4166 5.2405 0.2376 5 minutes 6.4166 5.4166 0.2305 Enterococcus 7 minutes 6.4166 5.4166 0.2736 faecalis 9 minutes 6.4166 5.4166 0.2895 (ATCC #29212) 11 minutes 6.4166 5.4166 0.2221 13 minutes 6.4166 5.4166 0.2783 15 minutes 6.4166 5.4166 0.2098 20 minutes 6.4166 5.4166 0.2847 30 seconds 6.1775 1.1130 0.0000 1 minute 6.1775 1.7650 0.0191 3 minutes 6.1775 4.3024 0.0000 5 minutes 6.1775 5.1775 0.0000 Staphylococcus 7 minutes 6.1775 5.1775 0.0000 aureus 9 minutes 6.1775 5.1775 0.0000 (ATCC #6538) 11 minutes 6.1775 5.1775 0.0267 13 minutes 6.1775 5.1775 0.0000 15 minutes 6.1775 5.1775 0.0191 20 minutes 6.1775 5.1775 0.0000 30 seconds 6.2405 0.9309 0.0000 _ 1 minute 6.2405 1.6173 0.0000 3 minutes 6.2405 3.8091 0.0460 5 minutes 6.2405 5.2405 0.0139 Staphylococcus -7 minutes 6.2405 5.2405 0.0000 aureus 9 minutes 6.2405 5.2405 0.0113 (ATCC #29213) 11 minutes 6.2405 5.2405 . 0.0283 13 minutes 6.2405 5.2405 0.0000 15 minutes 6.2405 5.2405 0.0000 .
20 minutes 6.2405 5.2405 0.0615 30 seconds 5.6385 5.0233 0.0456 1 minute 5.6385 5.0233 0.0410 3 minutes 5.6385 5.0233 0.0715 5 minutes 5.6385 5.0233 0.0888 Staphylococcus -7 minutes 5.6385 5.0233 0.0063 epidermiclis 9 minutes 5.6385 5.0233 0.0643 (ATCC #12228) 11 minutes 5.6385 5.0233 0.0211 13 minutes 5.6385 5.0233 0.1121 15 minutes 5.6385 5.0233 0.0321 20 minutes 5.6385 5.0233 0.1042 [0271] The results of this comparative In-Vitro Time-Kill evaluation demonstrate that Microcyn oxidative reductive potential water not only exhibits comparable antimicrobial activity to HISICLENS against Escherichia coli (ATCC #11229 and ATCC #25922), Pseudomonas aeruginosa (ATCC #15442 and ATCC #27853), Serratia marcescens (ATCC
#14756), and Micrococcus luteus (ATCC #7468), but provides more effective treatment against Enterococcus faecalis (ATCC #29212), Staphylococcus aureus (ATCC #6538 and ATCC #29213), and Staphylococ.cus eptclermidis (ATCC 412228). As shown in Table 9, TM
Microcyn exemplifies a more rapid antimicrobial response (i.e., less than 30 seeonds) in some species. Moreover, exposure to MicrocynImresults in a greater overall microbial reduction in all species listed in Table 9.

(02721 This example demonstrates the effectiveness of an ORP water solution against Penicillin Resistant Streptococcus pneumontae (ATCC 51915).
10273i A culture of Streptoeoceu..s pneumoniae was prepared by using a frozen culture to inoculate multiple BAP plates and incubating for 2-3 days at 35-37 C with CO2.
Following incubation 3-7 mL of sterile diluent/mediurn was transferred to each agar plate and swabbed to suspend the organism. The suspensions from all plates were collected and transferred to a sterile tube and compared to a 4.0 McFarland Standard. The suspension was filtered through sterile gauze and vortex mixed prior to use in the testing procedure. 1 102741 An inocuhins of 0.1 ml of the organism suspension was added to 49.9 ml of the Microcyrror control substance. At each exposure period, the test mixture was mixed by swirling. The test mixture was exposed for 15 seconds, 30 seconds, 60 seconds, seconds, 5 minutes, and 15 minutes at 25.0 C.
[02751 A 1.0 ml sample was removed from the test mixture and added to 9.0 ml of neutralizer representing a 100 dilution of the neutralized inoculated test mixture. A 5 ml aliquot of the 100 neutralized inoculated test mixture was transferred to a 0.45 microliter filter apparatus pre-wetted with 10 ml of Butterfield's Buffer. The filter was rinsed with approximately 50 mL of Butterfield's Buffer, aseptioaliy removed from the apparatus, and transferred to a BAP plate. Additional 1:10 serial dilutions were prepared and one (1.0) ml aliquots of the 10-3- 10-4 dilutions of neutralized inoculated test mixture were plated in duplicate on BAP.
[0276] The bacterial subculture plates were incubated for 4814 hours at 35-37 C in CO2. Subculture plates were refrigerated for two days at 2-8 C prior to examination Following incubation and storage, the agar plates were observed visually for the presence of growth. The colony forming units were enumerated and the number of survivors at each exposure time was determined Representative subcultures demonstrating growth were appropriately examined for confirmation of the test organisms.

TM
[0277] The exemplary OP,P water solution, Microcyn, demonstrated a >99.93197279%
reduction of Penicillin Resistant Streptococcus pnewnonicte (ATCC 51915) after 15 second, 30 second, 60 second, 120 second, 5 minute, and 15 minute contact times at 25.0 C.

[02781 The objective of this Example is to determine the microbial activity of an exemplary ORP water solution (Derrnacyn, versus Bacitracin using a bacterial suspension assay.
TM
(0279] Derrnacyn is a ready to use product, therefore performing dilutions during testing was not required. Bacitracin is a concentrated re-hydrated solution requiring a dilution to 33 Units/ml, [0280] A purchased spore suspension of B. atropheus at 2.5 x 107 Jrn1 was used for testing. In addition fresh suspensions of Pseudomonas aeruginosa, and Staphylococcus aureus were prepared and measured using a spectrophotometer to ensure the titer was acceptable [0281] Nine microliters of test substance was added to 100 ill of microbe suspen,sion.
The test mixture was held at 20 C for the contact times of 20 seconds, 5 minutes, and 20 minutes. 1.0 ml of the test mixture (entire mixture) was added to 9,0 ml of neutralizer for 20 minutes (this is the original neutralization tube or 01e1T) 1.0 ml of the neutralized test = mixture was plated on Tryptic Soy Agar in duplicate for the 5 minute and 20 minute contact times. Additional dilutions and spread plates were used for the 20 second time point, to = achieve countable plates.
0282] All plates were incubated at 30 C -35 C for a total of 3 days and were evaluated 11.1 after each day of incubation. To determine the number of microbes exposed to Demiacyri and Bacitracin during testing the suspensions Four 10-fold dilutions were performed and 1.0 ml of the final 2 dilutions was plated in duplicate, where applicable.
(0283] Dermacyn1M when challenged with the test organisms showed total eradication (>4 log reduction) of the vegetative bacteria at all time points and for spores at the 5, and 20 minute time points. Bacitracin only produced approximately 1 log reduction.
Microcyrimat the 20 second time point showed some reduction in spores. Bacitracin showed no evidence of lowering the bacterial or spore populations over the time periods. tested.

[0284] This example demonstrates the effectiveness of two exemplary ORP water solutions (M1 and M2) against bacteria in biofilms.

[0285) The parental strain for all studies is P. aeruginosa PAW All planktonic strains were grown aerobically in minimal medium (2.56 g Na2HPO4, 2.08 g KI-12PO4, 1.0 g NH4C1, 0.04 g CaC12. 2 H20, 0.5 g MgSO4 = 71320, 0.1 mg CuSO4' 51320) 0,1 mg ZnSO4 H20, 0.1 mg FeSO4 = 71420, and 0.004 mg MriC12 = 4H20 per liter, pH 7,2) at 22 C in shake flasks at 220 rpm. Bioillins were grown as described below at 22 C in minimal medium.
Glutamate (130 mg/liter) was used as the sole carbon source.
102861 Biofihns were grown as described previously (Sauer et.al., J.
Bacteriol.
184:1140-1154 (2002). Briefly, the interior surfaces of silicone tubing of a once-through continuous flow tube reactor system were used to cultivate biofilms at 22 C. Biofilms were harvested after 3 days (maturation4 stage), 6 days (maturation- 2 stage), and 9 days (dispersion stage) of growth under flowing conditions, Blofilm cells were harvested from the interior surface by pinching the tube along its entire length, resulting in extrusion of the cell material from the lumen. The resulting cell paste was collected on ice. Prior to sampling, the bulk liquid was purged from the tubing to prevent interference from detached, planktonic cells.
[02871 The population size of planktonic and biofilm cells was determined by the number of CFU by using serial dilution plate counts. To do so, biofilms were harvested from the interior surface after various periods of time of exposure to SOSs.
Images of biotilnis grown in once-through flow cells were viewed by transmitted light with an Olympus BX60 microscope (Olympus, Melville, NY) and a _100 magnification AlOOPI
objective lens. Images were captured using a IVIa.gnafire cooled three-chip charge-coupled device camera (Optronics Inc., Cialena, CA) and a 30-ms exposure. In addition, confocal scanning laser microscopy was performed with an LSM 510 Meta inverted microscope (Zeiss, Heidelberg, Germany). Images were obtained with a LD-Apochrome 40/0.6 lens and with the ISM 510 Meta software (Zeiss).
[02881 A 2-log reduction was observed for Ml-treated biofilms within 60 min of treatment. The finding indicates that every 10.8 min (+/- 2,8 min), treatment with M1 results in a 50% reduction in biofilm Table 10. M1 Killing.
Time (min) Viability (%) 34 12.5 47 6.25 54 3.125 [0289] However, overall M2 was somewhat more effective in killing biofilms than M1 because the results indicated that every 4.0 min (+/- 1.2 min), treatment with M2 results in a 50% reduction in biofilm viability.
Table 11. M2 Killing.
Viability Time (min) (%) 2.5 50 12 12.5 15 6.25 20 3.125 [0290] Thus, ORP water is effective against bacteria in bioflims.

[0291] [BLANK]
[02921 The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms "comprising," "having,"
"including," and "containing" are to be construed as open-ended terms (i.e., meaning "including, but not limited to,") unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, Or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the speoication should be construed as indicating any non-claimed element as essential to the practice of the invention.
[0293) Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention, Variations of those prefen-ed = embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context, =

Claims (21)

WE CLAIM:
1. Use of an oxidative reductive potential water solution, wherein the solution is stable for at least two months and the solution has a pH of from 6.4 to 7.8, wherein the oxidative reductive potential water solution comprises a mixture of cathode water and anode water, wherein the oxidative reductive potential water solution comprises free chlorine species at a level of 10 ppm to 400 ppm, wherein the free chlorine species is selected from the group consisting of hypochlorous acid, hypochlorite ions, sodium hypochlorite, chlorite ions, dissolved chlorine gas, and mixtures thereof, for the treatment of atopic dermatitis in a patient.
2. The use of claim 1, wherein the oxidative reductive potential water solution is for administration topically.
3. The use of claim 1, wherein the oxidative reductive potential water solution is for administration as a liquid, steam, aerosol, mist or spray.
4. The use of claim 1, wherein the oxidative reductive potential water solution is for administration by aerosolization, nebulization or atomization.
5. The use of claim 1, wherein the oxidative reductive potential water solution is for administration in the form of droplets having a diameter in the range of from 0.1 micron to 100 microns.
6. The use of claim 1, wherein the atopic dermatitis is associated with cellular histamine and pro-inflammatory cytokine release.
7. The use of claim 1, wherein the atopic dermatitis is cell-mediated.
8. The use of claim 1, wherein the oxidative reductive potential water solution inhibits mast cell degranulation.
9. The use of claim 1, wherein the oxidative reductive potential water solution inhibits mast cell cytokine secretion.
10. The use of claim 1, wherein the oxidative reductive potential water solution is stable for at least six months.
11. The use of claim 1, wherein the oxidative reductive potential water solution is stable for at least one year.
12. The use of claim 1, wherein the pH of the oxidative reductive potential water solution is from 7.4 to 7.6.
13. The use of claim 1, wherein the oxidative reductive potential water solution comprises cathode water in an amount of from 10% to 50% by volume of the solution.
14. The use of claim 1, wherein the oxidative reductive potential water solution comprises cathode water in an amount of from 20% to 40% by volume of the solution.
15. The use of claim 1, wherein the oxidative reductive potential water solution comprises anode water in an amount of from 50% to 90% by volume of the solution.
16. The use of claim 1, wherein the oxidative reductive potential water solution comprises from 10% by volume to 50% by volume of cathode water and from 50% by volume to 90% by volume of anode water.
17. The use of claim 1, wherein the free chlorine species comprises from 15 ppm to 35 ppm hypochlorous acid.
18. The use of claim 1, wherein the free chlorine species comprises from 25 ppm to 50 ppm sodium hypochlorite.
19. The use of claim 1, wherein the free chlorine species comprises from 15 ppm to 35 ppm hypochlorous acid, from 25 ppm to 50 ppm sodium hypochlorite, and a pH
of from 6.2 to 7.8.
20. The use of claim 1, wherein the oxidative reductive potential water solution has a potential between-400 mV and +1300 mV.
21. The use of claim 1, for administration with at least one therapeutic agent selected from the group consisting of antibiotics, anti-viral agents, and anti-inflammatory agents, and combinations thereof
CA2637175A 2006-01-20 2007-01-22 Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution Expired - Fee Related CA2637175C (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US76064506P 2006-01-20 2006-01-20
US76055706P 2006-01-20 2006-01-20
US76063506P 2006-01-20 2006-01-20
US76056706P 2006-01-20 2006-01-20
US60/760,635 2006-01-20
US60/760,557 2006-01-20
US60/760,645 2006-01-20
US60/760,567 2006-01-20
PCT/US2007/060854 WO2007085018A2 (en) 2006-01-20 2007-01-22 Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution

Publications (2)

Publication Number Publication Date
CA2637175A1 CA2637175A1 (en) 2007-07-26
CA2637175C true CA2637175C (en) 2015-07-14

Family

ID=38198117

Family Applications (3)

Application Number Title Priority Date Filing Date
CA2637175A Expired - Fee Related CA2637175C (en) 2006-01-20 2007-01-22 Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
CA2637197A Expired - Fee Related CA2637197C (en) 2006-01-20 2007-01-22 Methods of treating or preventing peritonitis with oxidative reductive potential water solution
CA2637178A Expired - Fee Related CA2637178C (en) 2006-01-20 2007-01-22 Methods of preventing or treating sinusitis with oxidative reductive potential water solution

Family Applications After (2)

Application Number Title Priority Date Filing Date
CA2637197A Expired - Fee Related CA2637197C (en) 2006-01-20 2007-01-22 Methods of treating or preventing peritonitis with oxidative reductive potential water solution
CA2637178A Expired - Fee Related CA2637178C (en) 2006-01-20 2007-01-22 Methods of preventing or treating sinusitis with oxidative reductive potential water solution

Country Status (9)

Country Link
US (7) US8147444B2 (en)
EP (3) EP1993570B1 (en)
JP (3) JP5723084B2 (en)
KR (3) KR20080093135A (en)
AU (3) AU2007205860B2 (en)
BR (3) BRPI0706677A2 (en)
CA (3) CA2637175C (en)
HK (1) HK1130675A1 (en)
WO (3) WO2007085018A2 (en)

Families Citing this family (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6702949B2 (en) 1997-10-24 2004-03-09 Microdiffusion, Inc. Diffuser/emulsifier for aquaculture applications
US9168318B2 (en) 2003-12-30 2015-10-27 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and methods of using the same
NO20043150D0 (en) * 2004-07-23 2004-07-23 Ntnu Technology Transfer As "Heat recovery method and equipment"
JP5816406B2 (en) 2005-03-23 2015-11-18 オキュラス イノヴェイティヴ サイエンシズ、インコーポレイテッド Method for treating skin ulcer using redox potential aqueous solution
CA2606734C (en) * 2005-05-02 2016-06-21 Oculus Innovative Sciences, Inc. Method of using oxidative reductive potential water solution in dental applications
EP1945576B8 (en) * 2005-10-28 2013-02-20 APR Nanotechnologies S.A. Device comprising an electrode with nanocoating for preparing a highly stable aqueous solution and method for making this aqueous solution
WO2007085018A2 (en) 2006-01-20 2007-07-26 Oculus Innovative Sciences, Inc. Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
EP1986952A4 (en) * 2006-02-22 2011-03-30 Puricore Inc Methods of treating cystic fibrosis
US8445546B2 (en) 2006-10-25 2013-05-21 Revalesio Corporation Electrokinetically-altered fluids comprising charge-stabilized gas-containing nanostructures
AU2007349224B2 (en) 2006-10-25 2014-04-03 Revalesio Corporation Methods of wound care and treatment
AU2007308838B2 (en) 2006-10-25 2014-03-13 Revalesio Corporation Mixing device and output fluids of same
AU2007308840C1 (en) 2006-10-25 2014-09-25 Revalesio Corporation Methods of therapeutic treatment of eyes and other human tissues using an oxygen-enriched solution
US8784898B2 (en) 2006-10-25 2014-07-22 Revalesio Corporation Methods of wound care and treatment
US8784897B2 (en) 2006-10-25 2014-07-22 Revalesio Corporation Methods of therapeutic treatment of eyes
US8609148B2 (en) 2006-10-25 2013-12-17 Revalesio Corporation Methods of therapeutic treatment of eyes
US9999635B2 (en) 2007-01-16 2018-06-19 Realm Therapeutics, Inc. Methods and compositions for treating inflammatory disorders
WO2008089268A2 (en) 2007-01-16 2008-07-24 Puricore, Inc. Methods and compositions for treating conditions associated with infection and/or inflammation
JP2010516428A (en) * 2007-01-31 2010-05-20 ヘラー, アダム Electrochemical management of pain
JP5313874B2 (en) * 2007-03-22 2013-10-09 大幸薬品株式会社 Allergen deactivator
CA2684807A1 (en) 2007-04-05 2008-10-16 Velomedix, Inc. Automated therapy system and method
MX2009011291A (en) * 2007-04-25 2010-02-18 Akuatech S R L Highly stable electrolytic water with reduced nmr half line width.
JP5196864B2 (en) * 2007-05-14 2013-05-15 キヤノン株式会社 Probe set, probe carrier and inspection method
US8439960B2 (en) 2007-07-09 2013-05-14 Velomedix, Inc. Hypothermia devices and methods
ES2671372T3 (en) * 2007-07-26 2018-06-06 Azad Pharma Ag Pharmaceutical preparations comprising electrochemically activated hypochlorite solutions
US8706211B2 (en) 2007-08-17 2014-04-22 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having self-cleaning surfaces
US8460229B2 (en) 2007-08-17 2013-06-11 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having components that are actively controllable between transmissive and reflective states
US8162924B2 (en) * 2007-08-17 2012-04-24 The Invention Science Fund I, Llc System, devices, and methods including actively-controllable superoxide water generating systems
US8366652B2 (en) 2007-08-17 2013-02-05 The Invention Science Fund I, Llc Systems, devices, and methods including infection-fighting and monitoring shunts
US8114346B2 (en) * 2007-08-17 2012-02-14 The Invention Science Fund I, Llc Event-triggered ultraviolet light sterilization of surfaces
US8753304B2 (en) 2007-08-17 2014-06-17 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having acoustically actuatable waveguide components for delivering a sterilizing stimulus to a region proximate a surface of the catheter
US8029740B2 (en) 2008-07-11 2011-10-04 The Invention Science Fund I, Llc Event-triggered self-sterilization of article surfaces
US8702640B2 (en) 2007-08-17 2014-04-22 The Invention Science Fund I, Llc System, devices, and methods including catheters configured to monitor and inhibit biofilm formation
US8647292B2 (en) 2007-08-17 2014-02-11 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having components that are actively controllable between two or more wettability states
US20090048648A1 (en) * 2007-08-17 2009-02-19 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Self-sterilizing device
US8734718B2 (en) 2007-08-17 2014-05-27 The Invention Science Fund I, Llc Systems, devices, and methods including catheters having an actively controllable therapeutic agent delivery component
US10125359B2 (en) 2007-10-25 2018-11-13 Revalesio Corporation Compositions and methods for treating inflammation
US9523090B2 (en) 2007-10-25 2016-12-20 Revalesio Corporation Compositions and methods for treating inflammation
US9745567B2 (en) 2008-04-28 2017-08-29 Revalesio Corporation Compositions and methods for treating multiple sclerosis
JP5483820B2 (en) * 2007-11-29 2014-05-07 株式会社マンダム Antibacterial composition and deodorant
DE102008011807A1 (en) * 2008-02-29 2009-09-10 Aquagroup Ag Process for in-process decontamination in food processing and treatment and for reducing the germ content of cosmetics, pharmaceuticals, daily care products and animal and vegetable foods, as well as for the treatment of surfaces
MX2010011856A (en) 2008-05-01 2011-02-15 Revalesio Corp Compositions and methods for treating digestive disorders.
JP2011528357A (en) * 2008-07-15 2011-11-17 ビー・エイ・エス・エフ、コーポレーション Non-cytotoxic chlorine dioxide fluid
WO2010011927A1 (en) 2008-07-25 2010-01-28 Noventis, Inc. Compositions and methods for the prevention and treatment of cardiovascular diseases
WO2010025305A1 (en) * 2008-08-27 2010-03-04 Drug Enhancement Company Of America, Llc Use of non-toxic antimicrobial, oxychlorine, hypochlorous acid and superoxidized water products
WO2010065135A1 (en) 2008-12-04 2010-06-10 Searete, Llc System, devices, and methods including actively-controllable sterilizing excitation delivery implants
US8585627B2 (en) 2008-12-04 2013-11-19 The Invention Science Fund I, Llc Systems, devices, and methods including catheters configured to monitor biofilm formation having biofilm spectral information configured as a data structure
US20120041285A1 (en) 2008-12-04 2012-02-16 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Systems, devices, and methods including implantable devices with anti-microbial properties
US10617715B2 (en) 2008-12-22 2020-04-14 Sonoma Pharmaceuticals, Inc. Methods of treating or preventing biofilm associated infections with free available chlorine free available chlorine water
US20100196512A1 (en) 2009-02-04 2010-08-05 Basf Catalyst Llc Treatment of Non-Oral Biological Tissue with Chlorine Dioxide
AU2010229042A1 (en) * 2009-03-23 2011-11-10 Kross-Link Laboratories Method for suppressing or preventing fibrous adhesion formation using a multicomponent aqueous oxychlorine composition prepared on-site
US8815292B2 (en) 2009-04-27 2014-08-26 Revalesio Corporation Compositions and methods for treating insulin resistance and diabetes mellitus
US20120207853A1 (en) * 2009-05-11 2012-08-16 Oculus Innovative Sciences, Inc. Methods of treating or preventing influenza associated illness with oxidative reductive potential water solutions
KR101781229B1 (en) 2009-06-15 2017-09-22 오클루스 이노바티브 사이언시즈 인코포레이티드 Solution containing hypochlorous acid and methods of using same
EP2954901A1 (en) * 2009-06-17 2015-12-16 APR Nanotechnologies S.A. Methods of treating outer eye disorders using high orp acid water and compositions thereof
CA2787512C (en) 2010-01-19 2021-10-19 Howard M. Alliger A method of treating sinusitis, including chronic sinusitis
CN102985096B (en) 2010-04-13 2014-12-10 凯希特许有限公司 Compositions with reactive ingredients, and wound dressings, apparatuses, and methods
KR20130092977A (en) 2010-04-14 2013-08-21 하이포-스트림 리미티드 Device for preparing dilute disinfectant solution
KR20130114581A (en) 2010-05-07 2013-10-18 레발레시오 코퍼레이션 Compositions and methods for enhancing physiological performance and recovery time
US9622670B2 (en) 2010-07-09 2017-04-18 Potrero Medical, Inc. Method and apparatus for pressure measurement
CN107670044A (en) * 2010-07-22 2018-02-09 雷文制药有限公司 Comprising the treatment using magnetic dipole stabilizing solutions or improve disease and strengthen the method for performance
JP2013533320A (en) 2010-08-12 2013-08-22 レバレジオ コーポレイション Compositions and methods for treating tauopathy
CN107439588B (en) 2011-03-18 2020-07-07 瑞尔姆治疗股份有限公司 Stable hypohalous acid solutions
US9381214B2 (en) 2011-03-18 2016-07-05 Puricore, Inc. Methods for treating skin irritation
US11452778B2 (en) 2011-03-18 2022-09-27 Urgo Us, Inc. Stabilized hypohalous acid solutions
ITRM20110390A1 (en) * 2011-07-21 2013-01-22 Eramo Alessandra D PRODUCT TO STIMULATE THE ENDOGENOUS PRODUCTION OF INFLAMMATION MEDIATORS.
KR101297712B1 (en) 2011-11-02 2013-08-20 (주)그린제약 Biocide/disinfection agent containing hypochlorous acid water and soy protein
US9101678B2 (en) 2011-11-03 2015-08-11 Elwha Llc Heat-sanitization of surfaces
US20150079039A1 (en) * 2012-04-13 2015-03-19 The Regents Of The University Of California Sinusitis diagnostics and treatments
US9815714B2 (en) * 2012-12-11 2017-11-14 Slate Group, Llc Process for generating oxygenated water
US9918477B2 (en) 2013-05-22 2018-03-20 Sonoma Pharmaceuticals, Inc. Stabilized hypochlorous acid solution and use thereof
WO2015061753A1 (en) * 2013-10-24 2015-04-30 Reoxcyn Discoveries Group, Inc. Redox signaling gel formulation
EP3062799A1 (en) 2013-10-29 2016-09-07 Hypo-Stream Limited Anti-inflammatory solution comprising sodium hypochlorite
DE102014204142B4 (en) * 2014-03-06 2016-05-25 Hans-Jürgen Dörfer Process for the preparation of aqueous chlorine dioxide solutions and use of an apparatus for carrying out the process
MX2017007852A (en) 2014-12-16 2018-01-11 Realm Therapeutics Inc Hypochlorous acid formulations and methods for treating skin conditions.
US10617716B2 (en) 2014-12-16 2020-04-14 Urgo Us, Inc. Hypochlorous acid formulations and methods for treating skin conditions
WO2016126855A1 (en) 2015-02-03 2016-08-11 Tipul Biotechnology, LLC Devices and methods for electrolytic production of disinfectant solution from salt solution in a container
WO2018109169A1 (en) 2016-12-15 2018-06-21 Adept Water Technologies A/S Device for producing aqueous liquid having free available chlorine (fac)
EP3645017A4 (en) 2017-06-28 2021-05-19 Collidion, Inc. Compositions, methods and uses for cleaning, disinfecting and/or sterilizing
US20190125825A1 (en) * 2017-10-12 2019-05-02 High Point University Small-molecule adjuvants for antibiotics to address antibiotic resistance
DE102017131104A1 (en) 2017-12-22 2019-06-27 B. Braun Avitum Ag Blood treatment machine with SOS generator and disinfection process
WO2019140408A1 (en) 2018-01-14 2019-07-18 Collidion, Inc. Compositions, kits, methods and uses for cleaning, disinfecting, sterilizing and/or treating
FR3088543B1 (en) * 2018-11-21 2021-03-19 Waterdiam France Treatment of skin conditions with electrolyzed water
CN109172601A (en) * 2018-08-20 2019-01-11 四川建元天地环保科技有限公司 Electrolyte is eliminating the purposes in pseudomonas aeruginosa
US20200281969A1 (en) * 2019-03-05 2020-09-10 Wonder Spray, LLC Inhibiting viral and bacterial activity using low concentration hypochlorous acid solutions
US11642372B2 (en) 2020-05-01 2023-05-09 Tygrus, LLC Therapeutic material with low pH and low toxicity active against at least one pathogen for addressing patients with respiratory illnesses
US11826382B2 (en) 2020-05-01 2023-11-28 Tygrus, LLC Therapeutic material with low pH and low toxicity active against at least one pathogen for addressing patients with respiratory illnesses
DE102020113383A1 (en) * 2020-05-18 2021-11-18 Cytopharma Gmbh Pharmaceutical preparation for use in the treatment of SIRS
JP7385616B2 (en) 2021-03-22 2023-11-22 株式会社日本トリム Electrolyzed water generator

Family Cites Families (417)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3066095A (en) 1959-09-30 1962-11-27 Hagan Chemicals & Controls Inc Water purification agents and method of using same
US3616355A (en) 1968-08-05 1971-10-26 Kdi Chloro Guard Corp Method of generating enhanced biocidal activity in the electroylsis of chlorine containing solutions and the resulting solutions
GB1367067A (en) 1970-10-06 1974-09-18 Wilkinson Sword Ltd Compositions containing a source of hypochlorite ions
GB1422795A (en) 1972-05-31 1976-01-28 Bunyan J Antiseptic and non-toxic substance and a method of making the same
US3975246A (en) * 1973-06-09 1976-08-17 Sachs-Systemtechnik Gmbh Method of disinfecting water
US3975247A (en) 1974-01-23 1976-08-17 Stralser Bernard J Treating sewage and recovering usable water and solids
DE2442078A1 (en) * 1974-09-03 1976-03-18 Sachs Systemtechnik Gmbh METHOD AND DEVICE FOR THE DISINICIATION AND DETOXIFICATION OF LIQUIDS BY ANODIC OXYDATION WITH THE ADDITION OF SILVER
US3949742A (en) 1974-09-20 1976-04-13 Frigitronics, Inc. Medical dressing
DE2607906A1 (en) * 1976-02-26 1977-09-01 Hans Einhell Inh Josef Thannhu ELECTROLYSIS CELL FOR WATER TREATMENT
US4146578A (en) 1977-12-27 1979-03-27 Olin Corporation Hypochlorous acid process
US4190638A (en) 1978-06-07 1980-02-26 Ppg Industries, Inc. Production of hypochlorous acid
US4242446A (en) * 1978-07-26 1980-12-30 Coulter Electronics, Inc. Method for determining a substance in a biological fluid and reagent combination for use in the method
WO1980001139A1 (en) * 1978-12-06 1980-06-12 Svedman Paul Device for treating tissues,for example skin
US4236992A (en) * 1979-08-06 1980-12-02 Themy Constantinos D High voltage electrolytic cell
JPS5684089U (en) 1979-11-30 1981-07-07
US4296103A (en) * 1980-08-08 1981-10-20 Felipe Laso Stabilized solution of chlorine oxides
DE3046324A1 (en) 1980-12-09 1982-12-16 Rudolf Dr.med. 6900 Heidelberg Anderson Microbicide effective against microbes which affect the lungs - contains calcium hypochlorite and water which produce hypochlorous acid, which produces iso:hypochlorous acid which releases active oxygen
SU1296156A1 (en) 1985-02-14 1987-03-15 Ю.Ф. Исаков, В.А. Михедьсон, С.А. Байдин, Г.А. Гинодман, А.А. Нурмухамедов, Д.Д. Казанский .А. ФатjaxoB Method of healing wounds
US4670252A (en) 1985-05-24 1987-06-02 The Procter & Gamble Company Treatment of oral diseases
US4615937A (en) * 1985-09-05 1986-10-07 The James River Corporation Antimicrobially active, non-woven web used in a wet wiper
US4693832A (en) 1985-11-27 1987-09-15 Quantum Technologies, Inc. Preparation of safe drinking water
US4970216A (en) 1986-03-17 1990-11-13 Richardson Vicks, Inc. Skin treatment composition and method
US4666621A (en) 1986-04-02 1987-05-19 Sterling Drug Inc. Pre-moistened, streak-free, lint-free hard surface wiping article
US4781974A (en) 1986-04-23 1988-11-01 James River Corporation Antimicrobially active wet wiper
IT1207620B (en) 1987-02-27 1989-05-25 Castellini Spa METHOD AND EQUIPMENT FOR COLD STELIZATION OF SURGICAL INSTRUMENTS, IN PARTICULAR FOR DENTAL INSTRUMENTS
US5906810A (en) * 1987-03-17 1999-05-25 Turner; Robert E. Formulations and uses thereof in the prevention and treatment of oral lesions
US4767511A (en) 1987-03-18 1988-08-30 Aragon Pedro J Chlorination and pH control system
US5388571A (en) * 1987-07-17 1995-02-14 Roberts; Josephine A. Positive-pressure ventilator system with controlled access for nebulizer component servicing
FI82808C (en) * 1987-12-31 1991-04-25 Etelae Haemeen Keuhkovammayhdi Ultraljudfinfördelningsanordning
JPH0249798B2 (en) 1988-01-28 1990-10-31 Kogyo Gijutsuin JUKIKAGOBUTSUGANJUSUINOSHORIHOHO
JPH01218682A (en) 1988-02-29 1989-08-31 Tatsuo Okazaki Method for regulating electrolytic water and apparatus thereof
JPH078768B2 (en) 1988-06-06 1995-02-01 ジプコム株式会社 Sterilized water
JP2638611B2 (en) 1988-06-17 1997-08-06 紀久雄 及川 Method for producing stabilized chlorine dioxide aqueous solution
DE3929411A1 (en) * 1988-09-22 1990-03-29 Siegfried Natterer Pharmaceutical preparation and process for its preparation
JPH0293088A (en) 1988-09-29 1990-04-03 Permelec Electrode Ltd Method and device for water electrolysis
US5037627A (en) 1988-10-31 1991-08-06 Olin Corporation Hypochlorous acid process
JPH02149395A (en) 1988-11-30 1990-06-07 Jipukomu Kk Apparatus and method of preparing aqueous disinfectant
US4979938A (en) * 1989-05-11 1990-12-25 Iomed, Inc. Method of iontophoretically treating acne, furuncles and like skin disorders
SE466196B (en) * 1989-06-13 1992-01-13 Pavel Voracek PROCEDURE AND DEVICE FOR ELECTRICAL TREATMENT OF AN ELECTROLYTIC SOLUTION AND SOLUTION FRACTIONS PREPARED ACCORDING TO THE PROCEDURE
US6638413B1 (en) 1989-10-10 2003-10-28 Lectro Press, Inc. Methods and apparatus for electrolysis of water
US5271943A (en) 1989-10-27 1993-12-21 Scott Health Care Wound gel compositions containing sodium chloride and method of using them
JPH03236315A (en) 1989-12-05 1991-10-22 Nippon Oil & Fats Co Ltd Antipsychotic agent
US5152757A (en) 1989-12-14 1992-10-06 Brigham And Women's Hospital System for diagnosis and treatment of wounds
US5126057A (en) 1990-01-22 1992-06-30 Auburn Research Foundation Disinfecting with N,N'-dihaloimidazolidin-4-ones
US5084011A (en) 1990-01-25 1992-01-28 Grady Daniel J Method for oxygen therapy using hyperbarically oxygenated liquid
JPH03247134A (en) 1990-02-26 1991-11-05 Nec Corp Pcm signal interface circuit
US5622848A (en) * 1990-05-23 1997-04-22 Medical Discoveries, Inc. Electrically hydrolyzed salines as microbiocides for in vitro treatment of contaminated fluids containing blood
US5334383A (en) * 1990-05-23 1994-08-02 Medical Discoveries, Inc. Electrically hydrolyzed salines as in vivo microbicides for treatment of cardiomyopathy and multiple sclerosis
US5128136A (en) 1990-07-16 1992-07-07 The Oregon Health Sciences University Wound healing kit comprised of gelable collagen
US5187154A (en) * 1990-12-13 1993-02-16 Board Of Regents, The University Of Texas System Diagnosis and treatment of humans with diabetes or at risk to develop diabetes
US5244768A (en) * 1991-02-15 1993-09-14 Fuji Xerox Co., Ltd. Manufacturing process for an electrophotographic toner
GB2253860B (en) 1991-03-12 1995-10-11 Kirk And Charashvili Internati The electrochemical treatment of water and a device for electrochemically treating water
JP3236315B2 (en) 1991-08-27 2001-12-10 修生 澄田 Pure water electrolysis tank with an intermediate chamber
US5152915A (en) 1991-09-03 1992-10-06 Olin Corporation Recovery of dichlorine monoxide from hypochlorous acid solutions
US5636643A (en) 1991-11-14 1997-06-10 Wake Forest University Wound treatment employing reduced pressure
DE69218901T2 (en) 1991-12-10 1997-07-17 Tdk Corp Ultrasonic atomizer
JP3247134B2 (en) 1991-12-27 2002-01-15 修生 澄田 Liquid in which hydrogen ion or hydroxide ion and redox substance coexist by electrolysis of pure water and method for producing the same
JP2615308B2 (en) 1992-02-20 1997-05-28 英雄 早川 Water reforming method
JP2611080B2 (en) 1992-02-20 1997-05-21 英雄 早川 Water reforming method
US5639441A (en) * 1992-03-06 1997-06-17 Board Of Regents Of University Of Colorado Methods for fine particle formation
US5622725A (en) * 1992-03-20 1997-04-22 Alcide Corporation Wound disinfection and repair
US5427667A (en) * 1992-04-03 1995-06-27 Bakhir; Vitold M. Apparatus for electrochemical treatment of water
US5287847A (en) * 1992-07-24 1994-02-22 Vortran Medical Technology, Inc. Universal nebulizer
US6544502B2 (en) * 1992-09-11 2003-04-08 Wasatch Pharmaceutical Inc. Skin treatment with a water soluble antibiotic dissolved in an electrolyzed water
JPH06206825A (en) 1992-12-11 1994-07-26 Nippon Rooshiyon Kk Agent for treatment of dermatosis caused by trichophyton, eczema, etc., and agent for skin disinfection and activation
JPH06182345A (en) 1992-12-21 1994-07-05 Tonen Corp Water purification device
GB9227020D0 (en) 1992-12-24 1993-02-17 Solvay Interox Ltd Microbicidual compositions
DE69409996T2 (en) 1993-02-22 1999-01-14 Nippon Intek Co Method and device for producing electrolytic water
JP2623204B2 (en) 1993-02-26 1997-06-25 英雄 早川 Water reforming method
JPH06312183A (en) 1993-04-28 1994-11-08 Shinko Pantec Co Ltd Method and apparatus for producing electrolytic ion water
DE4317174A1 (en) * 1993-05-22 1994-11-24 Bosch Gmbh Robert Composite system with at least two inorganic ceramic layers and process for their production
JPH06335685A (en) 1993-05-28 1994-12-06 Funai Electric Co Ltd Ionized water generator
JP2652609B2 (en) * 1993-05-31 1997-09-10 ミズ株式会社 Electrolyzed water generator
JPH0710762A (en) 1993-06-25 1995-01-13 Miura Denshi Kk Gargle for stomatitis
JP3325081B2 (en) 1993-06-28 2002-09-17 英雄 早川 Water purification method and apparatus
KR100314138B1 (en) 1993-06-29 2001-12-28 마틴 보게스 로버트 Metering device
JP3458341B2 (en) 1993-07-12 2003-10-20 有限会社コヒーレントテクノロジー Method for producing washing water containing hydrogen ions or hydroxyl ions in excess of counter ions and obtained washing water
JP3321256B2 (en) 1993-07-30 2002-09-03 ミズ株式会社 Water storage type electrolyzed water generator
JP3504344B2 (en) * 1993-08-31 2004-03-08 三浦電子株式会社 Livestock skin disease treatment water
JPH07104221A (en) 1993-10-01 1995-04-21 Tomey Technol Corp Method for cleaning and sterizing contact lens
US5965452A (en) 1996-07-09 1999-10-12 Nanogen, Inc. Multiplexed active biologic array
JP3500173B2 (en) 1993-12-01 2004-02-23 ホシザキ電機株式会社 Electrolyzed water production equipment
JP3420820B2 (en) 1994-02-05 2003-06-30 ペルメレック電極株式会社 Method and apparatus for producing electrolytic acidic water
JP3324260B2 (en) 1994-02-28 2002-09-17 宮▲崎▼ 浩司 Corner building tile roofing structure
JP2830733B2 (en) 1994-03-25 1998-12-02 日本電気株式会社 Electrolytic water generation method and electrolysis water generation mechanism
US5599438A (en) * 1994-03-25 1997-02-04 Nec Corporation Method for producing electrolyzed water
JP2737643B2 (en) 1994-03-25 1998-04-08 日本電気株式会社 Method and apparatus for producing electrolytically activated water
JP3468835B2 (en) * 1994-05-09 2003-11-17 ホシザキ電機株式会社 Electrolyzed water generator
JPH07323289A (en) 1994-05-30 1995-12-12 Janome Sewing Mach Co Ltd Superoxidized water and method therefor
JPH07328640A (en) 1994-06-02 1995-12-19 Nippon Intec Kk Effect judging method of electrolytic sterilization washing water and device therefor
JP3396853B2 (en) 1994-06-21 2003-04-14 有限会社コヒーレントテクノロジー Water production method and obtained water
JP2949322B2 (en) 1994-07-19 1999-09-13 株式会社アロンワールド Ionized water, its production method and production apparatus
WO1996003881A1 (en) 1994-07-29 1996-02-15 Toto Ltd. Strongly acidic sterile water containing low-concentration hypochlorous acid, method of forming strongly acidic sterile water containing low-concentration hypochlorous acid, apparatus therefor, and apparatus for forming and delivering strongly acidic sterile water containing low-concentration hypochlorous acid
JPH0852476A (en) 1994-08-12 1996-02-27 Janome Sewing Mach Co Ltd Superoxidized water forming device
JP3465367B2 (en) * 1994-08-23 2003-11-10 東陶機器株式会社 Ion-rich water generator
JPH0861788A (en) 1994-08-25 1996-03-08 Kozo Yamanoi Solar heat water heater
US6117285A (en) 1994-08-26 2000-09-12 Medical Discoveries, Inc. System for carrying out sterilization of equipment
US5507932A (en) * 1994-08-26 1996-04-16 Schlumberger Technology Corporation Apparatus for electrolyzing fluids
JP3488294B2 (en) * 1994-10-14 2004-01-19 忠正 中村 Water treatment method and treatment device
JP2857334B2 (en) * 1994-10-18 1999-02-17 行正 佐藤 Electrolytic ionic water generator
KR100227969B1 (en) * 1994-10-20 1999-11-01 사카모토 시게토시 Production system of electrolyzed water
JPH08126873A (en) 1994-10-28 1996-05-21 Nec Corp Washing and device for electronic parts and the like
JPH08126886A (en) 1994-10-28 1996-05-21 Japan Organo Co Ltd Production of ultrapure water and device therefor
JP3181796B2 (en) * 1994-10-28 2001-07-03 日本電気株式会社 Electrolyzed water production equipment
JP3181795B2 (en) 1994-10-28 2001-07-03 オルガノ株式会社 Electrolyzed water production equipment
ES2125672T3 (en) 1994-11-09 1999-03-01 Procter & Gamble CLEANSING TISUS TREATED WITH A WATER EMULSION IN LIPID.
DK173485B1 (en) * 1994-12-02 2000-12-18 Thure Barsoee Carnfeldt Process for disinfecting or sterilizing food, feed, machinery and equipment for food and feedstuffs
JPH08164192A (en) 1994-12-15 1996-06-25 Life Sapooto:Kk Vegetative deodorant
CA2209315C (en) 1995-01-06 2009-12-15 Australian Biomedical Company Pty.Ltd. Gibberellins compounds for veterinary and medicinal applications
JPH08252310A (en) * 1995-01-17 1996-10-01 Miura Denshi Kk Washing and disinfecting method of artificial dialyzing device using electrolytically generated acidic water, and device therefor
EP0723936B1 (en) 1995-01-30 1999-05-12 First Ocean Co., Ltd. A composite electrode construction for electrolysis of water
US5876743A (en) 1995-03-21 1999-03-02 Den-Mat Corporation Biocompatible adhesion in tissue repair
US6007693A (en) 1995-03-30 1999-12-28 Bioquest Spa halogen generator and method of operating
US5578022A (en) * 1995-04-12 1996-11-26 Scherson; Daniel A. Oxygen producing bandage and method
JP3313263B2 (en) 1995-04-15 2002-08-12 株式会社東芝 Electrolytic water generation method, its generation apparatus, and semiconductor manufacturing apparatus
HU213450B (en) 1995-04-26 1997-06-30 Ladanyi Jozsef Gel contains gelatin and process for producing it
JP2832171B2 (en) 1995-04-28 1998-12-02 信越半導体株式会社 Apparatus and method for cleaning semiconductor substrate
US5871623A (en) * 1995-05-31 1999-02-16 Rscecat, Usa, Inc. Apparatus for electrochemical treatment of water and/or water solutions
JPH08326124A (en) 1995-05-31 1996-12-10 Isao Ueno Toilet with washer
US5628888A (en) 1996-03-28 1997-05-13 Rscecat, Usa, Inc. Apparatus for electrochemical treatment of water and/or water solutions
US5792090A (en) * 1995-06-15 1998-08-11 Ladin; Daniel Oxygen generating wound dressing
JP3193295B2 (en) * 1995-07-07 2001-07-30 株式会社日本トリム Dialysis machine
JPH0925236A (en) 1995-07-12 1997-01-28 Koichiro Kameyama Alkaline electrolytic water containing zinc
JP3153473B2 (en) * 1995-08-17 2001-04-09 株式会社カイゲン Disinfection device for medical equipment
FR2740335B1 (en) 1995-10-26 1997-12-19 Oreal USE OF LANTHANIDE, LITHIUM, TIN, ZINC, MANGANESE OR YTTRIUM SALT AS A SUBSTANCE P ANTAGONIST
JPH09157173A (en) * 1995-11-02 1997-06-17 Asahi Glass Eng Kk Acidic water containing hydroxy free radical
JP3312837B2 (en) * 1995-11-30 2002-08-12 アルプス電気株式会社 Ion water production apparatus and production method, electrolytic ionic water production system and production method
US5928491A (en) * 1996-01-23 1999-07-27 Maruko & Co., Ltd. Plant compatible electrolyte composition as well as electrolyte ionized water production device and methods
US5858202A (en) * 1996-01-30 1999-01-12 Zenkoku-Mokko-Kikai-Kan, Inc. Method for producing electrolytic water and apparatus for producing the same
JP3481761B2 (en) * 1996-01-30 2003-12-22 忠正 中村 Method of generating electrolyzed water
US5783052A (en) * 1996-03-11 1998-07-21 Rscecat, Usa, Inc. Electrochemical cell
US5635040A (en) 1996-03-11 1997-06-03 Rscecat, Usa, Inc. Electrochemical cell
JP3785219B2 (en) * 1996-03-27 2006-06-14 ペルメレック電極株式会社 Method for producing acidic water and alkaline water
US5985110A (en) 1996-03-28 1999-11-16 Bakhir; Vitold M. Apparatus for electrochemical treatment of water and/or water solutions
JP3716042B2 (en) 1996-04-24 2005-11-16 ペルメレック電極株式会社 Acid water production method and electrolytic cell
WO1997040814A1 (en) 1996-04-30 1997-11-06 The Procter & Gamble Company Cleaning articles treated with a high internal phase inverse emulsion
US5662625A (en) 1996-05-06 1997-09-02 Gwr Medical, L.L.P. Pressure controllable hyperbaric device
JPH11169856A (en) 1996-06-04 1999-06-29 Mizu Kk Electrolytic water producing device
EP0912447A1 (en) 1996-06-26 1999-05-06 IST Instant Surface Technology S.A. Method and device for the activation of liquids
US20040171998A1 (en) 2000-07-21 2004-09-02 Marasco Patrick V. Isolated wound-treatment arrangement
US6028018A (en) 1996-07-24 2000-02-22 Kimberly-Clark Worldwide, Inc. Wet wipes with improved softness
JPH1043764A (en) 1996-08-06 1998-02-17 First Ocean Kk Electrode for electrolizing water and sterilizing method of water using the same
US5728274A (en) * 1996-08-13 1998-03-17 Hoshizaki Denki Kabushiki Kaisha Production system of electrolyzed water
JP3408394B2 (en) 1996-08-27 2003-05-19 株式会社日本トリム Method for producing electrolytic hydrogen dissolved water and apparatus for producing the same
JPH1080686A (en) 1996-09-09 1998-03-31 Takazono Sangyo Kk Strong acidic water generating device
GB2316090B (en) * 1996-09-26 1998-12-23 Julian Bryson Method and apparatus for producing a sterilising solution
US6007696A (en) * 1996-09-28 1999-12-28 Kabushiki Kaisha Toshiba Apparatus and method for manufacturing electrolytic ionic water and washing method using electroyltic ionic water
WO1998017588A1 (en) 1996-10-18 1998-04-30 Miz Co., Ltd. Reducing electrolytic water and method for preparing the same
GB2316091B (en) * 1996-10-23 1999-06-16 Julian Bryson Electrolytic treatment of aqueous salt solutions
US5728287A (en) * 1996-10-31 1998-03-17 H2 O Technologies, Ltd. Method and apparatus for generating oxygenated water
JPH10128331A (en) 1996-11-05 1998-05-19 Coherent Technol:Kk Method and apparatus for producing sterilized water
JPH1157720A (en) 1996-11-07 1999-03-02 Honda Motor Co Ltd Electrolytic functional water, its production method and device
JP3455035B2 (en) * 1996-11-14 2003-10-06 株式会社東芝 Electrolytic ionic water generation device and semiconductor manufacturing device
US5908707A (en) 1996-12-05 1999-06-01 The Procter & Gamble Company Cleaning articles comprising a high internal phase inverse emulsion and a carrier with controlled absorbency
RU2119802C1 (en) 1996-12-18 1998-10-10 Стерилокс Текнолоджиз, Инк. Device for performing electrochemical treatment of liquid media
JP3366986B2 (en) 1996-12-27 2003-01-14 高橋金属株式会社 Washing water
US5919129A (en) 1997-01-09 1999-07-06 The United States Of America As Represented By The Secretary Of The Army Fiber optic periodontal endoscope
US5941859A (en) 1997-03-17 1999-08-24 Lerman; Benjamin S. Wound irrigation shield with fluid scavenging
RU2110483C1 (en) 1997-03-24 1998-05-10 Стерилокс Текнолоджиз, Инк. Electrochemical water treatment apparatus
US5963435A (en) * 1997-03-25 1999-10-05 Gianna Sweeney Apparatus for coating metal with oxide
US5759478A (en) * 1997-03-27 1998-06-02 Viskase Corporation Mandrel structure for use in manufacture of cellulose food casing
JPH10286571A (en) 1997-04-16 1998-10-27 Permelec Electrode Ltd Electrolytic cell for acidic water and alkaline water preparation
JPH10314740A (en) 1997-05-19 1998-12-02 Permelec Electrode Ltd Electrolytic bath for acidic water production
US5888528A (en) 1997-05-19 1999-03-30 Bernard Technologies, Inc. Sustained release biocidal powders
RO117540B1 (en) 1997-06-13 2002-04-30 Scchimcomplex Sa Borzesti Disinfectant and detergent composition
US6093292A (en) 1997-06-17 2000-07-25 Shimadzu Corporation Electrolyte producing apparatus with monitoring device
TW502130B (en) 1997-06-17 2002-09-11 Toshiba Corp Cleaning method of substrate
RU2132821C1 (en) 1997-06-25 1999-07-10 Стерилокс Текнолоджиз, Инк. Electrolytic water treatment device
US5964089A (en) 1997-06-27 1999-10-12 Lynntech, Inc Diagnostics and control of an on board hydrogen generation and delivery system
WO1999000588A2 (en) 1997-06-27 1999-01-07 Lynntech, Inc. Method and apparatus for injecting hydrogen into a catalytic converter
WO1999002157A1 (en) * 1997-07-09 1999-01-21 Toray Industries, Inc. Antitussives
US6143163A (en) 1997-08-06 2000-11-07 Permelec Electrode Ltd. Method of water electrolysis
US5932171A (en) * 1997-08-13 1999-08-03 Steris Corporation Sterilization apparatus utilizing catholyte and anolyte solutions produced by electrolysis of water
US5944978A (en) 1997-08-21 1999-08-31 Omco Co., Ltd. Cleaning method of an electrolyzed water forming apparatus and an electrolyzed water forming apparatus having mechanism for conducting the method
US5928488A (en) * 1997-08-26 1999-07-27 David S. Newman Electrolytic sodium sulfate salt splitter comprising a polymeric ion conductor
CA2270199C (en) 1997-08-27 2005-08-09 Miz Co., Ltd. Electrolytic cell and electrolyzed water generating device
US6197814B1 (en) 1997-10-10 2001-03-06 Nvid International, Inc. Disinfectant and method of making
US6294073B1 (en) 1997-10-22 2001-09-25 Chemicoat & Co., Ltd. Manufacturing method and apparatus of alkaline ionized water
US20040131695A1 (en) 1997-10-23 2004-07-08 Radical Waters Ip (Pty) Ltd. Use of an aqueous solution in the treatment of live animals
CN1136153C (en) 1997-10-23 2004-01-28 星崎电机株式会社 Electrolyzed water production apparatus
JPH10113664A (en) 1997-11-13 1998-05-06 Aiken Kogyo Kk Sterilized water having high electron activity
JP3952228B2 (en) 1997-11-19 2007-08-01 有限会社コヒーレントテクノロジー Electrolysis apparatus and electrolysis method
DE69821973T2 (en) 1997-12-04 2004-11-11 Steris Corp., Mentor CHEMICAL CHANGE OF ELECTROCHEMICALLY ACTIVATED WATER
JPH11172482A (en) 1997-12-10 1999-06-29 Shinko Plant Kensetsu Kk Ozonized water producing device and production of ozonized water with the device
DE69833038T2 (en) * 1997-12-26 2006-08-03 Morinaga Milk Industry Co. Ltd. METHOD FOR STERILIZING AND PACKAGING ARTICLES
US6171551B1 (en) * 1998-02-06 2001-01-09 Steris Corporation Electrolytic synthesis of peracetic acid and other oxidants
JPH11226092A (en) 1998-02-12 1999-08-24 Trp:Kk Sterile washing method for medical implement and apparatus therefor
JPH11239790A (en) * 1998-02-25 1999-09-07 Yoshiya Okazaki Electrolytic water-making apparatus and washing method therefor
US6200434B1 (en) * 1998-02-27 2001-03-13 Amano Corporation Apparatus for producing electrolytic water
US5948220A (en) * 1998-02-27 1999-09-07 Hoshizaki Denki Kabushiki Kaisha Production system of electrolyzed water
JP3896210B2 (en) * 1998-03-06 2007-03-22 ホシザキ電機株式会社 Electrolyzed water generator
JP3443310B2 (en) 1998-03-09 2003-09-02 ホシザキ電機株式会社 Electrolyzed water generator
JPH11269686A (en) 1998-03-18 1999-10-05 Permelec Electrode Ltd Production of hydrogen peroxide and electrolytic cell for production of hydrogen peroxide
US20020025921A1 (en) 1999-07-26 2002-02-28 Petito George D. Composition and method for growing, protecting, and healing tissues and cells
US20030089618A1 (en) 1998-04-10 2003-05-15 Miz Co., Ltd. Reducing electrolyzed water and method for producing same
CN1231994A (en) 1998-04-13 1999-10-20 王守林 Domestic ozone water and oxygen-enriched water producer
JP3783150B2 (en) 1998-04-24 2006-06-07 株式会社オメガ Ice having sterilizing power and method for producing the same
US6126810A (en) 1998-04-27 2000-10-03 Steris Corporation Generation of active chlorine in the presence of an organic load from sodium chloride in water
JPH11320259A (en) 1998-05-15 1999-11-24 Michiko Suzuki Electric discharge machining method and device for performing the same
JP2000042556A (en) * 1998-05-28 2000-02-15 Shimadzu Corp Electrolytic water production device
RU2142917C1 (en) 1998-06-30 1999-12-20 Попов Алексей Юрьевич Method and device for electrochemical treatment of water
US6368592B1 (en) 1998-07-17 2002-04-09 Massachusetts Institute Of Technology Method of delivering oxygen to cells by electrolyzing water
US6458109B1 (en) 1998-08-07 2002-10-01 Hill-Rom Services, Inc. Wound treatment apparatus
JP2000051858A (en) 1998-08-10 2000-02-22 Osamu Miyake Electrolytic ionic water maker
US6033539A (en) * 1998-08-21 2000-03-07 Gablenko; Viacheslav G. Units for electro-chemical synthesis of water solution
JP2000070171A (en) * 1998-08-26 2000-03-07 Trp:Kk Wet wiper for disinfection and its feeding device
JP2000084559A (en) 1998-09-09 2000-03-28 Hideo Hayakawa Water treatment
US6342150B1 (en) 1998-09-09 2002-01-29 Thomas Clay Sale Redox water treatment system
TW523547B (en) 1998-10-05 2003-03-11 Miz Co Ltd Method of producing detergent and the apparatus thereof
US6663306B2 (en) 1998-11-09 2003-12-16 The Procter & Gamble Company Cleaning composition, pad, wipe, implement, and system and method of use thereof
US6280594B1 (en) 1998-12-01 2001-08-28 Tateki Yamaoka Device for producing ion water and partition wall for device for producing ion water
WO2000033757A1 (en) 1998-12-09 2000-06-15 Advanced H¿2?O Inc. A system for decontamination of dental unit waterlines using electrolyzed water
JP2000189972A (en) 1998-12-28 2000-07-11 Nippon Intek Kk Oxidation-reduction potential water, production device of oxidation-reduction potential water, and additive
DE10011829C2 (en) 1999-03-15 2002-08-08 Nhk Spring Co Ltd Actuator for a vehicle occupant restraint system
JP4583530B2 (en) 1999-03-19 2010-11-17 オルガノ株式会社 Heat exchange water and its supply device
US6258225B1 (en) 1999-03-23 2001-07-10 Tateki Yamaoka Device for producing ion water
US6277266B1 (en) 1999-03-23 2001-08-21 Tateki Yamaoka Device for producing ion water and partition wall for device for producing ion water
US6436445B1 (en) 1999-03-26 2002-08-20 Ecolab Inc. Antimicrobial and antiviral compositions containing an oxidizing species
US7011940B1 (en) * 1999-04-14 2006-03-14 Medical Discovery Partners Llc Quality control for cytochemical assays
US20020006961A1 (en) 1999-05-14 2002-01-17 Katz Stanley E. Method and composition for treating mammalian nasal and sinus diseases caused by inflammatory response
CA2377232A1 (en) 1999-06-11 2000-12-21 Henceforth Hibernia, Inc. Prophylactic, therapeutic and industrial antioxidant compositions enhanced with stabilized atomic hydrogen/free electrons and methods to prepare and use such compositions
US6462250B1 (en) 1999-06-22 2002-10-08 Canon Kabushiki Kaisha Method for decomposing halogenated aliphatic hydrocarbon compounds having adsorption process and apparatus for decomposition having adsorption means
EP1064845B1 (en) 1999-06-30 2003-09-10 Kao Corporation Virucidal and sporicidal composition
DE60040317D1 (en) 1999-06-30 2008-11-06 Kao Corp Germicidal detergent composition
SE9902627D0 (en) 1999-07-08 1999-07-08 Siemens Elema Ab Medical nebulizer
GB9918458D0 (en) 1999-08-06 1999-10-06 Sterilox Med Europ Ltd Method and apparatus for the electrochemical processing of aqueous salt solutions
DE60036582T2 (en) 1999-08-06 2008-06-26 Puricore International Ltd. Electrochemical treatment of an aqueous solution
GB2355190B (en) 1999-08-23 2004-07-28 Sterilox Medical Improvements in or relating to sterilising preparations
TW546257B (en) 1999-09-01 2003-08-11 Nihon Trim Co Ltd Method and apparatus for producing electrolytic reduced water
JP2001079548A (en) 1999-09-16 2001-03-27 Kaneko Agricult Mach Co Ltd Ionized water and, oxidation and reduction water production device
US6852205B1 (en) 1999-09-27 2005-02-08 Shinko-Pantec Co., Ltd. Water-electrolysis-device-use electrode plate, unit, solid electrolytic membrane unit and electrolytic cell
US6592907B2 (en) 1999-10-04 2003-07-15 Hampar L. Karagoezian Synergistic antimicrobial ophthalmic and dermatologic preparations containing chlorite and hydrogen peroxide
JP2001113276A (en) 1999-10-14 2001-04-24 Kaneko Agricult Mach Co Ltd Ion water and oxidation-reduction water making apparatus
US6333054B1 (en) 1999-10-21 2001-12-25 Amuchina S.P.A. Topical, non-cytotoxic, antimicrobial hydrogel with thixotropic properties
JP2001139477A (en) 1999-11-17 2001-05-22 Coherent Technology:Kk Tissue cell growth-promoting liquid for wounded part
US6340663B1 (en) * 1999-11-24 2002-01-22 The Clorox Company Cleaning wipes
US6793846B2 (en) 1999-12-10 2004-09-21 Kao Corporation Microbicide compositions
US6426066B1 (en) 2000-01-12 2002-07-30 California Pacific Labs, Inc. Use of physiologically balanced, ionized, acidic solution in wound healing
US20030185704A1 (en) 2000-01-12 2003-10-02 Suzanne Bernard Physiologically balanced, ionized, acidic solution and methodology for use in wound healing
US7393522B2 (en) 2000-01-12 2008-07-01 Novabay Pharmaceuticals, Inc. Physiologically balanced, ionized, acidic solution and methodology for use in wound healing
JP4462513B2 (en) 2000-01-12 2010-05-12 有限会社コヒーレントテクノロジー Electrolyzed water manufacturing method, cleaning water, and cleaning method
AU4600201A (en) 2000-02-04 2001-08-14 Jacobus Theodorus Marais Dental equipment and method of operating such equipment
JP2004129314A (en) * 2000-03-17 2004-04-22 Soichi Sato Cogeneration system equipped with capacitor device
JP2001259640A (en) 2000-03-23 2001-09-25 Asahi Pretec Corp Device and method for manufacturing and supplying electrolyzed water having bactericidal property
JP4090665B2 (en) 2000-04-11 2008-05-28 ファースト・オーシャン株式会社 Electrolyzed water production method
IT1317329B1 (en) * 2000-04-13 2003-06-16 Nottington Holding Bv BREATHABLE FOOTWEAR.
JP3888835B2 (en) 2000-05-10 2007-03-07 花王株式会社 Disinfection and deodorization method and disinfection and deodorization tool
JP3602773B2 (en) 2000-06-08 2004-12-15 株式会社ミクニ Anode electrolyzed water and method for producing the same
EP1162179B1 (en) 2000-06-10 2004-09-15 Degussa AG Photocatalytic process
WO2001096527A2 (en) 2000-06-14 2001-12-20 Chanda Zaveri Peptides with physiological activity
US20020023847A1 (en) * 2000-06-23 2002-02-28 Shinichi Natsume Cleansing system and method using water electrolysis
CN1449295A (en) 2000-07-07 2003-10-15 水基Ip股份有限公司 Method of and equipment for washing, disinfecting and/or sterilizing health care devices
US20020168418A1 (en) 2000-08-04 2002-11-14 H20 Technologies, Ltd. Method and apparatus for treating water for use in improving the intestinal flora of livestock and poultry
DE10038457B4 (en) 2000-08-07 2008-09-25 Cognis Ip Management Gmbh Process for the production of sterols and tocopherols
JP4607296B2 (en) 2000-08-08 2011-01-05 ジャパンマテックス株式会社 Reduced water production equipment
US6531158B1 (en) 2000-08-09 2003-03-11 Impax Laboratories, Inc. Drug delivery system for enhanced bioavailability of hydrophobic active ingredients
US20030019764A1 (en) 2000-08-11 2003-01-30 H20 Technologies, Ltd. Under the counter water treatment system
US6358395B1 (en) * 2000-08-11 2002-03-19 H20 Technologies Ltd. Under the counter water treatment system
WO2003103522A1 (en) 2002-06-10 2003-12-18 Map Technologies Llc Methods and devices for electrosurgical electrolysis
JP3299250B2 (en) 2000-08-21 2002-07-08 修生 澄田 Pure water electrolysis tank with an intermediate chamber
KR100389917B1 (en) * 2000-09-06 2003-07-04 삼성전자주식회사 Wet process for fabrication of semiconductor device using electrolytically ionized anode water and/or cathod water and electrolytically ionized anode water and/or cathode water used therein
JP2002079248A (en) * 2000-09-06 2002-03-19 Tominaga Oil Pump Mfg Co Ltd Electrolytic water making apparatus
US6638364B2 (en) 2000-09-08 2003-10-28 Electric Aquagenics Unlimited System to clean and disinfect carpets, fabrics, and hard surfaces using electrolyzed alkaline water produced from a solution of NaCl
US20020032141A1 (en) * 2000-09-08 2002-03-14 Gene Harkins System and method to clean and disinfect hard surfaces using electrolyzed acidic water produced from a solution of NaCl
JP2004510312A (en) 2000-09-28 2004-04-02 プロトン エネルギー システムズ,インク. Regenerative electrochemical cell system and operating method thereof
US20020090428A1 (en) * 2000-10-12 2002-07-11 Warf C. Cayce Treatment fluid application apparatus for foodstuffs and methods related thereto
JP2002145787A (en) * 2000-11-02 2002-05-22 Kanebo Ltd External preparation
PT1342462E (en) * 2000-11-22 2007-06-29 Otsuka Pharma Co Ltd O/w emulsion composition and method of preparing the same
JP2002165868A (en) * 2000-12-05 2002-06-11 Kanebo Ltd Aerosol product
JP3805621B2 (en) 2000-12-19 2006-08-02 株式会社富永製作所 Electrolyzed water generator
US7122018B2 (en) 2000-12-26 2006-10-17 Sensormedics Corporation Device and method for treatment of wounds with nitric oxide
US6432077B1 (en) 2000-12-26 2002-08-13 Sensormedics Corporation Device and method for treatment of surface infections with nitric oxide
JP2002201296A (en) 2000-12-28 2002-07-19 Yunippu:Kk Method for modifying surface of plastic base material
US6540733B2 (en) * 2000-12-29 2003-04-01 Corazon Technologies, Inc. Proton generating catheters and methods for their use in enhancing fluid flow through a vascular site occupied by a calcified vascular occlusion
WO2002079392A2 (en) 2001-01-16 2002-10-10 Tomco2 Equipment Company Pathogen management system _______________________________
US6620315B2 (en) 2001-02-09 2003-09-16 United States Filter Corporation System for optimized control of multiple oxidizer feedstreams
US6844026B2 (en) 2001-02-12 2005-01-18 Rhodia Chimie Preparation of particles by hydrolysis of a metal cation in the presence of a polymer
US7011739B2 (en) 2001-03-22 2006-03-14 Gene Harkins Method for sanitizing shells of eggs using electrolyzed oxidizing water
US6921743B2 (en) 2001-04-02 2005-07-26 The Procter & Gamble Company Automatic dishwashing compositions containing a halogen dioxide salt and methods for use with electrochemical cells and/or electrolytic devices
US6866756B2 (en) * 2002-10-22 2005-03-15 Dennis Klein Hydrogen generator for uses in a vehicle fuel system
US20040172985A1 (en) 2001-04-05 2004-09-09 Haruo Mamiya Electric washing machine
US6767342B1 (en) 2001-04-23 2004-07-27 Evelyna D. Cantwell Oxygen bandage system
GB2374819B (en) 2001-04-27 2004-12-01 Sterilox Tech Int Ltd Oxidation-resistant coatings
JP2002338498A (en) 2001-05-17 2002-11-27 Takeda Chem Ind Ltd Internal liquid medicine
JP2002363017A (en) 2001-06-01 2002-12-18 Tadashi Inoue Disinfection liquid
TWI320713B (en) 2001-06-01 2010-02-21 Neochemir Inc
JP3338435B2 (en) 2001-06-04 2002-10-28 修生 澄田 Method for producing liquid in which hydrogen ion or hydroxide ion and redox substance coexist by electrolysis of pure water
CN1252318C (en) * 2001-06-21 2006-04-19 三洋电机株式会社 ELectrolyzing electrode and prodn. method therefor and electrolysis method using electrolyzing, electrode and electrolysis solution producing device
CA2452682A1 (en) 2001-06-29 2003-01-09 Miz Co., Ltd. Method for antioxidation and antioxidative functional water
KR20030005777A (en) * 2001-07-10 2003-01-23 삼성전자 주식회사 Semiconductor cleaning process using electrolytic ionized water and diluted HF solution at the same time
JP2003093479A (en) * 2001-07-18 2003-04-02 Sanyo Electric Co Ltd Sterilizing method and electrolyzed water producing device
US6656334B2 (en) 2001-07-23 2003-12-02 Skydon Corp. Modified electrolysis cell and a housing for the same
WO2003010094A1 (en) 2001-07-26 2003-02-06 H20 Technologies, Ltd. Apparatus and methods for cleaning and controlling bacteria growth in fluid supply lines
JP5140218B2 (en) 2001-09-14 2013-02-06 有限会社コヒーレントテクノロジー Electrolyzer for producing charged anode water suitable for surface cleaning and surface treatment, method for producing the same, and method of use
EP1443973B1 (en) * 2001-09-15 2008-07-09 ICF Technologies, Inc. Kits and methods for determining the effectiveness of sterilization or disinfection processes
US6833206B2 (en) 2001-09-28 2004-12-21 Daimlerchrysler Ag Auxiliary power supply for a vehicle with a combustion engine and method for operating same
US20040171701A1 (en) 2001-11-02 2004-09-02 Technology Convergence Inc. Methanol production process
US7323118B2 (en) * 2001-10-01 2008-01-29 Aquilabs S.A. Composition of hypochlorous acid and its applications
US6756142B2 (en) 2001-10-31 2004-06-29 Motorola, Inc. Fuel cell using non-linear positive temperature coefficient material
JP2003144001A (en) 2001-11-08 2003-05-20 Hoshizaki Electric Co Ltd Raising water for marine organism, method and apparatus for producing raising water
US6833207B2 (en) 2001-11-09 2004-12-21 Hydrogenics Corporation Unitized regenerative fuel cell with bifunctional fuel cell humidifier and water electrolyzer
WO2003042111A2 (en) 2001-11-13 2003-05-22 Radical Waters (Ip) (Pty) Limited An electrochemical activation system suitable for producing electrochemically-activated solutions through use of an electrolytic cell exchange module
JP2003145153A (en) 2001-11-13 2003-05-20 Sugano Minoru Method and apparatus for producing electrolyzed water
DE60128815T2 (en) 2001-11-23 2008-02-07 Hoshizaki Denki K.K., Toyoake A method of cleaning a device for producing electrolyzed water
US6624135B2 (en) 2001-11-26 2003-09-23 S.C. Johnson & Son, Inc. Cleaning sheet
MXPA03007923A (en) 2001-12-05 2005-02-17 Micromed Lab Inc Method and apparatus for producing negative and positive oxidative reductive potential (orp) water.
US20030175220A1 (en) 2001-12-11 2003-09-18 Wang Xiao Bing Effect of electrolyzed solutions on acidogenesis of plaque
TW562673B (en) 2001-12-14 2003-11-21 Sanyo Electric Co Foot warming bath device
US20050178920A1 (en) 2002-01-22 2005-08-18 Wilson Thomas R. Spacecraft propulsion system
CA2446563A1 (en) 2002-01-29 2003-08-07 Mitsubishi Corporation High-pressure hydrogen producing apparatus and producing method
JP3848882B2 (en) * 2002-02-13 2006-11-22 ニッポン高度紙工業株式会社 High ion conductive solid electrolyte and electrochemical system using the solid electrolyte
JP2003236315A (en) 2002-02-14 2003-08-26 Daiki Kogyo Kk Cleaning apparatus for rotation type drum screen
JP2003236543A (en) 2002-02-14 2003-08-26 Hideo Hayakawa Alternating current electrolysis method and apparatus for liquid
JP2003247134A (en) 2002-02-21 2003-09-05 Toray Textile Inc Slitted film yarn for magnetic recording
EP1579037A4 (en) 2002-03-06 2008-02-13 Univ Georgia Res Found Method and apparatus for electrolyzing water
US6733435B2 (en) 2002-03-08 2004-05-11 Canedo Luis Electromagnetic method of treatment of lesions associated with inadequate blood perfusion, partial denervation, tissue loss, pain, edema, inflammation and infection
JP2003299250A (en) 2002-03-29 2003-10-17 Hitachi Ltd Power supply unit and elevator apparatus
JP2003311270A (en) 2002-04-18 2003-11-05 First Ocean Kk Liquid distributor
WO2003095102A1 (en) 2002-05-08 2003-11-20 Mikuni Corporation Electrolyzed water spraying device
JP2003338435A (en) 2002-05-21 2003-11-28 Meidensha Corp Electric double layered capacitor
KR100479627B1 (en) 2002-05-25 2005-04-06 유니셈 주식회사 Wet pre-treatment apparatus and method for treating an effluent gas
US6899903B2 (en) * 2002-06-25 2005-05-31 Patrick Quillin Composition for cleansing the sinuses
JP2004047827A (en) * 2002-07-12 2004-02-12 Mec Kk Method for manufacturing printed circuit board
JP3954458B2 (en) 2002-07-16 2007-08-08 株式会社コスモス.エンタープライズ Water purification pot and water purification method using the same
JP2004058006A (en) 2002-07-31 2004-02-26 First Ocean Kk Method of manufacturing electrolytic water
US20040081705A1 (en) * 2002-09-18 2004-04-29 Mana Gotou Digestion promoter for ruminant animal and breeding method of ruminant animal
US20040055896A1 (en) * 2002-09-20 2004-03-25 Sterilox Technologies, Inc. Biocidal solution
US6843448B2 (en) 2002-09-30 2005-01-18 Daniel W. Parmley Lighter-than-air twin hull hybrid airship
JP2004121607A (en) 2002-10-03 2004-04-22 Harasawa Pharmaceutical Co Ltd Bactericidal and deodorant agent composition
DE10250926A1 (en) 2002-10-31 2004-05-19 Microlas Lasersystem Gmbh Laser system which combines beams of two or more lasers, has beams directed onto beam splitter and mirror, so that reflected radiation passes through beam splitter and back to resonator
US6855233B2 (en) 2002-11-15 2005-02-15 Kinji Sawada Apparatus for production of strong alkali and acid electrolytic solution
JP2004173628A (en) 2002-11-28 2004-06-24 Fuso Pharmaceutical Industries Ltd Probe for detecting haemophilus influenzae and detection method using the same
JP3849644B2 (en) 2003-01-17 2006-11-22 松下電工株式会社 Electrolyzed water generator
JP3891119B2 (en) 2003-01-17 2007-03-14 松下電工株式会社 Electrolyzed water generator
JP3891120B2 (en) 2003-01-17 2007-03-14 松下電工株式会社 Electrolyzed water generator
JP3891118B2 (en) 2003-01-17 2007-03-14 松下電工株式会社 Electrolyzed water generator
JP4244645B2 (en) 2003-01-31 2009-03-25 パナソニック電工株式会社 Electrolyzed water generator
CN1751139B (en) 2003-02-21 2010-12-08 阿维伦斯有限责任公司 Electrolyzer apparatus and method for hydrogen production
US20040168909A1 (en) 2003-02-28 2004-09-02 Larson Arden L. Three-dimensional flow-through electrode and electrochemical cell
WO2004079051A1 (en) 2003-03-04 2004-09-16 FRS WATERWATER, INC. (d.b.a. WATERWARE, INC.) High electric field electrolysis cell
WO2004078654A2 (en) 2003-03-04 2004-09-16 Frs Waterware, Inc. (D.B.A. Waterware, Inc.) Free radical solution water
JP2004267956A (en) * 2003-03-11 2004-09-30 Mikuni Corp Method for producing mixed electrolytic water
JP2004273961A (en) 2003-03-12 2004-09-30 Ebara Corp Cleaning device of metal wiring forming substrate
JP4597123B2 (en) 2003-03-13 2010-12-15 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Purification method of bacterial cytolysin
US7923030B2 (en) 2003-03-14 2011-04-12 Sol-Gel Technologies, Inc. Agent-encapsulating micro- and nanoparticles, methods for preparation of same and products containing same
US7014932B2 (en) 2003-03-19 2006-03-21 Proton Energy Systems, Inc. Drainage system and process for operating a regenerative electrochemical cell system
AU2003901316A0 (en) 2003-03-21 2003-04-03 The Royal Alexandra Hospital For Children Regulation of cell surface proteins
JP4041422B2 (en) 2003-03-26 2008-01-30 ニッポン高度紙工業株式会社 Solid electrolyte and electrochemical system using the solid electrolyte
GB2400611B (en) 2003-04-15 2006-03-15 Empower Corp H An integrated renewable energy system
JP4040028B2 (en) 2003-04-16 2008-01-30 三洋電機株式会社 Method and system for treating water to be treated containing organic matter and nitrogen compound
US8912174B2 (en) * 2003-04-16 2014-12-16 Mylan Pharmaceuticals Inc. Formulations and methods for treating rhinosinusitis
JP2007502861A (en) 2003-05-01 2007-02-15 レプリダイン・インコーポレーテッド Antibacterial methods and compositions
US20040226894A1 (en) 2003-05-12 2004-11-18 Tatsuo Okazaki Method of preparing a sterile water containing hypochlorous or chlorous acid, package of sterile source materials, and sterile water preparation kit
MXPA05012437A (en) 2003-05-19 2006-01-30 Procter & Gamble Compositions, devices and methods for stabilizing and increasing the efficacy of halogen dioxide solutions.
US20040244537A1 (en) 2003-06-05 2004-12-09 Runyon Dale Leroy Processing coal combustion products (CCP), ore, or other material using functionally electrolyzed water (FEW)
US6856916B2 (en) 2003-06-13 2005-02-15 Wen-Shing Shyu Locating system of oxidation/reduction potential of electrolysis water and the constant output method of calibration and compensation thereof
US7453164B2 (en) 2003-06-16 2008-11-18 Polestar, Ltd. Wind power system
US7445800B2 (en) 2003-06-25 2008-11-04 Eau Technologies, Inc. Method for remediating mold and mildew using acidic electrolyzed water
JP2005013520A (en) 2003-06-26 2005-01-20 Spring:Kk Bathtub apparatus and method of operating bathtub
KR100412258B1 (en) 2003-07-01 2003-12-31 주식회사 에스에프에이 The process for cleaning a glass surface in manufacturing lcd
ITPD20030166A1 (en) 2003-07-22 2005-01-23 Geox Spa BREATHABLE AND WATERPROOF SOLE FOR FOOTWEAR, PARTICULARLY BUT NOT ONLY FOR OPEN-SHOE FOOTWEAR SUCH AS SANDALS, SABO 'AND SIMILAR AND SHOE MADE WITH SUCH SOLE
JP2005058848A (en) * 2003-08-08 2005-03-10 Spring:Kk Production method for water used for washing, disinfecting, and wound healing, its production apparatus, and water used for washing, disinfecting, and wound healing
SG149043A1 (en) 2003-08-18 2009-01-29 Novacal Pharmaceuticals Inc N,n-dihalogenated amino acids and derivatives
US20050064259A1 (en) 2003-09-24 2005-03-24 Protonetics International, Inc. Hydrogen diffusion electrode for protonic ceramic fuel cell
US20050067300A1 (en) * 2003-09-25 2005-03-31 The Procter & Gamble Company Electrolysis device for treating a reservoir of water
DE10349158A1 (en) 2003-10-22 2005-06-16 Airbus Deutschland Gmbh Device for water supply in aircraft
JP4653945B2 (en) 2003-10-24 2011-03-16 ミズ株式会社 Pharmacologically functional water and its use
GB0518826D0 (en) 2005-09-15 2005-10-26 Smith & Nephew Apparatus with actives from tissue - exudialysis
JP4353772B2 (en) 2003-11-07 2009-10-28 三洋電機株式会社 Electrolyzed water generator
US20050101838A1 (en) 2003-11-12 2005-05-12 Camillocci Philip L. Endoscope cover
US7592300B2 (en) 2003-11-24 2009-09-22 The Dial Corporation Antimicrobial compositions containing an aromatic carboxylic acid and a hydric solvent
GB0328124D0 (en) 2003-12-04 2004-01-07 Daly James Membrane electrolyser with a two part end design
KR100542895B1 (en) 2003-12-22 2006-01-11 재단법인 포항산업과학연구원 Method for controlling ballast water using effect of NaOCl produced electrolysis of natural seawater and an apparatus for the same
US20050196462A1 (en) 2003-12-30 2005-09-08 Oculus Innovative Sciences, Inc. Topical formulation containing oxidative reductive potential water solution and method for using same
US9168318B2 (en) * 2003-12-30 2015-10-27 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and methods of using the same
WO2005065383A2 (en) * 2003-12-30 2005-07-21 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution, processes for producing same and methods of using the same
US20050139808A1 (en) * 2003-12-30 2005-06-30 Oculus Innovative Sciences, Inc. Oxidative reductive potential water solution and process for producing same
US7374655B2 (en) 2004-01-12 2008-05-20 Novastron Electrochemical water purifier
JP4866741B2 (en) 2004-02-03 2012-02-01 アクゾ ノーベル コーティングス インターナショナル ビー ヴィ Antifouling composition comprising a polymer having a salt group
WO2005077056A2 (en) 2004-02-10 2005-08-25 Integrated Botanical Technologies Methods and compositions for the treatment of inflammation
US20050178349A1 (en) 2004-02-17 2005-08-18 Tse Kwong W. Multi-fueled internal combustion engine
US7510640B2 (en) 2004-02-18 2009-03-31 General Motors Corporation Method and apparatus for hydrogen generation
ATE399524T1 (en) 2004-02-19 2008-07-15 Reckitt & Colman Overseas SKIN CARE PRODUCTS THAT CONTAIN SALICYLIC ACID
FI20040279A (en) 2004-02-23 2005-08-24 Pohjoisen Kantaperuna Oy Method and apparatus for washing and disinfecting root vegetables
US7238272B2 (en) 2004-02-27 2007-07-03 Yoichi Sano Production of electrolytic water
US7000395B2 (en) 2004-03-11 2006-02-21 Yuan Ze University Hybrid clean-energy power-supply framework
US20050209518A1 (en) 2004-03-17 2005-09-22 Therafuse, Inc. Self-calibrating body analyte monitoring system
KR20070011394A (en) 2004-03-23 2007-01-24 더 클로록스 캄파니 Method for diluting hypochlorite
US7157412B2 (en) 2004-04-07 2007-01-02 Advanced Medical Optics, Inc. Alkylamine as an antimicrobial agent in ophthalmic compositions
US7758807B2 (en) 2004-04-20 2010-07-20 The Clorox Company Microbial control with reduced chlorine
WO2005117914A2 (en) 2004-06-04 2005-12-15 Sterilox Technologies, Inc. Biocidal solution
US20060008908A1 (en) 2004-07-08 2006-01-12 Giles Brian C Method and composition for longevity assurance
EP1650329A3 (en) 2004-10-21 2007-11-07 Trust Sterile Services Limited Apparatus and method for electrolytic cleaning
DE102004056456A1 (en) 2004-11-23 2006-05-24 Eugen Malamutmann Treatment device for wounds
US7749370B2 (en) * 2005-02-03 2010-07-06 Osao Sumita Manufacturing method of oxidative water to be employed for sterilization
CA2597092A1 (en) 2005-02-04 2006-08-10 Coley Pharmaceutical Group, Inc. Aqueous gel formulations containing immune reponse modifiers
CN101163491B (en) 2005-03-23 2017-04-19 奥古露丝创新科学公司 Method of treating skin ulcers using oxidative reductive potential water solution
JP5816406B2 (en) * 2005-03-23 2015-11-18 オキュラス イノヴェイティヴ サイエンシズ、インコーポレイテッド Method for treating skin ulcer using redox potential aqueous solution
US20060241002A1 (en) 2005-04-26 2006-10-26 Rogozinski Wallace J Sodium hypochlorite gel composition
CA2606734C (en) * 2005-05-02 2016-06-21 Oculus Innovative Sciences, Inc. Method of using oxidative reductive potential water solution in dental applications
US20060263240A1 (en) 2005-05-06 2006-11-23 Electric Aquagenics Unlimited Electrolyzed water treatment for face and hands
US20060275387A1 (en) 2005-06-03 2006-12-07 BAGLEY David Processed water and therapeutic uses thereof
EP1933941A2 (en) 2005-08-25 2008-06-25 Philip R. Houle Treatment systems for delivery of sensitizer solutions
DK2010245T3 (en) 2005-11-21 2016-01-18 Joshua David Smith WOUND CARE SYSTEM
WO2007085018A2 (en) 2006-01-20 2007-07-26 Oculus Innovative Sciences, Inc. Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
US7779625B2 (en) 2006-05-11 2010-08-24 Kalypto Medical, Inc. Device and method for wound therapy
US20080243096A1 (en) 2006-10-05 2008-10-02 Paul Svedman Device For Active Treatment and Regeneration of Tissues Such as Wounds
US8273747B2 (en) 2006-11-02 2012-09-25 Curis, Inc. Small organic molecule regulators of cell proliferation
WO2008089268A2 (en) 2007-01-16 2008-07-24 Puricore, Inc. Methods and compositions for treating conditions associated with infection and/or inflammation
JP4598130B2 (en) 2007-02-16 2010-12-15 大幸薬品株式会社 Wide range antiviral pharmaceutical composition with excellent storage stability
CA2680483C (en) 2007-03-13 2017-12-12 Oculus Innovative Sciences, Inc. Antimicrobial solutions containing dichlorine monoxide and methods of making and using the same
US20090068255A1 (en) 2007-04-30 2009-03-12 Betty Yu Use of matrix metalloproteinase inhibitors in skin care
US20100106079A1 (en) * 2008-06-03 2010-04-29 Oculus Innovative Sciences, Inc. Method and apparatus for treating a wound
US10617715B2 (en) * 2008-12-22 2020-04-14 Sonoma Pharmaceuticals, Inc. Methods of treating or preventing biofilm associated infections with free available chlorine free available chlorine water
KR101781229B1 (en) 2009-06-15 2017-09-22 오클루스 이노바티브 사이언시즈 인코포레이티드 Solution containing hypochlorous acid and methods of using same
MX2012001415A (en) 2009-07-30 2012-06-19 Oculus Innovative Sciences Inc Hydrogel formulation comprising oxidative reductive potential water.
US9918477B2 (en) 2013-05-22 2018-03-20 Sonoma Pharmaceuticals, Inc. Stabilized hypochlorous acid solution and use thereof

Also Published As

Publication number Publication date
WO2007085018A3 (en) 2007-11-29
EP1993572A2 (en) 2008-11-26
CA2637197C (en) 2017-01-31
CA2637197A1 (en) 2007-07-26
KR20080093132A (en) 2008-10-20
US9072726B2 (en) 2015-07-07
EP1993570A2 (en) 2008-11-26
AU2007205861A1 (en) 2007-07-26
US20150306137A1 (en) 2015-10-29
US20100092399A1 (en) 2010-04-15
AU2007205863B2 (en) 2013-06-20
AU2007205863A1 (en) 2007-07-26
CA2637178C (en) 2018-09-04
US8834445B2 (en) 2014-09-16
US20070196357A1 (en) 2007-08-23
WO2007085018A2 (en) 2007-07-26
BRPI0706677A2 (en) 2011-04-05
US20180028562A1 (en) 2018-02-01
EP1993570B1 (en) 2018-07-25
JP2009523829A (en) 2009-06-25
US8147444B2 (en) 2012-04-03
BRPI0706671A2 (en) 2011-04-12
US9782434B2 (en) 2017-10-10
JP2009523832A (en) 2009-06-25
BRPI0706676B1 (en) 2020-03-10
EP1993571B1 (en) 2018-07-25
BRPI0706676A2 (en) 2011-04-05
WO2007085021A2 (en) 2007-07-26
AU2007205860A1 (en) 2007-07-26
HK1130675A1 (en) 2010-01-08
US20070196434A1 (en) 2007-08-23
AU2007205861B2 (en) 2013-05-09
WO2007085019A3 (en) 2007-11-29
US20120251631A1 (en) 2012-10-04
US20070173755A1 (en) 2007-07-26
JP5723084B2 (en) 2015-05-27
EP1993571A2 (en) 2008-11-26
JP5449780B2 (en) 2014-03-19
KR20080093135A (en) 2008-10-20
KR101499824B1 (en) 2015-03-06
KR20080093134A (en) 2008-10-20
WO2007085021A3 (en) 2007-11-01
KR101499822B1 (en) 2015-03-06
AU2007205860B2 (en) 2013-01-31
CA2637175A1 (en) 2007-07-26
CA2637178A1 (en) 2007-07-26
JP2009523830A (en) 2009-06-25
BRPI0706676B8 (en) 2021-05-25
WO2007085019A2 (en) 2007-07-26

Similar Documents

Publication Publication Date Title
US9782434B2 (en) Methods of treating or preventing inflammation and hypersensitivity with oxidative reductive potential water solution
US10016455B2 (en) Method of preventing or treating influenza with oxidative reductive potential water solution
CN101405012B (en) Use the method for oxidative reductive potential water solution treatment or prevention of inflammation and allergy

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20210831

MKLA Lapsed

Effective date: 20200122