Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TOPICAL DELIVERY OF BUFFERING AGENTS FOR PREVENTION AND TREATMENT OF VIRAL INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2021/163648
Kind Code:
A2
Abstract:
Provided herein are formulations for topical and/or transdermal administration, and methods of using these formulations for increasing resistance to viral infections and improving immune system activity. Also provided are formulations for topical and/or transdermal administration, and methods of using these formulations for modulating the pH (e.g., raising) of a tissue or microenvironment for the prevention of and treatment of a viral infection and improving the immune response by activating immune cells (e.g., neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells).

Inventors:
BEAL RYAN (US)
Application Number:
PCT/US2021/018064
Publication Date:
August 19, 2021
Filing Date:
February 13, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DYVE BIOSCIENCES INC (US)
Attorney, Agent or Firm:
WEINSTEIN, Peter D. et al. (US)
Download PDF:
Claims:
CLAIMS

1. A method of preventing or treating a viral infection, comprising administering a transdermal formulation containing a buffering or alkalinizing agent to a patient suffering from a viral infection or at risk of a viral infection.

2. The method of claim 1, wherein the transdermal dermal formulation is administered to a patient topically.

3. The method of claim 1, wherein, the transdermal formulation is effective in decreasing the severity, duration and/or extent of viral infection.

4. The method of claim 1, wherein the transdermal formulation raises the pH locally or systemically in one or more areas of the patient.

5. The method of claim 1, wherein the transdermal formulation reduces a viral infection rate by inhibiting viral transmission.

6. The method of claim 1, wherein the transdermal formulation increases or enhances activity of one or more of neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells.

7. The method of claim 1, wherein the buffering or alkalinizing agent is sodium bicarbonate.

8. The method of claim 7, wherein the sodium bicarbonate is at a concentration from about 30% to 35% w/w of the transdermal formulation.

9. The method of claim 7, wherein the sodium bicarbonate is at a concentration of 33% w/w of the transdermal formulation.

10. The method of claim 1, wherein the buffering or alkalinizing agent comprises a carbonate salt, lysine, tris, a phosphate buffer and/or 2-imidazole- 1-yl-3- ethoxycarbonylpropionic acid (IEPA), or a combination thereof.

11. The method of claim 1 , wherein the buffering or alkalinizing agent constitutes between 10% - 56% w/w of the transdermal formulation.

12. The method of claim 1, wherein the buffering or alkalinizing agent penetrates the skin of the patient.

13. The method of claim 1, wherein the transdermal formulation comprises a buffering or alkalinizing agent in an amount between about 10% - 45 % w/w; a penetrant portion in an amount between about 5% - 55% w/w; a detergent portion in an amount between about 1% to 15% w/w; and water in an amount between about 15% to 65% w/w.

14. The method of claim 1, wherein the virus is selected from the group consisting of Adeno-Associated Virus, Adenovirus, Arena virus (Lassa virus), Alpha virus, Astrovirus, Bacille Calmette-Guerin ‘BCG’, BK virus (including associated with kidney transplant patients), Papovavirus, Bunyavirus, Burkett’s Lymphoma (Herpes), Calicivirus, California, encephalitis (Bunyavirus), Colorado tick fever (Reovirus), Corona virus, Coronavirus, Coxsackie, Coxsackie virus A, B (Enterovirus), Crimea-Congo hemorrhagic fever (Bunyavirus), Cytomegalovirus, Cytomegaly, Dengue (Flavivirus), Diptheria (bacteria), Ebola, Ebola/Marburg hemorrhagic fever (Filoviruses), Epstein-Barr Virus ‘EBV’, Echovirus, Enterovirus, Eastern equine encephalitis ΈEE’, Togaviruses, Encephalitis, Enterovirus, Flavi virus, Hantavirus, Bunyavirus, Hepatitis A., (Enterovirus), Hepatitis B virus (Hepadnavirus), Hepatitis C (Flavivirus), Hepatitis E (Calicivirus), Herpes, Herpes Varicella-Zoster virus, HIV Human Immunodeficiency Virus (Retrovirus), HIV-AIDS (Retrovirus), Human Papilloma Virus ‘HPV’, Cervical cancer (Papovavirus), HSV 1 Herpes Simplex I, HSV 2 Herpes Simplex II, HTLV-T-cell leukemia (Retrovirus), Influenza (Orthomyxovirus), Japanese encephalitis (Flavivirus), Kaposi’s Sarcoma associated herpes virus KSHV (Herpes HHV 8), Kyusaki, Lassa Virus, Lentivirus, Lymphocytic Choriomeningitis Virus LCMV (Arenavirus), Measles (Rubella), Measels, Measles Micro (Paramyxovirus), Monkey Bites (Herpes strain HHV 7), Mononucleosis (Herpes), Morbilli, Mumps (Paramyxovirus), Newcastle’s diseases virus, Norovirus, Norwalk virus (Calicivirus), Orthomyxoviruses (Influenza virus A, B, C), Papillomavirus (warts), Papova (M.S.), Papovavirus (JC-progressive multifocal leukoencephalopathy in HIV) (Papovavirus), Parainfluenza Nonsegmented (Paramyxovirus), Paramyxovirus, Parvovirus (B19 virusaplastic crises in sickle cell disease), Picoma virus, Pertussus (bacteria), Polio (Enterovirus), Poxvirus (Smallpox), Prions, Rabies (Rhabdovirus), Reovirus, Retrovirus, Rhabdovirus (Rabies), Rhinovirus, Roseola (Herpes HHV 6), Rotavirus, Respiratory SyncitialVirus (Paramyxovirus), Rubella (Togaviruses), Bunyavirus, Flavivirus, Poxvirus, Vaccinia virus, Variola, Venezuelan Equine Encephalitis ‘VEE’ (Togaviruses), Wart virus (Papillomavirus), Western Equine Encephalitis “WEE’ (Togaviruses), West Nile Virus (Flavivirus), and Yellow fever (Flavivirus).

15. The method of claim 1, wherein the transdermal formulation also includes one or more other therapeutic agents.

16. The method of claim 15, wherein the one or more other therapeutic agents are an antiviral drugs or protease inhibitors.

17. A method of increasing resistance to viral infections and/or improving immune system activity in a patient, the method comprising administering topically and/or transdermally an effective amount of a formulation for transdermal delivery comprising one or more buffering or alkalinizing agent to a patient in need thereof, where the administration is effective to i) decrease viral infection rates by inhibiting viral transmission; ii) improve the immune response by activating immune cells (e.g. neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells); and/or iii) decrease the severity, duration and/or extent of viral infection.

18. A transdermal delivery formulation comprising the following components: a. a buffering or alkalinizing agent at a concentration from 0.5% to 5%; b. isopropyl palmitate at a concentration from 5% to 20%; c. benzyl alcohol at a concentration of 0.5% to 5%; d. stearic acid at a concentration from 0.5% to 5%; e. safflower oil at 1% to 6%; f. oleic acid at 0.5% to 2%; and g. deionized water at 20% to 80%;

19. The transdermal delivery formulation of claim 18, wherein the transdermal formulation also includes: a. Aveeno® moisturizers, cream, oils, lotions; b. Jergens® moisturizers, cream, oils, lotions; c. Honest Company® moisturizers, cream, oils, lotions; d. Dermologica® moisturizers, cream, oils, lotions; or e. St. Ives™ moisturizers, cream, oils, lotions.

20. The transdermal delivery formulation of claim 18, wherein the transdermal formulation further comprises Phospholipon 90G at a concentration of 1% to 20%.

21. The transdermal delivery formulation of claim 18, wherein the transdermal formulation further comprises Durosoft PK-SG at a concentration of 0.5% to 5%.

22. The transdermal delivery formulation of claim 18, wherein the transdermal formulation further comprises Pluronic Gel at a concentration of 5% to 40%.

23. The transdermal delivery formulation of claim 18, wherein the transdermal formulation also includes a surfactant.

24. The transdermal delivery formulation of claim 18, wherein the transdermal formulation also includes a nonionic detergent.

25. The transdermal delivery formulation of claim 18, wherein the transdermal formulation also includes a polar gelling agent.

26. The transdermal delivery formulation of claim 24, wherein the nonionic detergent results in a more viscous and cream-like formulation.

27. The transdermal delivery formulation of claim 25, wherein the polar gelling agent results in a more viscous and gel-like formulation

28. The transdermal delivery formulation of claim 18, wherein the buffering or alkalinizing agent concentration is from 0.05% to 0.1%, from 0.1% to 0.5%, from 0.5% to 2%, from 0.5% to 1.5%, from 1% to 1.5%, from 0.5% to 1.5%, from 1% to 2.5%, from 1% to 3%, from 1.5% to 3%, from 1% to 4% or from 1% to 5%.

29. The transdermal delivery formulation of claim 18, wherein the isopropyl palmitate is from 1% to 15%, from 2.5% to 15%, from 4% to 15%, from 5% to 10%, from 10% to 15%, from 12% to 15%, from 5% to 8%, from 5% to 15% or from 10% to 20%.

30. The transdermal delivery formulation of claim 18, wherein the benzyl alcohol is from 0.5% to 1.5%, 0.5% to 4%, from 0.75% to 3%, from 1% to 2.5%, from 2% to 4% or from 2.5% to 5%.

31. The transdermal delivery formulation of claim 18, wherein the stearic acid is from 0.5% to 1.5%, from 1.5% to 2.5%, from 3.5% to 5%, from 2% to 5%, from 3% to 5% or from 4% to 5%.

32. The transdermal delivery formulation of claim 18, wherein the safflower oil is at a concentration from 1% to 3%, from 1.5% to 2.5%, from 3% to 5% from 4 to 6%, from 4.5% to 6% or from 5% to 6%.

33. The transdermal delivery formulation of claim 32, wherein the safflower oil is a linoleic acid.

34. The transdermal delivery formulation of claim 18, wherein the oleic acid is at a concentration from 0.5% to 1%, from 0.5% to 1.5%, from 1% to 1.5% or from 1% to 2%.

35. The transdermal delivery formulation of claim 18, wherein the deionized water is from 20% to 50%, from 25% to 75%, from 30% to 60%, from 40% to 60%, from 40% to 50%, or from 50% to 80%.

36. A method of administration of a buffering or alkalinizing agent to an individual using the transdermal delivery formulation of claim 18.

37. The transdermal delivery formulation of claim 18, wherein the formulation includes one or more anti -viral agents or protease inhibitors.

Description:
Topical Delivery of Buffering Agents For Prevention and Treatment of Viral Infections

FIELD OF INVENTION

[0001] This invention relates generally to formulations and methods to increase resistance to viral infections and improving immune system activity, and more specifically, to formulations and methods for transdermal or buffers or alkalinizing agents.

BACKGROUND

[0002] Acidosis exerts significant immunomodulatory effects. Both the innate and adaptive arms of the immune response appear to be finely regulated by extracellular acidosis in the range of pH values found at inflammatory sites and tumors. Innate immune cells (e.g., neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells) have all been shown to have functions that are downregulated by extracellular acidosis though some responses can also upregulated depending on the cell type involved. In contrast, it is clear that the adaptive T-cell response and corresponding T-cell mediated immunity is strongly suppressed at low pH. It is believed that low pH inhibits both T-cell activation and differentiation. Importantly, it has also been shown that this T-cell suppression is a reversible phenomenon if pH is normalized.

[0003] Further, most viruses (whether enveloped or not) use an endocytic entry mechanisms that require a drop in pH a trigger for host penetration. Some bacterial toxins also rely on low pH as a cue. Further, T-cell activation is inhibited at acidic pH weakening the innate immune response to a pathogen.

[0004] The first member of the coronavirus family was discovered in the 1930s but it has not been until recently that they gained notoriety. The severe acute respiratory syndrome (SARS) outbreak in 2002-2003 gained world- wide attention and the 2019-2020 novel coronavirus, COVID-19 has become a global health priority. Coronaviruses infect a wide variety of mammals and birds, causing respiratory and enteric diseases and, in some rarer cases, hepatitis and neurologic disease. Infection can be acute or persistent.

[0005] For COVID-19 there is currently no treatment. However, it has been shown in animal models that combatting the extracellular acidity associated with the acute immune response can thwart viral infection and enable a more robust immune response. Buffer or alkalinization therapy has also been shown to synergistically improve the efficacy of a number of immunotherapies.

[0006] Unfortunately there has not been a convenient nor effective way to deliver these alkalinizing therapies to patients and others who might benefit. Oral therapy is ineffective due to the acidic gastrointestinal tract and GI adverse events that prevent therapeutically meaningful doses to be delivered. Injectable approaches are similarly ineffective or impractical due to drug half-life limitations and or injection-related side effects and practical limitations.

[0007] The idea of using buffers to modulate the pH of the tissues presents problems related to administration. Oral and conventional administration of buffers has serious limitations in practice - specifically intolerance and side effects including diarrhea, gastric intolerance, nausea, vomiting and abdominal discomfort. Therapeutically effective amounts of pharmaceutical formulations comprising pH modulating buffers and the like cannot be administered, delivered, and tolerated orally. Intravenous i.e., systemic administration has also been discredited.

[0008] One approach that has demonstrated significant promise is topically delivered buffering therapies as described in U.S. Patent Application No. 16/456,256 and 16/456,260. This approach avoids the bioavailability and adherence challenges observed in orally and parenterally delivered buffer therapy.

[0009] New treatments, formulations and methods of administration of buffers or alkalinizing agents (e.g., sodium bicarbonate and others) are needed to overcome the current deficiencies. Another area of potential unmet need includes formulations of one or more buffering or alkalinizing agents and methods of use in combination with other agents or treatments such as anti-viral agents, immunotherapeutics, and/or other bioactive agents or Biologies such as antibody-based therapies or therapeutics. The inventions described herein address these unmet needs.

SUMMARY

[0010] The inventions described and claimed herein have many attributes and embodiments including, but not limited to, those set forth or described or referenced in herein. The inventions described and claimed herein are not limited to, or by, the features or embodiments identified in this Summary, which is included for purposes of illustration only and not restriction.

[0011] Studies have demonstrated that multiple disease states create an acidic extracellular environment due to increased glycolysis in situations of biologic stress. These acidic extracellular microenvironments lead to increased susceptibility to viral infection that then exacerbates the biologic stress and associated acidosis. This acidosis further leaves the biologic system susceptible to additional infection due to acid-induced viral to cell membrane fusion, and in a position of suboptimal adaptive immune response due to T-cell inhibition at low pH.

[0012] Accordingly, in one aspect a method of increasing resistance to viral infections and improving immune system activity in a patient is provided. In some embodiments the method comprises administering topically and/or transdermally an effective amount of a formulation for transdermal delivery comprising one or more buffering agent to a patient in need thereof, where the administration is effective to i) decrease viral infection rates by inhibiting viral transmission; ii) improve the immune response by activating immune cells (e.g. neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells); and/or iii) decrease the severity, duration and/or extent of viral infection.

[0013] In one aspect, a method of treating a viral infection in a patient is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to inhibit or prevent the viral activity and/or improve the patient’s immune response.

[0014] In one aspect, a method of preventing a viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to inhibit or prevent the virus from fusing with the patient and instigating infection.

[0015] In another aspect, a method of improving the immune response, decreasing the severity, duration or extent of viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof.

[0016] In another aspect, a method of treating a viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to increase or enhance immune system activity. In another aspect, the method includes administration of one or more anti-viral agents.

[0017] An exemplary embodiment of this aspect is a method of preventing or inhibiting a viral infection comprising i) selecting an antiviral agent (e.g. a biological agent, chemotherapeutic or immunotherapeutic agent), ii) formulating the therapeutic agent in a suitable formulation, iii) administering the formulation comprising the therapeutic agent, and iv) before, during or after step iii), administering a formulation comprising one or more buffering agent topically and/or transdermally in an amount effective to inhibit or prevent viral activity.

[0018] In another aspect, a method of treating a viral infection in a patient is provided where an effective amount of a formulation comprising one or more buffering agent is administering topically and/or transdermally to a patient in need thereof such that the administration is effective to alter the pH of a tissue or microenvironment proximal to an infected region in the patient.

[0019] In another aspect, a method of preventing viral activity is provided where an effective amount of a formulation comprising one or more buffering agent is administering topically and/or transdermally to a patient in need thereof such that the administration is effective to alter the pH of a tissue or microenvironment proximal to an area known to be infected by the virus (e.g., throat and sinus). The formulation can be administered, for example, to someone who is at risk of viral infection such as a health care worker or person in proximity to infected individuals.

[0020] In another aspect, methods of increasing the efficacy of conventional approaches to viral infection are provided such as co-administered with one or more antiviral agents (e.g. acyclovir, lamivudine or a protease inhibitor).

[0021] In another aspect, a method of treatment of a viral infection is provided comprising i) selecting a therapeutic agent (e.g. acyclovir, lamivudine or a protease inhibitor), ii) formulating the therapeutic agent in a suitable formulation, iii) administering the formulation comprising the therapeutic agent, and iv) before, during or after step iii), administering a formulation comprising one or more buffering agent topically and/or transdermally in an amount effective to inhibit or prevent viral activity. The therapeutic agent is typically formulated in a formulation suitable for a route of administration other than topical and/or transdermal, however in certain embodiments the therapeutic agent is formulated with the buffering agent in the same formulation.

[0022] For transdermal topical administration in particular for agents other than buffer, a suitable formulation typically involves a penetrant that enhances penetration of the skin and is, in some embodiments, composed of chemical permeation enhancers (CPEs). In some cases, it can also include peptides designed to penetrate cells i.e. cell penetrating peptides (CPPs) also known as skin penetrating peptides (SPPs). The formulation may be applied for example in the form of topical lotions, creams, and the like, as described herein.

[0023] If the active agent is a buffer, the choice of buffer system is based on the criteria of capability of buffering at a suitable pH typically between 7 and 10.5, as well as biocompatibility of the buffer system itself and the compatibility of the buffer system with the remaining components of the formulation. Conversely, the formulation is chosen to be compatible with the buffer selected; amounts of penetrants are generally less than those advantageous for therapeutic agents in general.

[0024] Some viruses may be resistant to lower doses, but treatable with higher doses. When the pH is adjusted for the purpose of inhibiting a virus, treatment can be followed by assessment of effectiveness. In a related aspect, particular types of viruses are evaluated for sensitivity/resistance to pH adjustment and capacity for treatment as a function of dosage and buffer formulation composition.

[0025] Other features and advantages of aspects of the present invention will become apparent from the following more detailed description, taken in conjunction with the accompanying drawings, which illustrate, by way of example, the principles of aspects of the invention.

Definitions

[0026] Reference in this specification to "one embodiment/aspect" or "an embodiment/aspect" means that a particular feature, structure, or characteristic described in connection with the embodiment/aspect is included in at least one embodiment/aspect of the disclosure. The use of the phrase "in one embodiment/aspect" or "in another embodiment/aspect" in various places in the specification are not necessarily all referring to the same embodiment/aspect, nor are separate or alternative embodiments/aspects mutually exclusive of other embodiments/aspects. Moreover, various features are described which may be exhibited by some embodiments/aspects and not by others. Similarly, various requirements are described which may be requirements for some embodiments/aspects but not other embodiments/aspects. Embodiment and aspect can in certain instances be used interchangeably.

[0027] The terms used in this specification generally have their ordinary meanings in the art, within the context of the disclosure, and in the specific context where each term is used. Certain terms that are used to describe the disclosure are discussed below, or elsewhere in the specification, to provide additional guidance to the practitioner regarding the description of the disclosure. It will be appreciated that the same thing can be said in more than one way.

[0028] Consequently, alternative language and synonyms may be used for any one or more of the terms discussed herein. Nor is any special significance to be placed upon whether or not a term is elaborated or discussed herein. Synonyms for certain terms are provided. A recital of one or more synonyms does not exclude the use of other synonyms. The use of examples anywhere in this specification including examples of any terms discussed herein is illustrative only and is not intended to further limit the scope and meaning of the disclosure or of any exemplified term. Likewise, the disclosure is not limited to various embodiments given in this specification.

[0029] Without intent to further limit the scope of the disclosure, examples of instruments, apparatus, methods and their related results according to the embodiments of the present disclosure are given below. Note that titles or subtitles may be used in the examples for convenience of a reader, which in no way should limit the scope of the disclosure. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure pertains. In the case of conflict, the present document, including definitions, will control.

[0030] As applicable, the terms "about" or "generally", as used herein in the specification and appended claims, and unless otherwise indicated, means a margin of +/- 20%. Also, as applicable, the term "substantially" as used herein in the specification and appended claims, unless otherwise indicated, means a margin of +/- 10%. It is to be appreciated that not all uses of the above terms are quantifiable such that the referenced ranges can be applied.

[0031] The term “active agent” or “active ingredient” refers to a substance, compound, or molecule, which is biologically active or otherwise, induces a biological or physiological effect on a subject to which it is administered to. In other words, “active agent” or “active ingredient” refers to a component or components of a composition to which the whole or part of the effect of the composition is attributed. A base or alkalinizing agent can be a primary active agent, or in other words, the component(s) of a composition to which the whole or part of the effect of the composition is attributed. An active agent can be a secondary agent, or in other words, the component(s) of a composition to which an additional part and/or other effect of the composition is attributed. [0032] The term “alkalinizing agent” refers to substances such as drugs used to manage disorders associated with low pH. For example, they can be used to treat acidosis due to kidney failure. Used for oral or parenteral therapy, sodium bicarbonate is the commonly preferred alkalinizing agent. Others include potassium citrate, calcium carbonate, sodium lactate and calcium acetate.

[0033] The term “antiviral drug” refers to one or more medicaments that inhibit a virus or viral activity. Such drugs are often nucleoside analogues which viruses incorporate into their genomes during replication. The life-cycle of the virus is then halted because the newly synthesized DNA is inactive. Examples of nucleoside analogues are aciclovir for Herpes simplex virus infections and lamivudine for HIV and hepatitis B virus infections. Other antiviral drugs in use target different stages of the viral life cycle (e.g., protease inhibitors).

[0034] The term “virus’” refers to a small infectious agent that replicates only inside the living cells of an organism. While not inside an infected cell or in the process of infecting a cell, viruses exist in the form of independent particles, or virions, that include (i) the genetic material (i.e. DNA or RNA that encode the structure of the proteins by which the virus acts); (ii) a protein coat, the capsid, which surrounds and protects the genetic material; and in some cases (iii) an outside envelope of lipids. The term “virus” may also encompass any chemical or biochemical component portion of a virus, including viral component preparations, a related particle (e.g. prion), or the like and it need not be infective or capable of self-replication.

[0035] The term “viral vector” refers to a viral genome that has been adapted into a plasmid- based technology and modified for safety through the removal of many essential genes and the separation of the viral components. The use of viral vectors is a means of gene transfer to modify a specific cell type or tissue and can be manipulated to express therapeutic genes.

[0036] Viral infectivity is defined as the number of virus particles capable to invade a host cell. This is determined by using susceptible cells to the specific virus by measuring the viral infectivity. The viral titer can be measured by such means as the TCID50 method or the plaque method, and the like, but these methods rely on the morphological change of the cell when it has been infected by the virus.

[0037] The term “vaccine” refers to a biological preparation that provides active acquired immunity to a particular disease. Some vaccines contain inactivated, but previously virulent, micro-organisms that have been destroyed with chemicals, heat, or radiation. Some vaccines contain live, attenuated microorganisms. Many of these are active viruses that have been cultivated under conditions that disable their virulent properties. Toxoid vaccines are made from inactivated toxic compounds that cause illness rather than the micro-organism. Other types of vaccines include protein subunit and conjugate vaccines.

[0038] All numerical designations, e.g. , pH, temperature, time, concentration, and molecular weight, including ranges, are to be understood as approximations in accordance with common practice in the art. When used herein, the term “about” may connote variation (+) or (-) 1%, 5% or 10% of the stated amount, as appropriate given the context. It is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.

[0039] Many known and useful compounds and the like can be found in Remington’s Pharmaceutical Sciences (13 th Ed), Mack Publishing Company, Easton, PA — a standard reference for various types of administration. As used herein, the term “formulation(s)” means a combination of at least one active ingredient with one or more other ingredient, also commonly referred to as excipients, which may be independently active or inactive. The term “formulation” may or may not refer to a pharmaceutically acceptable composition for administration to humans or animals and may include compositions that are useful intermediates for storage or research purposes.

[0040] In an embodiment, a “pharmaceutical composition” is intended to include the combination of a base with a carrier, inert or active, in a sterile composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo. In one aspect, the pharmaceutical composition is substantially free of endotoxins or is non-toxic to recipients at the dosage or concentration employed.

[0041] In an embodiment, “an effective amount” refers to the amount of the defined component sufficient to achieve the desired chemical composition or the desired biological and/or therapeutic result. In an embodiment, that result can be the desired pH or chemical or biological characteristic. In other embodiments, the desired result is the alleviation or amelioration of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. When the desired result is a therapeutic response, the effective amount will vary depending upon the specific disease or symptom to be treated or alleviated, the age, gender and weight of the subject to be treated, the dosing regimen of the formulation, the severity of the disease condition, the manner of administration and the like, all of which can be determined readily by one of skill in the art. A desired effect may, without necessarily being therapeutic, also be a cosmetic effect, in particular for treatment for disorders of the skin described herein.

[0042] In an embodiment, as used herein, the terms “treating,” “treatment” and the like are used herein to mean obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disorder or sign or symptom thereof, and/or may be therapeutic in terms of amelioration of the symptoms of the disease or infection, or a partial or complete cure for a disorder and/or adverse effect attributable to the disorder.

[0043] For purposes herein, a formulation, a formulation for transdermal delivery and a transdermal delivery formulation are each a formulation for transdermal delivery, including, the transdermal delivery of an active ingredient for the treatment of a syndrome and or a disease in an individual.

[0044] All numerical designations, e.g. , pH, temperature, time, concentration, and molecular weight, including ranges, are to be understood as approximations in accordance with common practice in the art. When used herein, the term “about” may connote variation ( + ) or ( - ) l%, 5% or 10% of the stated amount, as appropriate given the context. It is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.

[0045] The practices described herein employ, unless otherwise indicated, conventional techniques of tissue culture, immunology, molecular biology, microbiology, cell biology and recombinant DNA, which are within the skill of the art. See, e.g., Harlow and Lane eds. (1999) Antibodies, A Laboratory Manual and Herzenberg et al. eds (1996) Weir’s Handbook of Experimental Immunology.

DETAILED DESCRIPTION

[0046] Tissue acidosis (i.e., pH< 7.0) occurs over the course of the infectious processes. This acidosis has profound effects on how the infection takes hold and how the immune system responds and adapts. First, acidification is required for viruses to fuse with a host’s cell membrane (the initial infection). Second, T-cell activation and response to the infection is inhibited by acid environments. Further, patients in situations of biologic stress (e.g., the old, the ill, young, immunocompromised etc.) have been shown to have higher levels of extracellular acidosis (due to high glycolytic activity) leaving them more susceptible to infection and less able to combat infections once they occur. It has been shown in animal models that neutralizing extracellular acidosis using various buffering agents significantly decreases viral infection rates, and significantly increases t-cell activation and differentiation leading to a much more robust immune response. The animal model success has not translated into humans do to oral and parenteral drug delivery challenges. This invention allows transdermal delivery of these agents in humans.

[0047] Applicants have found that the drawbacks of intravenous and oral administration of buffers and other alkalinizing agents can be overcome by administering these agents topically and/or transdermally, but other types of administration are possible, including for example, intranasally or via transmembrane administration for example by suppository or intranasal application.

[0048] As noted above, one aspect of the invention is a method decrease viral infection rates by inhibiting viral transmission. Another aspect is improve the immune response by activating immune cells. For example, low pH can inhibits T Cell Responses. Studies demonstrate that extracellular acidosis can either stimulate or suppress innate immune responses depending on both the cell type involved and the particular response analyzed. The following cells can be activated to improve the immune response to decrease the severity and/or extent of viral infection:

Neutrophils play a critical role in host defense against bacterial and fungal infections and are also involved in the pathogenesis of a number of inflammatory conditions. Extracellular acidosis has been shown to induce either stimulatory or inhibitory effects on neutrophil responses, depending on the function analyzed.

Monocytes and Macrophages are also regulated by changes in the pH of the extracellular medium.

Natural Killer Cells or “NK” cells play an important role in the innate host defense against viruses and other intracellular pathogens as well as in antitumor immunity extracellular acidosis inhibits the antitumoral activity of NK cells.

Dendritic Cells conventional or myeloid dendritic cells (DCs) are highly specialized antigen- presenting cells with a unique ability to prime naive T cells inducing the activation of the adaptive immune response.

Platelets and Endothelial Cells are also sensitive to pH. Different groups have shown that low pH modulates the course of the innate immune response by acting not only on leukocytes but also on nonimmune cells. [0049] For non- systemic parenteral administration, such as intramuscular, intraperitoneal or subcutaneous administration standard formulations are sufficient. These formulations include standard excipients and other ancillary ingredients such as antioxidants, suitable salt concentrations and the like. Such formulations can be found, for example, in Remington’s Pharmaceutical Sciences (13 th Ed), Mack Publishing Company, Easton, PA — a standard reference for various types of administration. As used herein, the term “formulation(s)” means a combination of at least one active ingredient with one or more other ingredient, also commonly referred to as excipients, which may be independently active or inactive. The term “formulation” may or may not refer to a pharmaceutically acceptable composition for administration to humans or animals and may include compositions that are useful intermediates for storage or research purposes. In an embodiment, administration to humans or animals may include, without limitation, topical, sublingual, rectal, vaginal, transdermal, trancutaneous, oral, inhaled, intranasal, pulmonary, subcutaneous, pulmonary, intravenous, enteral or parenteral. Suitable topical formulations for transdermal administration of active agents for the methods provided herein are described in U.S.S.N. 14,757,703, to Sand B., et al., incorporated herein by reference in its entirety. Suitable penetrants are described, for example, in PCT publications WO/2016/105499 and W 0/2017/127834.

[0050] As the patients and subjects of the invention method are, in addition to humans, veterinary subjects, formulations suitable for these subjects are also appropriate. Such subjects include livestock and pets as well as sports animals such as horses, greyhounds, and the like.

[0051] In an embodiment, a “pharmaceutical composition” is intended to include, without limitation, the combination of an active agent with a carrier, inert or active, in a sterile composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo. In one aspect, the pharmaceutical composition is substantially free of endotoxins or is non-toxic to recipients at the dosage or concentration employed.

[0052] In an embodiment, “an effective amount” refers, without limitation, to the amount of the defined component sufficient to achieve the desired chemical composition or the desired biological and/or therapeutic result. In an embodiment, that result can be the desired pH or chemical or biological characteristic, e.g., stability of the formulation. In other embodiments, the desired result is the alleviation or amelioration of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. When the desired result is a therapeutic response, the effective amount will, without limitation, vary depending upon the specific disease or symptom to be treated or alleviated, the age, gender and weight of the subject to be treated, the dosing regimen of the formulation, the severity of the disease condition, the manner of administration and the like, all of which can be determined readily by one of skill in the art. A desired effected may, without necessarily being therapeutic, also be a cosmetic effect, in particular for treatment for disorders of the skin described herein.

[0053] In an embodiment, a “subject” of diagnosis or treatment is, without limitation, a prokaryotic or a eukaryotic cell, a tissue culture, a tissue or an animal, e.g., a mammal, including a human. Non-human animals subj ect to diagnosis or treatment include, for example, without limitation, a simian, a murine, a canine, a leporid, such as a rabbit, livestock, sport animals, and pets.

[0054] In an embodiment, as used herein, the terms “treating,” “treatment” and the like are used herein, without limitation, to mean obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disorder or sign or symptom thereof, and/or may be therapeutic in terms of amelioration of the symptoms of the disease or infection, or a partial or complete cure for a disorder and/or adverse effect attributable to the disorder.

Methods

[0055] Methods for treating, preventing or ameliorating a disease, disorder, a condition, or a symptom thereof or a condition related thereto are provided herein using formulations for transdermal delivery described herein below. The methods provided herein may comprise or consist of topically administering one or more of the formulations described herein to skin of a subject in need thereof. Preferred, but non-limiting embodiments are directed to methods for treating, preventing, inhibiting or ameliorating a disease, disorder, a condition, or a symptom described below.

Viruses

[0056] Many embodiments provided herein are directed to various methods of increasing resistance to viral infections and improving immune system activity. An exemplary embodiment of a method of treating a viral infection in a patient according to the invention comprises administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, wherein said administration is effective to increase resistance to viral infections and/or improve immune system activity. [0057] Accordingly, the method can comprise administering topically and/or transdermally an effective amount of a formulation for transdermal delivery comprising one or more buffering agent to a patient, where the administration is effective to i) decrease viral infection rates by inhibiting viral transmission; ii) improve the immune response by activating immune cells (e.g. neutrophils, monocytes and macrophages, natural killer cells, dendritic cells, and platelets and endothelial cells); and/or iii) decrease the severity, duration and/or extent of viral infection.

[0058] In one aspect, a method of treating a viral infection in a patient is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to inhibit or prevent the viral activity and/or improve the patient’s immune response.

[0059] In one aspect, a method of preventing a viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to inhibit or prevent the virus from fusing with the patient and instigating infection.

[0060] In another aspect, a method of improving the immune response, decreasing the severity, duration or extent of viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof.

[0061] In another aspect, a method of treating a viral infection is provided comprising administering topically and/or transdermally an effective amount of a formulation comprising one or more buffering agent to a patient in need thereof, where the administration is effective to increase or enhance immune system activity.

[0062] The methods can be used against viruses known in the art, including Adeno- Associated Virus, Adenovirus, Arena virus (Lassa virus), Alpha virus, Astrovirus, Bacille Calmette-Guerin ‘BCG’, BK virus (including associated with kidney transplant patients), Papovavirus, Bunyavirus, Burkett’s Lymphoma (Herpes), Calicivirus, California, encephalitis (Bunyavirus), Colorado tick fever (Reovirus), Corona virus, Coronavirus (including COVID- 19), Coxsackie, Coxsackie virus A, B (Enterovirus), Crimea-Congo hemorrhagic fever (Bunyavirus), Cytomegalovirus, Cytomegaly, Dengue (Flavivirus), Diptheria (bacteria), Ebola, Ebola/Marburg hemorrhagic fever (Filoviruses), Epstein-Barr Virus ‘EBV’, Echovirus, Enterovirus, Eastern equine encephalitis ΈEE’, Togaviruses, Encephalitis, Enterovirus, Flavi virus, Hantavirus, Hepatitis A, (Enterovirus), Hepatitis B virus (Hepadnavirus), Hepatitis C (Flavivirus), Hepatitis E (Calicivirus), Herpes, Herpes Varicella-Zoster virus, HIV Human Immunodeficiency Virus (Retrovirus), HIV — AIDS (Retrovirus), Human Papilloma Virus ‘HPV’, Cervical cancer (Papovavirus), HSV 1 Herpes Simplex I, HSV 2 Herpes Simplex II, HTLV — T-cell leukemia (Retrovirus), Influenza (Orthomyxovirus), Japanese encephalitis (Flavivirus), Kaposi’s Sarcoma associated herpes virus KSHV (Herpes HHV 8), Kyusaki, Lassa Virus, Lentivirus, Lymphocytic Choriomeningitis Virus LCV (Arenavirus), Measles (Rubella), Measels, Measles Micro (Paramyxovirus), Monkey Bites (Herpes strain HHV 7), Mononucleosis (Herpes), Morbilli, Mumps (Paramyxovirus), Newcastle’s diseases virus, Norovirus, Norwalk virus (Calicivirus), Orthomyxoviruses (Influenza virus A, B, C), Papillomavirus (warts), Papova (M.S.), Papovavirus (JC — progressive multifocal leukoencephalopathy in HIV) (Papovavirus), Parainfluenza Nonsegmented (Paramyxovirus), Paramyxovirus, Parvovirus (B19 virusaplastic crises in sickle cell disease), Picoma virus, Pertussus (bacteria), Polio (Enterovirus), Poxvirus (Smallpox), Prions, Rabies (Rhabdovirus), Reovirus, Retrovirus, Rhabdovirus (Rabies), Rhinovirus, Roseola (Herpes HHV 6), Rotavirus, Respiratory SyncitialVirus (Paramyxovirus), Rubella (Togaviruses), Flavivirus, Poxvirus, Vaccinia virus, Variola, Venezuelan Equine Encephalitis ‘VEE’ (Togaviruses), Wart virus (Papillomavirus), Western Equine Encephalitis “WEE’ (Togaviruses), West Nile Virus (Flavivirus), Yellow fever (Flavivirus) and Zika virus (ZIKV).

Coadministration with anti-viral agents

[0063] In another aspect, the method includes administration of one or more anti -viral agents (e.g. acyclovir, lamivudine or a protease inhibitor). The methods can increase the efficacy of conventional approaches.

[0064] An exemplary embodiment of this aspect is a method of preventing or inhibiting a viral infection comprising i) selecting an antiviral agent (e.g. a biological agent, chemotherapeutic or immunotherapeutic agent), ii) formulating the therapeutic agent in a suitable formulation, iii) administering the formulation comprising the therapeutic agent, and iv) before, during or after step iii), administering a formulation comprising one or more buffering agent topically and/or transdermally in an amount effective to inhibit or prevent viral activity.

[0065] In another aspect, a method of preventing viral activity is provided where an effective amount of a formulation comprising one or more buffering agent is administering topically and/or transdermally to a patient in need thereof such that the administration is effective to alter the pH of a tissue or microenvironment proximal to an area known to be infected by the virus (e.g., throat and sinus).

[0066] For transdermal topical administration in particular for agents other than buffer, a suitable formulation typically involves a penetrant that enhances penetration of the skin and is, in some embodiments, composed of chemical permeation enhancers (CPEs). In some cases, it can also include peptides designed to penetrate cells i.e., cell penetrating peptides (CPPs) also known as skin penetrating peptides (SPPs). The formulation may be applied for example in the form of topical lotions, creams, and the like, as described herein.

[0067] If the active agent is a buffer, the choice of buffer system is based on the criteria of capability of buffering at a suitable pH typically between 7 and 10.5, as well as biocompatibility of the buffer system itself and the compatibility of the buffer system with the remaining components of the formulation. Conversely, the formulation is chosen to be compatible with the buffer selected; amounts of penetrants are generally less than those advantageous for therapeutic agents in general.

[0068] Some viruses may be resistant to lower doses, but treatable with higher doses. When the pH is adjusted for the purpose of inhibiting a virus, treatment can be followed by assessment of effectiveness. In a related aspect, particular types of viruses are evaluated for sensitivity/resistance to pH adjustment and capacity for treatment as a function of dosage and buffer formulation composition.

[0069] In another aspect, formulations provided herein can be administered or co administered with diterpene compounds, including but not limited to paclitaxel, docetaxel, cabazitaxel, and the like.

[0070] In another aspect, formulations provided herein can be administered or co administered with compounds that inhibit topoisomerase II or compounds that otherwise interact with nucleic acids in cells. Such compounds include, for example, doxorubicin, epirubicin, etoposide, teniposide, mitoxantrone, and analogues thereof. In one example, this combination is used in treatment to reduce tumor cell contamination of peripheral blood progenitor cells (PBSC) in conjunction with high-dose chemotherapy and autologous stem cell support (HDC-ASCT). See U.S. Pat. No. 6,586,428 to Geroni et al.

[0071] Other diseases, conditions, and disorders described herein can be treated with formulations and methods provided herein. [0072] Other aspects include the parenterally or topically administered composition as a stand alone or in synergistic combination with traditional pharmacological agents. Topical administration is most conveniently transdermal, but further includes transmembrane administration, for example by suppository or intranasal application.

[0073] Other aspects include the topical administration of agents and drugs, with or without occlusion in any manner and which are not conjugated with or delivered by means of penetration enhancing formulations but are merely applied to the intact skin with or without massaging the skin for the purpose of breaching the skin’s permeation barrier.

[0074] The applicant surprisingly discovered that the combination of two hypothetical mechanisms, functioning in synergy, was successful in transdermal drug delivery (TDDD) of guest molecules of molecular weights exceeding 500 Da and, in fact, beyond 150 kDa. These two synergistic mechanisms involve different interactions between the SPPs and the cellular moiety in the "transcellular" mechanism and the CPEs in the "extracellular" mechanism.

[0075] In another aspect, the invention discloses and provides integrative and cooperative methods with compositions that are directed to the simultaneous and selective disruption of the cellular and lipid matrix contributions to the SC permeation barrier in conjunction with the transdermal delivery of agents. The mode of each physico-chemical component will be presented separately, although they may participate cooperatively in a chemical permeation enhancement (CPE) composition.

[0076] In another aspect a biochemical process, which is directed to the cellular component of the SC permeability barrier, is facilitated by a synergistic action of several biological processes, which combine to enhance transdermal drug delivery. In some embodiments, each of these processes are used individually.

[0077] Other embodiments include the use of TD-1, as well as the other cationic cyclo peptide variants identified as TDR-2, TDR-3 and TDR-7, in which arginine substitutions are made at N-4, N-5 and N-7, and TDK-2, TDK-3 and TDK-7, in which lysine substitutions are made at N-2, N-3 and N-7. Also embodied in this patent is cationic cyclo-peptide variant TD- 34 as bis-substitute peptide in N-5 and N-6. The cyclic structure and the disulfide constrained nature is critical for enhancement activity of the peptides. The TDS series of the same amino acid sequence of cyclic structure with TD-1 is further embodied as a modification via substitution of the N-terminal with three amino acids possessing the same cationic group with various side-chain lengths. The enhancement activity has been demonstrated to be proportional to side-chain length and identified as TDS-3 > TDS-2 > TDS-1.

[0078] While the exact mechanism is unclear, our studies have revealed the profound activity of cell penetrating peptides (CPPs) with special reference to TD-1, to be upon interactions with the skin cellular components. The CPPs function by permeating through the transcellular route passing through hydrophilic keratin-packed comeocytes that are embedded in multiple hydrophobic lipid bilayers. While partitioning into the keratin-rich comeocytes, they form bridges that bind with the filamentous keratin a-helices via hydrogen bonds in co administration as peptide-chaperones without interacting with the guest cargo or degrading the lipid matrix. SPPs, in fact, enhance the lipid organization while simultaneously increasing skin electrical conductivity. TD-1 is non-cytotoxic and non-irritating to skin.

[0079] It has been demonstrated that the CPPs also utilize the intercellular pathways via small gaps between the comeocytes by disrupting cell-to-cell junctional desmosomes expeditiously, thereby modifying the normal ultrastructural spacing from about 30 nm to about 466 nm in as little as 30 minutes from topical administration. Transmission electron microscopy has revealed that the intercellular gaps are a transient process that will escort macromolecules across the SC permeation barrier restoring the breaches in about one hour after application.

[0080] The co-administration of CPPs has been postulated to result in a statistically significant increase in percentage of a-helices of keratins, suggesting that CPPs stabilize these structural proteins (keratins). The intra-cellular keratins are stabilized by disulfide bonds, which are tightly packed either in a-chains (a-keratins) or in //-sheet ( /-keratins) structures. The high-degree of cross-linking by the disulfide bonds, hydrophobic interactions and hydrogen bonds between the keratin filament structures within the individual comeocytes confer its mechanical stability preventing free drug transport.

[0081] Keratolytic agents will disrupt the tertiary structure and hydrogen bonds between individual keratin filaments, thereby promoting penetration through intact skin. The administration of keratolytic agents will release keratin-bound active drug and enhance bioavailability.

[0082] One biochemical process is deployed to disrupt the disulfide linkage of the keratin filaments of which the comeocytes of the SC are comprised. This is contributed by means of a reducing agent containing a thiol moiety. Thiogly colic Acid (TGA) @ 5% concentration is the preferred embodiment. Other agents, such as Dithiothretol (DTT), B-Mercaptoethanol (B-ME) and Urea Hydrogen Peroxide @ 17.5% concentration might be similarly employed to act upon the hydrogen bonds, as well as the disulfide bonds.

[0083] An additional keratolytic agent or enzyme, such as Proteinase K might be employed to degrade the keratin substrate @ about 10 mg/mL The optimal pH of keratolytic activity is around pH 8, while activity is detected in a broad range of pH values between 6 to 11 for serine proteases. Chemical hydrolysis will further compromise the barrier property contributed by the comeocytes but the process is irreversible and concentration-dependent.

[0084] The simultaneous application of the reducing agent has been demonstrated to have no adverse effect on the keratolytic enzymes and, in fact, allows the preferential access of the enzymes to the substrate for enhanced proteolytic attack.

[0085] Sigma- Aldrich offers an appropriate keratinolytic product (K4519-500UN), which is a non-specific serine protease with the capability of degrading insoluble keratin substrates by cleaving non-terminal peptide bonds.

[0086] This patent further embodies an alternative to the reducing agent/keratolytic enzyme combination by means of two cooperating enzymes isolated from a keratin-degrading bacterium, Stenotrophomonas sp. strain D-l. These synergistic enzymes disrupt the disulfide bonds while simultaneously degrading the keratin substrate.

Formulations

[0087] A formulation for transdermal delivery may, for example, comprise two components or it may comprise one or more buffering agent and a penetrant. Typically, however, a penetrant is less than 85 %w/w. The formulation may have a detergent of at least 1 %w/w. For example, a suitable formulation may comprise about 10-56 %w/w buffering agent and a penetrant. In one aspect, disclosed herein is a formulation for transdermal delivery of one or more buffering agent through the skin of a subject, comprising: a buffering agent comprising a carbonate salt in an amount between about 10-56 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount of at least 1 %w/w; and wherein the formulation comprises water in an amount from none up to about 77 %w/w.

[0088] In another aspect, disclosed herein is a method for transdermal delivery of a carbonate salt of the formulation comprising: a buffering agent comprising a carbonate salt in an amount between about 10-45 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount between about 1 to 15 %w/w; and wherein the formulation comprises water in an amount between about 15 to 65 %w/w, through the skin of a subject, wherein the carbonate salt of the formulation is in an amount between about 15-32 %w/w of the formulation.

[0089] In yet another aspect, disclosed herein is a formulation for transdermal delivery of a therapeutic agent through the skin of a subject, wherein the formulation comprises at least one active agent in an amount effective for treatment of a condition in the subject and the formulation comprising: a buffering agent comprising a carbonate salt in an amount between about 10-45 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount between about 1 to 15 %w/w; wherein the formulation comprises water in an amount between about 15 to 65 %w/w, through the skin of a subject, wherein the carbonate salt of the formulation is in an amount between about 15-32 %w/w of the formulation, therapeutic, and wherein the alkalinity of the formulation enhances penetration of the therapeutic agent.

[0090] In one aspect, disclosed herein is a formulation for transdermal delivery of one or more buffering agent through the skin of a subject, comprising: a buffering agent comprising a carbonate salt in an amount between about 10-45 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount between about 1 to 15 %w/w; and wherein the formulation comprises water in an amount between about 15 to 65 %w/w, and wherein the formulation comprises less than about 12 %w/w lecithin.

[0091] In another aspect, disclosed herein is a method for transdermal delivery of a carbonate salt of the formulation comprising: a buffering agent comprising a carbonate salt in an amount between about 10-45 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount between about 1 to 15 %w/w; and wherein the formulation comprises water in an amount between about 15 to 65 %w/w, and wherein the formulation comprises less than about 12 %w/w lecithin, through the skin of a subject, wherein the carbonate salt of the formulation is in an amount between about 15-32 %w/w of the formulation, wherein the formulation comprises less than about 12 %w/w lecithin, and wherein the alkalinity of the formulation enhances penetration of the therapeutic agent.

[0092] In yet another aspect, disclosed herein is a formulation for transdermal delivery of a therapeutic agent through the skin of a subject, wherein the formulation comprises at least one active agent in an amount effective for treatment of a condition in the subject and the formulation comprising: a buffering agent comprising a carbonate salt in an amount between about 10-45 %w/w; a penetrant portion in an amount between about 5 to 55 %w/w; a detergent portion in an amount between about 1 to 15 %w/w; wherein the formulation comprises water in an amount between about 15 to 65 %w/w, through the skin of a subject, wherein the carbonate salt of the formulation is in an amount between about 15-32 %w/w of the formulation, and wherein the formulation comprises less than about 12 %w/w lecithin.

[0093] In some embodiments, a suitable formulation comprises: Lipmax™ in an amount between about 1-20 % w/w; benzyl alcohol in an amount between about 0.25 to 5 % w/w; menthol in an amount between about 0.1-5 %w/w; pluronic® in an amount between about 0.1- 5 %w/w; water in an amount between about 10-80 %w/w; sodium carbonate in an amount between about 1-32 %w/w; sodium bicarbonate in an amount between about 1-32 %w/w; ethylene glycol tetraacetic acid in an amount less than about 5 %w/w; propylene glycol in an amount between about 0.5-10 %w/w; almond oil in an amount between about 0.5-10 %w/w; cetyl alcohol in an amount between about 0.5-10 %w/w; lecithin in an amount less than about 12 %w/w; Cetiol Ultimate® in an amount less than about 10 %w/w; and ethanol in an amount between about 0.5-10 %w/w.

[0094] In some embodiments, a suitable formulation comprises: Lipmax™ in an amount between about 1-20 % w/w; benzyl alcohol in an amount between about 0.25 to 5 % w/w; menthol in an amount between about 0.1-5 %w/w; durasoft® in an amount between about 0.1- 5 %w/w; pluronic® in an amount between about 0.1-5 %w/w; water in an amount between about 10-80 %w/w; sodium carbonate in an amount less than about 32 %w/w; sodium bicarbonate in an amount between about 1-32 %w/w; ethylene glycol tetraacetic acid in an amount less than about 5 %w/w; sodium decanoate in an amount less than about 5 %w/w; propylene glycol in an amount between about 0.5-10 %w/w; almond oil in an amount between about 0.5-10 %w/w; zinc oxide in an amount less than about 2 %w/w; cetyl alcohol in an amount between about 0.5-10 %w/w; and ethanol in an amount between about 0.5-10 %w/w.

[0095] In some embodiments, a suitable formulation comprises: Water in an amount between about 10-80 %w/w; phospholipon® 90G in an amount between about 0.5-16 %w/w; myritol® 312 in an amount between about 0.5-10 %w/w; isopropyl palmitate in an amount between about 1-10 %w/w; Cetiol® Ultimate in an amount between about 0.25-5 %w/w; stearic acid in an amount between about 0.25-5 %w/w; cetyl alcohol in an amount between about 0.25-5 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; propylene glycol in an amount between about 0.25-5 %w/w; glycerin in an amount between about 0.25- 5 %w/w; ethanol in an amount between about 0.25-5 %w/w; Pluronic® in an amount between about 0.1-5 %w/w; Lipmax™ in an amount between about 1-20 %w/w; and sodium bicarbonate in an amount between about 1-32 %w/w.

[0096] In some embodiments, a suitable formulation comprises: Siligel™ in an amount between about 1-5 % w/w; water in an amount between about 10-80 %w/w; Phospholipon® 90G in an amount between about 0.5-16 %w/w; Myritol® 312 in an amount between about 0.5-10 %w/w; isopropyl palmitate in an amount between about 1-10 %w/w; Cetiol® Ultimate in an amount between about 0.25-5 %w/w; stearic acid in an amount between about 0.25-5 %w/w; cetyl alcohol in an amount between about 0.25-5 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; propylene glycol in an amount between about 0.25-5 %w/w; glycerin in an amount between about 0.25-5 %w/w; ethanol in an amount between about 0.25- 5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; Lipmax™ in an amount less than about 20 %w/w; and sodium bicarbonate in an amount between about 1- 32 %w/w.

[0097] In some embodiments, a suitable formulation comprises: water in an amount between about 10-80 %w/w; Phospholipon® 90G in an amount between about 0.5-10 %w/w; Myritol® 312 in an amount between about 0.5-10 %w/w; isopropyl palmitate in an amount between about 0.5-10 %w/w; Cetiol® Ultimate in an amount less than about 10 %w/w; stearic Acid in an amount between about 0.25-5 %w/w; cetyl alcohol in an amount between about 0.25-5 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; propylene glycol in an amount between about 0.25-5 %w/w; glycerin in an amount between about 0.25-5 %w/w; ethanol in an amount between about 0.25-5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-35 %w/w.

[0098] In some embodiments, a suitable formulation comprises: water in an amount between about 10-40 %w/w; Phospholipon® 90H in an amount between about 0.5-20 %w/w; Myritol® 312 in an amount between about 0.5-10 %w/w; isopropyl palmitate in an amount between about 0.5-20 %w/w; Cetiol® Ultimate in an amount less than about 10 %w/w; stearic acid in an amount between about 0.25-5 %w/w; cetyl alcohol in an amount between about 0.25-5 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; propylene glycol in an amount between about 0.25-5 %w/w; glycerin in an amount between about 0.25-5 %w/w; ethanol in an amount between about 0.25-5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-35 %w/w. [0099] In some embodiments, a suitable formulation comprises: water in an amount between about 10-40 %w/w; Phospholipon® 90H in an amount between about 0.5-20 %w/w; Phospholipon® 90G in an amount between about 0.5-20 %w/w; Myritol® 312 in an amount between about 0.5-10 %w/w; isopropyl palmitate in an amount between about 0.5-20 %w/w; Cetiol® Ultimate in an amount less than about 10 %w/w; stearic acid in an amount between about 0.25-5 %w/w; cetyl alcohol in an amount between about 0.25-5 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; propylene glycol in an amount between about 0.25- 5 %w/w; glycerin in an amount between about 0.25-5 %w/w; ethanol in an amount between about 0.25-5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-35 %w/w.

[00100] In some embodiments, a suitable formulation comprises: water in an amount between about 10-50 %w/w; Pluronic® gel 30% in an amount between about 5-30 %w/w; isopropyl palmitate in an amount between about 0.5-20 %w/w; stearic Acid in an amount between about 0.25-10 %w/w; cetyl alcohol in an amount between about 0.25-10 %w/w; benzyl alcohol in an amount between about 0.25-5 %w/w; almond oil in an amount between about 0.5-10 %w/w; propylene glycol in an amount between about 0.25-10 %w/w; ethanol in an amount between about 0.25-5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-32 %w/w.

[00101] In some embodiments, a suitable formulation comprises: Siligel™ in an amount less than about 5 % w/w; water in an amount between about 10-65 %w/w; isopropyl palmitate in an amount between about 0.5-10 %w/w; stearic Acid in an amount between about 0.25-10 %w/w; cetyl alcohol in an amount between about 0.25-10 %w/w; glycerin in an amount between about 0.25-5 %w/w; Lipmax™ in an amount between about 0.25-10 %w/w; ethanol in an amount less than about 5 %w/w; benzyl alcohol in an amount less than about 5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-32 %w/w.

[00102] In some embodiments, a suitable formulation comprises Aveeno® in an amount between about 20-85 %w/w; and sodium bicarbonate (3DF) in an amount between about 15- 45 %w/w.

[00103] In some embodiments, a suitable formulation comprises Aveeno® in an amount between about 20-85 %w/w; and sodium bicarbonate (Milled #7) in an amount between about 15-45 %w/w. [00104] In some embodiments, a suitable formulation comprises: Siligel™ in an amount less than about 5 % w/w; water in an amount between about 10-55 %w/w; isopropyl palmitate in an amount between about 0.5-10 %w/w; stearic Acid in an amount between about 0.25-5 %w/w; Cetyl alcohol in an amount between about 0.25-10 %w/w; almond oil in an amount between about 0.5-10 %w/w; propylene glycol in an amount between about 0.25-10 %w/w; ethanol in an amount less than about 5 %w/w; benzyl alcohol in an amount less than about 5 %w/w; sodium hydroxide 50 %w/v in an amount between about 0.1-5 %w/w; and sodium bicarbonate in an amount between about 1-32 %w/w.

[00105] The surprising effects achieved by the formulations and methods of the present invention are in part attributable to an improved formulation that enhances delivery of a carbonate salt through the skin. In some embodiments, the formulation employs penetrants described US2009/0053290 (’290), W02014/209910 (’910), and WO2017/127834. The present formulations may include a nonionic surfactant. Applicant has found that by employing carbonate salts with particle sizes as disclosed herein, delivered with the penetrants as disclosed herein, and in some embodiments providing a combination of a nonionic surfactant and a polar gelling agent, the penetration capabilities of the carbonate salts of the resulting formulation and the effective level of delivery of the carbonate salts has been enhanced. This enhanced level of penetration was also achieved using significantly less lecithin than anticipated or none at all. This result was completely unexpected as it was believed that relatively equal amounts of the benzyl alcohol and lecithin organogel especially a somewhat higher concentration of benzyl alcohol than lecithin organogel were responsible for the level of penetration achieved by prior art formulations.

[00106] Briefly, the penetrants described in the above-referenced US and PCT applications are based on combinations of synergistically acting components. Many such penetrants are based on combinations of an alcohol, such as benzyl alcohol to provide a concentration of 0.5- 20% w/w of the final formulation with lecithin organogel present in the penetrant to provide 25-70% w/w of the formulation. These penetrants are also useful when the agent is a buffer, such as sodium bicarbonate, but less lecithin organogel may be required - e.g. less than 12 %w/w when the sodium bicarbonate is present at high concentration as disclosed herein.

[00107] In some embodiments, the buffering component is any mildly basic compound or combination that will result in a pH of 7-8 in the microenvironment of the tumor cells. In some embodiments, the formulation has a pH of 7-10. Such buffers, in addition to carbonate and/or bicarbonate salts, include lysine buffers, chloroacetate buffers, tris buffers (i.e., buffers employing tris (hydroxymethyl) aminoethane), phosphate buffers and buffers employing non natural amino acids with similar pKa values to lysine. In some embodiments, the carbonate and/or bicarbonate salt is in an amount between about 7-32 %w/w of the formulation. For example, the enantiomers of native forms of such amino acids or analogs of lysine with longer or shorter carbon chains or branched forms thereof. Histidine buffers may also be used. Typically, the concentration of buffer in the compositions is in the range of 10-50 %w/w. More typical ranges for sodium bicarbonate or sodium carbonate or both are 10-35% by weight. In some embodiments, the carbonate salt is in an amount between about 15-32 %w/w of the formulation.

[00108] Alternatively, the penetrant component comprises a completion component as well as one or more electrolytes sufficient to impart viscosity and viscoelasticity, one or more surfactants and an alcohol. The completion component can be a polar liquid, a non-polar liquid or an amphiphilic substance.

[00109] The percentage of carbonate salt in the formulation will depend upon the amount required to be delivered in order to have a useful effect on treating the disorder. In general, the carbonate salt may be present in the formulation in an amount as low as 1 %w/w up to about 50 %w/w. Typical concentrations may include 15-32 %w/w. Since the required percentage of carbonate salt depends on the frequency of administration, as well as the time allotted for administration for each application, the level of carbonate salt may be varied over a wide range. In some embodiments, the carbonate salt is sodium carbonate and/or sodium bicarbonate milled to a particle size is less than 200 pm. In some embodiments, the carbonate salt is sodium carbonate and/or sodium bicarbonate milled to a particle size is less than 70 pm. In some embodiments, the carbonate salt is sodium carbonate and/or sodium bicarbonate milled to a particle size is less than 70 pm, wherein the sodium bicarbonate is solubilized in the formulation in an amount less than 20 %w/w of the formulation. In some embodiments, the carbonate salt is sodium carbonate and/or sodium bicarbonate milled to a particle size is less than 70 pm, wherein particle sizes less than about 10 pm have an enhanced penetration thru the skin of a subject. In some embodiments, the sodium carbonate and/or sodium bicarbonate are jet milled to a particle size less than about 70 pm. In some embodiments, the sodium bicarbonate is Sodium Bicarbonate USP Grade 3DF that has a particle size distribution less than 70 pm.

[00110] The formulations of the disclosure may be prepared in a number of ways. Typically, the components of the formulation are simply mixed together in the required amounts. However, it is also desirable in some instances to, for example, carry out dissolution of a carbonate salt and then add a separate preparation containing the components aiding the delivery of the carbonate salts in the form of a carrier. The concentrations of these components in the carrier, then, will be somewhat higher than the concentrations required in the final formulation. Thus, sodium bicarbonate may first be dissolved in water and then added to a carrier comprising an alcohol, lecithin and optionally a combination of a nonionic surfactant and polar gelling agent, or of ionic detergent. Alternatively, some subset of these components can first be mixed and then "topped off with the remaining components either simultaneously or sequentially. The precise manner of preparing the formulation will depend on the choice of carbonates and the percentages of the remaining components that are desirable with respect to that carbonate salt. In some embodiments, the water is in an amount between about 10-85 %w/w, 15-50 %w/w, or 15-45 %w/w of the formulation.

[00111] The penetrant portion is a multi-component mixture, whereby the particular concentrations of the penetration enhancers are informed in part by the molecular mass of the sodium bicarbonate, or sodium bicarbonate and the therapeutic agent to be transported. The formulation enables the sodium bicarbonate and/or therapeutic agent to become bio-available to the target site within minutes of topical administration. The formulations permit the use of minimal concentrations of therapeutic agents, as little as. 1/1000th of concentrations required of alternative processes, while enabling bioactivity and positive clinical outcomes simultaneously. In some embodiments, the penetrant portion comprises an alcohol in an amount less than 5 %w/w of the formulation.

[00112] One important aspect of the invention is based on the above-noted recognition that some tumors do not respond to buffer treatment as their microenvironment is not acidic and at least some of these tumors achieve metastasis by elevation of certain proteolytic enzymes that break down the extracellular matrix (ECM). If buffer treatment is contemplated, tumor cells from the biopsy of a solid tumor in a subject are therefore preferably cultured and tested in advance of treatment to insure responsiveness to buffer. Such evaluation can be carried out by any suitable means, including measurement of pH, assessment of the levels of relevant proteases, and invasion assays as impacted by buffer treatment as described in Bailey, K.M. et al (2014) supra. One important such assay is a glycolytic stress assay as described therein. Cell cultures of biopsied tumors that appear not to respond to buffer treatment as shown by such assays may benefit from administration of other antimetastatic agents and inclusion of such agents in the compositions of the invention that include buffers would also be beneficial. Thus, treatment with buffer-containing compositions alone may be contraindicated and the subject is not administered buffer as the sole active agent but diverted to alternative treatment. This does not mean, of course, that buffer is necessarily omitted from formulations used to administer alternative active agents.

[00113] The formulations comprise mixtures wherein the components interact synergistically and induce skin permeation enhancements better than that induced by the individual components. Synergies between chemicals can be exploited to design potent permeation enhancers that overcome the efficacy limitations of single enhancers. Several embodiments disclosed herein utilize three to five distinct permeation enhancers.

[00114] For topical administration, and in particular transdermal administration, the formulation will comprise penetrants including either or both chemical penetrants (CPEs) and peptide-based cellular penetrating agents (CPPs) that encourage transmission across the dermis and/or across membranes including cell membranes, as would be the case in particular for administration by suppository or intranasal administration, but for transdermal administration as well. Particularly suitable penetrants especially for those that contain at least one agent other than buffer include those that are described in the above-referenced US2009/0053290 ('290), W02014/209910 ('910), and WO2017/127834. In addition to formulations with penetrants, transdermal delivery can be affected by mechanically disrupting the surface of the skin to encourage penetration, or simply by supplying the formulation applied to the skin under an occlusive patch.

[00115] Alternatively, the penetrant portion comprises a completion component as well as one or more electrolytes sufficient to impart viscosity and viscoelasticity, one or more surfactants and an alcohol. The completion component can be a polar liquid, anon-polar liquid or an amphiphilic substance. The penetrant may further comprise a keratinolytic agent effective to reduce thiol linkages, disrupt hydrogen bonding and/or effect keratin lysis and/or a cell penetrating peptide (sometimes referred to as a skin-penetrating peptide) and/or a permeation enhancer.

[00116] Lecithin organogel is a combination of lecithin with a gelling component, which is typically amphiphilic. Suitable gelling components also include isopropyl palmitate, ethyl laurate, ethyl myristate and isopropyl myristate. In some embodiments, the formulation comprises a gelling agent in an amount less than 5 %w/w of the formulation. Certain hydrocarbons, such as cyclopentane, cyclooctane, trans- decalin, /ra -pinane. n-pentane, n- hexane, n-hexadecane may also be used. Thus, an important permeation agent is a lecithin organogel, wherein the combination resulting from lecithin and the organic solvent acts as a permeation agent. In some embodiments, the penetrant portion comprises lecithin organogel, an alcohol, a surfactant, and a polar solvent. In some embodiments, the lecithin organogel is a combination of soy lecithin and isopropyl palmitate. In some embodiments, the penetrant portion comprises lecithin and isopropyl palmitate, undecane, isododecane, isopropyl stearate, or a combination thereof. In some embodiments, the formulation comprises Lipmax™ (sold by Lucas Meyer Cosmetics) in an amount between about 1-20 % w/w or an equivalent 50/50 mixture of isopropyl palmitate and lecithin. Lecithin organogels are clear, thermodynamically stable, viscoelastic, and biocompatible jelly-like phases composed of hydrated phospholipids and appropriate organic liquid. An example of a suitable lecithin organogel is lecithin isopropyl palmitate, which is formed when isopropyl palmitate is used to dissolve lecithin. The ratio of lecithin to isopropyl palmitate may be 50:50. Illustrated below in the Examples is a formulation containing soy lecithin in combination with isopropyl palmitate; however, other lecithins could also be used such as egg lecithin or synthetic lecithins. Various esters of long chain fatty acids may also be included. Methods for making such lecithin organogels are well known in the art. In most embodiments, the lecithin organogel is present in the final formulation is less than about 20 %w/w. In those compositions used to dissolve fat deposits, to alleviate pain from fat removal or in anhydrous compositions, the concentration of lecithin organogel may be as low as 0.5% w/w, 1 % w/w, 5% w/w, 10% w/w or 20% w/w. In some embodiments, the penetrant portion comprises a mixture of xanthan gum, lecithin, sclerotium gum, pullulan, or a combination thereof in an amount less than 2 %w/w, 5 %w/w, or 10 %w/w of the formulation. In some embodiments, the formulation comprises Siligel™ in an amount between about 1-5 % w/w or 5-15 % w/w, or an equivalent mixture of xanthan gum, lecithin, sclerotium gum, and pullulan. In some embodiments, the penetrant portion comprises a mixture of caprylic triglycerides and capric triglycerides in amount less than 2 %w/w, 8 %w/w, or 10 %w/w of the formulation. In some embodiments, the formulation comprises Myritol® 312 in an amount between about 0.5-10 %w/w, or an equivalent mixture of caprylic triglycerides and capric triglycerides.

[00117] In some embodiments, the penetrant portion comprises phosphatidyl choline in amount less than 12 %w/w or 18 %w/w of the formulation. In some embodiments, the penetrant portion comprises a phospholipid in amount less than 12 %w/w or 18 %w/w of the formulation. In some embodiments, the penetrant portion comprises a mixture of tridecane and undecane in amount less than 2 %w/w, 5 %w/w, or 8 %w/w of the formulation. In some embodiments, the formulation comprises Cetiol Ultimate® in an amount less than about 2 %w/w, 5 %w/w, or 10 %w/w, or an equivalent mixture of tridecane and undecane. In some embodiments, the penetrant portion comprises cetyl alcohol in amount less than 2 %w/w, 5 %w/w, or 8 %w/w of the formulation. In some embodiments, the penetrant portion comprises benzyl alcohol in an amount less than about 2 %w/w, 5 %w/w, or 8 %w/w. In some embodiments, the penetrant portion comprises stearic acid in an amount less than 2 %w/w, 5 %w/w, or 8 %w/w of the formulation.

[00118] Lecithin organogels may be in the form of vesicles, microemulsions and micellar systems. In the form of self-assembled structures, such as vesicles or micelles, they can fuse with the lipid bilayers of the stratum comeum, thereby enhancing partitioning of encapsulated drug, as well as a disruption of the ordered bilayers structure. An example of a phospholipid- based permeation enhancement agent comprises a micro-emulsion-based organic gel defined as a semi-solid formation having an external solvent phase immobilized within the spaces available of a three-dimensional networked structure. This micro-emulsion-based organic gel in liquid phase is characterized by l,2-diacyl-sn-glycero-3-phosphatidyl choline, and an organic solvent, which is at least one of: ethyl laureate, ethyl myristate, isopropyl myristate, isopropyl palmitate; cyclopentane, cyclooctane, trans- decalin, /ra/ v-pinane. «-pentane, n- hexane, «-hexadecane, and tripropylamine.

[00119] The lecithin organogels are formulated with an additional component to assist in the formation of micelles or vascular structures. In one approach, the organogels are formulated with a polar component such as water, glycerol, ethyleneglycol or formamide, in particular with water. In general, a nonionic detergent such as a poloxamer in aqueous solution is used to top off. Alternatively, an anhydrous composition may be obtained by using, instead of a polar component, a material such as a bile salt. When formulated with bile salts, the mi cellular nature of the composition is altered so that rather than a more or less spherical vesicular form, the vesicles become wormlike and are able to accommodate larger guest molecules, as well as penetrate the epidermis more effectively. Suitable bile salts include salts of deoxy cholic acid, taurocholic acid, glycocholic acid, taurochenodeoxycholic acid, glycochenodeoxycholic acid, cholic acid and the like. Certain detergents, such as Tween® 80 or Span® 80 may be used as alternatives. The percentage of these components in the anhydrous forms of the composition is in the range of 1 % w/w - 15% w/w. In some embodiments, the range of bile salt content is 2% - 6% w/w or 1 % - 3.5% w/w. In these essentially anhydrous forms, powdered or micronized nonionic detergent is used to top off, typically in amounts of 20% - 60% w/w. In one approach to determine the amount of bile salt, the% is calculated by dividing the %w/w of lecithin by 10

[00120] An additional component in the formulations of the disclosure is an alcohol. Benzyl alcohol and ethanol are illustrated in the Examples in particular, derivatives of benzyl alcohol which contain substituents on the benzene ring, such as halo, alkyl and the like. The weight percentage of benzyl or other related alcohol in the final composition is 0.5-20% w/w, and again, intervening percentages such as 1 % w/w, 2% w/w, 5% w/w, 7% w/w, 10% w/w, and other intermediate weight percentages are incl tided. Due to the aromatic group present in a permeation enhancement formulation such as benzyl alcohol, the molecule has a polar end (the alcohol end) and a non-polar end (the benzene end). This enables the agent to dissolve a wider variety of drugs and agents. The alcohol concentration is substantially lower than the concentration of the lecithin organogel in the composition.

[00121] In some embodiments, as noted above, the performance of the formulations is further improved by including a nonionic detergent and polar gelling agent or including bile salts and a powdered surfactant. In both aqueous and anhydrous forms of the composition, detergents, typically nonionic detergents are added. In general, the nonionic detergent should be present in an amount of at least 2% w/w to 60% w/w. Typically, in the compositions wherein the formulation is topped off with a polar or aqueous solution containing detergent, the amount of detergent is relatively low - e.g., 2%-25% w/w, or 5-15% w/w or 7-12% w/w. However, in compositions comprising bile salts that are essentially anhydrous and are topped-off by powdered detergent, relatively higher percentages are usually used - e.g., 20%-60% w/w.

[00122] In some embodiments, the nonionic detergent provides suitable handling properties whereby the formulations are gel-like or creams at room temperature. To exert this effect, the detergent, typically a poloxamer, is present in an amount between about 2-12 %w/w, preferably between about 5-25 %w/w in polar formulations. In the anhydrous forms of the compositions, the detergent is added in powdered or micronized form to bring the composition to 100% and higher amounts are used. In compositions with polar constituents, rather than bile salts, the nonionic detergent is added as a solution to bring the composition to I 00%. If smaller amounts of detergent solutions are needed due to high levels of the remaining components, more concentrated solutions of the nonionic detergent are employed. Thus, for example, the percent detergent in the solution may be 10% to 40% or 20% or 30% and intermediate values depending on the percentages of the other components. [00123] Suitable nonionic detergents include poloxamers such as Pluronic® and any other surfactant characterized by a combination of hydrophilic and hydrophobic moieties. Poloxamers are triblock copolymers of a central hydrophobic chain of polyoxypropylene flanked by two hydrophilic chains of polyethyleneoxide. Other nonionic surfactants include long chain alcohols and copolymers of hydrophilic and hydrophobic monomers where blocks of hydrophilic and hydrophobic portions are used.

[00124] In some embodiments, the formulation also contains surfactant, typically, nonionic surfactant at 2-25% w/w along with a polar solvent wherein the polar solvent is present in an amount at least in molar excess of the nonionic surfactant. In these embodiments, typically, the composition comprises the above-referenced amounts of lecithin organogel and benzyl alcohol along with a carbonate salt with a sufficient amount of a polar solution, typically an aqueous solution or polyethylene glycol solution that itself contains 10%-40% of surfactant, typically nonionic surfactant to bring the composition to 100%.

[00125] Other examples of surfactants include polyoxyethylated castor oil derivatives such as HCO-60 surfactant sold by the HallStar Company; nonoxynol; octoxynol; phenylsulfonate; poloxamers such as those sold by BASF as Pluronic® F68, Pluronic® F127, and Pluronic® L62; polyoleates; Rewopal® HVIO, sodium laurate, sodium lauryl sulfate (sodium dodecyl sulfate); sodium oleate; sorbitan dilaurate; sorbitan dioleate; sorbitan monolaurate such as Span® 20 sold by Sigma-Aldrich; sorbitan monooleates; sorbitan trilaurate; sorbitan trioleate; sorbitan monopalmitate such as Span® 40 sold by Sigma-Aldrich; sorbitan stearate such as Span® 85 sold by Sigma-Aldrich; polyethylene glycol nonylphenyl ether such as Synperonic® NP sold by Sigma-Aldrich; p-(l,l,3,3-tetramethylbutyl)-phenyl ether sold as Triton™ X-100 sold by Sigma-Aldrich; and polysorbates such as polyoxyethylene (20) sorbitan monolaurate sold as Tween® 20, polysorbate 40 (polyoxyethylene (20) sorbitan monopalmitate) sold as Tween® 40, polysorbate 60 (polyoxyethylene (20) sorbitan monostearate) sold as Tween® 60, polysorbate 80 (polyoxyethylene (20) sorbitan monooleate) sold as Tween® 80, and polyoxyethylenesorbitan trioleate sold as Tween® 85 by Sigma-Aldrich. The weight percentage range of nonionic surfactant is in the range of 3% w/w-15% w/w, and again includes intermediate percentages such as 5% w/w, 7% w/w, 10% w/w, 12% w/w, and the like. In some embodiments, the detergent portion comprises a nonionic surfactant in an amount between about 2-25 %w/w of the formulation; and a polar solvent in an amount less than 5 %w/w of the formulation. In some embodiments, the nonionic surfactant is a poloxamer and the polar solvent is water, an alcohol, or a combination thereof. In some embodiments, the detergent portion comprises poloxamer, propylene glycol, glycerin, ethanol, 50 % w/v sodium hydroxide solution, or a combination thereof. In some embodiments, the detergent portion comprises glycerin in an amount less than 3 %w/w of the formulation.

[00126] In the presence of a polar gelling agent, such as water, glycerol, ethyleneglycol or formamide, a micellular structure is also often achieved. Typically, the polar agent is in molar excess of the nonionic detergent. The inclusion of the nonionic detergent/polar gelling agent combination results in a more viscous and cream-like or gel-like formulation which is suitable for application directly to the skin. This is typical of the aqueous forms of the composition.

[00127] In some embodiments other additives are included such as a gelling agent, a dispersing agent and a preservative. An example of a suitable gelling agent is hydroxypropylcellulose, which is generally available in grades from viscosities of from about 5 cps to about 25,000 cps such as about 1500 cps. All viscosity measurements are assumed to be made at room temperature unless otherwise stated. The concentration of hydroxypropylcellulose may range from about 1% w/w to about 2% w/w of the composition. Other gelling agents are known in the art and can be used in place of, or in addition to hydroxypropylcellulose. An example of a suitable dispersing agent is glycerin. Glycerin is typically included at a concentration from about 5% w/w to about 25% w/w of the composition. A preservative may be included at a concentration effective to inhibit microbial growth, ultraviolet light and/or oxygen-induced breakdown of composition components, and the like. When a preservative is included, it may range in concentration from about 0.01 % w/w to about 1.5% w/w of the composition.

[00128] Typical components that may also be included in the formulations are fatty acids, terpenes, lipids, and cationic, and anionic detergents. In some embodiments, the formulation further comprises tranexamic acid in an amount less than 2 % w/w, 5 % w/w, or 10 % w/w of the formulation. In some embodiments, the formulation further comprises a polar solvent in an amount less than 2 % w/w, 5 % w/w, 10 % w/w, or 20 % w/w of the formulation. In some embodiments, the formulation further comprises a humectant, an emulsifier, an emollient, or a combination thereof. In some embodiments, the formulation further comprises ethylene glycol tetraacetic acid in an amount less than about 2 % w/w, 5 % w/w, or 10 % w/w. In some embodiments, the formulation further comprises almond oil in an amount less than about 5 % w/w. In some embodiments, the formulation further comprises a mixture of thermoplastic polyurethane and polycarbonate in an amount less than about 5 % w/w. In some embodiments, the formulation further comprises phosphatidylethanolamine in an amount less than about 5 % w/w. In some embodiments, the formulation further comprises an inositol phosphatide in an amount less than about 5 % w/w.

[00129] Other solvents and related compounds that may be used in some embodiments include acetamide and derivatives, acetone, n-alkanes (chain length between 7 and 16), alkanols, diols, short chain fatty acids, cyclohexyl-l,l-dimethylethanol, dimethyl acetamide, dimethyl formamide, ethanol, ethanol/d-limonene combination, 2-ethyl- 1,3-hexanediol, ethoxy diglycol (Transcutol® by Gattefosse, Lyon, France), glycerol, glycols, lauryl chloride, limonene N-methylformamide, 2-phenylethanol, 3 -phenyl- 1 -propanol, 3-phenyl-2-propen-l- ol, polyethylene glycol, polyoxyethylene sorbitan monoesters, polypropylene glycol 425, primary alcohols (tridecanol), 1,2-propane diol, butanediol, C3-C6 triols or their mixtures and a polar lipid compound selected from Ci6 or Cie monounsaturated alcohol, Ci6 or Cie branched saturated alcohol and their mixtures, propylene glycol, sorbitan monolaurate sold as Span® 20 by Sigma-Aldrich, squalene, triacetin, trichloroethanol, trifluoroethanol, trimethylene glycol and xylene.

[00130] Fatty alcohols, fatty acids, fatty esters, are bilayer fluidizers that may be used in some embodiments. Examples of suitable fatty alcohols include aliphatic alcohols, decanol, lauryl alcohol (dodecanol), unolenyl alcohol, nerolidol, 1-nonanol, «-octanol. and oleyl alcohol. Examples of suitable fatty acid esters include butyl acetate, cetyl lactate, decyl N,N- dimethylamino acetate, decyl N,N-dimethylamino isopropionate, diethyleneglycol oleate, diethyl sebacate, diethyl succinate, diisopropyl sebacate, dodecyl N,N-dimethy amino acetate, dodecyl (N,N-dimethylamino)-butyrate, dodecyl N,N-dimethylamino isopropionate, dodecyl 2-(dimethyamino) propionate, EO-5-oleyl ether, ethyl acetate, ethylaceto acetate, ethyl propionate, glycerol monoethers, glycerol monolaurate, glycerol monooleate, glycerol monolinoleate, isopropyl isostearate, isopropyl linoleate, isopropyl myristate, isopropyl myristate/fatty acid monoglyceride combination, isopropyl palmitate, methyl acetate, methyl caprate, methyl laurate, methyl propionate, methyl valerate, 1 -monocaproyl glycerol, monoglycerides (medium chain length), nicotinic esters (benzyl), octyl acetate, octyl N,N- dimethylamino acetate, oleyl oleate, n-pentyl N-acetylprolinate, propylene glycol monolaurate, sorbitan dilaurate, sorbitan dioleate, sorbitan monolaurate, sorbitan monolaurate, sorbitan trilaurate, sorbitan trioleate, sucrose coconut fatty ester mixtures, sucrose monolaurate, sucrose monooleate, tetradecyl N.N-dimethylamino acetate. Examples of suitable fatty acid· include alkanoic acids, caprid acid, diacid, ethyloctadecanoic acid, hexanoic acid, lactic acid, lauric acid, linoelaidic acid, linoleic acid, linolenic acid, neodecanoic acid, oleic acid, palmitic acid, pelargonic acid, propionic acid, and vaccenic acid. Examples of suitable fatty alcohol ethers include a-monoglyceryl ether, EO-2-oleyl ether, EO-5-oleyl ether, EO-10-oleyl ether, ether derivatives of polyglycerols and alcohols, and (l-0-dodecyl-3-0-methyl-2-0-(2',3 dihydroxypropyl glycerol).

[00131] Examples of completing agents that may be used in some embodiments include b- and g-cyclodextrin complexes, hydroxypropyl methylcellulose (e.g., Carbopol® 934), liposomes, naphthalene diamide diimide, and naphthalene diester diimide.

[00132] One or more anti-oxidants may be included, such as vitamin C, vitamin E, proanthocyanidin and a-lipoic acid typically in concentrations of 0.1 %-2.5% w/w.

[00133] In some applications, it is desirable to adjust the pH of the formulation to assist in permeation or to adjust the nature of the carbonate and/or of the target compounds in the subject. In some instances, the pH is adjusted to a level of pH 9-11 or 10-11 which can be done by providing appropriate buffers or simply adjusting the pH with base.

[00134] In some applications, in particular when the therapeutic agent includes an anesthetic, epinephrine or an alternate vasoconstrictor, such as phenylephrine or epinephrine sulfate may be included in the formulation if a stabilizing agent is present. Otherwise, the epinephrine should be administered in tandem since epinephrine is not stable at alkali pH.

[00135] In any of the anesthetic compositions, it may be desirable to administer the epinephrine in tandem with the transdermal anesthetic. Alternatively, treatment of the epinephrine with a chelator, such as the iron chelator Desferal® may stabilize the epinephrine sufficiently to include it in the transdermal formulation.

[00136] It is understood that some tumors do not respond to treatment with buffer, but apparently metastasize by virtue of elevated levels of proteases that attack the extracellular matrix surrounding the tumor. In any event, breakdown of the ECM would encourage metastasis. Therefore, an additional active agent that is optionally included in the compositions of the invention is one or more appropriate protease inhibitors. Particularly important are inhibitors of cathepsins, for example of cathepsin B, and inhibitors of matrix metalloproteinases (MMPs). These components are active alone or augment the effect of buffer for tumors that are not resistant to buffer treatment.

[00137] The formulations may include other components that act as excipients or serve purposes other than active anti-tumor effects. For example, preservatives like antioxidants e.g., ascorbic acid or a-lipoic acid and antibacterial agents may be included. Other components apart from therapeutically active ingredients and components that are the primary effectors of dermal penetration may include those provided for aesthetic purposes such as menthol or other aromatics, and components that affect the physical state of the composition such as emulsifiers, for example, Durasoft® (which is a mixture of thermoplastic polyurethane and polycarbonate). Typically, these ingredients are present in very small percentages of the compositions. It is understood that these latter ancillary agents are neither therapeutically ingredients nor are they components that are primarily responsible for penetration of the skin. The components that primarily effect skin penetration have been detailed as described above. However, some of these substances have some capability for effecting skin penetration. See, for example, Kunta, J.R. et al, J. Pharm. Sci. (1997) 86:1369-1373, describing penetration properties of menthol.

[00138] In embodiments where a bile salt is added to the combination of benzyl alcohol and lecithin organogel in lieu of topping off with an aqueous medium, micelles that would have been relatively spherical may become elongated and worm-like thus permitting superior penetration of the stratum comeum of the epidermis. The worm like formation of the micelles is particularly helpful in accommodating higher molecular weight therapeutic agents. As is known, bile salts are facial amphiphiles and include salts of taurocholic acid, glycocholic acid, taurochenodeoxycholic acid, glycochenodeoxycholic acid, cholic acid, deoxycholic acid. Detergents are also useful in lieu of bile salts and include Tween® 80 and Span® 80.

[00139] In another aspect, certain embodiments are directed to a sustained release drug delivery platform releases a therapeutic compound or compounds disclosed and made as a formulation described herein over a period of, without limitation, about 3 days after administration, about 7 days after administration, about 10 days after administration, about 15 days after administration, about 20 days after administration, about 25 days after administration, about 30 days after administration, about 45 days after administration, about 60 days after administration, about 75 days after administration, or about 90 days after administration. In other aspects of this embodiment, a sustained release drug delivery platform releases a therapeutic compound or compounds disclosed herein with substantially first order release kinetics over a period of, without limitation, at least 3 days after administration, at least 7 days after administration, at least 10 days after administration, at least 15 days after administration, at least 20 days after administration, at least 25 days after administration, at least 30 days after administration, at least 45 days after administration, at least 60 days after administration, at least 75 days after administration, or at least 90 days after administration. [00140] The formulation described in this specification may also comprise more than one therapeutic compound as desired for the particular indication being treated, preferably those with complementary activities that do not adversely affect the other proteins. The formulations to be used for in vivo administration can be sterile. This can be accomplished, for instance, without limitation, by filtration through sterile filtration membranes, prior to, or following, preparation of the formulation or other methods known in the art, including without limitation, pasteurization.

[00141] Packaging and instruments for administration may be determined by a variety of considerations, such as, without limitation, the volume of material to be administered, the conditions for storage, whether skilled healthcare practitioners will administer or patient self- compliance, the dosage regime, the geopolitical environment (e.g., exposure to extreme conditions of temperature for developing nations), and other practical considerations.

[00142] In certain embodiments, kits can comprise, without limitation, one or more cream or lotion comprising one or more formulations described herein. In various embodiments, the kit can comprise formulation components for transdermal, topical, or subcutaneous administration, formulated to be administered as an emulsion coated patch. In all of these embodiments and others, the kits can contain one or more lotion, cream, patch, or the like in accordance with any of the foregoing, wherein each patch contains a single unit dose for administration to a subject.

[00143] Imaging components can optionally be included, and the packaging also can include written or web-accessible instructions for using the formulation. A container can include, for example, a vial, bottle, patch, syringe, pre-filled syringe, tube or any of a variety of formats well known in the art for multi-dispenser packaging.

Administration and Dosing

[00144] The formulations provided herein can be topically administered in any form. For administration for the treatment of skin conditions a sufficient amount of the topical composition can be applied onto a desired area and surrounding skin, for example, in an amount sufficient to cover a desired area plus a margin of healthy skin or tissue surrounding the desired area, if possible, for example, a margin of about 0.5 inches. A desired area can be an area of the skin affected by skin disorder in some embodiments. However, in other embodiments a desired area of the skin may be unaffected or healthy, e.g., skin not having any disorder or condition. Also, the formulations can be applied to any skin surface, including for example, facial skin, and the skin of the hands, neck, chest and/or scalp.

[00145] In applying the formulations of the invention, the formulation itself is simply placed on the skin and spread across the surface and/or massaged to aid in penetration. The amount of formulation used is typically sufficient to cover a desired surface area. In some embodiments, a protective cover is placed over the formulation once it is applied and left in place for a suitable amount of time, i.e., 5 minutes, 10 minutes, 20 minutes or more; in some embodiments an hour or two. The protective cover can simply be a bandage including a bandage supplied with a cover that is impermeable to moisture. This essentially locks in the contact of the formulation to the skin and prevents distortion of the formulation by evaporation in some cases. The composition may be applied to the skin using standard procedures for application such as a brush, a syringe, a gauze pad, a dropper, or any convenient applicator. More complex application methods, including the use of delivery devices, may also be used, but are not required. In an alternative to administering topically to intact skin, the surface of the skin may also be disrupted mechanically by the use of spring systems, laser powered systems, systems propelled by Lorentz force or by gas or shock waves including ultrasound and may employ microdermabrasion such as by the use of sandpaper or its equivalent or using microneedles or electroporation devices. Simple solutions of the agent(s) as well as the above-listed formulations that penetrate intact skin may be applied using occlusive patches, such as those in the form micro-patches. External reservoirs of the formulations for extended administration may also be employed.

[00146] In an alternative to administering topically to intact skin, the surface of the skin may also be disrupted mechanically by the use of spring systems, laser powered systems, use of iontophoresis, systems propelled by Lorentz force or by gas or shock waves including ultrasound and may employ microdermabrasion such as by the use of sandpaper or its equivalent or using microneedles or electroporation devices. Simple solutions of the agent(s) as well as the above-listed formulations that penetrate intact skin may be applied using occlusive patches, such as those in the form micro-patches. External reservoirs of the formulations for extended administration may also be employed.

[00147] Accordingly, in certain embodiments alternative methods of administering one or more buffering agent, therapeutic compounds, agents, drugs through intact skin are provided. As nonlimiting examples, these alternative methods might be selected from the following lists: on basis of working mechanism, spring systems, laser powered, energy-propelled, Lorentz force, gas/air propelled, shock wave (including ultrasound), on basis of type of load, liquid, powder, projectile, on basis of drug delivery mechanism, nano-patches, sandpaper (microdermabrasion), iontophoresis enabled, microneedles, on basis of site of delivery, intradermal, intramuscular, and subcutaneous injection. Other suitable delivery mechanisms include, without limitation, microneedle drug delivery, such as 3M Systems, Glide SDI (pushes drug as opposed to “firing”drug), MIT low pressure injectors, micropatches (single use particle insertion device), microelectro mechanical systems (MEMS), dermoelectroporation devices (DEP), transderm ionto system (DEP), TTS transdermal therapeutic systems, membrane moderated systems (drug reservoir totally encapsulated in a shallow compartment), adhesive diffusion-controlled system (drug reservoir in a compartment fabricated from drug-impermable metallic plastic backing), matrix dispersion type system (drug reservoir formed by homogeneously dispersing drug solids in a hydrophilic or lipophilic polymer matrix molder into medicated disc), and microreservoir system (combination of reservoir and matrix dispersion-type drug delivery system).

[00148] It has been found, generally, that the requirements for effective penetration of the skin in the case of buffers as active agents are less restrictive than those required for alternative agents. In addition, effective systemic pH alteration can be used as a way to diagnose the effectiveness of penetration when topical administration is employed.

[00149] The application method is determined by the nature of the treatment but may be less critical than the nature of the formulation itself. If the application is to a skin area, it may be helpful in some instances to prepare the skin by cleansing or exfoliation. In some instances, it is helpful to adjust the pH of the skin area prior to application of the formulation itself. The application of the formulation may be by simple massaging onto the skin or by use of devices such as syringes or pumps. Patches could also be used. In some cases, it is helpful to cover the area of application to prevent evaporation or loss of the formulation.

[00150] Where the application area is essentially skin, it is helpful to seal-off the area of application subsequent to supplying the formulation and allowing the penetration to occur so as to restore the skin barrier. A convenient way to do this is to apply a composition comprising linoleic acid which effectively closes the entrance pathways that were provided by the penetrants of the invention. This application, too, is done by straightforward smearing onto the skin area or can be applied more precisely in measured amounts. [00151] In some embodiments, the disclosure is directed to administering a local anesthetic to a subject transdermally and a formulation which contains an effective amount of anesthetic along with 25%-70% w/w or 30%-60% w/w or 30%-40% w/w of lecithin organogel typically wherein the lecithin organogel comprises soy lecithin in combination with isopropyl palmitate or isopropyl myristate and benzyl alcohol in the range of 0.5%-20% w/w or 0. 9%-2% w/w benzyl alcohol optionally including 1 %-5% w/w or 2%-4% w/w menthol wherein the composition is topped off with a polar solution, typically an aqueous solution comprising 15%- 50% w/w or 20%-40% w/w or 20%-30% w/w poloxamer, typically Pluronic® or alternatively may be an anhydrous composition comprising bile salts such as deoxycholic acid or sodium deoxycholate in the range of 4%-8% w/w, typically 6% w/w and the remainder of the composition powdered nonionic detergent, typically Pluronic®. The pH of the compositions is adjusted to 9-11, typically 10-11. The formulations are applied to the desired area of the skin and may be covered, for example, with Saran™ wrap for a suitable amount of time. Following the treatment, the skin can be repaired by applying a composition comprising linoleic acid.

[00152] A wide variety of therapeutic agents may be used in the formulations, including anesthetics, fat removal compounds, nutrients, nonsteroidal anti-inflammatory drugs (NSAIDs) agents for the treatment of migraine, hair growth modulators, antifungal agents, anti viral agents, vaccine components, tissue volume enhancing compounds, anti-cellulite therapeutics, wound healing compounds, compounds useful to effect smoking cessation, agents for prevention of collagen shrinkage, wrinkle relief compounds such as Botox®, skin- lightening compounds, compounds for relief of bruising, cannabinoids including cannabidiols for the treatment of epilepsy, compounds for adipolysis, compounds for the treatment of hyperhidrosis, acne therapeutics, pigments for skin coloration for medical or cosmetic tattooing, sunscreen compounds, hormones, insulin, com/callous removers, wart removers, and generally any therapeutic or prophylactic agent for which transdermal delivery is desired. As noted above, the delivery may simply affect transport across the skin into a localized subdermal location, such as treatment of nail fungus or modulation of hair growth or may effect systemic delivery such as is desirable in some instances where vaccines are used.

[00153] In addition to the compositions and formulations of the invention per se, the methods may employ a subsequent treatment with linoleic acid. As transdermal treatments generally open up the skin barrier, which is, indeed, their purpose, it is useful to seal the area of application after the treatment is finished. Thus, treatment with the formulation may be followed by treating the skin area with a composition comprising linoleic acid to seal off the area of application. The application of linoleic acid is applicable to any transdermal procedure that results in impairing the ability of the skin to act as a protective layer. Indeed, most transdermal treatments have this effect as their function is to allow carbonates to pass through the epidermis to the dermis at least, and, if systemic administration is achieved, through the dermis itself.

[00154] For administration of anesthetics as the therapeutic agent, the local anesthetic may be one or more of the following: benzocaine, lidocaine, tetracaine, bupivacaine, cocaine, etidocaine, mepivacaine, pramoxine, prilocaine, procaine, chloroprocaine, oxyprocaine, proparacaine, ropivacaine, dyclonine, dibucaine, propoxycaine, chloroxylenol, cinchocaine, dexivacaine, diamocaine, hexylcaine, levobupivacaine, propoxycaine, pyrrocaine, risocaine, rodocaine, and pharmaceutically acceptable derivatives and bioisosteres thereof. Combinations of anesthetic agents may also be used. The anesthetic agent{s) are included in the composition in effective amount(s). Depending on the anesthetic(s) the amounts of anesthetic or combination is typically in the range of 1 % w/w to 50% w/w. The compositions of the invention provide rapid, penetrating relief that is long lasting. The pain to be treated can be either traumatic pain and/or inflammatory pain.

[00155] In one embodiment, the anesthetic is administered to relieve the pain associated with invasive fat deposit removal. Specific removal of fat deposits has been attractive for both health and cosmetic reasons. Among the methods employed are liposuction and injection of a cytolytic agent for fat such as deoxy cholic acid (DCA). For example, a series of patents issued or licensed to Kythera Biopharmaceuticals is directed to methods and compositions for non- surgical removal of localized fat that involves injecting compositions containing DCA or a salt thereof. Representative issued patents are directed to formulation (8,367,649); method-of-use (8,846,066; 7,622, 130; 7, 754,230; 8,298,556); and synthetic DCA (7,902,387).

[00156] In this aspect of the invention, conventional invasive fat removal techniques are employed along with administering a pain-relieving effective agent - typically lidocaine or related anesthetics via transdermal administration. In some embodiments, the pain-relieving transdermal formulation is applied to the area experiencing pain immediately before, during or immediately after the invasive fat-removal procedure.

[00157] Additional therapeutic agents may be included in the compositions. For example, hydrocortisone or hydrocortisone acetate may be included in an amount ranging from 0.25% w/w to about 0.5% w/w. Menthol, phenol, and terpenoids, e.g., camphor, can be incorporated for cooling pain relief. For example, menthol may be included in an amount ranging from about 0.1 % w/w to about 1.0% w/w.

[00158] The compositions containing anesthetics are useful for temporary relief of pain and itching associated with minor bums, cuts, scrapes, skin irritations, inflammation and rashes due to soaps, detergents or cosmetics, or, as noted above, pain associated with removal of fat deposits.

[00159] The benefits of alkaline pH include higher penetration capability and adjustment of the active form of the fat dissolving compound when the anesthetic is used in conjugation therewith. For example, the pKa of the deoxy cholic acid is 6.58 and the pH of fat is neutral. When deoxy cholic acid (DC A) is injected without buffering, it is approximately an equilibrium between the protonated and unprotonated forms. Utilizing formulations with high pH buffering shifts the balance significantly to unprotonated form making the DCA more water soluble and more likely to emulsify fats.

[00160] The formulations can be applied in a single, one-time application, once a week, once a bi-week, once a month, or from one to twelve times daily, for a period of time sufficient to alleviate a condition, disease, disorder, symptoms, for example, for a period of time of one week, from 1 to 12 weeks or more, from 1 to 6 weeks, from 2 to 12 weeks, from 2 to 12 weeks, from 2 to 8 weeks, from 2 to 6 weeks, from 2 to 4 weeks, from 4 to 12 weeks, from 4 to 8 weeks, or from 4 to 6 weeks. The present compositions can be administered, for example, at a frequency of once per day to hourly if needed. The presently described formulations can be topically administered once or more per day for a period of time from 1 week to 4 weeks, of from 1 week to 2 weeks, for 1 week, for 2 weeks, for 3 weeks, for 4 weeks, or for 4 weeks or more. In some instances, it may also be desirable to continue treatment indefinitely for example to inhibit or prevent a viral infection. A suitable administration for a formulation comprising a skin cream, lotion or ointment, for example is once, twice, three, four times daily, or hourly if needed.

[00161] The formulations provided herein can be applied in a therapeutically effective amount. Suitable amounts, for example, per application can include, for example, from about 1 gram to about 500 grams; from about 1 gram to about 10 grams; from about 10 grams to about 25 grams; from about 10 grams to about 50 grams; from about 10 grams to about 100 grams; from about 10 grams to about 200 grams; from about 10 grams to about 350 grams; from about 10 grams to about 500 grams; from about 20 grams to about 500 grams; from about 20 grams to about 350 grams; from about 20 grams to about 200 grams; from about 20 grams to about 100 grams; from about 20 grams to about 90 grams; from about 20 grams to about 80 grams; from about 20 grams to about 70 grams; from about 20 grams to about 60 grams; from about 20 grams to about 50 grams; from about 30 grams to about 100 grams; from about 30 grams to about 80 grams; from about 30 grams to about 70 grams; or from about 30 grams to about 60 grams. Alternatively, suitable amounts, for example, per application can include, for example, at least 5 grams; at least 10 grams; at least 15 grams; at least 20 grams; at least 25 grams; at least 30 grams; at least 35 grams; at least 40 grams; at least 50 grams; at least 55 grams; at least 60 grams; at least 65 grams; at least 70 grams; at least 75 grams; at least 80 grams; at least 85 grams; at least 90 grams; at least 100 grams; or more.

[00162] If desired, other therapeutic agents can be employed in conjunction with those provided in the above-described compositions. The amount of active ingredients that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated, the nature of the disease, disorder, or condition, and the nature of the active ingredients.

[00163] It is understood that a specific dose level for any particular patient will vary depending upon a variety of factors, including the activity of the specific active agent; the age, body weight, general health, sex and diet of the patient; the time of administration; the rate of excretion; possible drug combinations; the severity of the particular condition being treated; the area to be treated and the form of administration. One of ordinary skill in the art would appreciate the variability of such factors and would be able to establish specific dose levels using no more than routine experimentation.

[00164] Pharmacokinetic parameters such as bioavailability, absorption rate constant, apparent volume of distribution, unbound fraction, total clearance, fraction excreted unchanged, first-pass metabolism, elimination rate constant, half-life, and mean residence time can be determined by methods well known in the art.

[00165] A formulation in accordance with the subject maher described herein may be a topical dosage form packaged in, for example, a multi-use or single-use package, including for example, a tube, a tohle, a pump, a container or bohle, a vial, a j ar, a packet, or a blister package.

[00166] Single dosage kits and packages containing a once per day amount of the topical formulation may be prepared. Single dose, unit dose, and once-daily disposable containers of the topical formulation are also provided. [00167] The present topical formulation remains stable in storage for periods including up to about 5 years, between about 3 months and about 5 years, between about 3 months and about 4 years, between about 3 months and about 3 years, and alternately any time period between about 6 months and about 3 years.

[00168] A topical formulation described herein remains stable for up to at least 3 years at a temperature of less than or equal to 40° C. In an embodiment, the presently described topical formulation remains stable for at least 2 years at a temperature of less than or equal to 40° C. In an embodiment, the presently described formulation or emulsion remains stable for at least 3 years at a temperature of less than or equal to 40° C and at a humidity of up to 75% RH, for at least 2 years at a temperature of less than or equal to 40° C and at a humidity of up to 75% RH, or for at least 3 years at a temperature of less than or equal to 30°C. and at a humidity of up to 75% RH. In a further embodiment, the presently described biocompatible composition in accordance with the subject matter described herein remains stable for an extended period of time when packaged in a multi-use container such as a bottle dispenser or the like and exhibits equal to or even greater stability when packaged in a single-use package.

[00169] In another aspect, the pharmaceutical composition of certain embodiments comprises a daily dose of a pH modulating composition or buffer (e.g., sodium bicarbonate as a topical formulation). A daily dose for topical or transdermal administration of any given pH modulating compound depends on the compound and animal and may be easily determined by the skilled artisan, a suitable amount is about 1 mg/kg to about 5g/kg, and more typically the daily dose is about lOmg/kg to about 5g/kg, about 25mg/kg to about 2000 mg/kg, about 50mg/kg to about 2000 mg/kg, about 25mg/kg to about lOOOmg/kg, about 50mg/kg to about lOOOmg/kg, about lOOmg/kg to about 700mg/kg, about lOOmg/kg to about 500mg/kg, about 150mg/kg to about 500mg/kg, about 150mg/kg to about 400mg/kg, about 200mg/kg to about 500mg/kg, about 200mg/kg to about 450mg/kg, about 200mg/kg to about 400mg/kg, about 250mg/kg to about 450mg/kg, about 250mg/kg to about 400mg/kg, about 250mg/kg to about 350mg/kg, and about 275mg/kg to about 325 mg/kg.

[00170] Alternatively, a suitable daily dose for topical or transdermal administration of a pH modulating composition or buffer (e.g. sodium bicarbonate) is at least about lmg/kg, at least about lOmg/kg, at least about 25mg/kg, at least about 30mg/kg, at least about 35mg/kg, at least about 40mg/kg, at least about 41mg/kg, at least about 42mg/kg, at least about 43mg/kg, at least about 44mg/kg, at least about 45mg/kg, at least about 46mg/kg, at least about 47mg/kg, at least about 48mg/kg, at least about 49mg/kg, at least about 50mg/kg, at least about 55mg/kg, at least about 60mg/kg, at least about 65mg/kg, at least about 70mg/kg, at least about 75mg/kg, at least about 80mg/kg, at least about 90mg/kg, at least about lOOmg/kg, at least about 125mg/kg, at least about 150mg/kg, at least about 160mg/kg, at least about 170mg/kg, at least about 175mg/kg, at least about 180mg/kg, at least about 190mg/kg, at least about 200mg/kg, at least about 225mg/kg, at least about 250mg/kg, at least about 275mg/kg, at least about 300mg/kg, at least about 325mg/kg, at least about 350mg/kg, at least about 375mg/kg, at least about 400mg/kg, at least about 425mg/kg, at least about 450mg/kg, at least about 475mg/kg, at least about 500mg/kg, at least about 550mg/kg, at least about 600mg/kg, at least about 700mg/kg, at least about 800mg/kg, at least about 900mg/kg, at least about lg/kg, at least about 2g/kg, at least about 3g/kg, or at least about 5g/kg.

[00171] Alternatively, a suitable dose for topical or transdermal administration of a pH modulating formulation or buffer (e.g. sodium bicarbonate) for subject (e.g. a human patient) is at least about lOOmg, at least about 500mg, at least about lg, at least about 5g, at least about lOg, at least about 15g, at least about 16g, at least about 17g, at least about 18g, at least about

19g, at least about 20g, at least about 21g, at least about 22g, at least about 23g, at least about

24g, at least about 25g, at least about 26g, at least about 27g, at least about 28g, at least about

29g, at least about 30g, at least about 35g, at least about 40g, at least about 45g, at least about

50g, at least about 60g, at least about 75g, at least about lOOg, at least about 200g, at least about 500g, or at least about 1.0kg. This does may be administered daily, twice a day, three times a day, four times a day, five times a day, or more than five times a day.

[00172] In another aspect, in certain embodiments a pH modulating composition or buffer (e.g. sodium bicarbonate) is administered topically or transdermally such that the dose results in a subject intake of at least about 0.1 nmol/hr/Kg, at least about 0.5 nmol/hr/Kg, at least about 0.7 nmol/hr/Kg, at least about 1.0 nmol/hr/Kg, at least about 1.1 nmol/hr/Kg, at least about 1.2 nmol/hr/Kg, at least about 1.3 nmol/hr/Kg, at least about 1.4 nmol/hr/Kg, at least about 1.5 nmol/hr/Kg, at least about 1.6 nmol/hr/Kg, at least about 1.7 nmol/hr/Kg, at least about 1.8 nmol/hr/Kg, at least about 1.9 nmol/hr/Kg, at least about 2.0 nmol/hr/Kg, at least about 2.5 nmol/hr/Kg, at least about 3.0 nmol/hr/Kg, at least about 3.5nmol/hr/Kg, at least about 4.0 nmol/hr/Kg, at least about 5 nmol/hr/Kg, at least about 10 nmol/hr/Kg, at least about 25 nmol/hr/Kg, at least about 50 nmol/hr/Kg, at least about 100 nmol/hr/Kg, at least about 500 nmol/hr/Kg, or at least about 1 pmol/hr/Kg, [00173] In another aspect, in certain embodiments a pH modulating composition or buffer (e.g. sodium bicarbonate) is administered topically or transdermally such that the dose results in a peak plasma concentration of a buffering or pH modulating compound ranges from about 1 μg/ml to 50 μg/ml, about 5 μg/ml to about 45 μg/ml, about 5 μg/ml to about 40 μg/ml, about 5 μg/ml to about 35 μg/ml, about 5 μg/ml to about 30 μg/ml, about 5 μg/ml to about 25 μg/ml, about 1 μg/ml to about 45 μg/ml, about 1 μg/ml to about 40 μg/ml, about 1 μg/ml to about 35 μg/ml, about 1 μg/ml to about 30 μg/ml, about 1 μg/ml to about 25 μg/ml, about 1 μg/ml to about 20 μg/ml, about 1 μg/ml to about 15 μg/ml, about 1 μg/ml to about 10 μg/ml, about 1 μg/ml to about 9 μg/ml, about 1 μg/ml to about 8 μg/ml, about 1 μg/ml to about 7 μg/ml, about 1 μg/ml to about 6 μg/ml, and about 1 μg/ml to about 5 μg/ml.

[00174] In another aspect, in certain embodiments a pH modulating composition or buffer (e.g. sodium bicarbonate) is administered topically or transdermally so that plasma concentration ranges from about 1 ng/ml to 500 μg/ml, about 10 ng/ml to 500 μg/ml, about 100 ng/ml to 500 μg/ml, about 1 μg/ml to 500 μg/ml, about 10 μg/ml to 500 μg/ml, about 25 μg/ml to 500 μg/ml, about 25 μg/ml to about 450 μg/ml, about 25 μg/ml to about 400 μg/ml, about 25 μg/ml to about 350 μg/ml, about 25 μg/ml to about 300 μg/ml, about 25 μg/ml to about 250 μg/ml, about 50 μg/ml to about 500 μg/ml, about 55 μg/ml to about 500 μg/ml, about 60 μg/ml to about 500 μg/ml, about 65 μg/ml to about 500 μg/ml, about 70 μg/ml to about 500 μg/ml, about 75 μg/ml to about 500 μg/ml, about 80 μg/ml to about 500 μg/ml, about 85 μg/ml to about 500 μg/ml, about 90 μg/ml to about 500 μg/ml, about 95 μg/ml to about 500 μg/ml, about 100 μg/ml to about 500 μg/ml, about 110 μg/ml to about 500 μg/ml, about 120 μg/ml to about 500 μg/ml, about 130 μg/ml to about 500 μg/ml, about 140 μg/ml to about 500 μg/ml about 150 μg/ml to about 500 μg/ml, about 160 μg/ml to about 500 μg/ml, about 170 μg/ml to about 500 μg/ml, about 180 μg/ml to about 500 μg/ml, about 200 μg/ml to about 500 μg/ml, about 200 μg/ml to about 490 μg/ml, about 200 μg/ml to about 480 μg/ml, about 200 μg/ml to about 470 μg/ml, about 200 μg/ml to about 460 μg/ml, about 200 μg/ml to about 450 μg/ml, about 200 μg/ml to about 440 μg/ml, about 200 μg/ml to about 430 μg/ml, or about 200 μg/ml to about 400 μg/ml.

[00175] In further embodiments, a pH modulating composition or buffer (e.g. sodium bicarbonate) is administered topically or transdermally so that plasma concentration is at least 10 ng/ml, at least 25 ng/ml, at least 50 ng/ml, at least 100 ng/ml, at least 250 ng/ml, at least 0.5 μg/ml, at least 0.75 μg/ml, at least 1 μg/ml, at least 2 μg/ml, at least 3 μg/ml, at least 4 μg/ml, at least 5 μg/ml, at least 6 μg/ml, at least 7 μg/ml, at least 8 μg/ml, at least 9 μg/ml, at least 10 mg/ml, at least 15 μg/ml, at least 20 μg/ml. at least 25 μg/ml. at least 30 μg/ml. at least 35 μg/ml. at least 40 μg/ml. at least 45 μg/ml. at least 50 μg/ml. at least 55 μg/ml. at least 60 μg/ml. at least 65 μg/ml. at least 70 μg/ml. at least 75 μg/ml. at least 80 μg/ml. at least 85 μg/ml. at least 90 μg/ml. at least 95 μg/ml. at least 100 μg/ml or more than 100 μg/ml.

[00176] In another aspect, a pH modulating compound or buffer (e.g., sodium bicarbonate) is administered topically or transdermally so that peak plasma concentration is reached in lOmin, 15min, 20min, 30min, 45min, 60min, 75min, 90min, 2hr, 3hr, 4hr, 5hr, 6 hr, 7hr, 8hr, lOhr, 12hr or 24hr after administration.

[00177] Aspects of the present specification disclose that the symptoms associated with a disease or disorder described herein are reduced by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% and the severity associated with a disease or disorder described herein is reduced by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. Aspects of the present specification disclose the symptoms associated with disease or disorder are reduced by about 10% to about 100%, about 20% to about 100%, about 30% to about 100%, about 40% to about 100%, about 50% to about 100%, about 60% to about 100%, about 70% to about 100%, about 80% to about 100%, about 10% to about 90%, about 20% to about 90%, about 30% to about 90%, about 40% to about 90%, about 50% to about 90%, about 60% to about 90%, about 70% to about 90%, about 10% to about 80%, about 20% to about 80%, about 30% to about 80%, about 40% to about 80%, about 50% to about 80%, or about 60% to about 80%, about 10% to about 70%, about 20% to about 70%, about 30% to about 70%, about 40% to about 70%, or about 50% to about 70%.

[00178] The formulations as described herein can be used in the manufacture of medicaments and for the treatment of humans and other animals by administration in accordance with conventional procedures.

EXPERIMENTAL EXAMPLES

[00179] The compositions and methods described herein will be further understood by reference to the following examples, which are intended to be purely exemplary. The compositions and methods described herein are not limited in scope by the exemplified embodiments, which are intended as illustrations of single aspects only. Any methods that are functionally equivalent are within the scope of the invention. Various modifications of the compositions and methods described herein in addition to those expressly described herein will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications fall within the scope of the invention.

[00180] The following examples are intended to illustrate but not to limit the invention.

Example 1 - Specific Formulations for Administration of Buffering Agents

[00181] The following compositions have been prepared and are found useful in the methods of the invention. In the tables below, “LIP” represents lecithin organogel comprised of a 1 : 1 molar mixture of soy lecithin containing 96% phosphatidyl choline and isopropyl palmitate; “BA” represents benzyl alcohol; PLU-F127 represents the detergent poloxamer F127 granules; PLU-Water represents PLU-F127 dissolved in deionized water. (Alternatively, commercially available Pluronic F12730% gel could be used) and Durasoft® is a commercially available form of emulsifier.

[00182] In the tables below, sodium bicarbonate and sodium carbonate were supplied as such. Tris buffer at pH 8.1 was used. The phosphate “buffer” was supplied as Na H2 P04. The percentages are wt/wt i.e., weight percentages.

[00183] Table 1 - Bicarbonate Formulations [00184] Table2-CarbonateFormulations

[00185] Table3- Tris Formulations

[00186] Table4-PhosphateFormulations [00187] Table 5 -Carbonate, TRIS, Phosphate Combination Formulations

[00188] Table 6 - Alternative Buffers [00189] Table7-AnyhrousFormulations

Example 2 - Topical Administration of Bicarbonate

[00190] 24 NCR nude 5 week old male mice were used in this study, divided into four groups of six mice each. Topical compositions were applied to the back of each mouse from hip to shoulder three times per day for 8 successive days for a total of 24 applications. A control group was administered sodium bicarbonate in water by mouth. The groups are as follows:

[00191] The transdermai formulations comprise penetrants to result in the formulations as follows: LIP-30.0%, EtOH-1.5%, BA-1.0%, menthol-0.5%, sodium bicarbonate-33.5%, PLU-

F127-10.1%, PLU-water-23.5%; i.e.. Formulation IB in Table 1. The concentration of sodium bicarbonate in the control group was 200 mM and the consumption was ad libidum. The concentration of sodium bicarbonate in the transdermai formulations was 33.5% wt/wt.

[00192] Urine samples were collected at one hour, three hours and six hours after the first drug application and stored at 4°C for subsequent pH determination. On days 2-12 urine was collected twice daily — prior to the first application and 15 minutes after the last application. The mice were sacrificed one hour after the last drug application on day 8 and the back skin was harvested and placed on bibulous paper.

[00193] To set a base line for pH without dosing, prior to beginning the protocol, urinary pH was measured at three time points during a single day in seven mice at 0900, five mice at 1300 and four mice at 1630. The pH of the urine had an overall mean value of 5.57 which did not vary over this time period.

[00194] As shown in Figure 1, all of the groups showed an increase in urine pH over the first six hours of treatment. The most significant increase occurred in Group 3 which was administered 220 microliters of the formulation.

[00195] Although the study was designed to be conducted for two weeks (Mon-Fri), it was terminated after day eight because the members of Group 2 (1,110 microliter) developed skin irritation; this was shown even in the low dose group receiving 30 microliters, i.e., Group 2.

[00196] Figure 2 shows the urine values of pH over the course of the eight day study. Although there was some variation, the group receiving the highest dosage (Group 4) was able to maintain a high pH over the course of the study.

[00197] The study was repeated using the formulations 25, 28 and 29 in Table 6 with the results shown in Table 8 and using the formulations in Table 7 with results shown in

[00198] Figure 3.

[00199] Table 8 - Mean urine pH at two collection time points on day one and overall.

[00200] As shown, transdermal administration was more effective than oral administration.

Example 3 -Transdermal Absorption in Humans

[00201] Healthy human Subjects Aged 18-60 were enrolled in a double-blinded, placebo controlled, randomized and cross-over in designed study. The subjects applied 0.6g/kg of body weight of formulation of Table 9, per randomization group, as follows: legs from ankle to top of thighs; arms from wrist to shoulder (including the deltoids) or drank 0.13 ounces of water per kg body weight (equates to about 8 ounces for a 140 pound subject) at 15 min, 1 hr 15 min, 2 hours and 15 min, 3 hours and 15 mins to control for dilution of urine. [00202] Table 9

[00203] At hourly intervals (at 1, 2, 3, and 4 hours) from start time of application, subjects collected 10-20 ml (approximately a Tablespoon) of urine and the pH was determined. The results are shown in Table 10.

[00204] Table 10 - Average Urine pH values for each treatment and time point

[00205] No adverse effects were shown and the transdermal formulation out-performed oral administration.

Example 4 - Administration of Bicarbonate Topically Using Selected Formulations

[00206] An objective of this series of studies was to compare different transdermal compounds to determine which compound, dose, concentration combinations induce the highest urine with the lowest levels of skin irritation. Presented below are data and results from several experiments that determine and compare various formulations in terms of effectiveness for treatment and side effects, and in particular tolerance from skin irritation.

[00207] In the first study, twenty-four 6 week old female SCID mice were used in this study, divided into four groups of six mice each. The backs of mice were treated with hair removal compound before topical formulations were applied. Topical formulations were then applied to the back of each mouse from hip to shoulder three times per day at a dose of 50pl, totaling 150m1 for 3 consecutive days. Urine samples were collected twice daily, one in the morning and one in the afternoon and stored at 4°C for subsequent pH determination. The transdermal formulations from Study 1 comprise formulations as follows: Group A, 15.8% sodium bicarbonate in water; Group B, 29% lysine in water; Group C, 9.3% Sodium Phosphate in water; Group D, 23.4% Tris in water (see Table 15). Pre-dosing urine pH from 15 mice was 5.97± 0.06 (mean± SEM). The transdermal formulations from cohort Groups A-D are shown in Table 15.

[00208] Table 11 - Buffer Formulations T ested

[00209] In all cohort groups the formulations absorbed quickly and easily. Formula A caused severe skin irritation after the first day and scabbing by the end of the third day in the Group A cohort mice (not shown). However, formulation A is the only one tested in Table 15 that significantly increased urine pH after the second day of dosing, and cyclic dinural variation was evident by day 2 (see Fig. 6).

[00210] In a second study, a variation of the above study was performed with the exclusion of Formula A in which the mice received larger doses to test the effect of larger doses (100 pl/dose, 3 times a day). The total dose/day in these cohort mice was 300 mΐ/day. Formula A was excluded from further testing because, while it was effective in increasing urine pH, it caused sever skin irritation and intolerance in mice. The transdermal formulations from Study 2 included the following formulations: Group B, 29% lysine in water; Group C, 9.3% Sodium Phosphate in water; Group D, 23.4% Tris in water (see Table 16).

[00211] Table 12 - Buffer Formulations Tested

[00212] Pre-dosing urine pH from 10 mice was 5.58± 0.04 (mean± SEM). The transdermal agent absorbed quickly and easily in all cohort groups (A-D). No skin irritation observed in any of the cohorts. As illustrated in Fig. 7, the results show that compounds C and D increased urine pH at various intervals. Urine collected for the 2nd time on day 1 was highest in pH in mice treated with compound C (Fig 7). Urine collected for the 2nd time on day 2 was highest in pH in mice treated with compound D (Fig. 7). [00213] In the third study, a variation of study II above was then performed with a higher total dose/day to test the effect of larger doses (100 μl/dose, 3 times a day). The total does/day in these cohort mice was 300 mΐ/day. Formula A was excluded from further testing because, while it was effective in increasing urine pH, it caused sever skin irritation and intolerance in mice. The transdermal formulations from Study 2 comprised formulations as follows: Group B, 29% lysine in water; Group C, 9.3% Sodium Phosphate in water; Group D, 23.4% Tris in water (see Table 16). Pre-dosing urine from 10 mice was 5.58± 0.04 (mean± SEM). The transdermal agent absorbed quickly and easily in all cohort groups (B,C,D). There was no skin irritation observed in any of the cohorts. The results are shown in Fig. 7, where it can be seen that compounds C and D increased urine pH at various intervals. Urine was collected for the 2nd time on day 1 was highest in pH in mice treated with compound C (Fig 7). Urine collected for the 2nd time on day 2 was highest in pH in mice treated with compound D (Fig. 7).

[00214] Table 13 - Buffer Formulations Tested

[00215] In a fourth study, a variation of the second study above was preformed where mice received a total dose of 300μl/day of the formulations shown in Table 17. Pre-dosing urine levels from 4 mice was 5.73± 0.02 (mean± SEM). As shown in Fig. 8, all compounds were able to increase urine pH significantly. Urine pH increases were observed after the 2nd collection of urine towards the end of the study day. Compound F (7.5% Sodium Carbonate with modified penetration enhancers) consistently induced the highest urine pH (Fig. 8).

[00216] In a fifth study, compound F (7.5% Sodium Carbonate with selected penetration enhancers) were tested in three doses of cohorts as follows: 150μL/day, 200μL/day, and 300μL/day. One group was treated with 200mM sodium bicarbonate drinking water as a positive control. The formulations used in this study are shown in Table 18. [00217] Table 14 - Buffer Formulations Tested

[00218] Pre-dosing urine from 6 mice was 5.6± 0.03 (mean± SEM). The pH measurement results are presented in Fig.8, which also shows that the response of compound F at a dose of 150 μL/day was similar to that at 300 μL/day (Fig. 8)

[00219] In another study, three doses of compound F (7.5% Sodium Carbonate with selected penetration enhancers) were test in three of the cohorts 30μL/day, 75μL/day, and 150μL/day. One group was treated with 200mM sodium bicarbonate drinking water as a positive control. The formulations tested are shown in Table 19.

[00220] Table 15 - Buffer Formulations Tested

[00221] The pre-dosing urine pH from 3 mice was 5.57 ± 0.06 (mean± SEM). As shown in Fig.9, urine pH values trended lower in all groups in this round of studies. Higher urine pH levels were observed in the 150 μL/day cohort and was most similar to the sodium bicarbonate water positive control group (Fig. 9).

[00222] In another study, three doses of compound F (7.5% Sodium Carbonate) was tested in three of the cohorts at doses of 100μL/day, 125μL/day, and 150μL/day. One group was treated with 200mM sodium bicarbonate drinking water as a positive control. The formulations tested are shown in Table 20 and results of the dose response curve are shown in Fig. 10. With reference to the selected penetration enhancers referenced in the formulations of Table 19 and 20, these were 0.11% diethanolamine, 1.94% sodium caprate, and 0.20% sodium lauryl sulfate. EDTA in an amount of 0.30% was also added to the formulations of Table 19 and 20. Other penetration enhancers as well as different amount of penetration enhancers are suitable in other embodiments.

[00223] Table 16 - Buffer Formulations Tested

[00224] Table 17 - Formulation Compositions Study

[00225] Table 18 - Formulation Compositions Study

[00226] The data and results from the studies reported in Example 4 can be summarized as follows. A formulation of 150 μL/day of 29.0% Lysine HCL, 9.3% Trisodium Phosphate, and 23.4% TRIS did not significantly raise urine pH. Doubling the doses (300 μL/day) of 9.3%

Trisodium Phosphate and 23.4% TRIS did not significantly raise urine pH. Doubling dose (300 μL/day) of 29.0% Lysine had no impact on urine pH. All formulations of sodium carbonate tested consistently increases urine pH. A formulation of 15.8% sodium carbonate is highly effective in raising urine pH, but caused skin irritation marked by redness and scabbing within 3 days. A formulation of 7.5% sodium carbonate (with selected penetration enhancers) achieved similar outcomes as 15.8%, but with no skin irritation. A formulation of 7.5% sodium carbonate (with modified penetration enhancers) appeared to have a greater impact on increasing urine pH than 7.5% sodium carbonate. Changes in urine pH induced by formulations comprising 7.5% sodium carbonate (with selected penetration enhancers) appears to be dose dependent. Doses of 7.5% sodium carbonate (with selected penetration enhancers) higher than 150 μL/day were not any more effective in increasing urine pH. The dose of 150 μL/day of 7.5% sodium carbonate (with selected penetration enhancers described herein) was more optimal in raising urine pH than lower doses. Urine pH increased after application during the day, then returned to baseline levels overnight.

Example 5 - CPE Formulation

[00227] This is an example of an integrative cooperative CPE formulation directed to the extra-cellular matrix to which might be added selected cysteine cathepsin protease-inhibitors, with or without a suitable buffering agent to NHE1 isoform inhibitor as a synergistic composition.

[00228] 1. Cetyltrimethyl ammonium bromide (from about 2.0% to about 10.0 %)

[00229] 2. Sodium cholate : Lecithin (96% pure ): Isopropyl myristate (equi-molar 1:1:1

(from about 10% to about 40.0 %)

[00230] 3. Sodium citrate (titrate to transparency/incr. viscosity of #2.)

[00231] 4. Benzyl alcohol (from about 2.0% to about 30.0 %)

[00232] 5. Cis-Palmitoleic acid (from about 20.0% to about 30% of BA)

[00233] 6. Methyl pyrrolidone (0.4%) /Dodecyl pyridinium (1.1%) (from about 0.5% to about 5.0%)

[00234] 7. Pluronic 127 (qs to 100%)

Example 6 - Penetration Enhancing Formulation

[00235] This is an example of the formulation, which is directed to the cellular component of the SC permeability barrier to which might be added selected cysteine cathepsin protease- inhibitors, with or without a suitable buffering agent, and NHE1 isoform inhibitor as a synergistic composition.

[00236] 1. ACSSSPSKHCG, [ alanine-cysteine-serine-serine-serine-proline-serine-lysine- hisitidine-cysteine-glycine ] identified as TD-1

[00237] 2. Thioglycolic Acid (TGA) (from about 2.0 % to about 7.0 % concentration)

[may be substituted by other reducing agents]

[00238] 3. Proteinase K (from about 5mg/mL to about 15 mg/mL)

Example 9 - Use of Topical Buffering Agents to Increase Effectiveness of Weak Base Chemotherapeutics without Adverse Events

[00239] In this experiment, doxorubicin in conjunction with a formulations of the invention are tested for their ability to enhance the cytotoxicity without adverse events observed with buffering therapies are administered orally.

[00240] In vitro test were first performed and demonstrated significant increases in drug uptake and cytotoxicity. Experiments were performed using MCF-7 cells grown to log phase in 96-well plates and medium was exchanged for one at either pH 6.8 or 7.4 containing 0.208 μCi per well of 14C doxorubicin. Medium pH was buffered using non-volatile buffers (10 mM MES, 20 mM HEPES and 10 mM TRICINE) in combination with bicarbonate concentrations that were adjusted to be in equilibrium with 5% ambient carbon dioxide. Twenty-four hours later, regular growth medium was replaced, and cells allowed to grow a subsequent 72 h, after which time they were fixed and stained with crystal violet for determination of cell number. In-vitro, raising the pHe from 6.8 to 7.4 resulted in a 2.25-fold enhancement of cytotoxicity and a 2.56-fold increase in intracellular doxorubicin concentrations.

[00241] In vivo tests were performed and doxorubicin in conjunction with buffering formulations of the invention demonstrated a significantly lowered tumor growth rate compared to treatment with doxorubicin alone and without adverse events observed when buffering therapies were administered orally. In this experiment MCF-7 tumors were grown in the mammary fat pads of 6-week-old female SCID mice, to sizes of 50-200 mm3, and were forcibly randomized according to tumor size into six groups:

Group A and B: Control

Group C and D: 200mM sodium bicarbonate drinking water ad libitum

Group E and F: 50μL x 3 doses approximately Q8 hours (total daily dose of 150iiL) of formulation in Table 21 below as follows:

[00242] The start of the bicarbonate treatment was designated as day 1. On days 3, 7 and 11 , animals in groups B, D, and F were injected i.p. with 1.6 mg kg-1 doxorubicin while animals in groups A, C, and E (n = 10 each) received saline injections of the same volume, as per established protocols. On day 15, animals in Groups C and D were placed back on normal drinking water and animals in Groups E and F ceased received t.i.d. topical applications.

[00243] Tumor volumes and animal weights were monitored every 2 days. Doxorubicin had a significant effect on the tumor growth rate (Groups B growth rate decreased -30% over assessment period). This effect was greater in animals co-treated with bicarbonate whether by oral or topical administration (With both Groups D and F observing a 55% reduction in growth rate). In this embodiment, bicarbonate alone (Groups C and E) had no significant effect on the growth rate alone. Other characteristics of tumors that increase its potential for carcinogenesis other than growth rates are measured by other parameters as well, including inhibition metastasis. In the inventor’s experience, different embodiments disclosed herein may have differential effects on different characteristics of tumors (e.g., growth rate, volume, metastasis, pH microenvironment, etc.).

[00244] In addition to tumor volumes, animals in group D experienced anal discharge suggesting increased GI tract cytotoxicity of doxorubicin when orally dosed. This adverse event was not noted in topically dosed animals that received doxorubicin (Group F).

[00245] Certain embodiments of the present invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the present invention to be practiced otherwise than 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 embodiments in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

[00246] Groupings of alternative embodiments, elements, or steps of the present invention are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other group members disclosed herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.

[00247] Unless otherwise indicated, all numbers expressing a characteristic, item, quantity, parameter, property, term, and so forth used in the present specification and claims are to be understood as being modified in all instances by the term “about.” As used herein, the term “about” means that the characteristic, item, quantity, parameter, property, or term so qualified encompasses a range of plus or minus ten percent above and below the value of the stated characteristic, item, quantity, parameter, property, or term. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical indication should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and values setting forth the broad scope of the invention are approximations, the numerical ranges and values set forth in the specific examples are reported as precisely as possible. Any numerical range or value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Recitation of numerical ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate numerical value falling within the range. Unless otherwise indicated herein, each individual value of a numerical range is incorporated into the present specification as if it were individually recited herein.

[00248] The terms “a,” “an,” “the” and similar referents used in the context of describing the present 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. 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 present invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the present specification should be construed as indicating any non-claimed element essential to the practice of the invention.

[00249] Specific embodiments disclosed herein may be further limited in the claims using consisting of or consisting essentially of language. When used in the claims, whether as filed or added per amendment, the transition term “consisting of’ excludes any element, step, or ingredient not specified in the claims. The transition term “consisting essentially of’ limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristic(s). Embodiments of the present invention so claimed are inherently or expressly described and enabled herein.

[00250] All patents, patent publications, and other publications referenced and identified in the present specification are individually and expressly incorporated herein by reference in their entirety for the purpose of describing and disclosing, for example, the compositions and methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.