WO2022265960A1 - Prevention and treatment of neuronal damage with pyridoindolobenz[b, d] azepine compositions - Google Patents

Prevention and treatment of neuronal damage with pyridoindolobenz[b, d] azepine compositions Download PDF

Info

Publication number
WO2022265960A1
WO2022265960A1 PCT/US2022/033189 US2022033189W WO2022265960A1 WO 2022265960 A1 WO2022265960 A1 WO 2022265960A1 US 2022033189 W US2022033189 W US 2022033189W WO 2022265960 A1 WO2022265960 A1 WO 2022265960A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
neuroprotective
group
ddd
neuronal damage
Prior art date
Application number
PCT/US2022/033189
Other languages
French (fr)
Inventor
Lorenzo MANNELLI
Raghavan Rajagopalan
Carla Ghelardini
Parthasarathi Rajagopalan
Original Assignee
Daya Drug Discoveries, 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
Application filed by Daya Drug Discoveries, Inc. filed Critical Daya Drug Discoveries, Inc.
Publication of WO2022265960A1 publication Critical patent/WO2022265960A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/06Peri-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • 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

Definitions

  • This invention relates to the suppression, prevention, or treatment of nerve and glial cell damages induced by physical (e.g., injury, trauma, or drug abuse), pathological (e.g., cancer, diabetes, or microbial infection), or clinical (e.g., chemo- or radiation therapy).
  • physical e.g., injury, trauma, or drug abuse
  • pathological e.g., cancer, diabetes, or microbial infection
  • clinical e.g., chemo- or radiation therapy
  • the word ‘neuropathy’ signifies inflammation of the nervous system
  • the word, ‘neurotoxicity’ or ‘neuronal damage’ signifies damages to central or peripheral nervous system that includes both neurons and glial cells
  • the word, ‘neuroprotection’ or ‘neuroprotective’ signifies prevention of damages to the structure and/or function of neurons or glial cells, or restoration of structure and function after said damages had occurred
  • the phrase, ‘disease modifying,’ signifies alleviation of root cause(s) of pain, namely, nerve damage or inflammation.
  • the term “prevent or preventing” is meant either the reduction of or the complete blocking of a symptom and/or damage.
  • treatment signifies the repair or restoration of damaged cells to their normal or near normal condition.
  • Pain is essentially a defense mechanism to protect the body from further damage, and typically persists until the healing process is completed.
  • neuropathic pain is pathological and is only one among many cause of pain.
  • Neuropathy is a consequence of nerve damage caused by various ailments including injuries, trauma, infections, cancer, cancer therapies, and metabolic disorders. It is a progressive, enduring, and often irreversible condition featuring pain, numbness, tingling, and sensitivity to cold in the hands and feet, which sometimes progresses to the arms and legs. It is a serious medical condition, affecting a significant portion of cancer and 25 % of diabetic patients.
  • Chemotherapy induced neuropathy is a painful and a major dose-limiting side effect of cancer treatment that can interrupt, delay, or compel premature cessation of therapy [3, 4 ].
  • DPN diabetic peripheral neuropathy
  • Chemotherapeutic drugs include paclitaxel, alumin-bound paclitaxel, docetaxel, doxorubicin, doxil, avastin, epirubicin, bortezomib, 5-fluorouracil, cyclophosphamide, cisplatin, oxaliplatin, carboplatin, vinorelbine, capecitabine, gemcitabine, ixabepilone, eribulin, irinotecan, etoposide, vinblastine, vincristine, tamoxifen, methotrexate, and pemetrexed.
  • Neuropathy is a highly complex event involving degradation of both neurons and glial cells, and is mediated by numerous receptors, enzymes, transporters, cell signaling molecules, and other factors [5]
  • Mika [d] demonstrated that modulation of glial cells can attenuate neuropathic pain symptoms. In necrosis, proinflammatory cytokines are released which produces neural inflammation and pain [7]
  • Chemotherapeutic drugs produce reactive species such as reactive oxygen species (hydroxyl radicals, hydroperoxyl radicals, and superoxide), nitric oxide, and the like that are injurious to both neurons and glial cells.
  • Chronic neuropathic pain along with comorbid depression and anxiety are debilitating consequences of cancer, cancer therapy, diabetes, and viral infection [5]
  • Nerve degradation and pain are independent events. Although nerve damage leads to neuropathic pain, the perception of pain does not imply the existence of nerve damage. Further, in drug-induced neuropathies, some of the drugs are known to degrade mitochondria whereas others are known to damage axons, dendrites, or glial cells ultimately leading to necrotic cell death. At present, neuropathic pain is, in general, being managed with diverse CNS drugs such as opioids, cannabinoids (medical marijuana), gabapentin/pregabalin (epilepsy), duloxetine (anxiety), and amytryptiline (depression).
  • opioids cannabinoids (medical marijuana), gabapentin/pregabalin (epilepsy), duloxetine (anxiety), and amytryptiline (depression).
  • DDD-028 (4) has been shown to be a potent non-opioid analgesic in various pain models [73], viz., chronic constriction injury (CCI), spinal nerve ligation (SNL), complete Freund’s adjuvant (CFA) induced inflammation, and monosodium iodoacetate (MIA) induced osteoarthritis.
  • CCI chronic constriction injury
  • SNL spinal nerve ligation
  • CFA complete Freund’s adjuvant
  • MIA monosodium iodoacetate
  • the present invention relates to the method of treating or preventing neuronal damage in patients afflicted with or potentially encounter neuropathic condition by administering effective amount of pharmaceutically acceptable compositions of Formula 1, wherein Y is a single bond or a double bond; A and B are independently -(CH2) n- ; and subscript ‘n’ varies from 0 to 3.
  • Each of the substituents, R 1 to R 10 is independently selected to optimize efficacy and/or safety.
  • the present invention further relates to the method of treating or preventing neuronal damage comprising administering an effective amount of compounds of Formula I in combination with other neuroprotective agents (e.g. RXDAx accelerants), analgesics (e.g. opioids), anti-epileptics (pregabalin), or antidepressants (e.g., duloxetine) to elicit additive or synergistic effect.
  • neuroprotective agents e.g. RXDAx accelerants
  • analgesics e.g. opioids
  • FIG. 1 Acute analgesic effect in paclitaxel CIN model.
  • FIG. 3 Neuroprotective effect: oxidative stress.
  • FIG. 4. Neuroprotective effect: reactive oxygen species (ROS).
  • ROS reactive oxygen species
  • FIG. 5 Neuroprotective effect: glial cell proliferation and morphology, spinal cord.
  • FIG. 6 Neuroprotective effect: glial cell proliferation, brain.
  • FIG. 7 Neuroprotective effect: intraepidermal nerve fiber (iENF) density.
  • FIG. 8 Neuroprotective effect: neurofilament heavy and light chain assays.
  • FIG. 9 Neuroprotective effect: myelin degradation.
  • FIG. 10 Pharmacodynamic mechanism of DDD-028. DETAILED DESCRIPTION
  • the present invention relates to the method of preventing or treating neuronal damage comprising administering an effective dose of Formula 1, wherein Y is a single bond or a double bond; A and B are independently -(CH 2 ) n- ; subscript ‘n’ varies from 0 to 3; and each of R 1 to R 10 is independently selected from the group consisting of hydrogen, Ci-Cio alkyl, Ci-Cio acyl, hydroxyl, amino, halo, cyano, carboxyl, Ci-Cio alkoxyl, Ci-Cio hydroxyalkyl, Ci-Cio alkoxycarbonyl, Ci-Cio alkoxycarbonylalkyl; Ci-Cio carbamoylalkyl C 5 -C 10 aryl unsubstituted or substituted with electron donating groups (EDG) or electron withdrawing groups (EWG), C 5 -C 10 arylalkyl wherein the aryl portion is unsubstituted or
  • EDG electrospray donating group
  • EWG electrospray withdrawing group
  • EDG comprises alkyl, hydroxyl, alkoxyl, amino, acyloxy, acylamino, mercapto, alkylthio, and the like.
  • EWG comprises halogen, acyl, nitro, cyano, carboxyl, alkoxycarbonyl, and the like.
  • the present invention further relates to the method of preventing or treating neuronal damage comprising administering an effective dose of a combination of Formula 1 and another neuroprotective and/or analgesic agent, wherein said neuroprotective agents comprise RXDAx accelerants, SARMl NADase inhibitors, Hsp modulators, and the like; and analgesic agents comprise opioids such as, but not limited to, morphine or oxycodone; cannabinoids such as, but not limited to, cannabidiol; anti-epileptic agents such as, but not limited to, pregabalin or gabapentin, anti-depressants such as, but not limited to duloxetine.
  • One embodiment of the present invention is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 5 of Formula I, wherein,
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 10 alkyl, C 1 -C 10 hydroxyalkyl, C 5 -C 10 , C 1 -C 15 aroylalkyl, C 1 -C 10 alkoxycarbonylalkyl, C 5 -C 10 arylalkyl, and C 1 -C 10 carbamoylalkyl;
  • R 2 is selected from the group consisting of hydrogen, Ci-Cio alkyl, C 5 -C 10 aryl, and C5-C10 arylalkyl; each of R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, -NR U R 12 , C1-C10 hydroxyalkyl, halogen, trihaloalkyl, cyano, carboxyl, C 1
  • R 11 and R 12 are independently hydrogen or C1-C10 alkyl.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 6 of Formula I, wherein
  • Y is a double bond
  • A is -CH2-
  • B is -CH2CH2-.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 7 of Formula I, wherein
  • Y is a double bond
  • A is -CH2-
  • B is -CH2CH2-
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 10 alkyl, C 1 -C 10 hydroxyalkyl, C 5 -C 10 arylalkyl, and C 1 -C 10 carbamoylalkyl;
  • R 2 and R 3 are hydrogens; and each of R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C 1 -C 10 alkyl, hydroxyl, C 1 -C 10 alkoxyl, and halogen.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 8 of Formula I, wherein
  • Y is a double bond
  • A is -CH2-
  • R B is -CH2CH2-; each of R 4 , R 5 , R 6 , and R 7 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen;
  • R 8 , R 9 , and R 10 are hydrogens; optionally wherein
  • R 1 is hydrogen, C 1 -C 10 alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 4 , R 5 , R 6 , and R 7 is hydrogen, CFb, hydroxyl, OCH 3 , F, or Cl.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 9 of Formula I, wherein
  • Y is a double bond
  • A is -CH 2- ;
  • B is -CH2CH2-
  • R 4 , R 5 , R 6 , and R 7 are hydrogens; and each of R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C1-C1 0 alkyl, hydroxyl, C1-C1 0 alkoxyl, and halogen, optionally wherein
  • R 1 is hydrogen, C 1 -C 10 alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 8 , R 9 , and R 10 is hydrogen, CH 3 , hydroxyl, OCH 3 , F, or Cl, in particular optionally wherein R 1 is hydrogen or CFb; and R 8 , R 9 , and R 10 are hydrogens.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 10 of Formula I, wherein
  • Y is a single bond with cA-fused B
  • A is -CH2-
  • B is -CH2CH2-
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 11 of Formula I, wherein
  • Y is a single bond with cA-fused B
  • A is -CH2-
  • B is -CH2CH2-
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 10 alkyl, C 1 -C 10 hydroxyalkyl, C 5 -C 10 arylalkyl, and C 1 -C 10 carbamoylalkyl;
  • R 2 and R 3 are hydrogens; and each of R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C1-C1 0 alkyl, hydroxyl, C1-C1 0 alkoxyl, and halogen.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 12 of Formula I, wherein
  • Y is a single bond with cA-fused B
  • A is -CH2-
  • R B is -CH2CH2-; each of R 4 , R 5 , R 6 , and R 7 is independently selected from the group consisting of hydrogen, C 1 -C 10 alkyl, hydroxyl, C 1 -C 10 alkoxyl, and halogen; and R 8 , R 9 , and R 10 are hydrogens, optionally wherein
  • R 1 is hydrogen, Ci-Cio alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 and each of R 4 , R 5 , R 6 , and R 7 is hydrogen, CH 3 , hydroxyl, OCH 3 , F, or Cl, optionally wherein
  • R 4 , R 5 , R 6 , and R 7 are hydrogens; and each of R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C1-C1 0 alkyl, hydroxyl, C1-C1 0 alkoxyl, and halogen, optionally wherein
  • R 1 is hydrogen, C1-C10 alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 8 , R 9 , and R 10 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, in particular optionally wherein R 1 is hydrogen or CFb; and R 8 , R 9 , and R 10 are hydrogens.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 13 of Formula I, wherein
  • Y is a single bond with trans- fused B
  • A is -CH 2- ;
  • B is -CH 2 CH 2-.
  • Another embodiment is related to the prevention of neuronal damage by administering an effective amount of Compound 14 of Formula I, wherein
  • Y is a single bond with trans- fused B
  • A is -CH 2- ;
  • B is -CH 2 CH 2-.
  • R 1 is selected from the group consisting of hydrogen, C 1 -C 10 alkyl, C 1 -C 10 hydroxyalkyl, C 5 -C 10 arylalkyl, and C 1 -C 10 carbamoylalkyl;
  • R 2 and R 3 are hydrogens; and each of R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C 1 -C 10 alkyl, hydroxyl, C 1 -C 10 alkoxyl, and halogen.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 15 of Formula I, wherein
  • Y is a single bond with trans- fused B
  • A is -CH 2- ;
  • R B is -CH 2 CH 2-.
  • each of R 4 , R 5 , R 6 , and R 7 is independently selected from the group consisting of hydrogen, Ci-Cio alkyl, hydroxyl, Ci-Cio alkoxyl, and halogen; and R 8 , R 9 , and R 10 are hydrogens, optionally wherein
  • R 1 is hydrogen, Ci-Cio alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 4 , R 5 , R 6 , and R 7 is hydrogen, CH 3 , hydroxyl, OCH 3 , F, or Cl; wherein
  • R 4 , R 5 , R 6 , and R 7 are hydrogens; and each of R 8 , R 9 , and R 10 is independently selected from the group consisting of hydrogen, C1-C1 0 alkyl, hydroxyl, C1-C1 0 alkoxyl, and halogen, optionally wherein
  • R 1 is hydrogen, C 1 -C 10 alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 8 , R 9 , and R 10 is hydrogen, CH 3 , hydroxyl, OCH 3 , F, or Cl, in particular optionally wherein
  • R 1 is hydrogen, C 1 -C 10 alkyl, CH 2 Ph, CH 2 CH 2 OH, or CH 2 CONH 2 ; and each of R 8 , R 9 , and R 10 is hydrogen, CH 3 , hydroxyl, OCH 3 , F, or Cl, in particular optionally wherein R 1 is hydrogen or CFb; and R 8 , R 9 , and R 10 are hydrogens.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 16 of Formula I, wherein
  • R 1 is hydrogen, methyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, or iso butyl, preferably hydrogen, methyl, n-propyl, or iso-propyl, more preferably hydrogen or methyl, and most preferably methyl; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is hydrogen;
  • Y is a double bond
  • A is -CH 2- ;
  • B is -CH 2 CH 2-.
  • Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 4 (DDD-028) of Formula I, wherein R 1 is methyl; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is hydrogen;
  • Y is a double bond
  • A is -CH 2- ;
  • Another embodiment is related to the prevention of CIN and DPN by administering an effective amount of a combination of compound Formula I and a neuroprotective agent comprising RXDAx accelerants, SARM1 NADase inhibitors, or Hsp modulators.
  • Another embodiment is related to the prevention of CIN and DPN by administering an effective amount of a combination of compound Formula I and an analgesic agent comprising opioids, cannabinoids, antiepileptics, or antidepressants.
  • Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) to patients undergoing or about to start chemotherapy.
  • Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) and a neuroprotective agent comprising RXDAx accelerants, SARM1 NADase inhibitors, or Hsp modulators to patients undergoing or about to start chemotherapy.
  • DDD-028 compound 4
  • a neuroprotective agent comprising RXDAx accelerants, SARM1 NADase inhibitors, or Hsp modulators
  • Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) and an analgesic agent comprising opioids, cannabinoids, antiepileptics, or antidepressants.
  • DDD-028 potent, non-opioid analgesic in several models of neuropathic pain 13
  • DDD-028 is not only a potent analgesic in the CIN model, but, more importantly, it prevents both glial and nerve cell damages.
  • analgesic activity viz., Paw Pressure for mechanical hyperalgesia, Von Frey for mechanical allodynia, and Cold Plate for Thermal allodynia
  • DDD-028 was able to recover, dose dependently, paclitaxel -induced hypersensitivity as well as to prevent the development of neuropathic symptoms when it was co-administered with the chemotherapeutic drug (Fig. 1).
  • DDD-028 The neuroprotective activity of one of the compounds of Formula 1, viz., DDD-028, in rat model of CIN has been demonstrated using many well-accepted assays using the brain and spinal cord and plasma: these test comprise glial cell proliferation, glial cell damage, protein degradation, (i.e., oxidative stress), peroxisome activity, nerve conduction, and nerve fiber density.
  • DDD-028 prevented oxidative stress induced by paclitaxel (Figs. 3, 4).
  • DDD- 028 blocks the proliferation of glial cells both in the peripheral regions as well in the brain (Fig. 5), and prevents astrocyte damage (Fig. 5, 6).
  • DDD-028 blocks palitaxel induced nerve degration as shown by three immunohistopathologial studies (Fig. 7-9). Thus, collectively these data firmly demonstrate that DDD-028 is both an analgesic and a neuroprotective agent. It is believed that dual action of DDD-028 is likely mediated via nicotinic acetylcholine pathway, particularly the a7NAChR (Fig. 10).
  • DDD-028 is well tolerated, does not induce sedation, and appears to be safe at the expected dose regimen.
  • CNS MPO CNS multiparameter optimization
  • the compounds of the present invention represented by Formula I commonly referred to as ‘active pharmaceutical ingredient (API)’ or ‘drug substance’ is typically formulated as pharmaceutically acceptable salts.
  • pharmaceutically acceptable means those formulations which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • compositions include, but are not limited to acetate, adipate, citrate, tartarate, benzoate, phosphate, glutamate, gluconate, fumarate, maleate, succinate, oxalate, chloride, bromide, hydrochloride, sodium, potassium, calcium, magnesium, or ammonium.
  • drug product commonly referred to as ‘drug product,’ may be administered enterally, parenterally, or topically.
  • Enteral route includes oral, rectal, topical, buccal, ophthalmic, and vaginal administration.
  • Parenteral route includes intravenous, intramuscular, intraperitoneal, intrastemal, and subcutaneous injection or infusion.
  • the drug product may be delivered in solid, liquid, or vapor forms, or can be delivered through a catheter for local delivery at a target.
  • Topical delivery may also include transdermal patches or transdermal microneedles for subcutaneous delivery. Transdermal patch and microneedle technology described by Tran et al. ⁇ 14 ⁇ and is incorporated by reference in entirety.
  • the dosage levels of API in the drug product can be varied so as to achieve the desired therapeutic response for a particular patient.
  • the phrase "effective amount” or “effective dose” of the compound of the invention means a sufficient amount of the compound is administered to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated, the severity of the disorder; activity of the specific compound employed; the specific composition employed, age, body weight, general health, sex, diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed, and the duration of the treatment.
  • the total daily dose of the compounds of this invention administered may range from about 0.0001 to about 1000 mg/kg/day.
  • preferable doses can be in the range from about 0.001 to about 5 mg/kg/day. If desired, the effective daily dose can be divided into multiple doses for optimal therapeutic effect.
  • the compostions of the present invention may be administered to any patient experiencing or expected to encounter neuropathy regardless of its cause.
  • chemotherapy co-treatment means giving the anti-cancer compound to the patient before, during, or after administration of a dose of chemotherapy, in a manner that prevents CIN.
  • the phrase “before administration” of a dose of chemotherapy may include a several seconds to several hours (such as 24 hours) before the chemotherapy.
  • “During” administration of a dose of chemotherapy may include a specific administration given at any time during the chemotherapy, or co-administration for the duration of the chemotherapy administration (such as by combining the two drugs in the same infusion bag).
  • “After” administration of a dose of chemotherapy may include administration several minutes or hours after the chemotherapy, and may include administration on a schedule for a number of days after the dose of chemotherapy.
  • Formulations for oral administration include capsules (soft or hard), tablets, pills, powders, and granules.
  • Such formulations may comprise the API along with at least one inert, pharmaceutically acceptable ingredients selected from the following: (a) buffering agents such as sodium citrate or dicalcium phosphate; (b) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; (c) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants such as glycerol; (e) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; (f) solution retarding agents such as paraffin; (g) absorption accelerators such as quaternary ammonium compounds; (h) wetting agents such as cetyl alcohol and glyce
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents, solubilizing agents, wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents used in the art.
  • compositions suitable for parenteral injection may comprise physiologically acceptable, sterile aqueous or nonaqueous isotonic solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • the compositions may also optionally contain adjuvants such as preserving; wetting; emulsifying; dispensing, and antimicrobial agents, buffers, diluents, carriers, adjuvants, preservatives, and excipients.
  • Suitable carriers, diluents, solvents, vehicles, or adjuvants include, but are not limited to water; ethanol; polyols such as propyl enegly col, polyethyleneglycol, glycerol, and the like; vegetable oils such as cottonseed, groundnut, com, germ, olive, castor and sesame oils, and the like; organic esters such as ethyl oleate and the like; phenol, parabens, sorbic acid, and the like.
  • Injectable formulations may also be suspensions that contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • the absorption of the drug in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsulated matrices of the drug in biodegradable polymers. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, these compositions release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Thus, the rate of drug release and the site of delivery can be controlled.
  • embedding compositions include, but are no limited to polylactide-polyglycolide poly(orthoesters), and poly(anhydrides), and waxes. The technology pertaining to controlled release formulations are described in “Design of Controlled Release Drug Delivery Systems,” [75] incorporated herein by reference in its entirety.
  • Formulations for topical administration include powders, sprays, ointments and inhalants. These formulations include the API along with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients and the like.
  • the preferred lipids are natural and synthetic phospholipids and phosphatidyl cholines (lecithins) used separately or together. Methods to form liposomes are known in the art and are described in “Liposomes,” [76], which is incorporated herein by reference in its entirety.
  • the compounds of the present invention can also be administered to a patient in the form of pharmaceutically acceptable ‘prcrirugs.’
  • Prodrugs are generally used to enhance the bioavailability, solubility, in vivo stability, or any combination thereof of the API. They are typically prepared by linking the API covalently to a biodegradable functional group such as a phosphate that will be cleaved enzymatically or hydrolytically in blood, stomach, or GI tract to release the API.
  • a biodegradable functional group such as a phosphate that will be cleaved enzymatically or hydrolytically in blood, stomach, or GI tract to release the API.
  • the nociceptive threshold in the rat was determined with an analgesimeter (Ugo Basile, Varese, Italy) according to the method described by Leighton et al. [75] Briefly, a constantly increasing pressure was applied to a small area of the dorsal surface of the hind paw using a blunt conical mechanical probe. Mechanical pressure was increased until vocalization or a withdrawal reflex occurred while rats were lightly restrained. Vocalization or withdrawal reflex thresholds were expressed in grams. These limits assured a more precise determination of mechanical withdrawal threshold in experiments aimed to determine the effect of treatments. An arbitrary cut-off value of 100 g was adopted.
  • DDD-028 was acutely per os administered when neuropathy was well established (day 10), 48 h after the last chemotherapeutic drug injection.
  • day 10 paclitaxel -treated rats showed a significantly reduction of the weight tolerated on posterior paws with respect to the control animals (43.2 ⁇ 0.5 g vs 66.5 ⁇ 0.7 g, respectively).
  • Increasing doses of DDD-028 (1 - 25 mg kg 1 ) reduced mechanical hypersensitivity in a dose-dependent manner starting 30 min after treatment.
  • the highest dose of DDD-028 (25 mg kg 1 ) completely abrogated paclitaxel- induced mechanical hyperalgesia with maximum analgesic effect occurring at 30 min (Fig. la).
  • DDD-028 counteracted paclitaxel-induced mechanical allodynia in a dose-dependent manner in the Von Frey test (Fig. lb).
  • the highest dose again showed a complete reversal of paclitaxel-induced neuropathy with a long-lasting effect starting from 30 min up to 90 min after treatment.
  • All the lower doses of DDD-028 displayed a shorter anti-hyperalgesic efficacy (Fig. lb).
  • DDD-028 also counteracted paclitaxel-induced thermal allodynia in a dose-dependent manner in the Cold Plate test (Fig. lc).
  • Thermal allodynia was assessed using the Cold-plate test. With minimal animal- handler interaction, rats were taken from home-cages, and placed onto the surface of the cold- plate maintained at a constant temperature of 4 ⁇ 1 °C. Ambulation was restricted by a cylindrical Plexiglas chamber (diameter: 10 cm, height: 15 cm), with open top. A timer controlled by foot pedal began timing response latency from the moment the mouse was placed onto the cold-plate. Pain-related behavior (licking of the hind paw) was determined by recording the time (seconds) of the first sign of licking of the hind paw.
  • the cut-off time of the latency of paw lifting or licking was set at 30 s.
  • DDD-028 was able to recover, dose dependently, paclitaxel-induced hypersensitivity and prevent the development of neuropathic symptoms when it was co-administered with the chemotherapeutic drug (Fig. lc).
  • DDD-028 Prophylactic Analgesic Effect of DDD-028 in ON [0058] Thereafter, to assess the protective profile of DDD-028, the compound was subjected to repeated administrations over an 18-day period.
  • Paclitaxel-treated animals were administered daily with DDD-028 (10 mg kg 1 , p.o.) starting from the same day of paclitaxel injection.
  • the response to mechanical noxious stimulus was measured on days 10, 12 and 18, 24 h after the last treatment.
  • DDD-028 significantly increased the pain threshold of paclitaxel -injected rats at all time points considered without development of tolerance to the anti-hypersensitivity effect.
  • Example 5 Neuroprotective Effect of DDD-028 in ON - Oxidative Stress [0059] Among the multiplicity of the pathophysiologic mechanisms, this oxidative stress plays a crucial role in the generation of paclitaxel-induced neuropathy.
  • day 18 i.e., the end of the experiments on sub-chronic analgesic effect of DDD-028), animals were sacrificed by decapitation.
  • L4-L5 dorsal root ganglia (DRG), sciatic nerve, lumbar spinal cord and brain were collected, frozen using liquid nitrogen or fixed by immersion in 4% neutral buffered formalin. Sciatic nerve and DRGs protein extracts were quantified by bicinchoninic acid.
  • the membranes were incubated for 1 hour in PBST containing the appropriate horseradish peroxidase-conjugated secondary antibody (1:5000; Cell Signalling, USA) and again washed. ECL (Pierce, USA) was used to visualize the peroxidase-coated bands. Densitometric analysis was performed using the “Image J” analysis software and the density of all bands displayed in the lane is reported as a mean.
  • Ponceau-stained membranes were used as loading control [4]
  • Formalin fixed cryostat sections (10 pm for brain and 5 pm for spinal cord) were incubated for 1 h in blocking solution (Bio-Optica; Italy) at room temperature, and thereafter sections were incubated for 24 h at 4 °C in PBST containing primary antisera and 5% normal donkey serum.
  • the primary antibody was directed against Ibal (rabbit antiserum, 1:500; Wako Chemicals, USA; [23]) for microglial staining and against glial fibrillary acidic protein (GFAP; rabbit antiserum, 1:500; Dako, USA;[28]) for astrocyte staining.
  • Ibal rabbit antiserum, 1:500; Wako Chemicals, USA; [23]
  • GFAP glial fibrillary acidic protein
  • Morphological examination of microglia and astrocytes was assessed by inspection of at least three fields (40 c 0.75NA objective) in the dorsal horn of the spinal cord and brain areas per section.
  • the full specimen thicknesses were acquired as z-stack series, deconvolved using Huygens Professional software (SVI, The Netherlands) and displayed using ImageJ software.
  • Huygens Professional software SVI, The Netherlands
  • ImageJ software Huygens Professional software
  • Oxidative stress a typical signature of chemotherapy-induced neurotoxicity, was measured in the peripheral nervous system.
  • paclitaxel induced an oxidative damage of dorsal root ganglia (DRG) as indicated by three-fold increase in carbonylation of proteins.
  • DDD-028 resulted in a significant prevention of the damage as evidenced by protein carbonylation values similar to the control group (Fig. 3).
  • DDD-028 was able to prevent the development of hypersensitivity, and to reduce paclitaxel-related damage to PNS and CNS when repeatedly administered with the anticancer drug.
  • DDD-028 demonstrated detoxifying properties as indicated by the enhancing the activity of the peroxisomal enzyme, catalase, and reducing the protein oxidation in DRGs (Fig. 4).
  • paclitaxel + DDD-028 showed a significantly lower number of astrocytes characterized by a reactive phenotype. Moreover, spinal astrocytes presented altered morphology showing hypertrophy of the cell body and processes (Fig. 5c). Microglia activation was measured by the quantification of Ibal positive cells in the spinal cord of treated rats. On day 18, paclitaxel treatment produced increased density of Ibal positive cells in the dorsal horns of the lumbar spinal cord (Fig. 5b). Animals treated with paclitaxel + DDD-028 showed a significant prevention of microglia activation (Figure 5b).
  • day 18 can be considered a late phase for microglia activation that is, according to the literature, strongly involved in the first days of treatment.
  • DDD-028 Neuroprotective Effect of DDD-028 in CIN - Astrocyte Proliferation, Brain [0062]
  • PAG periacqueductal gray
  • SI somatosensory cortex
  • Paclitaxel induced a significant numerical increase of both glial cell populations in brain areas, but a higher astrocyte activation was observed (Figs. 6a, b).
  • DDD-028 reduced both microglia and astrocyte cell number increase (numerical activation) in PAG, thalamus and SI.
  • IENF density fibers/mm
  • IENF density fibers/mm
  • NF-H neurofilament heavy chain
  • NF-L Neurofilament light chain
  • DDD-028 in CIN - Myelin Degradation
  • Paclitaxel 2.0 mg kg 1 was dissolved in a mixture of 10% Cremophor EL and saline solution and intraperitoneally (i.p.) administered on four alternate days (1, 3, 5 and 8).
  • DDD-028 (10 mg kg 1 ) was suspended in 1% carboxymethylcellulose sodium salt (CMC) and daily administered per os starting the first day of paclitaxel treatment and continuing 1 week after the end of treatment (from day 1 to day 17). Animals were sacrificed on day 18, 24h after the last treatment.
  • CMC carboxymethylcellulose sodium salt
  • Sciatic nerves and DRGs were fixed in 2.5% glutaraldehyde in cacodylate buffer, pH 7.4, for 24 hours. Tissues were embedded in Epoxy resin; sections (0.8 pm) were cut using an ultramicrotome, stained with toluidine blue. DRGs were studied measuring the number of neurons showing multinucleolated nuclei as sign of toxic alteration; the soma area was also assessed stratifying neurons in small ( ⁇ 600 microm diameter), medium (>600 ⁇ 1200 microm diameter) and large (>1200 microm diameter). In the sciatic nerve, myelin area, number of fibers (separated by axon diameter) and axon diameters were analyzed. The results indicate that DDD-028 prevents axon degradtion induced by paclictaxel (Fig. 10).
  • DDD-028 was subjected to the modulation of the s receptor-mediated neck dystonia. Accordingly, the s agonist DTG was injected in the red nucleus. DTG induced postural changes characterized by a marked deviation in the head angle (neck dystonia), peaking at 25-30 min after microinjection. DDD-028 (25 mg kg 1 ) was administered per os 15 min before DTG infusion without preventing DTG-induced neck dystonia (Fig. 1 lb).
  • DDD-028 25 mg kg 1 , per os
  • MECA 2 mg kg 1 , i.p.

Abstract

A method of preventing or treating neuronal damage in a patient being administered a therapy that can cause nerve damage, comprising administering to said patient an effective amount of a neuroprotective compounds for Formula 1, (I) wherein Y is a single bond or a double bond; A and B are independently -(CH2)n-; subscript 'n' varies from 0 to 3.

Description

PREVENTION AND TREATMENT OF NEURONAL DAMAGE WITH PYRIDOINDOLOBENZ [b, JAZEPINE
This application claims benefit of priority based on provisional application No. 63/210,063 filed on June 13, 2021, and said application is incorporated herein by reference in its entirety.
CROSS-REFERENCE TO RELATED APPLICATIONS Not applicable.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR
DEVELOPMENT
Not applicable.
THE NAMES OF THE PARTIES TO A JOINT RESEARCH AGREEMENT Not applicable.
INCORPORATION-BY-REFERECE OF MATERIAL SUBMITTED ON A COMPACT DISC
Not Applicable.
BACKGROUND OF THE INVENTION Field of the Invention.
[0001] This invention relates to the suppression, prevention, or treatment of nerve and glial cell damages induced by physical (e.g., injury, trauma, or drug abuse), pathological (e.g., cancer, diabetes, or microbial infection), or clinical (e.g., chemo- or radiation therapy).
Description of the Related Art.
[0002] Various prior art references in the specification are indicated by italicized Arabic numerals in brackets. Full citation corresponding to each reference number is listed at the end of the specification and is herein incorporated by reference in its entirety to describe fully and clearly the state of the art to which this invention pertains.
[0003] Unless otherwise specified, all technical terms and phrases used herein conform to standard organic and medicinal chemistry nomenclature established by International Union of Pure and Applied Chemistry (IUPAC), the American Chemical Society (ACS), and other international professional societies. The rules of nomenclature are described in various publications, including, “Nomenclature of Organic Compounds,” [7], and “Systematic Nomenclature of Organic Chemistry” [2], which are herein incorporated by reference in their entireties. For the purpose of this application, the word ‘neuropathy’ signifies inflammation of the nervous system; the word, ‘neurotoxicity’ or ‘neuronal damage’ signifies damages to central or peripheral nervous system that includes both neurons and glial cells; the word, ‘neuroprotection’ or ‘neuroprotective’ signifies prevention of damages to the structure and/or function of neurons or glial cells, or restoration of structure and function after said damages had occurred; the phrase, ‘disease modifying,’ signifies alleviation of root cause(s) of pain, namely, nerve damage or inflammation. The term “prevent or preventing” is meant either the reduction of or the complete blocking of a symptom and/or damage. The term “treatment” signifies the repair or restoration of damaged cells to their normal or near normal condition. [0004] Pain is essentially a defense mechanism to protect the body from further damage, and typically persists until the healing process is completed. However, neuropathic pain is pathological and is only one among many cause of pain. Neuropathy is a consequence of nerve damage caused by various ailments including injuries, trauma, infections, cancer, cancer therapies, and metabolic disorders. It is a progressive, enduring, and often irreversible condition featuring pain, numbness, tingling, and sensitivity to cold in the hands and feet, which sometimes progresses to the arms and legs. It is a serious medical condition, affecting a significant portion of cancer and 25 % of diabetic patients. Chemotherapy induced neuropathy (CIN) is a painful and a major dose-limiting side effect of cancer treatment that can interrupt, delay, or compel premature cessation of therapy [3, 4 ]. Likewise, diabetic peripheral neuropathy (DPN) is also a debilitating condition that impairs quality of life. Chemotherapeutic drugs include paclitaxel, alumin-bound paclitaxel, docetaxel, doxorubicin, doxil, avastin, epirubicin, bortezomib, 5-fluorouracil, cyclophosphamide, cisplatin, oxaliplatin, carboplatin, vinorelbine, capecitabine, gemcitabine, ixabepilone, eribulin, irinotecan, etoposide, vinblastine, vincristine, tamoxifen, methotrexate, and pemetrexed. [0005] Neuropathy is a highly complex event involving degradation of both neurons and glial cells, and is mediated by numerous receptors, enzymes, transporters, cell signaling molecules, and other factors [5] Mika [d] demonstrated that modulation of glial cells can attenuate neuropathic pain symptoms. In necrosis, proinflammatory cytokines are released which produces neural inflammation and pain [7] Chemotherapeutic drugs produce reactive species such as reactive oxygen species (hydroxyl radicals, hydroperoxyl radicals, and superoxide), nitric oxide, and the like that are injurious to both neurons and glial cells. Chronic neuropathic pain along with comorbid depression and anxiety are debilitating consequences of cancer, cancer therapy, diabetes, and viral infection [5]
[0006] Nerve degradation and pain are independent events. Although nerve damage leads to neuropathic pain, the perception of pain does not imply the existence of nerve damage. Further, in drug-induced neuropathies, some of the drugs are known to degrade mitochondria whereas others are known to damage axons, dendrites, or glial cells ultimately leading to necrotic cell death. At present, neuropathic pain is, in general, being managed with diverse CNS drugs such as opioids, cannabinoids (medical marijuana), gabapentin/pregabalin (epilepsy), duloxetine (anxiety), and amytryptiline (depression). Currently prescribed medications, oxycodone and duloxetine only relieve pain symptoms of CIPN; they do not block or suppress underlying nerve or glial damage. Scott et al. [9] disclose non-opioid analgesic 1 for neuropathic pain by activating the reactive species decomposition accelerant (RSDAx) pathway, but this compound has not demonstrated to be neuroprotective. Hoke et al. [70] disclose ethoxyquin (2) as a neuroprotective analgesic against cisplatin-induced CIPN that functions via the modulation of heat shock protein (Hsp). Melbrandt et al. [77] disclose neuroprotective agent 3 that prevents axonal degradation by inhibiting the SARM1 NADase
Figure imgf000004_0001
enzyme. However, no neuroprotective drugs are currently available for the treatment of neuropathic pain, including CIN and DPN. Hence, there is a considerable need for drugs that not only alleviate pain, but, more importantly, prevent neuron and glial cell damages.
SUMMARY
[0007] It is well recognized in the art that relief of pain and prevention/treatment of nerve damage are independent, but related events. Pain sensations depend on specific physical, clinical, or pathological causes, and the perception of pain does not necessarily imply the existence of nerve damage, i.e., an individual may still experience non-neuropathic pain even after treating the nerve damage. Rajagopalan [72], which is incorporated herein by reference it its entirety, discloses pridoindolobenz[Z>,i/]azepine derivatives of Formula 1 for the treatment of CNS disorders. One of the benzazepine derivatives, DDD-028 (4) has been shown to be a potent non-opioid analgesic in various pain models [73], viz., chronic
Figure imgf000005_0001
constriction injury (CCI), spinal nerve ligation (SNL), complete Freund’s adjuvant (CFA) induced inflammation, and monosodium iodoacetate (MIA) induced osteoarthritis. However, Rajagopalan neither demonstrated nor disclosed the use of said benzazepine derivatives for the prevention and treatment of nerve or glial cell damage. Clearly, prevention of neuronal damage cannot be predicted from their analgesic effect. Therefore, the present invention relates to the method of treating or preventing neuronal damage in patients afflicted with or potentially encounter neuropathic condition by administering effective amount of pharmaceutically acceptable compositions of Formula 1, wherein Y is a single bond or a double bond; A and B are independently -(CH2)n-; and subscript ‘n’ varies from 0 to 3. Each of the substituents, R1 to R10, is independently selected to optimize efficacy and/or safety. [0008] The present invention further relates to the method of treating or preventing neuronal damage comprising administering an effective amount of compounds of Formula I in combination with other neuroprotective agents (e.g. RXDAx accelerants), analgesics (e.g. opioids), anti-epileptics (pregabalin), or antidepressants (e.g., duloxetine) to elicit additive or synergistic effect.
BRIEF DESCRIPTION OF FIGURES
[0009] FIG. 1. Acute analgesic effect in paclitaxel CIN model.
[0010] FIG. 2. Prophylactic analgesic effect in CIN pain model.
[0011] FIG. 3. Neuroprotective effect: oxidative stress.
[0012] FIG. 4. Neuroprotective effect: reactive oxygen species (ROS).
[0013] FIG. 5. Neuroprotective effect: glial cell proliferation and morphology, spinal cord. [0014] FIG. 6. Neuroprotective effect: glial cell proliferation, brain.
[0015] FIG. 7. Neuroprotective effect: intraepidermal nerve fiber (iENF) density.
[0016] FIG. 8 Neuroprotective effect: neurofilament heavy and light chain assays.
[0017] FIG. 9 Neuroprotective effect: myelin degradation.
[0018] FIG. 10 Pharmacodynamic mechanism of DDD-028. DETAILED DESCRIPTION
[0019] The present invention relates to the method of preventing or treating neuronal damage comprising administering an effective dose of Formula 1,
Figure imgf000006_0001
wherein Y is a single bond or a double bond; A and B are independently -(CH2)n-; subscript ‘n’ varies from 0 to 3; and each of R1 to R10 is independently selected from the group consisting of hydrogen, Ci-Cio alkyl, Ci-Cio acyl, hydroxyl, amino, halo, cyano, carboxyl, Ci-Cio alkoxyl, Ci-Cio hydroxyalkyl, Ci-Cio alkoxycarbonyl, Ci-Cio alkoxycarbonylalkyl; Ci-Cio carbamoylalkyl C5-C10 aryl unsubstituted or substituted with electron donating groups (EDG) or electron withdrawing groups (EWG), C5-C10 arylalkyl wherein the aryl portion is unsubstituted or substituted with EDG or EWG); C5-C10 aroylalkyl wherein the aryl portion is unsubstituted or substituted with EDG or EWG). The phrase, ‘electron donating group (EDG)’ and ‘electron withdrawing group (EWG)’ are well understood in the art. EDG comprises alkyl, hydroxyl, alkoxyl, amino, acyloxy, acylamino, mercapto, alkylthio, and the like. EWG comprises halogen, acyl, nitro, cyano, carboxyl, alkoxycarbonyl, and the like. [0020] The present invention further relates to the method of preventing or treating neuronal damage comprising administering an effective dose of a combination of Formula 1 and another neuroprotective and/or analgesic agent, wherein said neuroprotective agents comprise RXDAx accelerants, SARMl NADase inhibitors, Hsp modulators, and the like; and analgesic agents comprise opioids such as, but not limited to, morphine or oxycodone; cannabinoids such as, but not limited to, cannabidiol; anti-epileptic agents such as, but not limited to, pregabalin or gabapentin, anti-depressants such as, but not limited to duloxetine. [0021] One embodiment of the present invention is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 5 of Formula I, wherein,
R1 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 hydroxyalkyl, C5-C10, C1-C15 aroylalkyl, C1-C10 alkoxycarbonylalkyl, C5-C10 arylalkyl, and C1-C10 carbamoylalkyl; R2 is selected from the group consisting of hydrogen, Ci-Cio alkyl, C5-C10 aryl, and C5-C10 arylalkyl; each of R3, R4, R5, R6, R7, R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, -NRUR12, C1-C10 hydroxyalkyl, halogen, trihaloalkyl, cyano, carboxyl, C1-C10 acyl, C1-C10 alkoxyalkyl; C1-C10 alkoxycarbonyl, C5-C10 aryl, and C5-C10; and
R11 and R12 are independently hydrogen or C1-C10 alkyl.
[0022] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 6 of Formula I, wherein
Y is a double bond;
A is -CH2-; and
B is -CH2CH2-.
[0023] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 7 of Formula I, wherein
Y is a double bond;
A is -CH2-;
B is -CH2CH2-;
R1 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 hydroxyalkyl, C5-C10 arylalkyl, and C1-C10 carbamoylalkyl;
R2 and R3 are hydrogens; and each of R4, R5, R6, R7, R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen.
[0024] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 8 of Formula I, wherein
Y is a double bond;
A is -CH2-;
B is -CH2CH2-; each of R4, R5, R6, and R7 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen;
R8, R9, and R10 are hydrogens; optionally wherein
R1 is hydrogen, C1-C10 alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R4, R5, R6, and R7 is hydrogen, CFb, hydroxyl, OCH3, F, or Cl.
[0025] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 9 of Formula I, wherein
Y is a double bond;
A is -CH2-;
B is -CH2CH2-;
R4, R5, R6, and R7 are hydrogens; and each of R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen, optionally wherein
R1 is hydrogen, C1-C10 alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R8, R9, and R10 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, in particular optionally wherein R1 is hydrogen or CFb; and R8, R9, and R10 are hydrogens.
[0026] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 10 of Formula I, wherein
Y is a single bond with cA-fused B|C rings;
A is -CH2-;
B is -CH2CH2-;
[0027] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 11 of Formula I, wherein
Y is a single bond with cA-fused B|C rings;
A is -CH2-;
B is -CH2CH2-;
R1 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 hydroxyalkyl, C5-C10 arylalkyl, and C1-C10 carbamoylalkyl;
R2 and R3 are hydrogens; and each of R4, R5, R6, R7, R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen.
[0028] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 12 of Formula I, wherein
Y is a single bond with cA-fused B|C rings;
A is -CH2-;
B is -CH2CH2-; each of R4, R5, R6, and R7 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen; and R8, R9, and R10 are hydrogens, optionally wherein
R1 is hydrogen, Ci-Cio alkyl, CH2Ph, CH2CH2OH, or CH2CONH2 and each of R4, R5, R6, and R7 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, optionally wherein
R4, R5, R6, and R7 are hydrogens; and each of R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen, optionally wherein
R1 is hydrogen, C1-C10 alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R8, R9, and R10 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, in particular optionally wherein R1 is hydrogen or CFb; and R8, R9, and R10 are hydrogens.
[0029] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 13 of Formula I, wherein
Y is a single bond with trans- fused B|C rings;
A is -CH2-;
B is -CH2CH2-.
[0030] Another embodiment is related to the prevention of neuronal damage by administering an effective amount of Compound 14 of Formula I, wherein
Y is a single bond with trans- fused B|C rings;
A is -CH2-;
B is -CH2CH2-.
R1 is selected from the group consisting of hydrogen, C1-C10 alkyl, C1-C10 hydroxyalkyl, C5-C10 arylalkyl, and C1-C10 carbamoylalkyl;
R2 and R3 are hydrogens; and each of R4, R5, R6, R7, R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen.
[0031] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 15 of Formula I, wherein
Y is a single bond with trans- fused B|C rings;
A is -CH2-;
B is -CH2CH2-. each of R4, R5, R6, and R7 is independently selected from the group consisting of hydrogen, Ci-Cio alkyl, hydroxyl, Ci-Cio alkoxyl, and halogen; and R8, R9, and R10 are hydrogens, optionally wherein
R1 is hydrogen, Ci-Cio alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R4, R5, R6, and R7 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl; wherein
R4, R5, R6, and R7 are hydrogens; and each of R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, and halogen, optionally wherein
R1 is hydrogen, C1-C10 alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R8, R9, and R10 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, in particular optionally wherein
R1 is hydrogen, C1-C10 alkyl, CH2Ph, CH2CH2OH, or CH2CONH2; and each of R8, R9, and R10 is hydrogen, CH3, hydroxyl, OCH3, F, or Cl, in particular optionally wherein R1 is hydrogen or CFb; and R8, R9, and R10 are hydrogens.
[0032] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 16 of Formula I, wherein
R1 is hydrogen, methyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, or iso butyl, preferably hydrogen, methyl, n-propyl, or iso-propyl, more preferably hydrogen or methyl, and most preferably methyl; each of R2, R3, R4, R5, R6, R7, R8, R9, and R10 is hydrogen;
Y is a double bond;
A is -CH2-; and
B is -CH2CH2-.
[0033] Another embodiment is related to the prevention or treatment of neuronal damage by administering an effective amount of Compound 4 (DDD-028) of Formula I, wherein R1 is methyl; each of R2, R3, R4, R5, R6, R7, R8, R9, and R10 is hydrogen;
Y is a double bond;
A is -CH2-; and
B is -CH2CH2-. [0034] Another embodiment is related to the prevention of CIN and DPN by administering an effective amount of a combination of compound Formula I and a neuroprotective agent comprising RXDAx accelerants, SARM1 NADase inhibitors, or Hsp modulators.
[0035] Another embodiment is related to the prevention of CIN and DPN by administering an effective amount of a combination of compound Formula I and an analgesic agent comprising opioids, cannabinoids, antiepileptics, or antidepressants.
[0036] Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) to patients undergoing or about to start chemotherapy.
[0037] Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) and a neuroprotective agent comprising RXDAx accelerants, SARM1 NADase inhibitors, or Hsp modulators to patients undergoing or about to start chemotherapy.
[0038] Another embodiment is related to the prevention of CIN and DPN by administering effective amounts of DDD-028 (compound 4) and an analgesic agent comprising opioids, cannabinoids, antiepileptics, or antidepressants.
[0039] We had previously reported that DDD-028 potent, non-opioid analgesic in several models of neuropathic pain 13 We now demonstrate that DDD-028 is not only a potent analgesic in the CIN model, but, more importantly, it prevents both glial and nerve cell damages. In all the three well-established tests for analgesic activity (viz., Paw Pressure for mechanical hyperalgesia, Von Frey for mechanical allodynia, and Cold Plate for Thermal allodynia), DDD-028 was able to recover, dose dependently, paclitaxel -induced hypersensitivity as well as to prevent the development of neuropathic symptoms when it was co-administered with the chemotherapeutic drug (Fig. 1). Moreover, in the sub-chronic administration of DDD-028 (Fig. 2), indicated that the animals did not develop tolerance to the antinociceptive effect exerted in paclitaxel-treated mice, in contrast to other known antinociceptive or analgesic drugs, such as morphine, which induces tolerance after repeated administration both in naive animals and in mice and rats treated with paclitaxel representing one of the most limiting side effects of opioids.
[0040] The neuroprotective activity of one of the compounds of Formula 1, viz., DDD-028, in rat model of CIN has been demonstrated using many well-accepted assays using the brain and spinal cord and plasma: these test comprise glial cell proliferation, glial cell damage, protein degradation, (i.e., oxidative stress), peroxisome activity, nerve conduction, and nerve fiber density. DDD-028 prevented oxidative stress induced by paclitaxel (Figs. 3, 4). DDD- 028 blocks the proliferation of glial cells both in the peripheral regions as well in the brain (Fig. 5), and prevents astrocyte damage (Fig. 5, 6). DDD-028 blocks palitaxel induced nerve degration as shown by three immunohistopathologial studies (Fig. 7-9). Thus, collectively these data firmly demonstrate that DDD-028 is both an analgesic and a neuroprotective agent. It is believed that dual action of DDD-028 is likely mediated via nicotinic acetylcholine pathway, particularly the a7NAChR (Fig. 10).
[0041] Early toxicity studies indicated that DDD-028 is well tolerated, does not induce sedation, and appears to be safe at the expected dose regimen. Finally, the Lipinski-Veber rules for drug-like properties, and the CNS multiparameter optimization (CNS MPO) value of 4.2 places DDD-028 in the high desirability range for CNS drugs. In addition to the fact the compounds of the present invention are structurally different from those disclosed by others, the neuroprotective activity of DDD-028 is also unlike those of the prior art in that DDD-028 elicits neuroprotective effect by inhibiting the damage to both nerve and glial cells.
[0042] The compounds of the present invention represented by Formula I, commonly referred to as ‘active pharmaceutical ingredient (API)’ or ‘drug substance’ is typically formulated as pharmaceutically acceptable salts. The phrase "pharmaceutically acceptable" means those formulations which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts include, but are not limited to acetate, adipate, citrate, tartarate, benzoate, phosphate, glutamate, gluconate, fumarate, maleate, succinate, oxalate, chloride, bromide, hydrochloride, sodium, potassium, calcium, magnesium, or ammonium. [0043] The final formulated product, commonly referred to as ‘drug product,’ may be administered enterally, parenterally, or topically. Enteral route includes oral, rectal, topical, buccal, ophthalmic, and vaginal administration. Parenteral route includes intravenous, intramuscular, intraperitoneal, intrastemal, and subcutaneous injection or infusion. The drug product may be delivered in solid, liquid, or vapor forms, or can be delivered through a catheter for local delivery at a target. Topical delivery may also include transdermal patches or transdermal microneedles for subcutaneous delivery. Transdermal patch and microneedle technology described by Tran et al. \14\ and is incorporated by reference in entirety.
[0044] The dosage levels of API in the drug product can be varied so as to achieve the desired therapeutic response for a particular patient. The phrase "effective amount" or “effective dose” of the compound of the invention means a sufficient amount of the compound is administered to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated, the severity of the disorder; activity of the specific compound employed; the specific composition employed, age, body weight, general health, sex, diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed, and the duration of the treatment. The total daily dose of the compounds of this invention administered may range from about 0.0001 to about 1000 mg/kg/day. For oral administration, preferable doses can be in the range from about 0.001 to about 5 mg/kg/day. If desired, the effective daily dose can be divided into multiple doses for optimal therapeutic effect.
[0045] The compostions of the present invention may be administered to any patient experiencing or expected to encounter neuropathy regardless of its cause. The term “chemotherapy co-treatment” means giving the anti-cancer compound to the patient before, during, or after administration of a dose of chemotherapy, in a manner that prevents CIN. The phrase “before administration” of a dose of chemotherapy may include a several seconds to several hours (such as 24 hours) before the chemotherapy. “During” administration of a dose of chemotherapy may include a specific administration given at any time during the chemotherapy, or co-administration for the duration of the chemotherapy administration (such as by combining the two drugs in the same infusion bag). “After” administration of a dose of chemotherapy may include administration several minutes or hours after the chemotherapy, and may include administration on a schedule for a number of days after the dose of chemotherapy.
[0046] Formulations for oral administration include capsules (soft or hard), tablets, pills, powders, and granules. Such formulations may comprise the API along with at least one inert, pharmaceutically acceptable ingredients selected from the following: (a) buffering agents such as sodium citrate or dicalcium phosphate; (b) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; (c) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants such as glycerol; (e) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; (f) solution retarding agents such as paraffin; (g) absorption accelerators such as quaternary ammonium compounds; (h) wetting agents such as cetyl alcohol and glycerol monostearate; (i) absorbents such as kaolin and bentonite clay and (j) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof; (k) coatings and shells such as enteric coatings, flavoring agents, and the like.
[0047] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the API, the liquid dosage forms may contain inert diluents, solubilizing agents, wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents used in the art.
[0048] Compositions suitable for parenteral injection may comprise physiologically acceptable, sterile aqueous or nonaqueous isotonic solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. The compositions may also optionally contain adjuvants such as preserving; wetting; emulsifying; dispensing, and antimicrobial agents, buffers, diluents, carriers, adjuvants, preservatives, and excipients. Examples of suitable carriers, diluents, solvents, vehicles, or adjuvants include, but are not limited to water; ethanol; polyols such as propyl enegly col, polyethyleneglycol, glycerol, and the like; vegetable oils such as cottonseed, groundnut, com, germ, olive, castor and sesame oils, and the like; organic esters such as ethyl oleate and the like; phenol, parabens, sorbic acid, and the like.
[0049] Injectable formulations may also be suspensions that contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. In some cases, in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
[0050] Injectable depot forms are made by forming microencapsulated matrices of the drug in biodegradable polymers. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, these compositions release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Thus, the rate of drug release and the site of delivery can be controlled. Examples of embedding compositions include, but are no limited to polylactide-polyglycolide poly(orthoesters), and poly(anhydrides), and waxes. The technology pertaining to controlled release formulations are described in “Design of Controlled Release Drug Delivery Systems,” [75] incorporated herein by reference in its entirety.
[0051] Formulations for topical administration include powders, sprays, ointments and inhalants. These formulations include the API along with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[0052] Compounds of the present invention can also be administered in the form of liposomes. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients and the like. The preferred lipids are natural and synthetic phospholipids and phosphatidyl cholines (lecithins) used separately or together. Methods to form liposomes are known in the art and are described in “Liposomes,” [76], which is incorporated herein by reference in its entirety. [0053] The compounds of the present invention can also be administered to a patient in the form of pharmaceutically acceptable ‘prcrirugs.’ Prodrugs are generally used to enhance the bioavailability, solubility, in vivo stability, or any combination thereof of the API. They are typically prepared by linking the API covalently to a biodegradable functional group such as a phosphate that will be cleaved enzymatically or hydrolytically in blood, stomach, or GI tract to release the API. A detailed discussion of the prodrug technology is described in “Prodrugs: Design and Clinical Applications,” [77] incorporated herein by reference.
[0054] The following examples illustrate specific embodiments and utilities of the invention, and are not meant to limit the invention. As would be apparent to skilled artisans, various modifications in the composition, operation, and method are possible, and are contemplated herein without departing from the concept and scope of the invention as defined in the claims. For all the experiments described in the following paragraphs, male Sprague-Dawley rats weighing approximately 200 - 2,50 g at the beginning of the experimental procedure were used. Animals were housed in CeSAL and used at least one week later after their arrival. Four rats were housed per cage (size 26x41 era2), kept at 23.-i.-l °C with a 12 h ligbt/dark cycle, light at 7 a m, and were fed with standard laboratory diet and tap water ad libitum. Experiments involving animals have been reported according to ARRIVE guidelines. Ail efforts were made to minimize animal suffering and to reduce the number of animals used. Paclitaxel was dissolved in a mixture of 10% saline solution and Chremophor EL, a derivative of castor oil and ethylene oxide that is clinically used as paclitaxel vehicle. Control animals received an equivalent volume of the vehicle. The results were expressed as mean ± S.E.M. Statistical analysis was performed using one-way ANOVA followed by post-hoc Bonferroni’s significant difference procedure. The ‘p’ values of less than 0.05, 0.01 or 0.001 were considered significant. All data were collected by an observer who was blinded to the treatments.
Example 1
Acute Analgesic Effect of DDD-028 in CIN - Mechanical Hyperalgesia [0055] The nociceptive threshold in the rat was determined with an analgesimeter (Ugo Basile, Varese, Italy) according to the method described by Leighton et al. [75] Briefly, a constantly increasing pressure was applied to a small area of the dorsal surface of the hind paw using a blunt conical mechanical probe. Mechanical pressure was increased until vocalization or a withdrawal reflex occurred while rats were lightly restrained. Vocalization or withdrawal reflex thresholds were expressed in grams. These limits assured a more precise determination of mechanical withdrawal threshold in experiments aimed to determine the effect of treatments. An arbitrary cut-off value of 100 g was adopted. For these studies, DDD-028 was acutely per os administered when neuropathy was well established (day 10), 48 h after the last chemotherapeutic drug injection. On day 10, paclitaxel -treated rats showed a significantly reduction of the weight tolerated on posterior paws with respect to the control animals (43.2 ± 0.5 g vs 66.5 ± 0.7 g, respectively). Increasing doses of DDD-028 (1 - 25 mg kg 1) reduced mechanical hypersensitivity in a dose-dependent manner starting 30 min after treatment. The highest dose of DDD-028 (25 mg kg 1) completely abrogated paclitaxel- induced mechanical hyperalgesia with maximum analgesic effect occurring at 30 min (Fig. la). The effect persisted for at least 90 min and then vanished 120 min after treatment. Strong, albeit slightly reduced, analgesic effected was also observed at the doses of 10, 5, and 1 mg kg 1, but the duration of the analgesic effect was reduced from about 30 - 90 min at 25 mg kg 1 to about 30 - 60 min at 1 mg kg 1 (Fig. 1).
Example 2
Acute Analgesic Effect of DDD-028 in CIN - Mechanical Allodynia [0056] In the same way, acute administration of DDD-028 counteracted paclitaxel-induced mechanical allodynia in a dose-dependent manner in the Von Frey test (Fig. lb). The highest dose again showed a complete reversal of paclitaxel-induced neuropathy with a long-lasting effect starting from 30 min up to 90 min after treatment. All the lower doses of DDD-028 displayed a shorter anti-hyperalgesic efficacy (Fig. lb). Finally, DDD-028 also counteracted paclitaxel-induced thermal allodynia in a dose-dependent manner in the Cold Plate test (Fig. lc). As shown in Fig. lc, paclitaxel alone significantly enhanced the sensitivity to cold after 10 days of treatment. Thermal allodynia was fully alleviated by DDD-028 (25 mg kg 1) administration. The result obtained with the higher dose was more effective and long-lasting with respect to the lower doses that were capable anyway to reach the statistical significance peaking 45 min after administration.
Example 3
Acute Analgesic Effect of DDD-028 in ON - Thermal Allodynia [0057] Thermal allodynia was assessed using the Cold-plate test. With minimal animal- handler interaction, rats were taken from home-cages, and placed onto the surface of the cold- plate maintained at a constant temperature of 4 ± 1 °C. Ambulation was restricted by a cylindrical Plexiglas chamber (diameter: 10 cm, height: 15 cm), with open top. A timer controlled by foot pedal began timing response latency from the moment the mouse was placed onto the cold-plate. Pain-related behavior (licking of the hind paw) was determined by recording the time (seconds) of the first sign of licking of the hind paw. The cut-off time of the latency of paw lifting or licking was set at 30 s. DDD-028 was able to recover, dose dependently, paclitaxel-induced hypersensitivity and prevent the development of neuropathic symptoms when it was co-administered with the chemotherapeutic drug (Fig. lc).
Example 4
Prophylactic Analgesic Effect of DDD-028 in ON [0058] Thereafter, to assess the protective profile of DDD-028, the compound was subjected to repeated administrations over an 18-day period. Paclitaxel-treated animals were administered daily with DDD-028 (10 mg kg 1, p.o.) starting from the same day of paclitaxel injection. The response to mechanical noxious stimulus was measured on days 10, 12 and 18, 24 h after the last treatment. As shown in Fig. 2a, DDD-028 significantly increased the pain threshold of paclitaxel -injected rats at all time points considered without development of tolerance to the anti-hypersensitivity effect. Repeated administration of DDD-028 induced similar results in reducing paclitaxel-induced mechanical and thermal allodynia and mechanical hyperalgesia (Figs. 2a-c) at all time points as evidenced by the Paw Pressure, Von Frey, and the Cold plate tests. The efficacy of DDD-028 was not different among 30 min and 24 h after treatment suggesting a stable improvement of the pain threshold.
Example 5 Neuroprotective Effect of DDD-028 in ON - Oxidative Stress [0059] Among the multiplicity of the pathophysiologic mechanisms, this oxidative stress plays a crucial role in the generation of paclitaxel-induced neuropathy. On day 18, (i.e., the end of the experiments on sub-chronic analgesic effect of DDD-028), animals were sacrificed by decapitation. L4-L5 dorsal root ganglia (DRG), sciatic nerve, lumbar spinal cord and brain were collected, frozen using liquid nitrogen or fixed by immersion in 4% neutral buffered formalin. Sciatic nerve and DRGs protein extracts were quantified by bicinchoninic acid. Five pg of each sample were denatured by 6% SDS and derivatized by 15-minute incubation with 2-4 dinitrophenyl hydrazine (DNPH; Sigma-Aldrich, Italy) at room temperature. Samples were separated on a 4-12% sodium dodecyl sulfate (SDS)-polyacrylamide gel by electrophoresis and transferred onto nitrocellulose membranes (Biorad, Italy). Membranes were blocked with 1% bovine serum albumin (BSA) in phosphate-buffered saline (PBS) containing .1% Tween 20 (PBST) and then probed overnight with primary antibody specific versus DNPH (Sigma-Aldrich, Italy) 1:5000 in PBST/1% BSA. After washing with PBST, the membranes were incubated for 1 hour in PBST containing the appropriate horseradish peroxidase-conjugated secondary antibody (1:5000; Cell Signalling, USA) and again washed. ECL (Pierce, USA) was used to visualize the peroxidase-coated bands. Densitometric analysis was performed using the “Image J” analysis software and the density of all bands displayed in the lane is reported as a mean. Ponceau-stained membranes were used as loading control [4] Formalin fixed cryostat sections (10 pm for brain and 5 pm for spinal cord) were incubated for 1 h in blocking solution (Bio-Optica; Italy) at room temperature, and thereafter sections were incubated for 24 h at 4 °C in PBST containing primary antisera and 5% normal donkey serum. The primary antibody was directed against Ibal (rabbit antiserum, 1:500; Wako Chemicals, USA; [23]) for microglial staining and against glial fibrillary acidic protein (GFAP; rabbit antiserum, 1:500; Dako, USA;[28]) for astrocyte staining. After rinsing in PBST, sections were incubated in donkey anti-rabbit IgG secondary antibody labelled with Alexa Fluor 488 or 568 (1:1000, Invitrogen, USA) at room temperature for 1 h. For all immunohistochemical studies, negative control sections (no exposure to the primary antisera) were processed concurrently with the other sections. A single optical density value for the dorsal horns in each rat was obtained by averaging the two sides, and this value was compared to the homologous average values from the vehicle-treated animals. Images were acquired using a motorized Leica DM6000B microscope equipped with a DFC350FX camera. Morphological examination of microglia and astrocytes was assessed by inspection of at least three fields (40 c 0.75NA objective) in the dorsal horn of the spinal cord and brain areas per section. The full specimen thicknesses were acquired as z-stack series, deconvolved using Huygens Professional software (SVI, The Netherlands) and displayed using ImageJ software. To evaluate the capability of DDD-028 to intervene against direct damages and against the maladaptive plasticity of the nervous system induced by paclitaxel, nervous tissues (brain, spinal cord, DRGs and sciatic nerve) were collected and analyzed at the end of the repeated treatment with the compound (day 18). Oxidative stress, a typical signature of chemotherapy-induced neurotoxicity, was measured in the peripheral nervous system. As shown in Figure 5, paclitaxel induced an oxidative damage of dorsal root ganglia (DRG) as indicated by three-fold increase in carbonylation of proteins. Treatment with DDD-028 resulted in a significant prevention of the damage as evidenced by protein carbonylation values similar to the control group (Fig. 3).
Example 6
Neuroprotective Effect of DDD-028 in ON - Peroxisome Functionality [0060] Enzymatic activity in both DRGs and sciatic nerve was measured in PBS using the homogenated tissues: the suspension was sonicated in ice using three 10 s bursts at high intensity with a 10 s cooling period between each burst and then centrifuged (13.000 x g for 15 min at 4°C). Catalase activity was measured in the supernatant by Amplex Red Catalase Assay Kit (Invitrogen, Monza, Italy) following the manufacturer’s instructions. Protein concentration was quantified by bicinchoninic acid assay (Sigma-Aldrich, Milan, Italy). Catalase activity for each sample was normalized to protein concentration. Control conditions in the absence of treatment were set as 100%. DDD-028 was able to prevent the development of hypersensitivity, and to reduce paclitaxel-related damage to PNS and CNS when repeatedly administered with the anticancer drug. DDD-028 demonstrated detoxifying properties as indicated by the enhancing the activity of the peroxisomal enzyme, catalase, and reducing the protein oxidation in DRGs (Fig. 4).
Example 7
Neuroprotective Effect of DDD-028 in ON - Astrocyte Proliferation, Spinal Cord [0061] To determine whether neurochemical reorganization in the spinal cord occurs following DDD-028 repeated treatment, lumbar spinal cord sections we examined by immunohistochemistry using antibodies against GFAP and Ibal to label astrocytes and microglia, respectively, which are non-neuronal cells strongly involved in chemotherapy- induced neuropathic pain. Astrocyte activation was measured as an increase in the number of GFAP-expressing cells in the dorsal horn of the spinal cord of treated rats. GFAP-positive cell number in superficial laminae of paclitaxel -treated rats was significantly greater than vehicle-treated number at day 18 (Fig. 5a). Animals treated with paclitaxel + DDD-028 showed a significantly lower number of astrocytes characterized by a reactive phenotype. Moreover, spinal astrocytes presented altered morphology showing hypertrophy of the cell body and processes (Fig. 5c). Microglia activation was measured by the quantification of Ibal positive cells in the spinal cord of treated rats. On day 18, paclitaxel treatment produced increased density of Ibal positive cells in the dorsal horns of the lumbar spinal cord (Fig. 5b). Animals treated with paclitaxel + DDD-028 showed a significant prevention of microglia activation (Figure 5b). However, no hypertrophy of this type of glia cells was observed, and microglia possessed a highly ramified morphology similar to those in saline-treated rats. On the other hand, day 18 can be considered a late phase for microglia activation that is, according to the literature, strongly involved in the first days of treatment.
Example 8
Neuroprotective Effect of DDD-028 in CIN - Astrocyte Proliferation, Brain [0062] To probe the effect of DDD-028 in the brain regions involved in pain sensation, a topographic analysis of microglia and astrocyte cells in three regions, the periacqueductal gray (PAG; involved in endogenous pain modulatory system), the thalamus and the somatosensory cortex (SI) were examined. Paclitaxel induced a significant numerical increase of both glial cell populations in brain areas, but a higher astrocyte activation was observed (Figs. 6a, b). DDD-028 reduced both microglia and astrocyte cell number increase (numerical activation) in PAG, thalamus and SI.
Example 9
Neuroprotective Effect of DDD-028 in CIN - Intraepi dermal Nerve Fiber Density [0063] Mice hind limb paw skin was placed overnight at 4 °C in 4% paraformaldehyde in PBS IX, transferred to 30% sucrose overnight, frozen and cryosectioned at 50 pm transversal to long paw axis. Free floating sections were incubated in PBS containing 0.3% Triton X-100 (TBS) 1 hour at room temperature, then in the primary antibody panaxonal marker PGP9.5 (abl08986, rabbit monoclonal [EPR4118] 1:600, Abeam) overnight at room temperature. Afterward, sections were rinsed in PBS IX and placed in a goat anti-rabbit IgG secondary antibody labelled with Alexa Fluor 488 (1:500), 2 hours at room temperature in the dark. To stain nuclei, sections were incubated with DAPI in PBS for 10 min. After three washes in PBS and a final wash in distilled water, slices were mounted using ProLong Gold (Life Technologies-ThermoFisher Scientific, Milan, Italy) as mounting medium. Digitalized images were collected at 200 c total magnification by a motorized Leica DM6000B microscope equipped with a DFC350FX. Quantitative analysis of intraepidermal nerve fibers (IENF) density (fibers/mm) was performed by collecting 6 independent fields in the skin of each animal and counting the number of single PGP9.5 -positive fibers crossing the epidermis-dermis boundary (basal membrane) by using the software ImageJ (NIH, Bethesda, MD, USA). Secondary branching is excluded from quantification. The administration of paclitaxel in mice determined a significant reduction of the intraepidermal nerve fibers (IENF) density, calculated as the number of PGP9.5 positive fibers per mm of skin (Fig. 7b). DDD-028 was able to effectively counteract the loss of IENF caused by paclitaxel (Fig. 7d). Indeed, the number of nerve fibers crossing the epidermis resulted significantly increased in paclitaxel -treated mice as a result of DDD-028 (10 mg kg 1) repeated administration. In contrast, the loss of IENF observed after paclitaxel administration in mice was unaffected by pregabalin (30 mg kg-1) treatment (Fig. 7c).
Example 10
Neuroprotective Effect of DDD-028 in CIN - NF-H Density [0064] Paclitaxel also induced a significantly reduction of the neurofilament heavy chain (NF-H) expression in the sciatic nerve (Fig. 8b) measured as a percent of fluorescence intensity in comparison to the control group (vehicle + vehicle-treated animals). Repeated daily administrations of DDD-028 were able to restore the loss of NF-H expression in both tissues analyzed (Fig. 8c). Pregabalin did not prevent the loss of NF-H expression in the sciatic nerve (Fig. 8d). Moreover, paclitaxel treatment determined a derangement of the nerve fibers disposition in the sciatic nerve that was restored by DDD-028 treatment.
Example 11
Neuroprotective Effect of DDD-028 in CIN - Plasma NF-L Levels [0065] Neurofilament light chain (NF-L), a well-accepted marker of neurotoxicity, was increased in plasma of paclitaxel -treated mice. In DDD-028 treated mice, the plasma NF-L concentration is substantially lower than in paclitaxel -treated mice (Fig. 9), whereas in pregabalin treated animals NF-L level was not significantly different from paclitaxel -treated animals.
Example 12
Neuroprotective Effect of DDD-028 in CIN - Myelin Degradation [0066] Effect of DDD-028 on morphology and morphometry of DRGs sciatic nerves of paclitaxel -treated mice. Paclitaxel 2.0 mg kg 1 was dissolved in a mixture of 10% Cremophor EL and saline solution and intraperitoneally (i.p.) administered on four alternate days (1, 3, 5 and 8). DDD-028 (10 mg kg 1) was suspended in 1% carboxymethylcellulose sodium salt (CMC) and daily administered per os starting the first day of paclitaxel treatment and continuing 1 week after the end of treatment (from day 1 to day 17). Animals were sacrificed on day 18, 24h after the last treatment. Sciatic nerves and DRGs were fixed in 2.5% glutaraldehyde in cacodylate buffer, pH 7.4, for 24 hours. Tissues were embedded in Epoxy resin; sections (0.8 pm) were cut using an ultramicrotome, stained with toluidine blue. DRGs were studied measuring the number of neurons showing multinucleolated nuclei as sign of toxic alteration; the soma area was also assessed stratifying neurons in small (<600 microm diameter), medium (>600<1200 microm diameter) and large (>1200 microm diameter). In the sciatic nerve, myelin area, number of fibers (separated by axon diameter) and axon diameters were analyzed. The results indicate that DDD-028 prevents axon degradtion induced by paclictaxel (Fig. 10).
Example 13
Possible Pharmacodynamic Mechanism of Compounds of Formula 1 [0067] To study the pharmacodynamics of DDD-028, three possible targets were evaluated: the nicotinic receptor (nAChR) of the cholinergic system, the voltage-gated potassium channel subtype Kv7, and sigma 1 (si) and 2 (s?) receptors. The relevance of the Kv7 was studied by using the selective Kv7 antagonist XE991 (1 mg/kg, i.p.). XE991 administered 15 min before DDD-028 was not able to alter the efficacy of DDD-028 over the time of observation (Fig. 1 la). B on previous studies that si receptor is involved in the pain pathway, DDD-028 was subjected to the modulation of the s receptor-mediated neck dystonia. Accordingly, the s agonist DTG was injected in the red nucleus. DTG induced postural changes characterized by a marked deviation in the head angle (neck dystonia), peaking at 25-30 min after microinjection. DDD-028 (25 mg kg 1) was administered per os 15 min before DTG infusion without preventing DTG-induced neck dystonia (Fig. 1 lb). Finally, in the Cold plate test, DDD-028 (25 mg kg 1, per os) increased the licking latency of paclitaxel treated animals starting 15 min after administration and lasting up to 105 min (Fig. 11c). Pre treatment of the animals with the nAChR antagonist MECA (2 mg kg 1, i.p.) 15 min before DDD-028 administration completely abolished the pain-relieving effect of the compound up to 60 min (Fig. 11c). However, when MECA was administered the second time at 45 min after DDD-028 treatment, the effect of DDD-028 was entirely blocked for all the times observed. The anti-hyperalgesic effect induced by DDD-028 was also substantially reduced by the pre-treatment with the a7 nAchR antagonist MLA (6 mg kg 1, i.p.) (Fig. 11c), which suggests that this receptor subtype is very likely involved in the mechanism of action of DDD-028. REFERENCES
E Fox, R.B.; Powell, W.H. Nomenclature of Organic Compounds: Principles and Practice, Second Edition. Oxford University Press, Oxford, 2001.
2. Hellwinkel, D. Systematic Nomenclature of Organic Chemistry: A Directory of Comprehension and Application of its Basic Principles. Springer- Verlag, Berlin, 2001.
3. Tzatha, E.; DeAngelis, L.M. Oncology 2016, 30, 240-244.
4. Poupon, L., et al. Expert Opinion on Drug Safety 2015, 14, 1269-1282.
5. Von Hehn, C.A., et al. Neuron 2012, 73, 638-652.
6. Mika, J. Phamacological Reports 2008, 60, 297-307.
7. Torrance, N., et al. JPain 2006, 7: 281-289.
8. Brzezinski, K. et al. Wspolczesna Onkologia 2011, 15, 246-250.
9. Scott, D.; Confalone, P. U.S. Patent 10,844,054; 2020.
10. Hoke, A. et al. U.S. Patent 9,527,817; 2019.
11. Melbrandt et al. U.S. Patent Application Publication 2020/0129493; 2020.
12. Rajagopalan, P. US Patent 9,822,117; 2014.
13. Bandyopadhyaya, A., et al. Bioorganic and Medicinal Chemistry Letters 2014, 24, 3088-3091.
14. Tran, T.M.K. et al. Nature Biomedical Engineering 2020, https://doi.org/10.1038/ s41551-020-00650-4.
15. Li, X. Design of Controlled Release Drug Delivery Systems. McGraw-Hill, New York, 2006.
16. Weissig, V. Liposomes: Methods and Protocols Volume 1: Pharmaceutical Nanocarriers. Humana Press, New York, 2009.
17. Rautio, J. et al. Nature Reviews Drug Discovery 2008, 7, 255-270.
18. Leighton G.E., et al. Br. J. Pharmacol. 1988, 93(3), 553-60.

Claims

We claim:
1. A method of preventing or treating neuronal damage in a patient being administered a therapy that can cause neuronal damage, comprising administering to said patient an effective amount of a neuroprotective composition of Formula 1, wherein
Figure imgf000024_0001
Y is a single bond or a double bond;
A and B are independently -(CFh)n-; subscript ‘n’ varies from 0 to 3;
R1 is selected from the group consisting of hydrogen, Ci-Cio alkyl, Ci-Cio hydroxyalkyl, C5-C10 aryl, C1-C15 aroylalkyl, C1-C10 alkoxycarbonylalkyl, C5-C10 arylalkyl, and C1-C10 carbamoylalkyl;
R2 is selected from the group consisting of hydrogen, C1-C10 alkyl, C5-C10 aryl, and C5- C10 arylalkyl; each of R3, R4, R5, R6, R7, R8, R9, and R10 is independently selected from the group consisting of hydrogen, C1-C10 alkyl, hydroxyl, C1-C10 alkoxyl, -NRUR12, C1-C10 hydroxyalkyl, halogen, trihaloalkyl, cyano, carboxyl, C1-C10 acyl, C1-C10 alkoxyalkyl; C1-C10 alkoxycarbonyl, C5-C10 aryl, and C5-C10 arylalkyl; and
R11 and R12 are independently hydrogen or C1-C10 alkyl.
2. The method of claim 1, wherein Y is a double bond; A is -CH2-; and B is -CH2CH2-.
3. The method of claim 2, wherein said neuroprotective compound is selected from the group consisting of compounds 4, 6, 7, 8, and 9.
4. The method of claim 3, wherein said neuroprotective compound is 4, DDD-028.
5. The method of claim 1, wherein Y is a single bond; A is -CH2-; and B is -CH2CH2-.
6. The method of claim 5, wherein said neuroprotective compound is selected from the group consisting of compounds 10, 11, 12, 13, 14, and 15
7. The method of claim 1, wherein the neuronal damage is caused by cancer.
8. The method of claim 1, wherein the neuronal damage is caused by the administration of a chemotherapeutic agent to a patient undergoing cancer chemotherapy.
9. The method of claim 8, wherein the chemotherapeutic agent is paclitaxel, albumin- bound paclitaxel, doxorubicin, cisplatin, or carboplatin.
10. The method of claim 1, wherein the neuronal damage is caused by diabetes.
11. The method of claim 1, wherein the neuronal damage is caused by microbial infection.
12. The method of claim 1, wherein said neuroprotective composition comprises compounds Formula 1 and a second neuroprotective compound is selected from the group consisting of RXDAx accelerants, SARM1 NADase inhibitors, and Hsp modulators.
13. The method of claim 12, wherein said second neuroprotective compound is selected from the group consisting of compounds 1, 2, and 3.
14. The method of claim 13, wherein said neuroprotective composition consists of compound 4 and compound 1.
15. The method of claim 13, wherein said neuroprotective composition consists of compound 4 and compound 2.
16. The method of claim 13, wherein said neuroprotective composition consists of compound 4 and compound 3.
17. The method of claim 1, wherein said neuroprotective composition comprises compounds Formula 1 and an analgesic compound selected from the group consisting of opioids, cannabinoids, antiepileptics, or antidepressants.
18. The method of claim 17, wherein said analgesic compound is selected from the group consisting of morphine, oxycodone, pregabalin, gabapentin, and duloxetine.
19. The method of claim 18, wherein said neuroprotective composition comprises compound 4 and oxycodone.
20. The method of claim 18, wherein said neuroprotective composition comprises compound 4 and pregabalin.
PCT/US2022/033189 2021-06-13 2022-06-13 Prevention and treatment of neuronal damage with pyridoindolobenz[b, d] azepine compositions WO2022265960A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163210063P 2021-06-13 2021-06-13
US63/210,063 2021-06-13

Publications (1)

Publication Number Publication Date
WO2022265960A1 true WO2022265960A1 (en) 2022-12-22

Family

ID=84526624

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/033189 WO2022265960A1 (en) 2021-06-13 2022-06-13 Prevention and treatment of neuronal damage with pyridoindolobenz[b, d] azepine compositions

Country Status (1)

Country Link
WO (1) WO2022265960A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014137848A1 (en) * 2013-03-04 2014-09-12 Daya Drug Discoveries, Inc PENTACYCLIC PYRIDOINDOLOBENZ[b,d]AZEPINE DERIVATIVES AND USES THEREOF
US20160039820A1 (en) * 2013-03-04 2016-02-11 Daya Drug Discoveries, Inc. Pyridoindolobenz[b,e]azepine derivatives and uses thereof
US9527817B2 (en) * 2011-06-21 2016-12-27 The Johns Hopkins University Compounds for treating peripheral; neuropathies and other neurodegenerative; disorders
US20190367499A1 (en) * 2018-05-29 2019-12-05 Cersci Therapeutics, Inc. Compounds for Pain Treatment, Compositions Comprising Same, and Methods of Using Same
US20200121651A1 (en) * 2018-10-18 2020-04-23 Stc.Unm Pramipexole for Use in the Treatment of Pain
US20200129493A1 (en) * 2016-09-24 2020-04-30 Washington University Inhibitors of sarm1 nadase activity and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9527817B2 (en) * 2011-06-21 2016-12-27 The Johns Hopkins University Compounds for treating peripheral; neuropathies and other neurodegenerative; disorders
WO2014137848A1 (en) * 2013-03-04 2014-09-12 Daya Drug Discoveries, Inc PENTACYCLIC PYRIDOINDOLOBENZ[b,d]AZEPINE DERIVATIVES AND USES THEREOF
US20160039820A1 (en) * 2013-03-04 2016-02-11 Daya Drug Discoveries, Inc. Pyridoindolobenz[b,e]azepine derivatives and uses thereof
US20200129493A1 (en) * 2016-09-24 2020-04-30 Washington University Inhibitors of sarm1 nadase activity and uses thereof
US20190367499A1 (en) * 2018-05-29 2019-12-05 Cersci Therapeutics, Inc. Compounds for Pain Treatment, Compositions Comprising Same, and Methods of Using Same
US20200121651A1 (en) * 2018-10-18 2020-04-23 Stc.Unm Pramipexole for Use in the Treatment of Pain

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RAJAGOPALAN ET AL.: "DDD-028: A potent potential non-opioid, non-cannabinoid analgesic for neuropathic and inflammatory pain", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 24, 14 May 2014 (2014-05-14), pages 3088 - 3091, XP029033872, DOI: 10.1016/j.bmcl.2014.05.016 *

Similar Documents

Publication Publication Date Title
US20230285329A1 (en) Methods and Compositions for the Treatment of Steatosis-Associated Disorders
Zhou et al. PPARγ activation mitigates mechanical allodynia in paclitaxel-induced neuropathic pain via induction of Nrf2/HO-1 signaling pathway
Zhu et al. Neuroprotective effects of Astilbin on MPTP-induced Parkinson's disease mice: Glial reaction, α-synuclein expression and oxidative stress
US20020128299A1 (en) Methods of treatment using MAO-A and MAO-B inhibitors such as L-deprenyl
US11607389B2 (en) Delayed release deferiprone tablets and methods of using the same
JPH11504044A (en) Treatment method and pharmaceutical preparation
EP2882495A1 (en) Laquinimod for treatment of cannabinoid receptor type 1 (cb1) mediated disorders
Dunne et al. A phase 1 study to assess the pharmacokinetics of sulopenem etzadroxil (PF-03709270)
WO2014028397A2 (en) Laquinimod for treatment of gaba mediated disorders
WO2005079792A1 (en) Preventive or therapeutic agents for severe diabetic retinopathy
WO2022265960A1 (en) Prevention and treatment of neuronal damage with pyridoindolobenz[b, d] azepine compositions
TW201618760A (en) Methods of treating huntington&#39;s disease using cysteamine compositions
TW201609640A (en) Use of indolyl and indolinyl hydroxamates for treating neurodegenerative disorders or cognitive deficits
TW201900163A (en) Pharmaceutical combination of ZONISAMIDE and PRAMIPEXOLE for treating synuclearopathy and related methods
KR20150058159A (en) Baclofen and acamprosate based therapy of macular degeneration disorders
CA3123524C (en) Synergistic nutritional compositions for pain management
RU2805061C2 (en) Treatment of demyelinating diseases
US20240058308A1 (en) Treatment and prevention of dry macular degeneration
US20060128738A1 (en) Treatment of interstitial cystitis using cannabinoid analogs
WO2018057933A1 (en) Compounds, compositions, and methods for reducing oxidative stress in cardiomyocytes
EP4313067A1 (en) Treatment of immune-related disorders, kidney disorders, liver disorders, hemolytic disorders, and oxidative stress-associated disorders using nrh, narh and reduced derivatives thereof
NZ787785A (en) Delayed release deferiprone tablets and methods of using the same
EP3265086A1 (en) A lysosomal inhibitor and a multikinase inhibitor in the treatment of liver cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22825586

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE