US20230293538A1 - Methods for the Prevention and Treatment of Hearing Loss - Google Patents

Methods for the Prevention and Treatment of Hearing Loss Download PDF

Info

Publication number
US20230293538A1
US20230293538A1 US18/117,852 US202318117852A US2023293538A1 US 20230293538 A1 US20230293538 A1 US 20230293538A1 US 202318117852 A US202318117852 A US 202318117852A US 2023293538 A1 US2023293538 A1 US 2023293538A1
Authority
US
United States
Prior art keywords
hearing
momelotinib
treatment
compounds
hearing loss
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.)
Granted
Application number
US18/117,852
Other versions
US11766440B1 (en
Inventor
Jian Zuo
Zhuo Li
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.)
Ting Therapeutics LLC
Original Assignee
Ting Therapeutics LLC
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
Application filed by Ting Therapeutics LLC filed Critical Ting Therapeutics LLC
Priority to US18/117,852 priority Critical patent/US11766440B1/en
Publication of US20230293538A1 publication Critical patent/US20230293538A1/en
Application granted granted Critical
Publication of US11766440B1 publication Critical patent/US11766440B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61FFILTERS IMPLANTABLE INTO BLOOD VESSELS; PROSTHESES; DEVICES PROVIDING PATENCY TO, OR PREVENTING COLLAPSING OF, TUBULAR STRUCTURES OF THE BODY, e.g. STENTS; ORTHOPAEDIC, NURSING OR CONTRACEPTIVE DEVICES; FOMENTATION; TREATMENT OR PROTECTION OF EYES OR EARS; BANDAGES, DRESSINGS OR ABSORBENT PADS; FIRST-AID KITS
    • A61F11/00Methods or devices for treatment of the ears or hearing sense; Non-electric hearing aids; Methods or devices for enabling ear patients to achieve auditory perception through physiological senses other than hearing sense; Protective devices for the ears, carried on the body or in the hand
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/60Salicylic acid; Derivatives thereof
    • A61K31/609Amides, e.g. salicylamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/7036Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin having at least one amino group directly attached to the carbocyclic ring, e.g. streptomycin, gentamycin, amikacin, validamycin, fortimicins
    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals

Definitions

  • the present invention relates to therapeutic uses of compounds for treating, inhibiting, and/or preventing loss of hearing.
  • NIHL Antibiotic-induced hearing impairment
  • Neonates admitted to the neonatal intensive care unit have ten times the rate of hearing loss as neonates not admitted, often associated with the administration of antibiotics.
  • the spiral-shaped cochlea of the inner ear is responsible for detecting sound.
  • Inner hair cells lining the cochlea transform the mechanical vibrations of sound waves into chemical signals. These chemicals are then released from the hair cells and received by receptors on the auditory nerve fibers that send electrical impulses to the brain.
  • the inner ear cells lining the cochlea can be destroyed by the use of antibiotics, leading to a condition called sensorineural hearing impairment.
  • hearing aid which functions to amplify sound.
  • Hearing aids are non-invasive and can improve an individual's ability to hear.
  • hearing aids can often be quite conspicuous and embarrassing to the wearer and hearing aids do not return hearing to normal levels.
  • hearing aids amplify sound indiscriminately, sometimes amplifying sounds that an individual does not wish to hear, such as environmental noise.
  • the invention provides a method to prevent or treat hearing loss including the steps of administering to an animal in need thereof an effective amount of a pharmaceutical composition containing a therapeutically active agent, wherein the therapeutically active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1.
  • the therapeutically active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD
  • the inventive subject matter also includes a composition for use in preventing or treating hearing loss by protecting inner ear cells from death wherein the composition is an effective amount of an active agent, wherein the active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1 or a pharmaceutically acceptable salt thereof.
  • the active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib,
  • the inventive subject matter also includes a kit made of an active agent, wherein the active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1 or a pharmaceutically acceptable salt thereof; and one or more of: (A) at least one antibiotic; (B) at least one cancer drug (C) instructions for preventing a hearing impairment.
  • the active agent includes: momelotinib, fedratinib, axitinib, sunitinib,
  • FIG. 1 is a schematic diagram showing how compounds of this invention were identified based on a comparison of data sets from drug screen connectivity maps and pathway enrichment analysis revealing drugs acting against hearing loss.
  • FIG. 2 shows outer hair cell protection by Momelotinib against aminoglycosides.
  • B) shows outer hair cell (OHC) survival (OHC number per 100 ⁇ m length of cochlear middle turn) after treatment with various concentration of Momelotinib and 50 mg/mL gentamicin or 600 mM kanamycin. (Mann-Whitney U Test, *P ⁇ 0.05, **P ⁇ 0.01) shown in a micromolar concentration.
  • FIG. 3 A-C show Momelotinib (MMB) protected zebrafish mechanosensory hair cells against gentamicin (GM) and kanamycin (KM) in vivo. 5 dpf Tg(Brn3c:mGFP) larvae were used for the experiments. 3 A and 3 B were treated with KM (200 uM for 1 hour and 150 ⁇ M for 6 hours, respectively) and 3 C was treated with GM (100 uM for 1 hour). Each treatment was pretreated with MMB at the prescribed doses prior to co-treatment with the aminoglycoside. Control animal were treated with DMSO ( ⁇ ) or KM or GM alone (+) Animals were then fixed and immunostained for GFP and otoferlin.
  • MMB Momelotinib
  • GM gentamicin
  • KM kanamycin
  • FIG. 5 shows Momelotinib is not toxic in C57BL/6 mice (4-week-old) in vivo up to a dose of 100 mg/kg/day over a 14-day treatment regimen. Oral administration of Momelotinib did not result in any general or inner ear toxicity, measured as and hearing function in 28-day old C57BL/6 mice. Auditory brainstem response (ABR) thresholds were recorded for 3 frequencies (8 kHz, 16 kHz, and 32 kHz) before and after a 14-day MMB treatment protocol. No differences were observed across all frequencies tested. Statistical analysis was done with repeated measures ANOVA between groups.
  • compounds can be used as a therapy for the treatment and/or prevention of hearing loss.
  • the compounds and compositions of the invention can be administered in pharmaceutical compositions, which are formulated according to the intended method of administration.
  • the compounds of this invention are defined as a therapeutically active agent in a treatment regimen or procedure that is intended for preventing hearing loss by noise or aging by protecting inner ear cells from death and in preventing hearing loss by chemotherapy or antibiotics induced hearing loss.
  • Therapeutic agent means a chemical substance that is used for the treatment or mitigation of a disease condition or ailment.
  • compounds can be used as a therapy for the treatment and/or prevention of hearing loss.
  • the compounds and compositions of the invention can be administered in pharmaceutical compositions, which are formulated according to the intended method of administration.
  • the compounds of this invention are defined as a therapeutically active agent in a treatment regimen or procedure that is intended for preventing hearing loss by noise or aging by protecting inner ear cells from death and in preventing hearing loss by chemotherapy or antibiotics induced hearing loss.
  • Therapeutic agent means a chemical substance that is used for the treatment or mitigation of a disease condition or ailment.
  • FIG. 1 compounds were identified based on a comparison of data sets from drug screen connectivity maps and pathway enrichment analysis revealing compounds acting against hearing loss. This method was developed to derive drug candidates from a diverse chemical space, covering a wide range of biological pathways, avoiding bias associated with focusing on previously reported pathways. The resulting compounds exhibited overlaps in gene expression transcriptomic profiles between at least one of a plurality of cell-lines or mouse strains treated with ototoxic insults (cisplatin FIG. 5 A /D, noise FIG. 5 B , or antibiotic exposure FIG. 5 C ) and at least one of a plurality of cell-lines or mouse strains treated with one of the compounds.
  • ototoxic insults cisplatin FIG. 5 A /D, noise FIG. 5 B , or antibiotic exposure FIG. 5 C
  • the data set sought was conformity to a NIHL-resistant mouse strains to NIHL sensitive mouse strains (129 SvJ and CAST).
  • the data set sought was conformity to cisplatin-resistant and sensitive cancer cell lines, HEI-OC1 cell line, in vivo mouse cochlear single cell RNA seq with and without cisplatin treatment.
  • the data set greatest weigh was transcriptome perturbation of neonatal mouse organ of Corti exposed to gentamicin for damage related to antibiotic treatment.
  • These compounds with an overlap of the data sets include as the therapeutically active agent: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1.
  • Compounds are revealed to protect against hair cell apoptosis. Compounds are identified as acting against hair cell loss in animals by the models and data presented. Models reveal properties necessary for an otoprotective compound such as high efficacy against hair cell loss, relatively low toxicity. Compounds are revealed to have high efficacy and high affinity in mouse and zebrafish models used to demonstrate protection against hair cell loss. The lateral-line neuromasts of zebrafish are a valuable model for testing compounds protective against hearing loss in vivo, as their HCs are considered homologous to those in the mammalian inner ear and are readily accessible to drugs in vivo. Teitz et al., J. Exp. Med. 2; 215(4):1187-1203 (2016).
  • compositions described herein can be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients.
  • a pharmaceutical composition can be formulated for local or systemic administration, e.g., administration by drops or injection into the ear, insufflation (such as into the ear), intravenous, topical, or oral administration.
  • Compounds can be synthesized by a variety of methods known in the art.
  • Fedratinib is an FDA-approved therapy for patients with myeloproliferative neoplasm-associated myelofibrosis.
  • Momelotinib is in phase 3 clinical trials for the treatment of patients with intermediate- or high-risk myelofibrosis who previously received a JAK inhibitor.
  • AZD1480 a novel ATP-competitive JAK2 inhibitor with IC50 of 0.26 nM in a cell-free assay, selectivity against JAK3 and Tyk2, and to a smaller extent against JAK1.
  • AT9283 is a potent JAK2/3 inhibitor with IC50 of 1.2 nM/1.1 nM in cell-free assays; also potent to Aurora A/B, Abl1(T315I).
  • WP1066 is a novel inhibitor of JAK2 and STAT3 with IC50 of 2.30 ⁇ M and 2.43 ⁇ M in HEL cells; shows activity to JAK2, STAT3, STAT5, and ERK1/2 not JAK1 and JAK3. WP1066 induces apoptosis.
  • WHI-P154 is a potent JAK3 inhibitor with IC50 of 1.8 ⁇ M, no activity against JAK1 or JAK2, also inhibits EGFR, Src, Abl, VEGFR and MAPK, prevents Stat3, but not Stat5 phosphorylation.
  • ZM 39923 HCl is an JAK1/3 inhibitor with pIC50 of 4.4/7.1, almost no activity to JAK2 and modestly potent to EGFR; and is sensitive to transglutaminase.
  • Oclacitinib maleate (PF-03394197) is a novel inhibitor of JAK family members with IC50 ranging from 10 to 99 nM and JAK1-dependent cytokines with IC50 ranging from 36 to 249 nM. It does not inhibit a panel of 38 non-JAK kinases.
  • Deucravacitinib (BMS-986165) is a highly potent and selective allosteric inhibitor of Tyk2 with a Ki value of 0.02 nM for binding to the Tyk2 pseudokinase domain. It is highly selective against a panel of 265 kinases and pseudokinases.
  • Itacitinib is an orally bioavailable inhibitor of Janus-associated kinase 1 (JAK1) with potential antineoplastic activity.
  • Solcitinib (GLPG0778, GSK2586184) is an inhibitor of JAK1 with an IC50 of 8-9 nM, and shows 11-, 55- and 23-fold selectivity over JAK2, JAK3 and TYK2, respectively.
  • Ritlecitinib (PF-06651600) is a potent and irreversible JAK3-selective inhibitor with an IC50 of 33.1 nM but without activity (IC50>10,000 nM) against JAK1, JAK2, and TYK2.
  • WHI-P97 is a potent inhibitor of JAK-3 with an estimated Ki value of 0.09 ⁇ M in modeling studies and a measured IC50 value of 2.5 ⁇ M in EGFR kinase inhibition assays.
  • SAR-20347 is a potent inhibitor of TYK2, JAK1, JAK2 and JAK3 with IC50 of 0.6 nM, 23 nM, 26 nM, and 41 nM, respectively.
  • SAR-20347 inhibits TYK2- and JAK1-mediated IL-12 and IFN- ⁇ signaling.
  • Filgotinib (GLPG0634) is a selective JAK1 inhibitor with IC50 of 10 nM, 28 nM, 810 nM, and 116 nM for JAK1, JAK2, JAK3, and TYK2, respectively.
  • Decernotinib (VX-509) is a potent and selective JAK3 inhibitor with Ki of 2.5 nM, >4-fold selectivity over JAK1, JAK2,and TYK2, respectively.
  • GLPG0634 analogue is a selective JAK1 inhibitor with IC50 of 10 nM, 28 nM, 810 nM, and 116 nM for JAK1, JAK2, JAK3, and TYK2, respectively.
  • PF-06826647 (Tyk2-IN-8, compound 10) is a selective and orally administered inhibitor of tyrosine kinase 2 (TYK2) with IC50 of 17 nM for binding to TYK2 catalytically active JH1 domain.
  • PF-06826647 (Tyk2-IN-8, compound 10) also inhibits JAK1 and JAK2 with IC50 of 383 nM and 74 nM, respectively.
  • PF-06826647 (Tyk2-IN-8, compound 10) is used in the treatment of psoriasis (PSO).
  • JANEX-1 (WHI-P131) is a small molecule inhibitor of JAK3 that selectively inhibits JAK3 at an IC50 of 78 ⁇ M without altering the activity of JAK1 or JAK2, or any other protein tyrosine kinases (IC50 ⁇ 350 ⁇ M).
  • the pharmaceutical compositions for administration is dependent on the mode of administration and can readily be determined by one of ordinary skill in the art.
  • the pharmaceutical composition is sterile or sterilizable.
  • the therapeutic compositions featured in the invention can contain carriers or excipients, many of which are known to skilled artisans. Excipients that can be used include buffers (for example, citrate buffer, phosphate buffer, acetate buffer, and bicarbonate buffer), amino acids, urea, alcohols, ascorbic acid, phospholipids, polypeptides (for example, serum albumin), EDTA, sodium chloride, liposomes, mannitol, sorbitol, water, and glycerol.
  • nucleic acids, polypeptides, small molecules, and other modulatory compounds featured in the invention can be administered by any standard route of administration.
  • administration can be parenteral, intravenous, subcutaneous, or oral.
  • a modulatory compound can be formulated in various ways, according to the corresponding route of administration.
  • liquid solutions can be made for administration by drops into the ear, for injection, or for ingestion; gels or powders can be made for ingestion or topical application. Methods for making such formulations are well known and can be found in, for example, Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa. 1990.
  • the disclosed pharmaceutical compositions include the disclosed compounds (including pharmaceutically acceptable salt(s) thereof) as an active ingredient, a pharmaceutically acceptable carrier, and, optionally, other therapeutic ingredients or adjuvants.
  • the instant compositions include those suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • the pharmaceutical compositions of this invention can include a pharmaceutically acceptable carrier and a compound or a pharmaceutically acceptable salt of the compounds of the invention.
  • the compounds of the invention, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.
  • the pharmaceutical carrier employed can be, for example, a solid, liquid, or gas.
  • compositions of the present invention include the compound of the invention (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier, and optionally one or more additional therapeutic agents or adjuvants.
  • the instant compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered.
  • the pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • compositions of the present invention suitable for parenteral administration can be prepared as solutions or suspensions of the active compounds in water.
  • a suitable surfactant can be included such as, for example, hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
  • compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions.
  • the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions.
  • the final injectable form must be sterile and must be effectively fluid for easy syringability.
  • the compounds of this invention can be used in combination with one or more other drugs, in the form of a kit, to prevention, control, amelioration, or reduction of risk of hearing impairments, when the other drugs can have been known to impair hearing such as an antibiotic.
  • antibiotics especially aminoglycosides (such as gentamicin, streptomycin, and neomycin). Hearing-related side effects from these antibiotics are most common in people who have kidney disease or who already have ear or hearing problems
  • FIG. 2 A-B Momelotinib protects from aminoglycoside toxicity in cochlear explants of P3 FVB mice.
  • FVB mice were bred in the Creighton University animal facility. Procedures were approved by the IACUC committee of the Creighton University.
  • the culturing medium is DMEM (10565-018; gibco) with 1% FBS (16000-044; gibco), 500 ⁇ L ampicillin (A5354; Sigma), 100 ⁇ L N-2 supplement (17502-048, gibco) and 100 ⁇ L B-27 supplement (17504-044, gibco).
  • DMEM fetal calf serum
  • gentamicin (G1397, Sigma) was chosen to induce around 35% hair cell death after incubation for 24 h at 37° C.
  • MMB MMB
  • AdooQ AdooQ
  • the procedures were performed under sterile condition in the culture hood.
  • Cochleae were then fixed with 4% PFA for 30 min and stained with Alexa Fluor 555 phalloidin in to determine the survival rate of the hair cells. All the cochleae images were taken with Zeiss 700 confocal microscopy. Immunofluorescent images of cochlea stained with phalloidin (red) at the middle turn region shows minimal levels of hair cell loss when cotreated with MMB and GM.
  • OHC survival OHC number per 100 ⁇ m length of cochlear middle turn
  • Momelotinib at 0.0016, 0.008, 0.04, 0.2, 1.0, 5.0, 10.0 and 20.0 ⁇ M for 1 hour and co-incubated with 100 ⁇ M GM for 1 hour, or co-incubated with 600 ⁇ M kanamycin (K0254, Sigma) for 1 hour.
  • Momelotinib protects from aminoglycoside induced hair cell loss in zebrafish lateral line neuromasts in vivo.
  • Zebrafish lateral line neuromast HC counts are commonly used in vivo models for screening otoprotectants.
  • Danio rerio experimental larvae were obtained by pair mating of adult fish maintained at Creighton University by standard methods approved by the Institutional Animal Care and Use Committee.
  • Experimental fish were maintained at 28.5° C.
  • E3 media 5 mM NaCl, 0.17 mM KCl, 0.33 mM CaCl2 and 0.33 nM MgSO4, pH 7.2.
  • Animals were cryoanaesthetized after drug treatment and prior to fixation.
  • the neuromasts inspected, SO3 and O1-2, were part of the cranial system and included the otic, middle, and opercular neuromasts.
  • the lateral-line neuromasts of zebrafish are a valuable system for testing protectivity of compounds against aminoglycoside toxicity in vivo, as their HCs are considered homologous to those in the mammalian inner ear and are readily accessible to drugs.
  • mice were transferred to E3 water for 5 hours and fixed in 4% paraformaldehyde (PFA) overnight (26).
  • Neuromast HCs were immunolabeled with anti-otoferlin (HCS-1, DSHB) and anti-GFP (NB100-1614, Novus Biologicals). These two markers were used to detect and count neuromast HCs to reduce the chances of missing some of the HCs after the treatment since we previously noticed that incubation with the compounds can affect GFP expression more difficult to detect under a fluorescence microscope (26).
  • Fedratinib protects from aminoglycoside induced hair cell loss in zebrafish lateral line neuromasts in vivo.
  • FED 5-day post-fertilization
  • dpf 5-day post-fertilization
  • Tg(brn3c:GFP) larvae were pre-incubated with Fedratinib at 0.002, 0.018, 0.165, 1.48, and 13.3 ⁇ M for 1 hour followed by co-incubation with either 300 ⁇ M KM for 1 hour or 150 ⁇ M KM for 6 hours.
  • DMSO at ⁇ M was used as a control.
  • Momelotinib showed no inner ear toxicity in C57BL/6 mice. 4-week-old C57BL/6 mice obtained from The Jackson Laboratory were used, with a mix of males and females across experiments. Mice were administered a 14-day Momelotinib treatment protocol. Auditory Brainstem Response thresholds were recorded at 8 kHz, 16 kHz, and 32 kHz for a baseline sample of mice and the mice administered Momelotinib. No differences were observed across all frequencies tested.

Abstract

In one aspect, use of compounds as active agents to treat a hearing impairment and to prevent a hearing impairment, and methods of treating and/or preventing hearing impairments or disorders using the compositions are disclosed. Momelotinib and Fedratinib exhibit excellent protection against antibiotic-induced hearing loss in zebrafish and mice when administered prophylactically. In one aspect, Momelotinib or Fedratinib can be used as a therapy for the treatment and/or prevention of hearing loss. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. provisional patent application Ser. No. 63/050,568 filed Jul. 10, 2020, under 35 USC § 119(e) and 35 U.S.C. § 111(a) (hereby specifically incorporated herein by reference).
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • NIH/NIDCD 1R43DC019065, NIH/NIDCD R01DC015444, Office of Naval Research (ONR) N00014-18-1-2507, Department of Defense (DoD)/USAMRMC-RH170030.
  • BACKGROUND OF THE INVENTION (1) Field of the Invention
  • The present invention relates to therapeutic uses of compounds for treating, inhibiting, and/or preventing loss of hearing.
  • (2) Description of Related Art Including Information Disclosed under 37 CFR 1.97 and 37 CFR 1.98
  • Over 35 million Americans suffer from hearing impairment. In mammals, hair cell impairment is permanent. Antibiotic-induced hearing impairment (NIHL) is a permanent side effect from often necessary medical care. Neonates admitted to the neonatal intensive care unit have ten times the rate of hearing loss as neonates not admitted, often associated with the administration of antibiotics. The spiral-shaped cochlea of the inner ear is responsible for detecting sound. Inner hair cells lining the cochlea transform the mechanical vibrations of sound waves into chemical signals. These chemicals are then released from the hair cells and received by receptors on the auditory nerve fibers that send electrical impulses to the brain. The inner ear cells lining the cochlea can be destroyed by the use of antibiotics, leading to a condition called sensorineural hearing impairment.
  • Currently, there are no clinically proven medications for the treatment of hearing impairment (sensorineural and neural), or tinnitus associated with the inner ear, and a medication that could be used to prevent, alleviate, or eliminate hearing impairment (or tinnitus) would thus be very desirable. The most common remedy for individuals suffering from severe sensorineural hearing impairment is a hearing aid, which functions to amplify sound. Hearing aids are non-invasive and can improve an individual's ability to hear. However, hearing aids can often be quite conspicuous and embarrassing to the wearer and hearing aids do not return hearing to normal levels. Furthermore, hearing aids amplify sound indiscriminately, sometimes amplifying sounds that an individual does not wish to hear, such as environmental noise.
  • There exists a need in the art for a solution to hearing impairment due to antibiotics.
  • BRIEF SUMMARY OF THE INVENTION
  • The invention provides a method to prevent or treat hearing loss including the steps of administering to an animal in need thereof an effective amount of a pharmaceutical composition containing a therapeutically active agent, wherein the therapeutically active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1.
  • The inventive subject matter also includes a composition for use in preventing or treating hearing loss by protecting inner ear cells from death wherein the composition is an effective amount of an active agent, wherein the active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1 or a pharmaceutically acceptable salt thereof.
  • The inventive subject matter also includes a kit made of an active agent, wherein the active agent includes: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1 or a pharmaceutically acceptable salt thereof; and one or more of: (A) at least one antibiotic; (B) at least one cancer drug (C) instructions for preventing a hearing impairment.
  • BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING
  • The accompanying figures, which are incorporated in and constitute a part of this specification, illustrate several aspects and together with the description serve to explain the principles of the invention.
  • FIG. 1 is a schematic diagram showing how compounds of this invention were identified based on a comparison of data sets from drug screen connectivity maps and pathway enrichment analysis revealing drugs acting against hearing loss.
  • FIG. 2 shows outer hair cell protection by Momelotinib against aminoglycosides. A) Representative confocal images of the middle turn of whole-mount cochlear explants from P3 FVB mice treated with medium alone, 50 mg/mL gentamicin, 0.2 μM Momelotinib and 50 mg/mL gentamicin for 24 h. 0.2 μM Momelotinib alone did not show toxicity to HCs. HCs are labeled with phalloidin. B) shows outer hair cell (OHC) survival (OHC number per 100 μm length of cochlear middle turn) after treatment with various concentration of Momelotinib and 50 mg/mL gentamicin or 600 mM kanamycin. (Mann-Whitney U Test, *P<0.05, **P<0.01) shown in a micromolar concentration.
  • FIG. 3A-C show Momelotinib (MMB) protected zebrafish mechanosensory hair cells against gentamicin (GM) and kanamycin (KM) in vivo. 5 dpf Tg(Brn3c:mGFP) larvae were used for the experiments. 3A and 3B were treated with KM (200 uM for 1 hour and 150 μM for 6 hours, respectively) and 3C was treated with GM (100 uM for 1 hour). Each treatment was pretreated with MMB at the prescribed doses prior to co-treatment with the aminoglycoside. Control animal were treated with DMSO (−) or KM or GM alone (+) Animals were then fixed and immunostained for GFP and otoferlin. Quantification was done by assessing three neuromasts in the same anatomic location of each animal (n=5). The data is plotted as mean+SD. Statistical testing was done with One-way ANOVA and Dunnett post-hoc test (*P<0.05, **P<0.01, ***P<0.001).
  • FIG. 4A-B show Fedratinib (FED) protected zebrafish mechanosensory hair cells against kanamycin (KM) in vivo. 5 dpf Tg(Brn3c:mGFP) larvae were used for the experiments. Each treatment was pretreated FED at the prescribed doses prior to co-treatment with the aminoglycoside. 3A shows co-treatment with KM at 300 um for 1 hour. 3B shows co-treatment with KM at 150 uM for 6 hours. Control animal were treated with DMSO (−) or KM alone (+) Animals were then fixed and immunostained for GFP and otoferlin. Quantification was done by assessing three neuromasts in the same anatomic location of each animal (n=5). The data is plotted as mean+SD. Statistical testing was done with One-way ANOVA and Dunnett post-hoc test (*P<0.05, **P<0.01, ***P<0.001).
  • FIG. 5 shows Momelotinib is not toxic in C57BL/6 mice (4-week-old) in vivo up to a dose of 100 mg/kg/day over a 14-day treatment regimen. Oral administration of Momelotinib did not result in any general or inner ear toxicity, measured as and hearing function in 28-day old C57BL/6 mice. Auditory brainstem response (ABR) thresholds were recorded for 3 frequencies (8 kHz, 16 kHz, and 32 kHz) before and after a 14-day MMB treatment protocol. No differences were observed across all frequencies tested. Statistical analysis was done with repeated measures ANOVA between groups.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention can be understood more readily by reference to the following detailed description of the invention and the Examples included therein. Before the present compounds, compositions, articles, systems, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, example methods and materials are now described.
  • While aspects of the present invention can be described and claimed in a particular statutory class, such as the system statutory class, this is for convenience only and one of skill in the art will understand that each aspect of the present invention can be described and claimed in any statutory class. Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification.
  • In one aspect, compounds can be used as a therapy for the treatment and/or prevention of hearing loss. In various aspects, the compounds and compositions of the invention can be administered in pharmaceutical compositions, which are formulated according to the intended method of administration. The compounds of this invention are defined as a therapeutically active agent in a treatment regimen or procedure that is intended for preventing hearing loss by noise or aging by protecting inner ear cells from death and in preventing hearing loss by chemotherapy or antibiotics induced hearing loss. Therapeutic agent means a chemical substance that is used for the treatment or mitigation of a disease condition or ailment.
  • In one aspect, compounds can be used as a therapy for the treatment and/or prevention of hearing loss. In various aspects, the compounds and compositions of the invention can be administered in pharmaceutical compositions, which are formulated according to the intended method of administration. The compounds of this invention are defined as a therapeutically active agent in a treatment regimen or procedure that is intended for preventing hearing loss by noise or aging by protecting inner ear cells from death and in preventing hearing loss by chemotherapy or antibiotics induced hearing loss. Therapeutic agent means a chemical substance that is used for the treatment or mitigation of a disease condition or ailment.
  • Now referring to FIG. 1 compounds were identified based on a comparison of data sets from drug screen connectivity maps and pathway enrichment analysis revealing compounds acting against hearing loss. This method was developed to derive drug candidates from a diverse chemical space, covering a wide range of biological pathways, avoiding bias associated with focusing on previously reported pathways. The resulting compounds exhibited overlaps in gene expression transcriptomic profiles between at least one of a plurality of cell-lines or mouse strains treated with ototoxic insults (cisplatin FIG. 5A/D, noise FIG. 5B, or antibiotic exposure FIG. 5C) and at least one of a plurality of cell-lines or mouse strains treated with one of the compounds.
  • Here specifically, the data set sought was conformity to a NIHL-resistant mouse strains to NIHL sensitive mouse strains (129 SvJ and CAST). Here specifically the data set sought was conformity to cisplatin-resistant and sensitive cancer cell lines, HEI-OC1 cell line, in vivo mouse cochlear single cell RNA seq with and without cisplatin treatment. However, the data set greatest weigh was transcriptome perturbation of neonatal mouse organ of Corti exposed to gentamicin for damage related to antibiotic treatment. These compounds with an overlap of the data sets include as the therapeutically active agent: momelotinib, fedratinib, axitinib, sunitinib, ruxolitinib, AZD1480, AT9283, WP1066, WHI-P154, ZM 39923 HCL, oclacitinib maleate, deucravacitinib, itacitinib, solcitinib, ritlecitinib, WHI-P97, SAR-20347, filgotinib, decernotibnib, glpg0634, PF-06826647, and JANEX-1.
  • Compounds are revealed to protect against hair cell apoptosis. Compounds are identified as acting against hair cell loss in animals by the models and data presented. Models reveal properties necessary for an otoprotective compound such as high efficacy against hair cell loss, relatively low toxicity. Compounds are revealed to have high efficacy and high affinity in mouse and zebrafish models used to demonstrate protection against hair cell loss. The lateral-line neuromasts of zebrafish are a valuable model for testing compounds protective against hearing loss in vivo, as their HCs are considered homologous to those in the mammalian inner ear and are readily accessible to drugs in vivo. Teitz et al., J. Exp. Med. 2; 215(4):1187-1203 (2018). Mouse models involving HEI-OC1 have shown effective in validating therapeutic uses of compounds against hearing loss due to cancer treatment such as cisplatin, noise, antibiotics, and aging. Teitz et al., J. Exp. Med. 2; 215(4):1187-1203 (2018).
  • The compounds and compositions described herein can be formulated in a conventional manner using one or more physiologically acceptable carriers or excipients. For example, a pharmaceutical composition can be formulated for local or systemic administration, e.g., administration by drops or injection into the ear, insufflation (such as into the ear), intravenous, topical, or oral administration. Compounds can be synthesized by a variety of methods known in the art.
  • Fedratinib is an FDA-approved therapy for patients with myeloproliferative neoplasm-associated myelofibrosis. Momelotinib is in phase 3 clinical trials for the treatment of patients with intermediate- or high-risk myelofibrosis who previously received a JAK inhibitor.
  • AZD1480 a novel ATP-competitive JAK2 inhibitor with IC50 of 0.26 nM in a cell-free assay, selectivity against JAK3 and Tyk2, and to a smaller extent against JAK1. AT9283 is a potent JAK2/3 inhibitor with IC50 of 1.2 nM/1.1 nM in cell-free assays; also potent to Aurora A/B, Abl1(T315I). WP1066 is a novel inhibitor of JAK2 and STAT3 with IC50 of 2.30 μM and 2.43 μM in HEL cells; shows activity to JAK2, STAT3, STAT5, and ERK1/2 not JAK1 and JAK3. WP1066 induces apoptosis. WHI-P154 is a potent JAK3 inhibitor with IC50 of 1.8 μM, no activity against JAK1 or JAK2, also inhibits EGFR, Src, Abl, VEGFR and MAPK, prevents Stat3, but not Stat5 phosphorylation. ZM 39923 HCl is an JAK1/3 inhibitor with pIC50 of 4.4/7.1, almost no activity to JAK2 and modestly potent to EGFR; and is sensitive to transglutaminase.
  • Oclacitinib maleate (PF-03394197) is a novel inhibitor of JAK family members with IC50 ranging from 10 to 99 nM and JAK1-dependent cytokines with IC50 ranging from 36 to 249 nM. It does not inhibit a panel of 38 non-JAK kinases. Deucravacitinib (BMS-986165) is a highly potent and selective allosteric inhibitor of Tyk2 with a Ki value of 0.02 nM for binding to the Tyk2 pseudokinase domain. It is highly selective against a panel of 265 kinases and pseudokinases. Itacitinib (INCB39110) is an orally bioavailable inhibitor of Janus-associated kinase 1 (JAK1) with potential antineoplastic activity. Solcitinib (GLPG0778, GSK2586184) is an inhibitor of JAK1 with an IC50 of 8-9 nM, and shows 11-, 55- and 23-fold selectivity over JAK2, JAK3 and TYK2, respectively. Ritlecitinib (PF-06651600) is a potent and irreversible JAK3-selective inhibitor with an IC50 of 33.1 nM but without activity (IC50>10,000 nM) against JAK1, JAK2, and TYK2.
  • WHI-P97 is a potent inhibitor of JAK-3 with an estimated Ki value of 0.09 μM in modeling studies and a measured IC50 value of 2.5 μM in EGFR kinase inhibition assays. SAR-20347 is a potent inhibitor of TYK2, JAK1, JAK2 and JAK3 with IC50 of 0.6 nM, 23 nM, 26 nM, and 41 nM, respectively. SAR-20347 inhibits TYK2- and JAK1-mediated IL-12 and IFN-α signaling. Filgotinib (GLPG0634) is a selective JAK1 inhibitor with IC50 of 10 nM, 28 nM, 810 nM, and 116 nM for JAK1, JAK2, JAK3, and TYK2, respectively. Decernotinib (VX-509) is a potent and selective JAK3 inhibitor with Ki of 2.5 nM, >4-fold selectivity over JAK1, JAK2,and TYK2, respectively.
  • GLPG0634 analogue is a selective JAK1 inhibitor with IC50 of 10 nM, 28 nM, 810 nM, and 116 nM for JAK1, JAK2, JAK3, and TYK2, respectively. PF-06826647 (Tyk2-IN-8, compound 10) is a selective and orally administered inhibitor of tyrosine kinase 2 (TYK2) with IC50 of 17 nM for binding to TYK2 catalytically active JH1 domain. PF-06826647 (Tyk2-IN-8, compound 10) also inhibits JAK1 and JAK2 with IC50 of 383 nM and 74 nM, respectively. PF-06826647 (Tyk2-IN-8, compound 10) is used in the treatment of psoriasis (PSO). JANEX-1 (WHI-P131) is a small molecule inhibitor of JAK3 that selectively inhibits JAK3 at an IC50 of 78 μM without altering the activity of JAK1 or JAK2, or any other protein tyrosine kinases (IC50≥350 μM).
  • The nature of the pharmaceutical compositions for administration is dependent on the mode of administration and can readily be determined by one of ordinary skill in the art. In various aspects, the pharmaceutical composition is sterile or sterilizable. The therapeutic compositions featured in the invention can contain carriers or excipients, many of which are known to skilled artisans. Excipients that can be used include buffers (for example, citrate buffer, phosphate buffer, acetate buffer, and bicarbonate buffer), amino acids, urea, alcohols, ascorbic acid, phospholipids, polypeptides (for example, serum albumin), EDTA, sodium chloride, liposomes, mannitol, sorbitol, water, and glycerol. The nucleic acids, polypeptides, small molecules, and other modulatory compounds featured in the invention can be administered by any standard route of administration. For example, administration can be parenteral, intravenous, subcutaneous, or oral. A modulatory compound can be formulated in various ways, according to the corresponding route of administration. For example, liquid solutions can be made for administration by drops into the ear, for injection, or for ingestion; gels or powders can be made for ingestion or topical application. Methods for making such formulations are well known and can be found in, for example, Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa. 1990.
  • In various aspects, the disclosed pharmaceutical compositions include the disclosed compounds (including pharmaceutically acceptable salt(s) thereof) as an active ingredient, a pharmaceutically acceptable carrier, and, optionally, other therapeutic ingredients or adjuvants. The instant compositions include those suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • In various aspects, the pharmaceutical compositions of this invention can include a pharmaceutically acceptable carrier and a compound or a pharmaceutically acceptable salt of the compounds of the invention. The compounds of the invention, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds. The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas.
  • The pharmaceutical compositions of the present invention include the compound of the invention (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier, and optionally one or more additional therapeutic agents or adjuvants. The instant compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions can be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • Pharmaceutical compositions of the present invention suitable for parenteral administration can be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.
  • Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability.
  • In various aspects, the compounds of this invention can be used in combination with one or more other drugs, in the form of a kit, to prevention, control, amelioration, or reduction of risk of hearing impairments, when the other drugs can have been known to impair hearing such as an antibiotic. Certain antibiotics, especially aminoglycosides (such as gentamicin, streptomycin, and neomycin). Hearing-related side effects from these antibiotics are most common in people who have kidney disease or who already have ear or hearing problems
  • Now referring to FIG. 2A-B, Momelotinib protects from aminoglycoside toxicity in cochlear explants of P3 FVB mice. FVB mice were bred in the Creighton University animal facility. Procedures were approved by the IACUC committee of the Creighton University. 2A) Inner ears were collected from the post-natal day 3 (P3) FVB mice and transferred onto a filter (Millicell, PICM03050; Millipore) soaked in culturing medium. The filter was placed in 6-well culture plates with 1.2 mL medium outside the filter. The culturing medium is DMEM (10565-018; gibco) with 1% FBS (16000-044; gibco), 500 μL ampicillin (A5354; Sigma), 100 μL N-2 supplement (17502-048, gibco) and 100 μL B-27 supplement (17504-044, gibco). After culturing for 24 h at 37° C. in 5% CO2, 1 mL culturing medium with MMB at 0.2 μM, 200 μM MMB alone or without MMB and added both inside and outside the filter to preincubate for another 1 h before adding gentamicin solution. A 50 μg/mL gentamicin (G1397, Sigma) was chosen to induce around 35% hair cell death after incubation for 24 h at 37° C. For compound CYT-387 or MMB (A10263, AdooQ). The procedures were performed under sterile condition in the culture hood. Cochleae were then fixed with 4% PFA for 30 min and stained with Alexa Fluor 555 phalloidin in to determine the survival rate of the hair cells. All the cochleae images were taken with Zeiss 700 confocal microscopy. Immunofluorescent images of cochlea stained with phalloidin (red) at the middle turn region shows minimal levels of hair cell loss when cotreated with MMB and GM. The survival OHCs were then normalized to 100 μm length to further compare between different experimental conditions. 2B) OHC survival (OHC number per 100 μm length of cochlear middle turn) after treatment with Momelotinib at 0.0016, 0.008, 0.04, 0.2, 1.0, 5.0, 10.0 and 20.0 μM for 1 hour and co-incubated with 100 μM GM for 1 hour, or co-incubated with 600 μM kanamycin (K0254, Sigma) for 1 hour. (Mann-Whitney U Test, *P<0.05, **P<0.01). Quantification of outer hair cells from immunofluorescent images shows that cotreatment with Momelotinib grants full protection against aminoglycoside-induced hair cell loss (n=5 per group, student's t-test). *P<0.05, data shown as mean±standard error in all panels.
  • Now referring to FIG. 3A-C, Momelotinib protects from aminoglycoside induced hair cell loss in zebrafish lateral line neuromasts in vivo. Zebrafish lateral line neuromast HC counts are commonly used in vivo models for screening otoprotectants. Danio rerio experimental larvae were obtained by pair mating of adult fish maintained at Creighton University by standard methods approved by the Institutional Animal Care and Use Committee. We used Tg(pou4f3:mGFP) expressing a membrane bound GFP in HCs. Experimental fish were maintained at 28.5° C. in E3 media (5 mM NaCl, 0.17 mM KCl, 0.33 mM CaCl2 and 0.33 nM MgSO4, pH 7.2). Animals were cryoanaesthetized after drug treatment and prior to fixation. The neuromasts inspected, SO3 and O1-2, were part of the cranial system and included the otic, middle, and opercular neuromasts. The lateral-line neuromasts of zebrafish are a valuable system for testing protectivity of compounds against aminoglycoside toxicity in vivo, as their HCs are considered homologous to those in the mammalian inner ear and are readily accessible to drugs. For the screenings, 5-day post-fertilization (dpf) Tg(brn3c:GFP) larvae were pre-incubated with Momelotinib at 0.002, 0.018, 0.165, 1.48, and 13.3 μM for 1 hour followed by co-incubation with either 200 μM KM for 1 hour, 150 μM KM for 6 hours, or 100 μM GM for 1 hour. DMSO at μM was used as a control.
  • Subsequently, animals were transferred to E3 water for 5 hours and fixed in 4% paraformaldehyde (PFA) overnight (26). Neuromast HCs were immunolabeled with anti-otoferlin (HCS-1, DSHB) and anti-GFP (NB100-1614, Novus Biologicals). These two markers were used to detect and count neuromast HCs to reduce the chances of missing some of the HCs after the treatment since we previously noticed that incubation with the compounds can affect GFP expression more difficult to detect under a fluorescence microscope (26). Otic, middle, and opercular neuromasts were identified, and HCs at SO3 (supraorbital line neuromast) and O1-2 (Otic line neuromasts) were manually counted using a Zeiss AxioSkop 2 fluorescence microscope with a 40× oil objective. Compounds were then evaluated on efficiency and potency, with the top-rated compounds showing high protection at lower concentrations.
  • Now referring to FIG. 4A-B, Fedratinib (FED) protects from aminoglycoside induced hair cell loss in zebrafish lateral line neuromasts in vivo. For the screenings, 5-day post-fertilization (dpf) Tg(brn3c:GFP) larvae were pre-incubated with Fedratinib at 0.002, 0.018, 0.165, 1.48, and 13.3 μM for 1 hour followed by co-incubation with either 300 μM KM for 1 hour or 150 μM KM for 6 hours. DMSO at μM was used as a control. Additionally, 5-day post-fertilization (dpf) Tg(brn3c:GFP) larvae were pre-incubated with Fedratinib at, 0.018, 0.165 μM for 1 hour without being followed by co-incubation with KM.
  • Now referring to FIG. 5 , Momelotinib showed no inner ear toxicity in C57BL/6 mice. 4-week-old C57BL/6 mice obtained from The Jackson Laboratory were used, with a mix of males and females across experiments. Mice were administered a 14-day Momelotinib treatment protocol. Auditory Brainstem Response thresholds were recorded at 8 kHz, 16 kHz, and 32 kHz for a baseline sample of mice and the mice administered Momelotinib. No differences were observed across all frequencies tested.
  • All publications, patents and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
  • Teitz T, Fang J, Goktug A N, Bonga J D, Diao S, Hazlitt R A, Iconaru L, Morfouace M, Currier D, Zhou Y, Umans R A, Taylor M R, Cheng C, Min J, Freeman B, Peng J, Roussel M F, Kriwacki R, Guy R K, Chen T, Zuo J. CDK2 Inhibitors as Candidate Therapeutics for Cisplatin- and Noise-Induced Hearing Impairment. J Exp Med. 2018 Apr. 2; 215(4):1187-1203.
  • While the invention has been described with reference to details of the illustrated embodiments, these details are not intended to limit the scope of the invention as defined in the appended claims. The embodiment of the invention in which exclusive property or privilege is claimed is defined as follows:

Claims (11)

1-5. (canceled)
6. A method to prevent or treat hearing loss comprising:
administering to a mammal in need thereof an effective amount of a pharmaceutical composition containing a therapeutically active agent, wherein the therapeutically active agent is momelotinib.
7. The method of claim 6, comprising protecting the inner ear cells from death caused by an antibiotic.
8. The method of claim 7, wherein the antibiotic is an aminoglycoside.
9. The method of claim 6, comprising protecting the inner ear cells from death caused by noise.
10. The method of claim 6, comprising protecting the inner ear cells from death caused by cisplatin treatment.
11. (canceled)
12. (currently mended) A kit comprising: an active agent, wherein the active agent is momelotinib or a pharmaceutically acceptable salt thereof; and one or more of:
(A) at least one antibiotic;
(B) at least one cancer drug;
(C) instructions for preventing a hearing impairment.
13. (canceled)
14. The kit of claim 12, wherein the at least one antibiotic is an aminoglycoside.
15. The kit of claim 12, wherein the at least one cancer drug is cisplatin.
US18/117,852 2020-07-10 2023-03-06 Methods for the prevention and treatment of hearing loss Active US11766440B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/117,852 US11766440B1 (en) 2020-07-10 2023-03-06 Methods for the prevention and treatment of hearing loss

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063050568P 2020-07-10 2020-07-10
PCT/US2021/041197 WO2022051029A2 (en) 2020-07-10 2021-07-10 Methods for the prevention and treatment of hearing loss
US18/084,033 US20230285401A1 (en) 2020-07-10 2022-12-19 Methods for the Prevention and Treatment of Hearing Loss
US18/117,852 US11766440B1 (en) 2020-07-10 2023-03-06 Methods for the prevention and treatment of hearing loss

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US18/084,033 Division US20230285401A1 (en) 2020-07-10 2022-12-19 Methods for the Prevention and Treatment of Hearing Loss

Publications (2)

Publication Number Publication Date
US20230293538A1 true US20230293538A1 (en) 2023-09-21
US11766440B1 US11766440B1 (en) 2023-09-26

Family

ID=79552120

Family Applications (4)

Application Number Title Priority Date Filing Date
US17/371,847 Active 2042-04-07 US11857551B1 (en) 2020-07-10 2021-07-09 Methods for the prevention and treatment of hearing loss
US18/081,951 Pending US20230285418A1 (en) 2020-07-10 2022-12-15 Methods for the Prevention and Treatment of Hearing Loss
US18/084,033 Pending US20230285401A1 (en) 2020-07-10 2022-12-19 Methods for the Prevention and Treatment of Hearing Loss
US18/117,852 Active US11766440B1 (en) 2020-07-10 2023-03-06 Methods for the prevention and treatment of hearing loss

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US17/371,847 Active 2042-04-07 US11857551B1 (en) 2020-07-10 2021-07-09 Methods for the prevention and treatment of hearing loss
US18/081,951 Pending US20230285418A1 (en) 2020-07-10 2022-12-15 Methods for the Prevention and Treatment of Hearing Loss
US18/084,033 Pending US20230285401A1 (en) 2020-07-10 2022-12-19 Methods for the Prevention and Treatment of Hearing Loss

Country Status (3)

Country Link
US (4) US11857551B1 (en)
CN (2) CN115884768A (en)
WO (3) WO2022020114A2 (en)

Family Cites Families (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7071230B2 (en) * 1997-10-02 2006-07-04 Board Of Trustees Of Southern Illinois University Therapeutic use of D-methionine to reduce the toxicity of noise
US6331289B1 (en) 1996-10-28 2001-12-18 Nycomed Imaging As Targeted diagnostic/therapeutic agents having more than one different vectors
GB9718913D0 (en) 1997-09-05 1997-11-12 Glaxo Group Ltd Substituted oxindole derivatives
US6268351B1 (en) 1997-09-26 2001-07-31 The Trustees Of The University Of Pennsylvania Methods for inducing proliferation in auditory receptor epithelium
JP2002525329A (en) 1998-09-25 2002-08-13 セフアロン・インコーポレーテツド Method for preventing / treating damage to sensory hair cells and cochlear neurons
AU2001251011A1 (en) 2000-03-27 2001-10-08 The General Hospital Corporation Treatments for immune-mediated ear disorders
CA2464214C (en) 2001-10-22 2011-02-08 The Research Foundation Of State University Of New York Protein kinase and phosphatase inhibitors, methods for designing them, and methods of using them
US6822097B1 (en) 2002-02-07 2004-11-23 Amgen, Inc. Compounds and methods of uses
GB0205693D0 (en) 2002-03-09 2002-04-24 Astrazeneca Ab Chemical compounds
US20060148829A1 (en) 2002-12-17 2006-07-06 Laurent Meijer Means for inducing differentiation of supernumerary hair cells and deiters' cells in the developing organ of corti
EP1594682A4 (en) 2003-01-27 2008-11-19 Shamrock Tech Inc Method for producing submicron polytetrafluoroethylene powder and products thereof
CA2532965C (en) 2003-07-22 2013-05-14 Astex Therapeutics Limited 3, 4-disubstituted 1h-pyrazole compounds and their use as cyclin dependent kinases (cdk) and glycogen synthase kinase-3 (gsk-3) modulators
CA2573103A1 (en) * 2004-07-06 2006-02-09 Angion Biomedica Corporation Quinazoline modulators of hepatocyte growth factor / c-met activity for the treatment of cancer
US8404718B2 (en) 2005-01-21 2013-03-26 Astex Therapeutics Limited Combinations of pyrazole kinase inhibitors
WO2007031098A1 (en) 2005-09-12 2007-03-22 Xigen S.A. Cell-permeable peptide inhibitors of the jnk signal transduction pathway
AU2006294565A1 (en) * 2005-09-30 2007-04-05 Perlegen Sciences, Inc. Methods and compositions for screening and treatment of disorders of blood glucose regulation
US20070078177A1 (en) 2005-09-30 2007-04-05 Washington University In St. Louis Methods and compositions for treating non age related hearing impairment in a subject
US7754700B2 (en) 2006-04-24 2010-07-13 Trager Seymour F Composition and methods for alleviating symptoms of neurotoxicity
US8329683B2 (en) 2006-06-02 2012-12-11 Nexgenix Pharmaceuticals, Llc Treatment of neurofibromatosis with radicicol and its derivatives
CA2658881C (en) 2006-07-25 2015-09-15 Hough Ear Institute Methods for treating acute acoustic trauma
US7935697B2 (en) 2006-12-28 2011-05-03 Kinex Pharmaceuticals, Llc Compositions for modulating a kinase cascade and methods of use thereof
US9259399B2 (en) 2007-11-07 2016-02-16 Cornell University Targeting CDK4 and CDK6 in cancer therapy
WO2009067555A1 (en) 2007-11-19 2009-05-28 Bionovo, Inc. Scutellaria barbata extract and combinations for the treatment of cancer
CA2720721A1 (en) 2008-04-07 2009-10-15 Interface Biologics, Inc. Combination therapy for the treatment of bacterial infections
DK2805720T3 (en) 2008-05-28 2019-09-02 Reveragen Biopharma Inc Non-hormonal steroid modulators of NF-κB for the treatment of disease
US8211656B2 (en) 2008-08-13 2012-07-03 The Invention Science Fund I, Llc Biological targeting compositions and methods of using the same
JP5809061B2 (en) 2008-11-24 2015-11-10 マサチューセッツ・アイ・アンド・イア・インファーマリー Pathways for producing hair cells
CA2772097C (en) 2009-08-24 2019-04-30 Hough Ear Institute Methods for treating acute acoustic trauma
KR20180053419A (en) 2010-05-26 2018-05-21 큐알엔에이, 인크. Treatment of atonal homolog 1 (atoh1) related diseases by inhibition of natural antisense transcript to atoh1
CA2801535C (en) 2010-06-04 2017-05-30 Hough Ear Institute Composition and method for inner ear sensory hair cell regeneration or replacement
WO2011159945A2 (en) 2010-06-16 2011-12-22 Afraxis, Inc. Methods for treating neurological conditions
US10105356B2 (en) 2011-01-31 2018-10-23 Avalyn Pharma Inc. Aerosol pirfenidone and pyridone analog compounds and uses thereof
AR085397A1 (en) 2011-02-23 2013-09-25 Intellikine Inc COMBINATION OF QUINASA INHIBITORS AND THEIR USES
GB201107176D0 (en) 2011-04-28 2011-06-15 Cxr Biosciences Ltd Pyrrolnitrin derivatives
CN104204804B (en) 2011-11-11 2016-09-28 因特利凯有限责任公司 Combination of inhibitors of kinases and application thereof
WO2013071415A1 (en) 2011-11-15 2013-05-23 University Health Network Targeting the rb pathway for the prevention of cancer
WO2013120107A1 (en) 2012-02-09 2013-08-15 University Of Rochester Methods and compositions for treating a subject to inhibit hearing loss
US20150050354A1 (en) 2012-04-02 2015-02-19 Moderna Therapeutics, Inc. Modified polynucleotides for the treatment of otic diseases and conditions
EP2833892A4 (en) 2012-04-02 2016-07-20 Moderna Therapeutics Inc Modified polynucleotides for the production of oncology-related proteins and peptides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
CA2880236C (en) * 2012-07-27 2022-09-13 Antonius Martinus Gustave Bunt Efflux inhibitor compositions and methods of treatment using the same
WO2014052836A2 (en) 2012-09-27 2014-04-03 Dunman Paul M Methods and compositions for treating infection
WO2014089177A2 (en) * 2012-12-04 2014-06-12 Massachusetts Institute Of Technology Compounds, conjugates and compositions of epipolythiodiketopiperazines and polythiodiketopiperazines
WO2014102590A1 (en) 2012-12-27 2014-07-03 Purdue Pharma L.P. Substituted piperidin-4-amino-type compounds and uses thereof
WO2014134184A1 (en) 2013-02-28 2014-09-04 Anida Pharma Inc. Methods of treating ototoxicity
DK2968377T3 (en) 2013-03-12 2020-08-24 Laszlo Sichtnik Compositions and methods for treating an infectious disease
US9572815B2 (en) 2013-03-15 2017-02-21 St. Jude Children's Research Hospital Methods and compositions of p27KIP1 transcriptional modulators
US20160089371A1 (en) 2013-03-15 2016-03-31 Intellikine, Llc Combination of Kinase Inhibitors and Uses Thereof
MX2016006316A (en) 2013-11-13 2016-07-26 Novartis Ag Mtor inhibitors for enhancing the immune response.
WO2015126192A1 (en) 2014-02-20 2015-08-27 아주대학교산학협력단 Pharmaceutical composition for treating or preventing sensorineural hearing loss, containing cysteinyl leukotriene receptor antagonist and ginkgo leaf extract
WO2015168149A2 (en) 2014-04-28 2015-11-05 Massachusetts Eye & Ear Infirmary Sensorineural hair cell differentiation
US9475854B2 (en) 2014-05-23 2016-10-25 Washington State University Lead compound for otoprotection: targeting HGF signaling with dihexa
US10010585B2 (en) 2014-06-16 2018-07-03 Massachusetts Eye And Ear Infirmary Methods of treating vestibular schwannoma and reducing hearing or neurite loss caused by vestibular schwannoma
US10391168B1 (en) 2014-08-22 2019-08-27 University Of Bern Anti-CD70 combination therapy
WO2016069906A1 (en) * 2014-10-29 2016-05-06 Massachusetts Eye And Ear Infirmary Efficient delivery of therapeutic molecules to cells of the inner ear
JP2017537946A (en) * 2014-12-11 2017-12-21 ゼニス・エピジェネティクス・リミテッドZenith Epigenetics Ltd. Substituted heterocycles as bromodomain inhibitors
WO2016114655A1 (en) * 2015-01-12 2016-07-21 Ry Pharma B.V. Treating neuromuscular or neurologic disease through reducing gabaergic and/or glycinergic inhibitory neurotransmitter overstimulation
US10399994B2 (en) 2015-02-06 2019-09-03 University Of Washington Compounds and methods for preventing or treating sensory hair cell death
WO2016205806A1 (en) 2015-06-18 2016-12-22 St. Jude Children's Research Hospital Methods and compositions for the prevention and treatment of hearing loss
KR20210143952A (en) * 2015-10-09 2021-11-30 의료법인 성광의료재단 Methods and compositions for increasing human somatic cell nuclear transfer (SCNT) efficiency by removal of histone H3-lysine trimethylation, and induction of human NT-ESCs
EP3390389B1 (en) 2015-12-17 2021-05-19 Merck Patent GmbH Polycyclic tlr7/8 antagonists and use thereof in the treatment of immune disorders
US10583140B2 (en) 2016-01-27 2020-03-10 Glaxosmithkline Intellectual Property Development Limited Ingenol analogs, pharmaceutical compositions and methods of use thereof
EP3943090A1 (en) 2016-01-29 2022-01-26 Massachusetts Eye & Ear Infirmary Expansion and differentiation of inner ear supporting cells and methods of use thereof
US10213511B2 (en) 2016-03-02 2019-02-26 Frequency Therapeutics, Inc. Thermoreversible compositions for administration of therapeutic agents
US10201540B2 (en) 2016-03-02 2019-02-12 Frequency Therapeutics, Inc. Solubilized compositions for controlled proliferation of stem cells / generating inner ear hair cells using GSK3 inhibitors: I
CN107345231A (en) 2016-05-05 2017-11-14 江苏命码生物科技有限公司 A kind of siRNA for suppressing EGFR gene expression and its precursor and application
CA3027498A1 (en) 2016-06-21 2017-12-28 X4 Pharmaceuticals, Inc. Cxcr4 inhibitors and uses thereof
US20180021315A1 (en) 2016-07-20 2018-01-25 Wayne State University Methods of treating hearing disorders
US11331373B2 (en) 2017-02-10 2022-05-17 St. Jude Children's Research Hospital Combination therapy for treating disorders of the ear
US10729692B2 (en) 2017-02-26 2020-08-04 Institute For Cancer Research Dual inhibition of CDK and HSP90 destabilize HIF1alpha and synergistically induces cancer cell death
SG11201909710XA (en) 2017-04-21 2019-11-28 Kyn Therapeutics Indole ahr inhibitors and uses thereof
US11883491B2 (en) 2017-05-03 2024-01-30 St. Jude Children's Research Hospital, Inc. Compositions and methods for prevention and treatment of hearing loss
WO2018204764A1 (en) * 2017-05-05 2018-11-08 Camp4 Therapeutics Corporation Identification and targeted modulation of gene signaling networks
US10665391B2 (en) 2017-11-13 2020-05-26 Micron Technology, Inc. Capacitor having bottom electrode comprising TiN
CN112105385A (en) 2017-12-26 2020-12-18 凯麦拉医疗公司 IRAK degrading agents and uses thereof
EP3743057A4 (en) 2018-01-26 2021-11-17 Massachusetts Eye & Ear Infirmary Treatment of hearing loss
US10988477B2 (en) 2018-01-29 2021-04-27 Merck Patent Gmbh GCN2 inhibitors and uses thereof
CN112292128A (en) 2018-04-16 2021-01-29 阿瑞斯医疗有限公司 EP4 inhibitors and uses thereof
EP3810146A4 (en) 2018-04-25 2022-08-17 Otonomy, Inc. Otic formulations for drug-induced ototoxicity
CR20200578A (en) 2018-05-01 2021-02-22 Revolution Medicines Inc C40-, c28-, and c-32-linked rapamycin analogs as mtor inhibitors
MX2021000395A (en) 2018-07-11 2021-05-12 Rubedo Life Sciences Inc Senolytic compositions and uses thereof.
US20200031873A1 (en) 2018-07-30 2020-01-30 Eternans Limited Repurposing cell penetrating peptides and their novel derivatives and iopromide and iodo-aryl carbonates for treatment of senescence-related diseases and disorders
US20230293524A1 (en) 2019-10-30 2023-09-21 Spiral Therapeutics Inc Otic formulations for drug-induced ototoxicity
US11000540B1 (en) 2019-11-22 2021-05-11 Al Siamon Treatment for reducing adverse events including chemotherapy discomfort and other conditions

Also Published As

Publication number Publication date
US11766440B1 (en) 2023-09-26
WO2022051029A3 (en) 2022-06-02
US11857551B1 (en) 2024-01-02
WO2022011325A1 (en) 2022-01-13
WO2022051029A2 (en) 2022-03-10
WO2022011325A8 (en) 2022-03-17
CN115884768A (en) 2023-03-31
US20230285401A1 (en) 2023-09-14
WO2022020114A3 (en) 2022-03-31
US20230285418A1 (en) 2023-09-14
WO2022020114A2 (en) 2022-01-27
CN116157131A (en) 2023-05-23

Similar Documents

Publication Publication Date Title
Li et al. Novel compounds protect auditory hair cells against gentamycin-induced apoptosis by maintaining the expression level of H3K4me2
CN102858157A (en) Combination pharmaceutical agents as inhibitors of hcv replication
CN106488769A (en) For treating Fibrotic Sai Nikeweiluo
CN107106530A (en) The method for treating multiple sclerosis
US10894035B2 (en) Use of indole compounds to stimulate the immune system
ES2853974T3 (en) Use of a composition containing benzoate in urea cycle disorders and neurodegenerative disorders
JP2018522938A (en) Setron family calcineurin inhibitors for the treatment of hearing loss
Marshall et al. TrkB-enhancer facilitates functional recovery after traumatic brain injury
US11766440B1 (en) Methods for the prevention and treatment of hearing loss
US20050131044A1 (en) Small molecule inhibitors of necrosis
US20220016118A1 (en) Combination of a mcl-1 inhibitor and midostaurin, uses and pharmaceutical compositions thereof
CN113855803B (en) Use of PRMT5 inhibitors for the preparation of hearing protection medicaments
US20230330097A1 (en) Compositions and Methods for the Prevention and Treatment of Hearing Loss
JP2004099560A (en) Medicine for prevention and/or treatment of drug-induced nephropathy
CN113648310A (en) Application of GSK872 in preparation of medicine for treating nerve cell injury after cerebral hemorrhage
US9000047B2 (en) Compound inhibiting activation of the enzyme Erk 1/2 to be used in the treatment of neurodegenerative illnesses
EP4028012A1 (en) Compound for inhibiting cell death
CN102046165A (en) Methods of treating fibrotic disorders
WO2018136933A1 (en) Inhibition of stromal interaction molecule 1 (stim1) as a co-treatment for adult onset polycystic kidney disease (adpkd)
ES2341517B1 (en) USE OF AN INHIBITOR COMPOUND OF THE ACTIVATION OF THE ERK 1/2 ENZYME IN THE TREATMENT OF NEURODEGENERATIVE DISEASES.
EP3344244A1 (en) Use of indole compounds to stimulate the immune system
KR20030046119A (en) A composition for treating hepatic fibrosis and cirrhosis containing butein
CA3214549A1 (en) Nitazoxanide in the treatment of sepsis
WO2023015197A1 (en) Treatment or prevention of sars-cov-2 infection using (s)-crizotinib
Han Interplay between metformin and serotonin transport in the gastrointestinal tract: A novel mechanism for the intestinal absorption and adverse effects of metformin

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE