CN114302647A - Otoprotectant methods against platinum-based antineoplastic agents - Google Patents

Otoprotectant methods against platinum-based antineoplastic agents Download PDF

Info

Publication number
CN114302647A
CN114302647A CN202080043662.0A CN202080043662A CN114302647A CN 114302647 A CN114302647 A CN 114302647A CN 202080043662 A CN202080043662 A CN 202080043662A CN 114302647 A CN114302647 A CN 114302647A
Authority
CN
China
Prior art keywords
thiosulfate
platinum
hours
administered
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202080043662.0A
Other languages
Chinese (zh)
Inventor
J·李
J·R·索利亚
Q·胡
F·施
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Decibel Therapeutics Inc
Original Assignee
Decibel Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/427,567 external-priority patent/US10813947B1/en
Application filed by Decibel Therapeutics Inc filed Critical Decibel Therapeutics Inc
Publication of CN114302647A publication Critical patent/CN114302647A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/04Sulfur, selenium or tellurium; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0046Ear
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Abstract

Disclosed herein are methods of otoprotection against platinum-based anti-tumor agents by administering thiosulfate to a subject in need thereof. Typically, the thiosulfate salt is administered to the subject who is scheduled to administer a platinum-based anti-neoplastic agent within 4 hours. Alternatively, the thiosulfate salt is administered within 7 hours after administration of the platinum-based neoplastic agent.

Description

Otoprotectant methods against platinum-based antineoplastic agents
Technical Field
The present invention provides methods of otoprotection against platinum-based antineoplastic agents.
Background
Platinum-based antineoplastic agents (e.g., cisplatin) are chemotherapeutic agents that are widely used in the treatment of cancers and tumors. These agents are toxic and are known to induce hearing loss in both human and animal models. Thus, patients undergoing chemotherapy with platinum-based antineoplastic agents may suffer from hearing loss. Otoprotectant compositions and methods are needed to prevent or reduce hearing loss associated with chemotherapy regimens, including platinum-based anti-neoplastic agents.
Disclosure of Invention
In general, the present invention provides methods for reducing platinum-induced ototoxicity in a subject in need thereof. The method involves administering to the subject an effective amount of thiosulfate.
In some embodiments, the subject is administered the platinum-based antineoplastic agent no more than 7 hours prior to administration of the thiosulfate salt, or is scheduled to be administered within 4 hours. In certain embodiments, the subject is administered the platinum-based oncology agent no more than 7 hours prior to administration of the thiosulfate salt. In a particular embodiment, the subject is scheduled to administer a platinum-based anti-neoplastic agent at 4.5 hours. In further embodiments, the subject is administered the platinum-based oncology agent no more than 2.5 hours prior to administration of the thiosulfate salt. In additional embodiments, the subject is administered the platinum-based oncology agent no more than 1 hour prior to administration of the thiosulfate salt.
In some embodiments, administration of an effective amount of thiosulfate to a subject produces plasma thiosulfate C that is 30 μ M or less at the time of administration of a platinum-based anti-neoplastic agentmax. In certain embodiments, an effective amount of thiosulfate-produced cochlear thiosulfate C is administered to a subjectmaxCochlea C that is a platinum-based anti-tumor agentmaxAt least 30 times larger. Cochlear platinum concentration and cochlear CmaxIt is usually modeled by pharmacokinetic simulations of intravenous infusions in a two-compartment model. For example, pharmacokinetic simulations can be performed using WinNonlin (Phoenix64) PK simulation model 9 (intravenous infusion, 2-compartment).
In further embodiments, the thiosulfate salt is administered otically. In certain embodiments, the thiosulfate salt is administered intratympanically, transtympanically, or by injection in the inner ear. In particular embodiments, the thiosulfate salt is administered via the tympanic cavity or by injection in the inner ear.
In some embodiments, the method further comprises administering a platinum-based anti-neoplastic agent.
In certain embodiments, the thiosulfate salt is an alkaline thiosulfate salt, an ammonium thiosulfate salt, or a solvate thereof. In a further embodiment, the effective amount of thiosulfate is administered as a hypertonic pharmaceutical composition comprising an effective amount of thiosulfate. In further embodiments, 200-.
In further embodiments, the calculated osmolarity of the hypertonic pharmaceutical composition is 500-5,000mOsm/L (e.g., 600-5,000mOsm/L, 700-5,000mOsm/L, 800-5,000mOsm/L, 900-5,000mOsm/L, 1,000-5,000mOsm/L, 1,500-5,000mOsm/L, 2,000-5,000mOsm/L, 2,500-5,000mOsm/L, 3,000-5,000mOsm/L, 500-4,000mOsm/L, 600-4,000mOsm/L, 700-4,000mOsm/L, 800-4,000mOsm/L, 900-4,000mOsm/L, 1,000mOsm/L, 500-4,000mOsm/L, 500-2,000mOsm/L, 2,000-2,000 mOsm/L, 500-4,000mOsm/L, 2,000-2-4,000-500-4,000 mOsm/L, 2-4,000-one, 600-rich 3,000mOsm/L, 700-rich 3,000mOsm/L, 800-rich 3,000mOsm/L, 900-rich 3,000mOsm/L, 1,000-rich 3,000mOsm/L, 1,500-rich 3,000mOsm/L, 2,000-rich 3,000mOsm/L, 2,500-rich 3,000mOsm/L, 500-rich 2,500mOsm/L, 600-rich 2,500mOsm/L, 700-rich 2,500mOsm/L, 800-rich 2,500mOsm/L, 900-rich 2,500mOsm/L, 1,000-rich 2,500mOsm/L, 1,500-rich 2,500mOsm/L, 2,000-rich 2,500mOsm/L, 500-rich 2,000mOsm/L, 500mOsm/L, 1,000-rich 2,000mOsm/L, 1,000-rich 2,000-rich 2,500mOsm/L, 500mOsm/L, 1,000-rich 2,000-rich 2,500mOsm/L, 500mOsm/L, 1,000-rich 2,000-Osm/L, 2,000-rich 2,000-Osm/L, 500-Osm/L, 2,000-rich, 600-type 1,500mOsm/L, 700-type 1,500mOsm/L, 800-type 1,500mOsm/L, 900-type 1,500mOsm/L, or 1,000-type 1,500 mOsm/L).
In some embodiments, the concentration of thiosulfate in the hypertonic pharmaceutical composition is 0.5M-2.5M (e.g., about 0.05M to about 1.5M, about 0.05M to about 0.5M, about 0.05M to about 0.2M, about 0.05M to about 0.1M, about 0.1M to about 1.5M, about 0.1M to about 0.5M, about 0.1M to about 0.2M, about 0.2M to about 1.5M, about 0.2M to about 0.5M, about 0.5M to about 1.5M, 0.05M to about 1.0M, about 0.05M to about 0.5M, about 0.05M to about 0.2M, about 0.05M to about 0.1M, about 0.1M to about 1.0M, about 0.1M to about 0.5M, about 0.0M to about 0.5M, about 0.1M to about 0.1M, about 0.2M, about 0.5M to about 0.1M, about 0.2M, about 0M to about 0.5M, about 0.1M, about 0M, about 0.2M, about 0.5M, about 0.1M, about 0M, about 0.2M, or about 0M, about 0.1M, about 0.5M, about 0M, about 0.1M, or about 0.1M, about 0M, about 0.1M, about 0.5M, about 0.1M, about 0.5M, about 0M, about 0.1M, about 0.5M, about 0M, about 0.1M, about 0.or about 0M, about 0.1M, about 0M, about 0.5M, about 0.1M, about 0M, about 0.5M, about 0.1M, about 0M, about 0.1M, about 0.5M, about 0.1M, about 0M, about 0.1M, about 0.or about 0.5M, about 0M, about 0.5M, about 0.1M, about 0.
In certain embodiments, an effective amount is an amount that results in a plasma thiosulfate concentration of 30 μ M or less when a platinum-based antineoplastic agent is administered. In certain embodiments, an effective amount is 0.1 to 2.5mmol of thiosulfate. In a particular embodiment, an effective amount is an amount that results in a maximum thiosulfate concentration of 0.6-10 mmol/L1 h after administration. In further embodiments, an effective amount is an amount that results in a thiosulfate concentration of 0.1-2 mmol/L7 hours after administration in the cochlea of the subject.
In some embodiments, the subject is scheduled to administer the platinum-based anti-neoplastic agent within about 1 hour to about 6 hours (e.g., within about 1 hour to about 5 hours, about 1 hour to about 4 hours, about 1 hour to about 3 hours, about 1 hour to about 2 hours, about 2 hours to about 3 hours, about 2 hours to about 4 hours, about 2 hours to about 5 hours, about 2 hours to about 6 hours, about 3 hours to about 4 hours, about 3 hours to about 5 hours, about 3 hours to about 6 hours, about 4 hours to about 5 hours, about 4 hours to about 6 hours, or about 5 hours to about 6 hours after administration of the thiosulfate).
In some embodiments, the subject administers the platinum-based anti-neoplastic agent within about 1 hour to about 6 hours after administration of the thiosulfate (e.g., within about 1 hour to about 5 hours, about 1 hour to about 4 hours, about 1 hour to about 3 hours, about 1 hour to about 2 hours, about 2 hours to about 3 hours, about 2 hours to about 4 hours, about 2 hours to about 5 hours, about 2 hours to about 6 hours, about 3 hours to about 4 hours, about 3 hours to about 5 hours, about 3 hours to about 6 hours, about 4 hours to about 5 hours, about 4 hours to about 6 hours, or about 5 hours to about 6 hours after administration of the thiosulfate).
In some embodiments, the subject administers the platinum-based anti-neoplastic agent within about 1 hour to about 6 hours prior to administration of the thiosulfate (e.g., within about 1 hour to about 5 hours, about 1 hour to about 4 hours, about 1 hour to about 3 hours, about 1 hour to about 2 hours, about 2 hours to about 3 hours, about 2 hours to about 4 hours, about 2 hours to about 5 hours, about 2 hours to about 6 hours, about 3 hours to about 4 hours, about 3 hours to about 5 hours, about 3 hours to about 6 hours, about 4 hours to about 5 hours, about 4 hours to about 6 hours, or about 5 hours to about 6 hours prior to administration of the thiosulfate).
In further embodiments, the invention is described by the items listed below.
1. A method of reducing platinum-induced ototoxicity in a subject in need thereof, the method comprising administering an effective amount of thiosulfate to the subject, wherein the subject is administered a platinum-based antineoplastic agent no more than 7 hours prior to administration of thiosulfate, or is scheduled to be administered a platinum-based antineoplastic agent within 4 hours.
2. The method of clause 1, wherein the effective amount is an amount that results in a plasma thiosulfate concentration of 30 μ Μ or less when the platinum-based antineoplastic agent is administered.
3. A method of reducing platinum-induced ototoxicity in a subject in need thereof, the method comprising administering to the subject an effective amount of thiosulfate to produce (i) plasma thiosulfate C of 30 μ M or less when administered a platinum-based anti-neoplastic agentmaxAnd (ii) cochlea C that is a platinum-based anti-neoplastic agentmaxAt least 30 times as large as cochlear thiosulfate CmaxWherein the cochlear platinum concentration and the cochlear CmaxThe modeling was performed by pharmacokinetic simulation of intravenous infusion in a two-compartment model.
4. The method of clause 2, wherein the subject is administered the platinum-based oncology agent no more than 7 hours prior to administration of the thiosulfate salt, or is scheduled to be administered the platinum-based antineoplastic agent within 4 hours.
5. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 7 hours prior to administration of the thiosulfate.
6. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 6 hours prior to administration of the thiosulfate.
7. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 5 hours prior to administration of the thiosulfate.
8. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 4 hours prior to administration of the thiosulfate.
9. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 3 hours prior to administration of the thiosulfate.
10. The method of any of clauses 1-3, wherein the subject is administered the platinum-based neoplastic agent no more than 2.5 hours prior to administration of the thiosulfate.
11. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 2 hours prior to administration of the thiosulfate.
12. The method of any of clauses 1-3, wherein the subject is administered the platinum-based oncology agent no more than 1 hour prior to administration of the thiosulfate.
13. The method of any of clauses 1 to 3, wherein the subject is scheduled to administer the platinum-based anti-neoplastic agent within 4.5 hours.
14. The method of any of clauses 1-3, wherein the subject is scheduled to administer a platinum-based anti-neoplastic agent within 4 hours.
15. The method of any of clauses 1-3, wherein the subject is scheduled to administer a platinum-based anti-neoplastic agent within 3 hours.
16. The method of any of clauses 1-3, wherein the subject is scheduled to administer a platinum-based anti-neoplastic agent within 2 hours.
17. The method of any of clauses 1 to 3, wherein the subject is scheduled to administer a platinum-based anti-neoplastic agent within 1 hour.
18. The method according to any of clauses 1 to 16, wherein the thiosulfate salt is administered otically.
19. The method of clause 17, wherein the thiosulfate is administered intratympanically.
20. The method of clause 17, wherein the thiosulfate salt is administered tympanically.
21. The method of clause 17, wherein the thiosulfate salt is administered by injection in the inner ear.
22. The method of any one of clauses 1 to 20, further comprising administering a platinum-based anti-neoplastic agent.
23. The method according to any one of clauses 1 to 21, wherein the thiosulfate salt is an alkaline thiosulfate salt, an ammonium thiosulfate salt, or a solvate thereof.
24. The method of any of clauses 1 to 22, wherein the effective amount of thiosulfate is administered as a hypertonic pharmaceutical composition comprising an effective amount of thiosulfate.
25. The method of clause 23, wherein 200-1,000 μ L of the hypertonic pharmaceutical composition is administered to the round window of the subject.
26. The method of clauses 23 or 24, wherein the calculated osmolality of the hypertonic pharmaceutical composition is 500-5,000 mOsm/L.
27. The method according to any of clauses 23 to 25, wherein the concentration of thiosulfate in the hypertonic pharmaceutical composition is 0.5M-2.5M.
28. The method according to any of clauses 23 to 25, wherein the concentration of thiosulfate in the hypertonic pharmaceutical composition is 0.5M-1.5M.
29. The method according to any of clauses 23 to 25, wherein the concentration of thiosulfate in the hypertonic pharmaceutical composition is 0.5M-1.0M.
30. The method of any of clauses 1 to 28, wherein the effective amount is at least 0.05mmol of thiosulfate.
31. The method of any of clauses 1 to 28, wherein the effective amount is at least 0.1mmol of thiosulfate.
32. The method of any of clauses 1 to 28, wherein the effective amount is at least 0.2mmol of thiosulfate.
33. The method of any of clauses 1 to 28, wherein the effective amount is at least 0.3mmol of thiosulfate.
34. The method of any of clauses 1 to 28, wherein the effective amount is at least 0.4mmol of thiosulfate.
35. The method of any of clauses 1 to 33, wherein the effective amount is 2.5mmol or less of thiosulfate.
36. The method of any of clauses 1 to 33, wherein the effective amount is 2.0mmol or less of thiosulfate.
37. The method of any of clauses 1 to 33, wherein the effective amount is 1.5mmol or less of thiosulfate.
38. The method of any of clauses 1 to 33, wherein the effective amount is 1.0mmol or less of thiosulfate.
39. The method of any of clauses 1 to 33, wherein the effective amount is 0.5mmol or less of thiosulfate.
40. The method of any of clauses 1 to 38, wherein the effective amount is an amount that results in a maximum thiosulfate concentration of 0.6-10 mmol/L1 h after administration.
41. The method according to any of clauses 1 to 39, wherein the effective amount is an amount that results in a thiosulfate concentration of 0.1-2 mmol/L7 h after administration in the cochlea of the subject.
Definition of
The term "about" as used herein means a value within ± 10% of the value after the term "about".
The term "basic salt" as used herein represents a sodium or potassium salt of a compound. The basic salt may be monobasic or if the acidic moiety (e.g., -COOH, -SO)3H or-P (O) (OH)nPortions) are allowed, then binary or ternary.
The term "ammonium salt" as used herein represents the NH of a compound4 +And (3) salt. The ammonium salt may be mono-basic or if the acidic moiety (e.g., -COOH, -SO)3H or-P (O) (OH)nPortions) are allowed, then binary or ternary.
The term "gelling agent" as used herein refers to a pharmaceutically acceptable excipient known in the art for producing a gel when mixed with a solvent (e.g., an aqueous solvent). Non-limiting examples of gelling agents include hyaluronic acid, polyoxyethylene-polyoxypropylene block copolymers (e.g., poloxamers), poly (lactic-co-glycolic acid), polylactic acid, polycaprolactone, alginic acid or a salt thereof, polyethylene glycol, cellulose ethers, carbomers (e.g.,
Figure BDA0003410553030000081
) Agar, gelatin, glucomannan, galactomannan (e.g., guar gum, locust bean gum, or tara gum), xanthan gum, chitosan, pectin, starch, tragacanth, carrageenan, polyvinylpyrrolidone, polyvinyl alcohol, paraffin, guar gum, gum,Petrolatum, silicates, silk proteins, and combinations thereof.
The term "hypertonic", as used herein in relation to a pharmaceutical composition, means that the pharmaceutical composition has a calculated osmolality of 300 to 7,000mOsm/L (e.g., 300 to 2,500mOsm/L) corresponding to 300 to 7,000mmol (e.g., 300 to 2,500mmol) of ionic and/or neutral molecules produced by dissolving an inactivating platinum agent and any ionic non-polymeric excipients in 1L of a solvent having a calculated osmolality of 0 mOsm/L. For the purposes of this disclosure, the calculated osmolarity excludes ionic and/or neutral molecules produced by polymeric excipients (e.g., by gelling agents). For the purposes of this disclosure, polymeric excipients (e.g., gelling agents) are considered to not contribute to the calculated osmolarity of the compositions disclosed herein.
The term "intratympanic" as used herein in relation to a route of administration refers to delivery to the round window by injection or infusion into the middle ear of a subject via the ear canal with the tympanic membrane temporarily removed or lifted or via a port formed through an auditory bulb (audio bulla).
The term "pharmaceutical composition" as used herein means a composition formulated with a pharmaceutically acceptable excipient and approved by a governmental regulatory agency for manufacture or sale as part of a therapeutic regimen for the treatment of a disease in a mammal.
The term "pharmaceutical dosage form" as used herein represents those pharmaceutical compositions intended for administration to a subject without further modification (e.g., without dilution, suspension or dissolution with a liquid solvent).
The term "pharmaceutically acceptable excipient" as used herein refers to any ingredient (e.g., vehicle capable of suspending or dissolving an active compound) other than the thiosulfate and gelling agents described herein, and which has the characteristics of being substantially non-toxic to a patient and substantially free of inflammation. Excipients may include, for example, antioxidants, disintegrants, dyes (colorants), emollients, emulsifiers, fillers (diluents), fragrances, preservatives, printing inks, adsorbents, suspending or dispersing agents, sweeteners, liquid solvents, and buffers.
The term "pharmaceutically acceptable salts" as used herein means those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: berge et al, J.pharmaceutical Sciences 66:1-19,1977 and Pharmaceutical Salts, Properties, Selection, and Use, (P.H.Stahl and C.G.Wermuth), Wiley-VCH, 2008. The salts may be prepared in situ during the final isolation and purification of the compounds described herein, or separately by reacting the free base group with a suitable organic acid. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate (glucoheptonate), glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxyethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, phosphate, benzoate, bisulfate, caproate, bromate, bisulfate, thymidate, salt, a salt of a salt, a salt of a salt, a salt, Picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, tosylate, undecanoate, valerate, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
The term "pharmaceutically acceptable solvate" as used herein refers to a compound as described herein, wherein molecules of a suitable solvent are incorporated into the crystal lattice. Suitable solvents are physiologically tolerable at the doses administered. For example, solvates may be prepared by crystallization, recrystallization or precipitation from solutions comprising organic solvents, water or mixtures thereof. Examples of suitable solvents are ethanol, water (e.g., monohydrate, dihydrate, trihydrate, tetrahydrate, and pentahydrate), N-methylpyrrolidone (NMP), dimethyl sulfoxide (DMSO), N '-Dimethylformamide (DMF), N' -Dimethylacetamide (DMAC), 1, 3-dimethyl-2-imidazolidinone (DMEU), 1, 3-dimethyl-3, 4,5, 6-tetrahydro-2- (1H) -pyrimidinone (DMPU), Acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When the solvate is water-based, the solvate is referred to as a hydrate.
The term "platinum-based antineoplastic agent" as used herein represents a coordination compound of pt (ii) or pt (iv). Platinum-based antineoplastic agents are known in the art as pladins (platins). Typically, platinum-based antineoplastic agents comprise at least two coordination sites occupied by one or more nitrogen-containing bystander ligands at the platinum center. The nitrogen-containing spectator ligand being a monodentate or bidentate ligand in which the donor atom is sp within the ligand3-or sp2-a hybridized nitrogen atom. Non-limiting examples of nitrogen-containing spectator ligands are ammonia, 1, 2-cyclohexanediamine, picoline, phenanthrene, or 1, 6-hexanediamine. Non-limiting examples of platinum-based antineoplastic agents include cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthroline, picoplatin, and satraplatin.
The term "subject" as used herein refers to an animal (e.g., a mammal, e.g., a human). A subject treated according to the methods described herein can be a subject treated with a treatment regimen that includes a platinum-based anti-neoplastic agent (e.g., a treatment regimen for treating a benign tumor, a malignant tumor, or a cancer). A subject may have been diagnosed as having a benign tumor, a malignant tumor, or a cancer by any method or technique known in the art. One skilled in the art will appreciate that a subject to be treated according to the present invention may have received standard testing, or may have been determined, without examination, to be at high risk due to receiving a treatment regimen comprising a platinum-based anti-neoplastic agent.
The term "substantially neutral" as used herein means a pH level of 5.5 to about 8.5 measured at 20 ℃.
The term "tonicity agent" as used herein refers to a class of pharmaceutically acceptable excipients used to control the osmolarity of a pharmaceutical composition. Non-limiting examples of tonicity agents include substantially neutral buffers (e.g., phosphate buffered saline, tris buffer, or artificial perilymph fluid), dextrose, mannitol, glycerol, potassium chloride, and sodium chloride (e.g., hypertonic saline, isotonic saline, or hypotonic saline). The artificial perilymph fluid contains NaCl (120-2(1.3-1.5mM)、MgCl2(1.2mM), glucose (5.0-11mM), and a buffer (e.g., NaHCO)3(25mM) and NaH2PO4(0.75mM), or HEPES (20mM) and NaOH (adjusted to a pH of about 7.5)).
The term "trans-tympanic" as used herein in reference to a route of administration refers to delivery to the round window by injection or infusion across the tympanic membrane. Trans-tympanic injection can be performed directly via the tympanic membrane or via a tube embedded in the tympanic membrane (e.g., via a tympanostomy tube or a tympanostomy tube).
The term "inner ear injection" as used herein refers to the injection of a drug directly into the inner ear space.
Drawings
Figure 1 is a graph showing the profile of the mean plasma thiosulfate concentration over time (0-24h) for each tested population group. The X-axis shows time (h) and the Y-axis shows mean plasma thiosulfate concentration (ng/mL).
Figure 2 is a graph showing the profile of the mean plasma thiosulfate concentration over time (0-4h) for each tested population group. The error bars shown are standard deviations. The X-axis shows time (h) and the Y-axis shows mean plasma thiosulfate concentration (ng/mL).
Figure 3 is a graph showing the profile of the mean plasma thiosulfate concentration over time (0-672h) for each tested population group. The X-axis shows time (h) and the Y-axis shows mean plasma thiosulfate concentration (ng/mL).
Figure 4 is a schematic showing the timing of thiosulfate administration relative to cisplatin.
Fig. 5A is a graph showing the average threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured during the Auditory Brainstem Response (ABR) test of control guinea pigs. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve. The X-axis shows the acoustic frequency in kHz, and the Y-axis shows the response threshold in decibels (dB SPL) of sound pressure level.
Fig. 5B is a graph showing the mean threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured during the Auditory Brainstem Response (ABR) test in cisplatin-challenged guinea pigs after sodium thiosulfate was administered to each ear. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve. The X-axis shows the acoustic frequency in kHz and the Y-axis shows the response threshold in decibels of sound pressure level (dB SPL). Data shown are mean ± Standard Error of Mean (SEM); two-way analysis of variance (ANOVA); p < 0.01; p < 0.001. Treated ears compared to untreated ears (compare with fig. 5A).
FIG. 6 is a graph showing the concentration dependence of thiosulfate (DB-020) in a human tumor cell line treated with 15 μ M cisplatin. The following tumor cell lines were used: SH-N-AS (brain, neuroblastoma), SNU899 (larynx, squamous cell carcinoma), NCI-H23 (lung, non-small cell), HLF (liver, undifferentiated hepatocellular carcinoma), and A2780 (ovary, malignancy).
Figure 7 is a graph showing the profile (0-8h) of mean thiosulfate concentrations (mM) in plasma (human) and perilymph fluid (guinea pig) over time after administration of hyaluronic acid gel 1 (12% w/v, 0.5M sodium thiosulfate). The horizontal solid line shows the level of 30 μ M below which plasma thiosulfate levels should be. The dotted horizontal line shows the level of 660. mu.M (0.66mM), above which the perilymph fluid thiosulfate level should be.
Figure 8A is a graph showing the change over time in thiosulfate concentration in plasma, perilymph fluid and cerebrospinal fluid of guinea pigs administered with a gel containing 0.1M sodium thiosulfate and 20% (w/v) poloxamer 407.
FIG. 8B is a graph showing the change over time of thiosulfate concentrations in plasma, perilymph fluid and cerebrospinal fluid of guinea pigs administered with a gel containing 0.5M sodium thiosulfate and 1% (w/v) hyaluronic acid.
FIG. 9A is a graph showing the change over time of thiosulfate concentrations in plasma, perilymph fluid and cerebrospinal fluid of guinea pigs administered with a gel containing 0.1M sodium thiosulfate and 2% (w/v) hyaluronic acid.
Figure 9B is a graph showing the change in thiosulfate concentration over time in plasma, perilymph fluid and cerebrospinal fluid of guinea pigs administered with a gel containing 0.5M sodium thiosulfate and 2% (w/v) hyaluronic acid.
Fig. 10A is a graph showing threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured in five guinea pig groups (n ═ 27 animals) during the auditory brainstem response test. All guinea pigs were injected intraperitoneally with cisplatin 7 days prior to the auditory brainstem response test. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve.
Fig. 10B is a graph showing threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured in five guinea pig groups (n ═ 18 animals) during the auditory brainstem response test. All guinea pigs were injected intraperitoneally with cisplatin 7 days prior to the auditory brainstem response test. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve.
Fig. 11A is a graph showing the mean threshold sound pressure levels at 4kHz, 24kHz and 32kHz measured during the auditory brainstem response test of guinea pigs (n ═ 18 animals) considered to have hearing loss. All guinea pigs were injected intraperitoneally with cisplatin 7 days prior to the auditory brainstem response test. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve.
Fig. 11B is a graph showing the mean threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured during the Auditory Brainstem Response (ABR) test in cisplatin-challenged guinea pigs after vehicle or sodium thiosulfate was applied to each ear. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve.
Figure 12 is a graph showing the cisplatin challenge test after vehicle or sodium thiosulfate administration to one ear of guinea pigs.
Figure 13 is a graph showing the mean threshold sound pressure levels at 4kHz, 24kHz, and 32kHz measured during the Auditory Brainstem Response (ABR) test in guinea pigs challenged with cisplatin following application of vehicle or sodium thiosulfate (0.1M, 0.5M, or 1M sodium thiosulfate gels) to each ear. Baseline thresholds were derived from historical auditory brainstem response tests on cisplatin-naive guinea pigs (n ═ 100 ears). The baseline threshold is shown as a shaded area curve.
Detailed Description
In general, the invention provides methods of reducing platinum-induced ototoxicity in a subject by administering an effective amount of thiosulfate to the subject. Preferably, the thiosulfate is administered otically (e.g., intratympanically or transtympanically).
Typically, thiosulfate is administered to a subject who is scheduled to administer a platinum-based anti-neoplastic agent within 4 hours (e.g., within 3 hours, within 2 hours, or within 1 hour). Alternatively, the thiosulfate salt is administered within 7 hours (e.g., within 6 hours, within 5 hours, within 4 hours, within 3 hours, within 2 hours, or within 1 hour) after the platinum-based neoplastic agent is administered. Preferably, thiosulfate is administered to a subject who is scheduled to administer a platinum-based anti-neoplastic agent within 3 hours. Alternatively, the thiosulfate salt is administered within 4 hours after administration of the platinum-based neoplastic agent. More preferably, thiosulfate is administered to a subject who is scheduled to administer a platinum-based anti-neoplastic agent within 1 hour. Alternatively, the thiosulfate salt is administered within 1 hour after administration of the platinum-based neoplastic agent.
An effective amount of thiosulfate typically results in a plasma thiosulfate concentration of 30 μ M or less (e.g.,20 μ M or less, 10 μ M or less, or close to endogenous concentrations) when a platinum-based antineoplastic agent is administered. Additionally or alternatively, an effective amount of thiosulfate typically produces cochlea C that is a platinum-based anti-neoplastic agentmaxAt least 30 times greater (e.g., 30 times to 1000 times greater, 30 times to 500 times greater, or 30 times to 150 times greater) of cochlear thiosulfate concentration, wherein the cochlear platinum concentration and the cochlear C aremaxThe modeling was performed by pharmacokinetic simulation of intravenous infusion in a two-compartment model.
Platinum-induced ototoxicity can occur in a subject (e.g., a subject having a tumor or cancer) that has received a platinum-based anti-neoplastic agent. Non-limiting examples of platinum-based antineoplastic agents include cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthroline, picoplatin, and satraplatin.
Thiosulfate can reduce (e.g., eliminate) hearing loss in a subject receiving a platinum-based anti-neoplastic agent, as measured by a reduction of at least 50% (e.g., at least 60%, at least 70%, or at least 80%) in the subject's sound pressure level threshold assessment at a frequency of 8kHz or higher (e.g., between 8kHz and 20 kHz) relative to a reference subject receiving the same platinum-based anti-neoplastic agent regimen but not thiosulfate.
Thiosulfate can exhibit otoprotective properties against platinum-based anti-neoplastic agents and can be used in methods for reducing (e.g., eliminating) platinum-induced ototoxicity in a subject in need thereof. Typically, the thiosulfate is administered to the round window of the subject. The subject may be undergoing therapy with a platinum-based antineoplastic agent (e.g., cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthratinum, picoplatin, or satraplatin).
The thiosulfate salt can be administered to the subject, for example, before or after the platinum-based anti-neoplastic agent is administered to the subject. Alternatively, the thiosulfate salt can be administered, for example, at the same time as the platinum-based antineoplastic agent. Thiosulfate can be administered to a subject who is scheduled to administer a platinum-based anti-neoplastic agent within 4 hours, such as within 3 hours, within 2 hours, or within 1 hour (e.g., at least 5 minutes, at least 15 minutes, or at least 30 minutes prior to administration of the platinum-based anti-neoplastic agent). Alternatively, the thiosulfate salt can be administered after (e.g., at least 5 minutes, at least 15 minutes, or at least 30 minutes after) the platinum-based antineoplastic agent is administered, such as no more than 7 hours (e.g., no more than 6 hours, no more than 5 hours, no more than 4 hours, no more than 3 hours, no more than 2 hours, or no more than 1 hour). Administration of platinum-based antineoplastic agents is typically performed prior to administration of a hydration composition (e.g., physiological saline optionally containing, for example, mannitol or furosemide). Further, administration of the platinum-based antineoplastic agent is typically continued for a period of at least 1 hour (e.g., 1 to 24 hours, such as 1 to 12 hours, 1 to 6 hours, 1 to 3 hours, or 1 to 2 hours). One skilled in the art will recognize that the timing between the administration of the thiosulfate and the platinum-based neoplastic agent is the time between the completion of the administration of one and the beginning of the administration of the other. For example, administration of a platinum-based neoplastic agent 30 minutes to 4 hours after the step of administering thiosulfate indicates that 30 minutes to 4 hours separate the end of thiosulfate administration from the beginning of platinum-based neoplastic agent administration (e.g., infusion of cisplatin).
In general, the pharmaceutical compositions of the present invention may be administered by a route other than platinum-based antineoplastic agents. The methods of the invention may utilize a topical route of administration, e.g., the pharmaceutical compositions of the invention may be administered intratympanically or transtympanically. Trans-tympanic administration can include injecting or infusing an effective amount of the pharmaceutical composition of the present invention into the tympanic cavity through the tympanic membrane, thereby providing the anti-platinum chemoprotectant to the round window.
In the methods of the present invention, a needle is typically used to pierce the tympanic membrane to instill the drug into the middle ear space, or through an existing PE tube or perforation of the ear drum to instill the drug. A separate vent may or may not be created in the tympanic membrane to allow air to escape the middle ear space. Subsequently, instilled drugs can be targeted to middle ear structures, cells, or designed to enter the inner ear through circular and oval membranes to affect specific targets. This may be achieved, for example, by instillation of the drug through the round window membrane, oval window, cochleostomy (cochleostomy), or labyrinthomy (labrinthomy). These surgeries can be performed by raising the tympanic membrane ear canal flap (lifting the tympanic membrane) and exposing the round window, the stapedial/oval window and promontory. A stapedial footplate porosimetry hole may be created in the stapedial fundus (footplate) and the drug instilled into the vestibule by pump, injection, or some other method. Alternatively, the bone lips of a Round Window (RW) are removed (typically by drilling) to expose the RW. The RW can then be punctured with a needle and the drug infused, or the RW can be fenestrated and the drug instilled directly through the fenestration. Finally, a completely separate cochlear inlet hole may be opened by drilling a cochlear ostomy hole in the cochlea and instilling the drug.
Alternatively, instead of lifting the tympanic ear canal flap, a mastoidectomy may be performed and the facial recess opened to provide direct access to the oval and round windows and promontory and semicircular canals. In this way, all three sites can be used as just described. Furthermore, the labyrinth can be opened for drug instillation much like a cochlear ostomy. To dissipate the fluid/pressure build-up in the inner ear, a separate opening into RW or OW may be created to allow excess perilymph fluid to escape.
The thiosulfate salt can be provided in a pharmaceutical composition. The pharmaceutical composition may be, for example, hypertonic. Without wishing to be bound by theory, the higher tonicity of the pharmaceutical compositions disclosed herein is believed to increase the bioavailability of thiosulfate at the round window of a subject relative to compositions having lower tonicity (e.g., hypotonic or isotonic). Bioavailability is typically calculated using thiosulfate exposure (AUC) after administration of thiosulfate to a subject. The calculated osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, at least 400mOsm/L (e.g., at least 500mOsm/L, at least 600mOsm/L, at least 700mOsm/L, at least 800mOsm/L, at least 900mOsm/L, at least 1,000mOsm/L, at least 1,500mOsm/L, at least 2,000mOsm/L, at least 2,500mOsm/L, or at least 3,000mOsm/L), and/or 5,000mOsm/L or less (e.g., 4,000mOsm/L or less, 3,000mOsm/L or less, 2,000mOsm/L or less, 1,900mOsm/L or less, 1,800mOsm/L or less, 1,700mOsm/L or less, 1,600mOsm/L or less, or 1,500mOsm/L or less). The calculated osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, 1,500-4,500 mOsm/L. The calculated osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, 3,000-4,500 mOsm/L. The measured osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, at least 0.3Osm/kg (e.g., at least 0.5Osm/kg, at least 0.6Osm/kg, at least 0.7Osm/kg, at least 0.8Osm/kg, at least 0.9Osm/kg, at least 1.0Osm/kg, at least 1.2Osm/kg, at least 1.4Osm/kg, or at least 1.8 Osm/kg). The measured osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, 2.5Osm/kg or less (e.g., 2.1Osm/kg or less). The measured osmolality of the pharmaceutical composition (e.g., pharmaceutical dosage form) can be, for example, 0.3-2.5Osm/kg (e.g., 0.5-2.5Osm/kg, 0.6-2.5Osm/kg, 0.7-2.5Osm/kg, 0.8-2.5Osm/kg, 0.9-2.5Osm/kg, 1.0-2.5Osm/kg, 1.2-2.5Osm/kg, 1.4-2.5Osm/kg, 1.8-2.5Osm/kg, 0.5-2.1Osm/kg, 0.6-2.1Osm/kg, 0.7-2.1Osm/kg, 0.8-2.1Osm/kg, 0.9-2.1Osm/kg, 1.0-2.1Osm/kg, 1.2-2.1Osm/kg, 1.4-2.1Osm/kg, 1.8-2.1Osm/kg, or 1 Osm/kg). "calculated osmolarity" refers to the number of millimoles of ionic and/or neutral molecules produced by dissolving one or more compounds in 1 liter of deionized or distilled water; the osmolarity is calculated to exclude ionic and/or neutral molecules produced by the polymeric excipient (e.g., by the gelling agent). "measured osmolarity" refers to the osmolarity of a composition measured using an osmometer (typically a membrane osmometer).
The preferred pharmaceutical dosage form of the present invention is a gel.
In some embodiments, at least 50 μ L (preferably, at least 100 μ L; more preferably, at least 200 μ L) of the pharmaceutical composition is administered to the round window of the subject. In particular embodiments, 1mL or less (e.g., 0.8mL or less, or 0.5mL or less) of the pharmaceutical composition is administered to the round window of the subject. In certain embodiments, 100 μ L to 1mL (e.g.,200 μ L to 1mL, 100 μ L to 0.8mL, 200 μ L to 0.8mL, 100 μ L to 0.5mL, 200 μ L to 0.5mL, 0.5mL to 1.0mL, 0.5mL to 0.8mL, or 0.8mL to 1.0mL) of the pharmaceutical composition is administered to the round window of the subject.
The thiosulfate salt can be the only compound that contributes to the osmolality of the pharmaceutical composition, for example. Alternatively, higher osmolalities than provided by the desired concentration of thiosulfate can be achieved, for example, by using tonicity agents. Tonicity agents may be present in a hypertonic, isotonic or hypotonic vehicle (e.g., hypotonic liquid solvents). Non-limiting examples of tonicity agents include substantially neutral buffers (e.g., phosphate buffered saline, tris buffer, or artificial perilymph fluid), dextrose, mannitol, glycerol, potassium chloride, and sodium chloride (e.g., hypertonic, isotonic, or hypotonic saline).
Thiosulfate salts
Without wishing to be bound by theory, thiosulfate is believed to reduce or eliminate the toxicity of platinum-based antineoplastic agents by competitively linking and substantially coordinately saturating the platinum centers present in the platinum-based antineoplastic agents. The concentration of thiosulfate in a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, at least about 0.05M (e.g., at least about 0.1M, at least about 0.2M, at least about 0.3M, at least about 0.4M, at least about 0.5M, or at least about 1M). The concentration of thiosulfate in a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be, for example, about 2.5M or less (e.g., 2.0M or less, 1.5M or less, 1.0M or less, 0.5M or less, about 0.3M or less, or about 0.2M or less). Non-limiting examples of the concentration of thiosulfate in a pharmaceutical composition (e.g., pharmaceutical dosage form) can be, for example, from about 0.05M to about 1.5M, from about 0.05M to about 0.5M, from about 0.05M to about 0.2M, from about 0.05M to about 0.1M, from about 0.1M to about 1.5M, from about 0.1M to about 0.5M, from about 0.1M to about 0.2M, from about 0.2M to about 1.5M, from about 0.2M to about 0.5M, from about 0.5M to about 1.5M, from 0.05M to about 1.0M, from about 0.05M to about 0.5M, from about 0.05M to about 0.2M, from about 0.05M to about 0.1M, from about 0.1M to about 0.5M, from about 0.1M to about 0.2M, from about 0.1M to about 0M, from about 0.1M, from about 0.2M, from about 0M to about 0M, from about 0.5M, or from about 0M to about 0.5M. Preferably, the concentration of the thiosulfate agent in the pharmaceutical composition (e.g., pharmaceutical dosage form) is from about 0.5M to about 1.5M. More preferably, the concentration of the thiosulfate agent in the pharmaceutical composition (e.g., pharmaceutical dosage form) is from about 0.5M to about 1.0M.
Preferably, the thiosulfate salt is an alkaline thiosulfate salt or an ammonium thiosulfate salt. More preferably, the thiosulfate salt is sodium thiosulfate.
Gelling agent
The pharmaceutical compositions disclosed herein comprise a gelling agent. The gelling agent may be used to increase the viscosity of the pharmaceutical composition, thereby improving the retention of the pharmaceutical composition at the target site. Relative to the solvent, the pharmaceutical composition (e.g., pharmaceutical dosage form) can contain, for example, about 0.1% to about 25% (w/v) (e.g., about 0.1% to about 20% (w/v), about 0.1% to about 10% (w/v), about 0.1% to about 2% (w/v), about 0.5% to about 25% (w/v), about 0.5% to about 20% (w/v), about 0.5% to about 10% (w/v), about 0.5% to about 2% (w/v), about 1% to about 20% (w/v), about 1% to about 10% (w/v), about 1% to about 2% (w/v), about 5% to about 20% (w/v), about 5% to about 10% (w/v), or about 7% to about 10% (w/v)) of the gelling agent. Preferably, a pharmaceutical composition (e.g., a pharmaceutical dosage form) may contain, for example, about 0.5% to about 25% (w/v) (e.g., about 0.5% to about 20% (w/v), about 0.5% to about 10% (w/v), about 0.5% to about 2% (w/v), about 1% to about 20% (w/v), about 1% to about 10% (w/v), about 1% to about 2% (w/v), about 5% to about 20% (w/v), about 5% to about 10% (w/v), or about 7% to about 10% (w/v)) of a gelling agent relative to the solvent.
Gelling agents that can be used in the pharmaceutical compositions disclosed herein are known in the art. Non-limiting examples of gelling agents include hyaluronic acid, polyoxyethylene-polyoxypropylene block copolymers (e.g., poloxamers), poly (lactic-co-glycolic acid), polylactic acid, polycaprolactone, alginic acid or a salt thereof, polyethylene glycol, cellulose, and mixtures thereof,Cellulose ethers, carbomers (e.g.,
Figure BDA0003410553030000201
) Agar, gelatin, glucomannan, galactomannan (e.g., guar gum, locust bean gum, or tara gum), xanthan gum, chitosan, pectin, starch, tragacanth gum, carrageenan, polyvinylpyrrolidone, polyvinyl alcohol, paraffin, petrolatum, silicates, silk proteins, and combinations thereof. The gelling agents described herein are known in the art. Preferably, the gelling agent is hyaluronic acid.
The pharmaceutical composition may contain, for example, about 0.5% to about 2% (w/v) (e.g., about 1% to about 2% (w/v)) of hyaluronic acid relative to the solvent. The pharmaceutical composition can contain, for example, about 5% to about 10% (w/v) (e.g., about 6% to about 8% (w/v)) of methylcellulose relative to the solvent. The pharmaceutical composition may contain, for example, hyaluronic acid and methylcellulose as gelling agents (e.g., about 0.5% to about 2% (w/v) hyaluronic acid and about 5% to about 10% (w/v) methylcellulose, relative to the solvent). The pharmaceutical composition may contain, for example, a polyoxyethylene-polyoxypropylene block copolymer (e.g., poloxamer) as a gelling agent. The pharmaceutical composition can contain, relative to the solvent, for example, about 1% to about 20% (w/v) (e.g., about 1% to about 15% (w/v), about 1% to about 10% (w/v), about 5% to about 20% (w/v), about 5% to about 15% (w/v), about 5% to about 10% (w/v), about 10% to about 20% (w/v), or about 10% to about 15% (w/v)) of a polyoxyethylene-polyoxypropylene block copolymer (e.g., a poloxamer). The poloxamer can be poloxamer 407, poloxamer 188, or a combination thereof. The pharmaceutical composition may contain, for example, about 0.5% (w/v) to about 20% (w/v) silk protein as a gelling agent with respect to the solvent.
Hyaluronic acid (Hyaluronan) is hyaluronic acid (hyaluronic acid) or a salt thereof (e.g., sodium hyaluronate). Hyaluronic acid (Hyaluronan) is known in the art and is typically isolated from various bacteria (e.g., Streptococcus zooepidemicus, Streptococcus equi, or Streptococcus pyogenes) or other sources (e.g., bovine vitreous humor or cockscomb)In (1). Weight average molecular weight (M) of hyaluronic acidW) Typically from about 50kDa to about 10 MDa. Preferably, M of hyaluronic acid (e.g., sodium hyaluronate)WIs about 500kDa to 6MDa (e.g., about 500kDa to about 750kDa, about 600kDa to about 1.1MDa, about 750kDa to about 1MDa, about 1MDa to about 1.25MDa, about 1.25 to about 1.5MDa, about 1.5MDa to about 1.75MDa, about 1.75MDa to about 2MDa, about 2MDa to about 2.2MDa, about 2MDa to about 2.4 MDa). More preferably, M of hyaluronic acid (e.g., sodium hyaluronate)WFrom about 620kDa to about 1.2MDa or from about 1.2MDa to about 1.9 MDa. Other preferred molecular weight ranges for hyaluronic acid include, for example, from about 600kDa to about 1.2 MDa.
Polyoxyethylene-polyoxypropylene block copolymers are known in the art. A non-limiting example of a polyoxyethylene-polyoxypropylene block copolymer is a poloxamer, where a single polyoxypropylene block is flanked by two polyoxyethylene blocks. Poloxamers are available under various trade names, e.g., poloxamer
Figure BDA0003410553030000221
And
Figure BDA0003410553030000222
and (4) carrying out commercial purchase. The pharmaceutical composition can contain, for example, a polyoxyethylene-polyoxypropylene block copolymer (e.g., a poloxamer) that includes a number average molecular weight (M)n) For example, from about 1,100g/mol to about 17,400g/mol (e.g., from about 2,090g/mol to about 2,360g/mol, from about 7,680g/mol to about 9,510g/mol, from 6,830g/mol to about 8,830g/mol, from about 9,840g/mol to about 14,600g/mol, or from about 12,700g/mol to about 17,400 g/mol). The polyoxyethylene-polyoxypropylene block copolymer (e.g., poloxamer) may comprise a number average molecular weight (M)n) A polyoxypropylene block of about 1,100g/mol to about 4,000g/mol and a calculated polyoxyethylene content of about 30% to about 85% (w/w). Preferably, the polyoxyethylene-polyoxypropylene block copolymer (e.g., poloxamer) may comprise a polyoxypropylene block having a calculated molecular weight of, for example, about 1,800g/mol to about 4,000 g/mol. Preferably, the calculated polyoxyethylene content of the polyoxyethylene-polyoxypropylene block copolymer (e.g., poloxamer) may be, for example, from about 70% to about 80% (w/w). Preference is given toAlternatively, the polyoxyethylene-polyoxypropylene block copolymer (e.g., poloxamer) may have a number average molecular weight of, for example, about 7,680g/mol to about 14,600 g/mol. Non-limiting examples of poloxamers are poloxamer 407 and poloxamer 188.
Cellulose and cellulose ethers are known in the art. Cellulose and cellulose ethers are available under various trade names, e.g.
Figure BDA0003410553030000223
MethocelTM
Figure BDA0003410553030000224
And
Figure BDA0003410553030000225
and (4) carrying out commercial purchase. Non-limiting examples of cellulose ethers include methyl cellulose, carboxymethyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methyl hydroxyethyl cellulose, hydroxypropyl methyl cellulose, or hydroxypropyl cellulose. Number average molecular weight (M) of cellulose ether (e.g., methylcellulose)n) May be, for example, from about 5kDa to about 300 kDa. Methyl-substituted celluloses (e.g., methylcellulose, hydroxypropyl methylcellulose, or methyl hydroxyethyl cellulose) can have a methyl content of, for example, 19% to 35% (e.g., 19% to 30%).
Silk protein is a protein present in silk produced by many insects. Silk proteins are known in the art and are commercially available from various suppliers, such as Jiangsu SOHO International Group; simatech, Suzhou, China; xi' an Lyphar Biotech, ltd.; xi' an Rongsheng Biotechnology; mulberry Farms, Treenway Silks, Sharda groups, Maniar Enterprises and Wild fibers. The molecular weight of silk proteins is typically from about 10kDa to about 500 kDa. Silk proteins are described in WO 2017/139684, the disclosure of which is incorporated herein by reference.
Crosslinked gelling agents
The pharmaceutical composition may contain non-crosslinked or crosslinked gelling agents. The gelling agent may be crosslinked using crosslinking agents known in the art. Preferably, the crosslinked gelling agent is covalentAnd (3) crosslinking. Pharmaceutical compositions (e.g., pharmaceutical dosage forms) comprising crosslinked gelling agents can be used to control the release profile of an anti-platinum chemoprotectant. For example, release of an anti-platinum chemoprotectant from a pharmaceutical composition (e.g., a pharmaceutical dosage form) containing a crosslinked gelling agent can be extended relative to a reference composition that differs from the pharmaceutical composition only in the absence of crosslinking of the gelling agent in the reference composition. T can be determined by comparing the T of a pharmaceutical composition with a reference compositionmaxValues were evaluated for the prolongation of the release of the anti-platinum chemoprotectant.
Certain gelling agents, such as those having carboxylate moieties (e.g., hyaluronic acid, alginic acid, and carboxymethylcellulose), may use ionic crosslinking agents (e.g., polyvalent metal ions, such as Mg)2+、Ca2+Or Al3+) And carrying out ionic crosslinking. Techniques for ionic crosslinking of gelling agents are known in the art (see, e.g., U.S. Pat. nos. 6,497,902 and 7,790,699, the disclosures of which are incorporated herein by reference). Generally, the gelling agent may use multivalent metal ions (e.g., Mg) in the aqueous solution2+、Ca2+Or Al3+) As an ionic crosslinking agent to carry out ionic crosslinking. Without wishing to be bound by theory, it is believed that the metal ion coordinates with different molecules of the gelling agent (e.g., with pendant carboxylic acid groups located on different molecules of the gelling agent) thereby forming a linkage between these different molecules of the gelling agent.
Certain functional groups having reactivity (e.g., -OH, -COOH, or-NH)2) The gelling agent(s) of (a) may be covalently cross-linked. Gelling agent covalent crosslinking techniques are known in the art (see, e.g., khunnanee et al, j.tissue eng.,8:2041731417726464,2017, the disclosure of which is incorporated herein by reference). Non-limiting examples of covalent crosslinking agents include: 1, 4-butanediol diglycidyl ether (BDDE), divinyl sulfone, glutaraldehyde, cyanogen bromide, octyl succinic anhydride, acid chloride, diisocyanate, methacrylic anhydride, boric acid, and sodium periodate/adipic dihydrazide.
Other excipients
The pharmaceutical composition may contain a pharmaceutically acceptable excipient other than a gelling agent. For example, the pharmaceutical composition may contain, for example, liquid solvents, tonicity agents, buffering agents and/or coloring agents. Certain excipients may serve multiple functions. For example, the liquid solvent may act as a tonicity agent and/or a buffer in addition to its function as a carrier. Such solvents are known in the art, such as saline (e.g., hypertonic saline, hypotonic saline, isotonic saline, or phosphate buffered saline) and artificial perilymph fluid.
Liquid solvents can be used as vehicles in pharmaceutical compositions (e.g., pharmaceutical dosage forms). Liquid solvents are known in the art. Non-limiting examples of liquid solvents include water, saline (e.g., hypertonic saline, hypotonic saline, isotonic saline, or phosphate buffered saline), artificial perilymph fluid, and tris buffer. The artificial perilymph fluid contains NaCl (120-2(1.3-1.5mM)、MgCl2(1.2mM), glucose (5.0-11mM), and a buffer (e.g., NaHCO)3(25mM) and NaH2PO4(0.75mM), or HEPES (20mM) and NaOH (adjusted to a pH of about 7.5)).
Tonicity agents may be included in pharmaceutical compositions (e.g., pharmaceutical dosage forms) to increase the osmolarity relative to the osmolarity provided by the anti-platinum chemoprotectant. Tonicity agents are known in the art. Non-limiting examples of tonicity agents include substantially neutral buffers (e.g., phosphate buffered saline, tris buffer, or artificial perilymph fluid), dextrose, mannitol, glycerol, potassium chloride, and sodium chloride (e.g., hypertonic saline, isotonic saline, or hypotonic saline). A pharmaceutical composition (e.g., a pharmaceutical dosage form) comprises a sufficient amount of a tonicity agent to provide a hypertonic pharmaceutical dosage form for administration to a subject (e.g., a pharmaceutical dosage form having a calculated osmolality of at least 400mOsm/L (e.g., at least 500mOsm/L, at least 600mOsm/L, or at least 700mOsm/L), and/or 2,500mOsm/L or less (e.g.,2,000 mOsm/L, 1,900mOsm/L or less, 1,800mOsm/L or less, 1,700mOsm/L or less, 1,600mOsm/L or less, or 1,500mOsm/L or less)). For example, the target concentration of a tonicity agent in a pharmaceutical composition (e.g., a pharmaceutical dosage form) can be determined, for example, by: (i) subtracting the calculated osmolarity contributions of the anti-platinum chemoprotectant and other non-polymeric excipients from the total calculated osmolarity of the target to obtain a calculated osmolarity contribution from the tonicity agent, and (ii) determining the concentration of the tonicity agent by dividing the calculated osmolarity contribution from the tonicity agent by the number of ions and/or molecules produced when the tonicity agent is dissolved in the liquid solvent. Thus, a suitable amount of a tonicity agent may be included in a pharmaceutical composition (e.g., a pharmaceutical dosage form).
Buffering agents can be used to adjust the pH of a pharmaceutical composition (e.g., a pharmaceutical dosage form) to a substantially neutral pH level. Buffers are known in the art. Non-limiting examples of buffers include, for example, phosphate buffers and gule buffers (e.g., tris, MES, MOPS, TES, HEPES, HEPPS, tris (hydroxymethyl) methylglycine (tricine), and bicine). In addition to pH control, buffering agents can also be used to control the osmolarity of a pharmaceutical composition (e.g., a pharmaceutical dosage form).
Preparation method
Pharmaceutical compositions (e.g., pharmaceutical dosage forms) of the invention can be prepared from an anti-platinum chemoprotectant, a gelling agent, and a liquid solvent. The present invention provides a method of preparing a pharmaceutical composition (e.g., a pharmaceutical dosage form) of the present invention by: (i) providing an anti-platinum chemoprotectant and a gelling agent, and (ii) mixing the anti-platinum chemoprotectant and the gelling agent with a liquid solvent to produce a hypertonic pharmaceutical composition.
The anti-platinum chemoprotectant and gelling agent may be provided, for example, as a mixture or as separate components. When the anti-platinum chemoprotectant and the gelling agent are provided separately, step (ii) may include, for example:
(a) first mixing a liquid solvent with a gelling agent to produce an intermediate mixture, and then mixing the intermediate mixture with an anti-platinum chemoprotectant;
(b) first mixing a liquid solvent with an anti-platinum chemoprotectant to produce an intermediate mixture, and then mixing the intermediate mixture with a gelling agent; or
(c) Mixing a portion of the liquid solvent with an anti-platinum chemoprotectant to produce a first mixture, mixing another portion of the liquid solvent with a gelling agent to produce a second mixture, and combining the first mixture and the second mixture.
The following examples are intended to illustrate the invention. They are not meant to limit the invention in any way.
Examples
EXAMPLE 1 preparation of thiosulfate formulations
Hyaluronic acid gel 1(0.5M STS, 1% (w/v) hyaluronic acid)
Sodium thiosulfate pentahydrate (619.75mg) was dissolved in sterile distilled water (5mL) in a sterile vial to produce a clear solution. Hyaluronic acid (50.30 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to the solution, and the resulting mixture was stirred at 4 ℃ for 8-10 minutes. The resulting solution was filtered through a 0.22 μm Millex-GV sterile filter.
Hyaluronic acid gel 2(0.1M STS, 2% (w/v) hyaluronic acid)
Sodium thiosulfate pentahydrate (124.87mg) was dissolved in sterile distilled water (3.031 mL). Methylcellulose (351.01 mg;
Figure BDA0003410553030000261
a15 Premium LV, Dow Chemical Company) was dissolved in sterile distilled water (2.0mL) and the resulting solution was mixed with a sodium thiosulfate solution. Hyaluronic acid (100.10 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to the resulting mixture and mixed at 4 ℃ for 10-15 minutes.
Hyaluronic acid gel 3(0.5M STS, 2% (w/v) hyaluronic acid)
Sodium thiosulfate pentahydrate (620.35mg) was dissolved in sterile distilled water (3 mL). Methylcellulose (350.23 mg;
Figure BDA0003410553030000262
a15 Premium LV, Dow Chemical Company) was dissolved in sterile distilled water (2.0mL) and the resulting solution was mixed with a sodium thiosulfate solution. Hyaluronic acid (100.65 mg; pharmaceutical grade 80, Kikkoman Biochemifa com)pany; 0.6-1.2mDa) was added to the resulting mixture and mixed at 4 ℃ for 10-15 minutes.
Hyaluronic acid gel 4(0.1M STS, 1% (w/v) hyaluronic acid, mannitol)
Hyaluronic acid (50.09 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to water (5 mL). Sodium thiosulfate pentahydrate (124.9mg) was added. The pH of the resulting mixture was adjusted to pH7.12 by the addition of sodium hydroxide (1N, ca. 0.5. mu.L). An appropriate amount of mannitol was added to the vial to adjust the osmolality to 1.046 Osm/kg. The viscous solution was filtered through a 0.22 μm Millex-GV filter.
Hyaluronic acid gel 5(0.1M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 5 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a concentration of 0.1M sodium thiosulfate.
Hyaluronic acid gel 6(0.2M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 6 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a concentration of 0.2M sodium thiosulfate.
Hyaluronic acid gel 7(0.3M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 7 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a concentration of 0.3M sodium thiosulfate.
Hyaluronic acid gel 8(0.4M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 8 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a concentration of 0.4M sodium thiosulfate.
Hyaluronic acid gel 9(0.5M STS, 1% (w/v) hyaluronic acid, Tris (5X))
Hyaluronic acid (79.99 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to Tris buffer (8mL, AMRESCO-0497-500G). The pH of the resulting mixture was adjusted to pH7.13 by the addition of HCl (5N). Sodium thiosulfate pentahydrate (992.60mg) was added to the above solution. The viscous solution was filtered through a 0.22 μm Millex-GV filter.
Hyaluronic acid gel 10(0.5M STS, 1% (w/v) hyaluronic acid, phosphate buffered saline (5X))
Hyaluronic acid (70.38 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to PBS buffer (7mL, 5X). Sodium thiosulfate pentahydrate (868.46mg) was added. The pH of the resulting mixture was adjusted to pH6.99 by the addition of NaOH (1N). The viscous solution was filtered through a 0.22 μm Millex-GV filter.
Hyaluronic acid gel 11(0.8M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 11 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a concentration of 0.8M sodium thiosulfate.
Hyaluronic acid gel 12(1M STS, 0.8% (w/v) hyaluronic acid)
Hyaluronic acid gel 12 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a 1M concentration of sodium thiosulfate, and the amount of hyaluronic acid was adjusted to provide a 0.8% (w/v) concentration of hyaluronic acid.
Hyaluronic acid gel 13(0.5M STS, 0.82% (w/v) hyaluronic acid (HYALGAN))
The hyaluronic acid gel 13 was prepared by: sodium thiosulfate pentahydrate was mixed with hyaluronic acid (HYALGAN, Fidia Pharma USA, Florham Park, NJ) to provide a final preparation with hyaluronic acid concentration of 0.82% (w/v).
Hyaluronic acid gel 14(0.5M STS, 1% (w/v) hyaluronic acid (SINGCLEAN))
Hyaluronic acid gel 14 was prepared according to the procedure described for hyaluronic acid gel 13, except that hyaluronic acid (SINGCLEAN, Hangzhouh Singclean Medical Products co., ltd., Hangzhou, China) was used in the preparation of the gel.
Hyaluronic acid gel 15(0.5M STS, 1% (w/v) hyaluronic acid (EUFLEXXA))
Hyaluronic acid gel 15 was prepared according to the procedure described for hyaluronic acid gel 13, except that hyaluronic acid (EUFLEXXA, Ferring Pharmaceuticals inc., Parsippany, NJ) was used in the preparation of the gel.
Hyaluronic acid gel 16(0.5M STS, 1% (w/v) hyaluronic acid (HEALON))
Hyaluronic acid gel 16 was prepared according to the procedure described for hyaluronic acid gel 13, except that hyaluronic acid (helon, Johnson & Johnson, New Brunswick, NJ) was used in the preparation of the gel.
Hyaluronic acid gel 17(1M STS, 1% (w/v) hyaluronic acid)
Hyaluronic acid gel 17 was prepared according to the procedure described for hyaluronic acid gel 1, except that the amount of sodium thiosulfate pentahydrate was adjusted to provide a 1M concentration of sodium thiosulfate.
Hyaluronic acid gel 18 (10% (w/v) N-acetyl-L-cysteine, 1% (w/v) hyaluronic acid)
Hyaluronic acid (39.38 mg; pharmaceutical grade 80, Kikkoman Biochemifa company; 0.6-1.2mDa) was added to water (4 mL). N-acetyl-L-cysteine (399.14mg) was added. The pH of the resulting mixture was adjusted to pH 7.21 by the addition of NaOH (10N, 240. mu.L). The viscous solution was filtered through a 0.22 μm Millex-GV filter. The osmotic pressure was measured to be 1.107 Osm/kg.
Other hyaluronic acid gels can be prepared using the procedures described herein. For example, 1M and 1.5M hyaluronic acid gels can be prepared according to the same procedures as described for, e.g., hyaluronic acid gel 1 and hyaluronic acid gel 12. In addition, the pH level of the gel may be adjusted to pH 6.5 to 8.5 with a bronsted acid (e.g., hydrochloric acid) and a base (e.g., sodium hydroxide).
Example 2 pharmacokinetic modeling of perilymph fluid concentration
Treatment with high dose cisplatin (100 mg/m) by Pharmacokinetic (PK) modeling using cisplatin distribution in humans2) Largest cochlear platinum water in the posteriorAnd carrying out modeling. Kinetic parameters for the simulations were obtained from literature reports on cisplatin population PK (Urien et al, Br. J. Clin. Pharmacol.,57: 756. Sci. 63,2004) and PK after high dose cisplatin treatment (Andersson et al, J. pharm. Sci.,85: 824. 27, 1996). PK simulations were performed using WinNonlin (Phoenix64) PK simulation model 9 (intravenous infusion, 2-chamber). Based on previous animal studies, the plasma to cochlear concentration ratio was assumed to be 1:1, and this hypothesis was further supported by literature reports showing tissue distribution characteristics of cisplatin (Johnsson et al, Cancer chemi.
The predicted maximum plasma platinum concentration following cisplatin treatment was determined to be about 22 μ M. The use of a 30-fold molar stoichiometric ratio resulted in a concentration of 660. mu.M (0.66mM), which when using the molecular weight of thiosulfate, resulted in a thiosulfate concentration of 74. mu.g/mL. Achieving this thiosulfate level in the human cochlea is expected to provide contrast at high doses (e.g., 100 mg/m)2) Complete (maximal) protection of cisplatin-induced ototoxicity following cisplatin treatment.
Example 3 in vivo pharmacokinetic Studies
Hyaluronic acid gel number 1 was administered to male Hartley guinea pigs at a dose of 12% w/v (0.5M,6.2 mg). The dose volume was fixed (10 μ L). Perilymph fluid was sampled at each time point (n ═ 5 animals/time point) and the concentration of thiosulfate was quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS).
Hyaluronic acid gel 1 reached a maximum perilymph fluid concentration of 868.5 μ g/mL (about 7.8mM) at the first sampling time point (1 h). This maximum perilymph fluid concentration was about 10-fold higher than the hyaluronic acid gel 1 concentration (minimum effective concentration; 74. mu.g/mL, 0.66mM) that was expected to provide 100% protection against cisplatin-induced ototoxicity. Perilymph fluid t1/2In the range of 2.7 hours to 6.4 hours. The combination of high perilymph fluid hyaluronic acid gel 1 concentration with a relatively long half-life provides a therapeutic window from 3 hours prior to cisplatin treatment to 4 hours post-cisplatin treatment. In another pharmacokinetic study, healthy subjects were divided into 4 dose groups: group 1, group 2, group 3, and group 4. Each one of which isThe dose cohort contained 8 human subjects randomized to receive either DB-020 or placebo (6/2 randomized; 6 subjects received DB-020, 2 subjects received placebo). Group 1 was administered to the unilateral intratympanic cavity 19mg of sodium thiosulfate, a 0.15M sodium thiosulfate/hyaluronic acid gel prepared as described for hyaluronic acid gel 1. Group 2 was administered to the unilateral intratympanic cavity 62mg of sodium thiosulfate as hyaluronic acid gel 1. Group 3 was administered into the unilateral intratympanic cavity 124mg of sodium thiosulfate as hyaluronic acid gel 17. Group 4 was administered into the unilateral intratympanic cavity 186mg of sodium thiosulfate, a 1.5M sodium thiosulfate/hyaluronic acid gel prepared as described for hyaluronic acid gel 1. A1% w/v aqueous solution of hyaluronic acid was administered to placebo subjects.
The results of the study are shown in tables 1 and 2 and fig. 1 to 3.
TABLE 1
Figure BDA0003410553030000311
TABLE 2
Figure BDA0003410553030000321
aIncreased to endogenous ═ by (average thiosulfate C)max) - (mean thiosulfate placebo level)
bThiosulfate having MW of 112g/mol
Example 4 in vivo pharmacodynamic Studies
To assess the therapeutic window associated with cisplatin administration, a single dose of 1.24mg hyaluronic acid gel 1 was administered Intratympanically (IT) to the left ear of guinea pigs 24 hours, 6 hours, 3 hours, or 1 hour prior to cisplatin administration, or 1 hour, 4 hours, or 24 hours after cisplatin administration (a single bolus of 10mg/kg intravenous; fig. 4). All control right ears not treated with hyaluronic acid gel 1 showed a significant threshold shift (i.e., hearing loss) compared to the non-experimentally treated animals (fig. 5A). Hyaluronic acid gel 1 administered 3 hours prior to cisplatin administration to 4 hours post-cisplatin administration provided protection against cisplatin-induced hearing loss relative to control ears not treated with hyaluronic acid gel 1 (figure 5B). When administered further distally in cisplatin treatment (e.g., > 6h before or 24h after cisplatin administration), hyaluronic acid gel 1 was slightly less effective in protecting against cisplatin-induced hearing loss. The greatest protection was observed when hyaluronic acid gel 1 was administered 1h before cisplatin administration, indicating that the highest level of protection can be achieved when hyaluronic acid gel 1 was administered before cisplatin treatment and preferably proximal to cisplatin treatment.
Example 5 pharmacokinetic Properties of exemplary hydrogels
Guinea pig, study 1
Studies were conducted using albino guinea pigs (Hartley) weighing 250-350 g. For administration, the animal is placed against its shoulder with the surgical ear facing up and the posterior auricular region is first used to access and expose the auditory bulb. A hole of 2-3mm diameter was drilled in the auditory bulb to provide direct visualization of the round window microenvironment. Then, 10 μ L of an aqueous composition of 0.5M sodium thiosulfate/2% (w/v) hyaluronic acid (STS composition) was applied to the RWM using a 10 μ L Hamilton syringe and 26 gauge needle. After application, guinea pigs were held in this position for 30 minutes to allow diffusion of the compound into the cochlea. The opening of the bulla was closed with a muscle graft and the incision was closed with sutures.
Briefly, the sampling procedure is as follows. All sampling procedures were end-point. With CO2Animals were euthanized. A0.5 mL blood sample was collected by cardiac puncture. Plasma was separated by centrifugation at 5,000rpm for 10 minutes at 4 ℃ and collected in separate tubes. 50 μ L of cerebrospinal fluid was collected through the cisterna magna. Perilymph fluid was collected ex vivo to avoid contamination from cerebrospinal fluid inflow via the cochlear aqueduct. The temporal bone was quickly detached and the auditory bulb was then removed to expose the cochlea. Any remaining dose of the composition that was visible was carefully removed under a surgical microscope with a moisture absorbing tip (absorbent point) before sampling of the perilymph fluid. Perforating the tip and then sampling 5-7. mu.L using a drawn glass pipetteThe external lymph is liquid. All samples were immediately frozen on dry ice and stored at-80 ℃ until analysis. The concentration of thiosulfate in a sample is measured using the method disclosed in Togawa et al, chem.pharm.Bull.,40:3000-3004,1992, the disclosure of which is incorporated herein by reference. The results of this study are shown in fig. 8A, 8B, 9A, 9B and table 3.
Macaca fascicularis
Touretidine (4mg/kg) was administered subcutaneously to cynomolgus monkeys. After 30 minutes, the animals were anesthetized by an intravenous bolus injection of propofol (5.5 mg/kg). The animals were then maintained under anesthesia using 2-3% isoflurane inhalation. The animals were then fixed and placed laterally in the anti-trendelenburg position to ensure a round window of access. During the surgical procedure, the animal was kept on a warm blanket.
When the animal reached anesthesia, an intratympanic injection was performed in the right ear. 1.1mL of epinephrine hydrochloride-saline (0.1mg dissolved in 10mL of saline) and 0.5mL of lidocaine hydrochloride (20mg/mL) were subcutaneously injected as local anesthetics into the skin of the posterior wall of the ear canal of each ear, respectively. An incision is then made in the posterior auricle skin and a portion of the temporal bone is drilled away to expose the middle ear. 50 μ L of STS composition was injected into the round window membrane using a 25G needle. After dosing, the animals were placed in line, head up, to allow the drug solution to remain in the tympanic cavity for 30 minutes. The same procedure was then repeated for the other ear.
Plasma and CSF were collected about 2 hours after administration to ear 1 (right). Sampling of the pericochlear lymph fluid from the right ear was performed approximately 3 hours after administration to the right ear. Animals were euthanized by intravenous injection of 11mg/kg propofol and then bled via the femoral artery. The animals were then placed in a lateral decubitus position. A posterior auricular skin incision was made and the external auditory meatus was removed to expose the middle ear. A portion of the temporal bone is then drilled away to expose the basal turn of the cochlea (basal turn). The remaining dose (if visible) in the middle ear was cleaned with a cotton swab. A drop of tissue glue was applied at the base of the cochlea to minimize contamination from the administered composition.
A round burr crochet (burr crochet) or a sharp crochet (sharp crochet) of 0.5-1mm is used to perforate the bottom of cochlea. Perilymph fluid (approximately 10 μ L) was then collected using a capillary tube inserted into the scala tympani of the cochlea. The same procedure was repeated for left ear pericochlear lymph fluid sampling approximately 2 hours after left ear administration. The results of this study are shown in table 3.
TABLE 3
Figure BDA0003410553030000341
In the above table, IT is intratympanic administration and TT is transtympanic administration.
Thiosulfate concentrations measured from test animal plasma samples.
Guinea pig, study 2
Male guinea pigs of about 5-7 weeks of age with a weight of 200-. Prior to any procedure, animals were anesthetized by intramuscular route with zolazepam hydrochloride (shutitan 50; 20mg/kg) 10 minutes prior to surgery. If desired, one tenth of the original dose of intraoperative reinforcement agent is administered intraperitoneally.
Intratympanic injection:
1. under microscopic magnification, a 0.5-1.5cm retroauricular skin incision was made with sharp scissors at about 6-8mm caudal to the ear-head fold. Care was taken to avoid cutting too deep to protect the underlying vascular structure.
2. Blunt dissection was performed carefully through the subcutaneous fat layer, muscle and tissue with forceps. The mastoid body was gently retracted until a smooth dome of bubbling periosteum was seen. At the tail of the auditory bulb, the deeper cervical muscle, the attachment of the mastoid muscle, can be seen. The facial nerve, which becomes visible at the back and at the lips of the alveolar dome, is preserved during surgery.
3. In the posterior portion of the bulla, a self-retaining retractor was placed before drilling a small hole (0.5 mm diameter) using a drill. The alveolar bone is cut in the dorsal and caudal directions with a pair of jewelry point forceps. At high magnification, the bones are removed one by one. Care was taken not to puncture the stapedial artery, which is located directly under the bulb cap (bulla cap), as bleeding from this artery may compromise the surgical procedure. The amount of bone removed is kept to a minimum to prevent excessive fluid from entering the middle ear while still allowing good visual results and access to the round window microenvironment.
4. 10 or 90 μ L of the gel formulation was delivered into a round window microenvironment using a sterile glass Hamilton syringe with a 25-26G blunt needle.
5. The delivered agent is allowed to reside within the round window microenvironment for up to 30 minutes. This aperture is covered by muscle tissue and tissue glue.
6. The incision is closed with sutures (4-0 non-absorbable monofilament or 5-0 non-absorbable nylon) and tissue glue or wound clips. The entire process takes about 3-5 minutes, depending on the formulation specifications.
7. During surgery and until recovery, the animals were placed on a temperature-controlled (38 ℃) heating pad until consciousness was restored, at which time the animals were returned to a home-cage.
Alternatively, the gel formulation is administered to the animal via the tympanic cavity.
Collecting samples:
blood collection:
1. without pre-aeration in the euthanizer, the guinea pigs were placed in the euthanizer and 100% carbon dioxide was introduced to unconscious and reduce pain in the animals. After cessation of breathing, the carbon dioxide flow was maintained for a minimum of 1 minute. After confirmation of death, the guinea pigs were removed from the euthanasia chamber.
2. Blood was collected immediately after euthanasia.
3. After the operator has fixated the animal in the dorsal position, the needle is inserted 4-6 or slightly anteriorly at the anterior portion of the sternal spine.
4. The blood is returned by pulling back the needle.
5. Volume: for each blood collection, approximately 1mL of blood was collected.
And (3) CSF collection:
CSF was collected after euthanasia. 0.5 x 20 intravenous infusion needle was slowly lowered from 90 ° relative to the foramen magnum. The needle was brought to a distance of 4.5-5mm under the skin and 50-200. mu.l of clear interstitial fluid was removed.
Collecting the perilymph fluid:
after euthanasia, animals were stripped of excess skin and muscle tissue to obtain a complete bulla, and the bulla wall was cut open with small forceps to expose the cochlea. The bottom rotation of the auditory bulb was cleaned with a small cotton ball. The cochlea diaphragma and round window were coated with biogum. After drying, unique microholes were manually drilled at the apical ring of the cochlea. Then 2 μ L of perilymph fluid was collected using a microcapillary tube inserted into the apical ring of the cochlea. The perilymph fluid samples were added to vials containing 18 μ L of bovine serum albumin (BSA, 1M) and stored at-80 ℃ until analysis.
Results of guinea pig study 2 are provided in tables 4 and 5.
TABLE 4
Figure BDA0003410553030000371
Figure BDA0003410553030000381
In this table, TT is administered trans-tympanic,
this test is a repeat of the previous test.
TABLE 5
Figure BDA0003410553030000382
In the table, IT is intratympanic administration and TT is transtympanic administration.
Example 6 pharmacodynamic Properties of exemplary hydrogels
Cisplatin was diluted with 0.9% (w/v) saline to a final concentration of 5 mg/mL. Albino guinea pigs (Hartley) weighing 250-350g were used for the study. After at least 3 days of acclimation, 28 animals were included in the study. Cisplatin was administered intraperitoneally using a bolus injection under sterile conditions. Five groups were staggered with different start dates for study.
7 days after cisplatin administration, Auditory Brainstem Responses (ABR) of animals were recorded using a TDT RZ6 multiple input multiple output processor. Historical ABR data is used to define the baseline. Animals were anesthetized with teletamine hydrochloride and zolazepam hydrochloride (shutitan). The sound stimulus is delivered via headphones. The needle electrodes are placed at the caudal-ventral position near the ear canal, at the vertex of the skull, and at the base of the calf (ground). The stimulation level was from 10dB to 90dB in 5dB steps and the short tone frequencies were 4kHz, 24kHz and 32 kHz. The upper limit of the sound pressure level is 90 dB. The ABR threshold is observed by visually inspecting the stacked waveforms where the waveform is above the noise floor as the lowest sound pressure level.
ABR data were recorded in both ears of each animal from 50 animals (non-experimentally treated n-100) prior to the cisplatin study. The threshold at 32kHz for the non-experimentally treated animals was 39.8 dB. The normal hearing range is defined as mean ± 2SD, 27.9 to 51.6 dB. Cisplatin mainly induces hearing loss at high frequencies. The clear pattern of post-cisplatin hearing loss is limited to a threshold of 60dB and above at 32 kHz.
In this study, 1 of 28 animals died before day 7 of the measurement. Of the remaining 27 animals, 18 had hearing loss at 32kHz with a threshold >60dB (fig. 10A). The hearing loss at 32kHz ranges from 65dB to a threshold of 90dB (fig. 10B). 90dB is the upper measurement limit. It should be noted that the threshold is defined as 90dB when there is no waveform or a waveform is seen only at 90 dB. The average threshold at 32kHz was 82dB, which corresponds to an average of 42.2dB from the initial threshold offset of 39.8dB (fig. 11A).
Local intratympanic drug delivery and cochlear sampling
Local intratympanic administration and cochlear sampling were performed as described in example 5.
Topically delivered anti-platinum chemoprotectants provide hearing protection from platinum-based antineoplastic agents.
Evaluation of the hearing protective effect of a topically delivered anti-platinum chemoprotectant against a platinum-based antineoplastic agent was performed as follows.
An aqueous composition of 0.5M sodium thiosulfate/2% (w/v) hyaluronic acid (STS composition) or vehicle was administered intraventricularly to the round window in the Left Ear (LE) as described above, and the Right Ear (RE) of the guinea pig was left untreated (fig. 12). Animals were injected intraperitoneally with 10mg/kg cisplatin 60 minutes after STS composition or vehicle administration. ABR at 4kHz, 24kHz, and 32kHz were measured in both ears 7 days after cisplatin administration.
Due to the heterogeneity of hearing loss after cisplatin challenge, untreated right ears were used to select animals with hearing loss. 21 animals had a right ear threshold >60dB at 32 kHz. Of these 21 animals, 3 animals with otitis media were excluded, and 18 animals were left for final analysis. Vehicle was administered to 10 animals and STS composition was administered to 8 animals (fig. 11B). In the untreated right ear, there was no difference in ABR thresholds for both the STS composition group and vehicle group, with an average threshold of 73dB at 4kHz, 71dB at 24kHz, and 80dB at 32 kHz. Vehicle-treated left ear was not significantly different from untreated right ear, showing a threshold of 74dB at 4kHz, 70dB at 24kHz, and 74dB at 32 kHz.
The STS composition treated ears had significantly lower thresholds at both 32KHz and 24KHz (×) P <0.001, two-way anova) compared to vehicle treated ears and untreated right ears. At 4kHz, the average threshold was 61dB in the STS composition treated ear and 75dB in the untreated contralateral right ear; protection was not statistically significant (P ═ 0.089). The average threshold at 24kHz and 32kHz in the STS composition treated ear was 40dB and 48dB, respectively, compared to 69dB and 80dB in the contralateral untreated right ear. The normal hearing thresholds in the non-experimentally treated animals were 35dB and 40dB at 24kHz and 32kHz, respectively. For the ears of the non-experimentally treated animals, the untreated ears after cisplatin had an average threshold rise of 34dB and 40dB at 24kHz and 32kHz, respectively, but the ears treated with the STS composition had only 5dB and 8dB shifts. Thus, sodium thiosulfate provided an average of 80% protection at both 24kHz and 32 kHz.
In a similarly designed study as described above, sound pressure levels at 4kHz, 24kHz and 32kHz were measured during ABR testing in guinea pigs administered either vehicle or sodium thiosulfate (0.1M, 0.5M or 1M sodium thiosulfate gel) per ear followed by cisplatin challenge (10MPK of cisplatin, intravenous injection). Different doses of hyaluronic acid gel were administered as 10 μ Ι _ IT injections into the left ear 1 hour before cisplatin administration. The contralateral ear (right ear) of the animal was untreated. Hyaluronic acid gel 5(0.1M), hyaluronic acid gel 1(0.5M) and hyaluronic acid gel 17(1M) were tested. The untreated ear showed a significant threshold shift compared to the non-experimentally treated animals (grey shaded area). At all frequencies tested, the groups treated with hyaluronic acid gel 1(0.5M) and hyaluronic acid gel 17(1M) showed hearing protection compared to untreated contralateral control ears. No protection was seen in vehicle treated ears. The results are summarized in FIG. 13.
Other embodiments
Various modifications and alterations of this invention will become apparent to those skilled in the art without departing from the scope and spirit of this invention. While the invention has been described in connection with specific embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention which are obvious to those skilled in the art are intended to be within the scope of the invention.
Other embodiments are within the scope of the following claims.

Claims (20)

1. A method of reducing platinum-induced ototoxicity in a subject in need thereof, the method comprising administering to the subject an effective amount of thiosulfate, wherein the subject is administered a platinum-based antineoplastic agent no more than 7 hours prior to administration of the thiosulfate, or is scheduled to be administered a platinum-based antineoplastic agent within 4 hours.
2. The method of claim 1, wherein the subject is administered a platinum-based oncology agent no more than 7 hours prior to administration of the thiosulfate salt.
3. The method of claim 1, wherein the subject is scheduled to administer a platinum-based anti-neoplastic agent within 4.5 hours.
4. The method of claim 1, wherein the subject is administered a platinum-based oncology agent no more than 2.5 hours prior to administration of the thiosulfate salt.
5. The method of claim 1, wherein the subject is administered a platinum-based oncology agent no more than 1 hour prior to administration of the thiosulfate salt.
6. The method of claim 1, wherein the thiosulfate salt is administered otically.
7. The method of claim 6, wherein the thiosulfate salt is administered intratympanically, transcompartually, or by injection in the inner ear.
8. The method of claim 1, further comprising administering the platinum-based anti-neoplastic agent.
9. The method of claim 1, wherein the thiosulfate salt is an alkaline thiosulfate salt, an ammonium thiosulfate salt, or a solvate thereof.
10. The method of claim 1, wherein the effective amount of thiosulfate is administered as a hypertonic pharmaceutical composition comprising the effective amount of thiosulfate.
11. The method of claim 10, wherein 200 μ L of the hypertonic pharmaceutical composition is administered to the round window of the subject.
12. The method of claim 10 wherein the calculated osmolality of the hypertonic drug composition is 500-5,000 mOsm/L.
13. The method of claim 10, wherein the concentration of the thiosulfate salt in the hypertonic pharmaceutical composition is 0.5M-2.5M.
14. The method of claim 1, wherein the effective amount is an amount that results in a plasma thiosulfate concentration of 30 μ Μ or less when the platinum-based anti-neoplastic agent is administered.
15. The method of claim 1, wherein said effective amount is 0.1-2.5mmol of said thiosulfate.
16. The method of claim 1, wherein the effective amount is an amount that results in a maximum thiosulfate concentration of 0.6-10 mmol/L1 h after administration.
17. The method of claim 1, wherein the effective amount is an amount that results in a thiosulfate concentration of 0.1-2 mmol/L7 hours after administration in the cochlea of the subject.
18. A method of reducing platinum-induced ototoxicity in a subject in need thereof, the method comprising administering to the subject an effective amount of thiosulfate to produce (i) plasma thiosulfate C that is 30 μ M or less at the time of administration of a platinum-based anti-neoplastic agentmaxAnd (ii) cochlea C that is the platinum-based anti-neoplastic agentmaxAt least 30 times as large as cochlear thiosulfate CmaxWherein the cochlear platinum concentration and the cochlea CmaxThe modeling was performed by pharmacokinetic simulation of intravenous infusion in a two-compartment model.
19. The method of claim 18, wherein the effective amount of thiosulfate is administered within 3 hours before administration of the platinum-based anti-neoplastic agent or within 4 hours after administration of the platinum-based anti-neoplastic agent.
20. The method of claim 18, wherein the effective amount of thiosulfate is administered within 4.5 hours before or within 2.5 hours after the platinum-based anti-neoplastic agent.
CN202080043662.0A 2019-05-31 2020-05-29 Otoprotectant methods against platinum-based antineoplastic agents Pending CN114302647A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US16/427,567 2019-05-31
US16/427,567 US10813947B1 (en) 2019-05-31 2019-05-31 Methods of otoprotection against platinum-based antineoplastic agents
US201962886865P 2019-08-14 2019-08-14
US62/886,865 2019-08-14
PCT/US2020/035271 WO2020243536A1 (en) 2019-05-31 2020-05-29 Methods of otoprotection against platinum-based antineoplastic agents

Publications (1)

Publication Number Publication Date
CN114302647A true CN114302647A (en) 2022-04-08

Family

ID=73553934

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080043662.0A Pending CN114302647A (en) 2019-05-31 2020-05-29 Otoprotectant methods against platinum-based antineoplastic agents

Country Status (11)

Country Link
EP (1) EP3975722A4 (en)
JP (1) JP2022534617A (en)
KR (1) KR20220047217A (en)
CN (1) CN114302647A (en)
AU (1) AU2020284122A1 (en)
BR (1) BR112021023684A2 (en)
CA (1) CA3142153A1 (en)
IL (1) IL288532A (en)
MX (1) MX2021014447A (en)
SG (1) SG11202113053RA (en)
WO (1) WO2020243536A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110008467A1 (en) * 2009-07-08 2011-01-13 Craig Sherman Sodium thiosulfate-containing pharmaceutical compositions
CN102083436A (en) * 2008-06-18 2011-06-01 帕特里克·普伦德加斯特 Anti-tumour compositions and methods
US9427472B2 (en) * 2008-07-21 2016-08-30 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
US20190160094A1 (en) * 2017-11-29 2019-05-30 Edward A. Neuwelt Method for reducing ototoxicity in pediatric patients receiving platinum-based chemotherapy

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220296637A1 (en) * 2019-06-19 2022-09-22 Stichting Het Nederlands Kanker Instituut-Antoni van Leeuwenhoek Ziekenhuis Sodium thiosulfate gel for preventing or reducing hearing loss

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102083436A (en) * 2008-06-18 2011-06-01 帕特里克·普伦德加斯特 Anti-tumour compositions and methods
US9427472B2 (en) * 2008-07-21 2016-08-30 Otonomy, Inc. Controlled release compositions for modulating free-radical induced damage and methods of use thereof
US20110008467A1 (en) * 2009-07-08 2011-01-13 Craig Sherman Sodium thiosulfate-containing pharmaceutical compositions
US20190160094A1 (en) * 2017-11-29 2019-05-30 Edward A. Neuwelt Method for reducing ototoxicity in pediatric patients receiving platinum-based chemotherapy

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BERGLIN ET AL.: "Prevention of cisplatin-induced hearing loss by administration of a thiosulfate-containing gel to the middle ear in a guinea pig model", 《CANCER CHEMOTHER PHARMACOL》, vol. 68, pages 1547, XP019980101, DOI: 10.1007/s00280-011-1656-2 *
MEYER ET AL.: "Randomized controlled trial to test the efficacy of trans-tympanic injections of a sodium thiosulfate gel to prevent cisplatin-induced ototoxicity", 《JOURNAL OF CLINICAL ONCOLOGY》, vol. 35, no. 15, pages 1 *

Also Published As

Publication number Publication date
IL288532A (en) 2022-01-01
MX2021014447A (en) 2022-02-11
JP2022534617A (en) 2022-08-02
EP3975722A4 (en) 2023-02-08
EP3975722A1 (en) 2022-04-06
SG11202113053RA (en) 2021-12-30
CA3142153A1 (en) 2020-12-03
AU2020284122A1 (en) 2021-12-23
KR20220047217A (en) 2022-04-15
WO2020243536A1 (en) 2020-12-03
BR112021023684A2 (en) 2022-04-12

Similar Documents

Publication Publication Date Title
US11857567B2 (en) Hypertonic pharmaceutical compositions containing an anti-platinum chemoprotectant agent
US10925889B2 (en) Method of treating, reducing, or alleviating a medical condition in a patient
EP3294212B1 (en) Drug delivery from hydrogels
US9066865B2 (en) Pharmaceutical compositions for the treatment of inner ear disorders
US11690870B2 (en) Methods of otoprotection against platinum-based antineoplastic agents
WO2011063606A1 (en) Ophthalmic gel of gatifloxacin and preparation method thereof
CN107848973A (en) The dimethylaniline formyl piperidine crystal of 1 propyl group of S () 2 &#39;, 6 &#39; and its sustained release preparation
US20220296637A1 (en) Sodium thiosulfate gel for preventing or reducing hearing loss
CN114302647A (en) Otoprotectant methods against platinum-based antineoplastic agents
KR101877894B1 (en) Injectable combination drug for treating hearing loss and method of preparing the same
US20240131055A1 (en) Methods of otoprotection against platinum-based antineoplastic agents
RU2323726C1 (en) Composition with local anesthetic activity
CN116196266A (en) Local anesthetic low molecular weight gel, liposome and preparation method thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination