WO2022263817A1 - Compounds and their use for the treatment of alpha1-antitrypsin deficiency - Google Patents

Compounds and their use for the treatment of alpha1-antitrypsin deficiency Download PDF

Info

Publication number
WO2022263817A1
WO2022263817A1 PCT/GB2022/051503 GB2022051503W WO2022263817A1 WO 2022263817 A1 WO2022263817 A1 WO 2022263817A1 GB 2022051503 W GB2022051503 W GB 2022051503W WO 2022263817 A1 WO2022263817 A1 WO 2022263817A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mixture
treatment
disorder
disease
Prior art date
Application number
PCT/GB2022/051503
Other languages
French (fr)
Inventor
Nigel Ramsden
David John Fox
Original Assignee
Z Factor Limited
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 Z Factor Limited filed Critical Z Factor Limited
Publication of WO2022263817A1 publication Critical patent/WO2022263817A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/92Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with a hetero atom directly attached to the ring nitrogen atom
    • C07D211/96Sulfur atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • oci-Antitrypsin is a member of the serpin superfamily produced by the liver and secreted into the blood. It inhibits a variety of serine proteases, especially neutrophil elastase. When blood levels of A1AT are low, excessive neutrophil elastase activity degrades lung tissue resulting in respiratory complications, such as chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • A1AT in blood is 0.9-2.3 g/L. Levels lower than this are typical of oci-antitrypsin deficiency (A1AD or AATD), a genetic disorder caused by mutations in the SERPINA1 gene, coding for A1AT.
  • the Z mutation the most common cause of AATD, is the substitution of glutamate to lysine at position 366 of A1AT (UniProtKB - P01009 (A1AT_HUMAN)), corresponding to position 342 in the mature protein (Z A1AT).
  • the Z mutation affects the folding of A1AT resulting in only a small fraction acquiring the native/active state. The remainder is either cleared as misfolded protein or accumulates in the liver as stable polymers.
  • homozygous carriers of the Z mutation have plasma levels of A1AT that are 10-15% of normal, predisposing carriers to COPD.
  • Accumulation of Z A1AT polymers in liver cells predisposes carriers to cirrhosis, liver cancer and other liver pathologies.
  • the current treatment for the lung manifestation of AATD involves augmentation therapy using A1AT concentrates prepared from the plasma of blood donors.
  • the US FDA has approved the use of four A1AT products: Prolastin, Zemaira, Glassia, and Aralast. Dosing is via once weekly intravenous infusion. Augmentation therapy has been demonstrated to slow progression of COPD.
  • the liver manifestations of AATD e.g. cirrhosis and cancer
  • Investigational approaches to improved treatment of the liver manifestations include inhibition of Z A1AT polymerisation and increased clearance of polymers through the activation of autophagy. Investigational approaches to improved treatment of both the lung and the liver manifestations are directed towards improvement of Z A1AT folding and secretion.
  • Ri , R2, R3 and R4 are each independently selected from chloride, fluoride, trifluoromethyl, -O-methyl and hydrogen,
  • Also provided according to the invention is a mixture of the two enantiomers of any compound according to formula (I), wherein the mixture is either racemic or has one enantiomer in excess of the other enantiomer.
  • the compound or mixture of the invention may be in a pharmaceutically acceptable salt form or crystalline form.
  • pharmaceutically acceptable salt refers to a pharmaceutically acceptable mono organic or inorganic salt of the compound of the invention. This may include those derived from bases such as sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, l-deoxy-2-(methylamino)-D-glucitol, magnesium hydroxide, zinc hydroxide, aluminium hydroxide, ferrous or ferric hydroxide, ammonium hydroxide or organic amines such as N-methylglucamine, choline, arginine and the like.
  • bases such as sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, l-deoxy-2-(methylamino)-D-glucitol, magnesium hydroxide, zinc hydroxide, aluminium hydroxide, ferrous or ferric hydroxide, ammonium hydroxide or organic amines such as N-methylglucamine, choline, arginine and the like.
  • bases such as sodium hydroxide, potassium hydroxide, lithium hydro
  • a pharmaceutical composition comprising the compound or mixture of the invention as described herein and a pharmaceutically or therapeutically acceptable excipient or carrier.
  • pharmaceutically or therapeutically acceptable excipient or carrier refers to a solid or liquid filler, diluent or encapsulating substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the host, which may be either humans or animals, to which it is administered.
  • a variety of pharmaceutically acceptable carriers such as those well known in the art may be used.
  • Non-limiting examples include sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water.
  • administration of the medicament may be via oral, subcutaneous, direct intravenous, slow intravenous infusion, continuous intravenous infusion, intravenous or epidural patient controlled analgesia (PCA and PCEA), intramuscular, intrathecal, epidural, intracistemal, intraperitoneal, transdermal, topical, transmucosal, buccal, sublingual, transmucosal, inhalation, intranasal, intra-atricular, intranasal, rectal or ocular routes.
  • the medicament may be formulated in discrete dosage units and can be prepared by any of the methods well known in the art of pharmacy.
  • Administration of the medicament may for example be in the form of oral solutions and suspensions, tablets, capsules, lozenges, effervescent tablets, transmucosal films, suppositories, buccal products, oral mucoretentive products, topical creams, ointments, gels, films and patches, transdermal patches, abuse deterrent and abuse resistant formulations, sterile solutions suspensions and depots for parenteral use, and the like, administered as immediate release, sustained release, delayed release, controlled release, extended release and the like.
  • Another aspect of the invention is the use of the compound or mixture of the invention as defined herein in the manufacture of a medicament for the treatment of a disease or disorder.
  • a further aspect of the invention is the compound or mixture of the invention for use as an inducer of Z A1AT secretion.
  • the invention also encompasses a method of treating a disease or disorder, comprising the step of administering the compound or mixture or the pharmaceutical composition of the invention as defined herein to a patient in need thereof.
  • the invention further encompasses the use of a compound or mixture of the invention as an inducer of Z A1AT secretion.
  • the use may be in the treatment of a disease or disorder. Additionally or alternatively, the use may be in vitro , for example in an in vitro assay.
  • a disease or disorder suitable for treatment according to the relevant aspects of the invention is one which is characterised by low plasma levels of A1AT, for example AATD.
  • the invention also provides the use of a racemic compound of formula (I) in the manufacture of a medicament for the treatment of a disease or disorder, wherein the disease or disorder is AATD.
  • racemic compound of formula (I) as an inducer of Z A1AT secretion.
  • the use of a numerical range in this description is intended unambiguously to include within the scope of the invention all individual integers within the range and all the combinations of upper and lower limit numbers within the broadest scope of the given range.
  • compositions comprising as active ingredient a compound
  • this terminology is intended to cover both compositions in which other active ingredients may be present and also compositions which consist only of one active ingredient as defined.
  • (S)- l-((3-chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid was prepared using the following synthesis procedure.
  • (S)-Piperidine-3-carboxylic acid (7.7 mmol), potassium hydroxide (7.7 mmol) and potassium carbonate (15.4 mmol) were added to water (20ml) and stirred.
  • 3-Chloro-2- (trifluoromethyl)benzenesulfonyl chloride (7.7 mmol) was added and the reaction was stirred at room temperature for 3 hours.
  • the reaction was cooled to 0°C and acidified with 2M hydrochloric acid to give a white precipitate. This precipitate was dried and triturated with n-pentane to give (S)-l-((3-chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3- carboxylic acid.

Abstract

The invention relates to compounds of formula (I) wherein R1-R4, m and n are as described in the application, and their medical uses, for example for use in the treatment of α1-antitrypsin deficiency (A1AD or AATD).

Description

Compounds and their Use for the Treatment of cci-Antitrypsin Deficiency
The invention relates to certain carboxylic acids and their medical use. oci-Antitrypsin (A1AT) is a member of the serpin superfamily produced by the liver and secreted into the blood. It inhibits a variety of serine proteases, especially neutrophil elastase. When blood levels of A1AT are low, excessive neutrophil elastase activity degrades lung tissue resulting in respiratory complications, such as chronic obstructive pulmonary disease (COPD).
The reference range of A1AT in blood is 0.9-2.3 g/L. Levels lower than this are typical of oci-antitrypsin deficiency (A1AD or AATD), a genetic disorder caused by mutations in the SERPINA1 gene, coding for A1AT. The Z mutation, the most common cause of AATD, is the substitution of glutamate to lysine at position 366 of A1AT (UniProtKB - P01009 (A1AT_HUMAN)), corresponding to position 342 in the mature protein (Z A1AT). The Z mutation affects the folding of A1AT resulting in only a small fraction acquiring the native/active state. The remainder is either cleared as misfolded protein or accumulates in the liver as stable polymers. As a consequence of the misfolding, homozygous carriers of the Z mutation (ZZ) have plasma levels of A1AT that are 10-15% of normal, predisposing carriers to COPD. Accumulation of Z A1AT polymers in liver cells predisposes carriers to cirrhosis, liver cancer and other liver pathologies.
The current treatment for the lung manifestation of AATD involves augmentation therapy using A1AT concentrates prepared from the plasma of blood donors. The US FDA has approved the use of four A1AT products: Prolastin, Zemaira, Glassia, and Aralast. Dosing is via once weekly intravenous infusion. Augmentation therapy has been demonstrated to slow progression of COPD. The liver manifestations of AATD (e.g. cirrhosis and cancer) are treated with steroids and liver transplantation. Investigational approaches to improved treatment of the liver manifestations include inhibition of Z A1AT polymerisation and increased clearance of polymers through the activation of autophagy. Investigational approaches to improved treatment of both the lung and the liver manifestations are directed towards improvement of Z A1AT folding and secretion.
A prior art search based on the structure of l-((2- (trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid was conducted. The closest prior art molecule identified by the search was the racemic compound, l-((2- (trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid (CAS registry number 891392-68-0). This compound is listed as commercially available from Aurora, ChemDiv and the FCH Group but no publications are recorded. No pharmaceutical compositions or uses are known.
According to one aspect of the present invention, there is provided a compound of formula
(I):
Figure imgf000003_0001
wherein
• Ri , R2, R3 and R4 are each independently selected from chloride, fluoride, trifluoromethyl, -O-methyl and hydrogen,
• m and n are independently 1, 2 or 3, but the combination where both n and m are 1 is excluded,
• when the combination of m and n results in a chiral centre, optionally both enantiomers and the racemic mixture are included, and the compound is
Figure imgf000004_0001
Also provided according to the invention is a mixture of the two enantiomers of any compound according to formula (I), wherein the mixture is either racemic or has one enantiomer in excess of the other enantiomer.
The compound or mixture of the invention may be in a pharmaceutically acceptable salt form or crystalline form.
The term “pharmaceutically acceptable salt” refers to a pharmaceutically acceptable mono organic or inorganic salt of the compound of the invention. This may include those derived from bases such as sodium hydroxide, potassium hydroxide, lithium hydroxide, calcium hydroxide, l-deoxy-2-(methylamino)-D-glucitol, magnesium hydroxide, zinc hydroxide, aluminium hydroxide, ferrous or ferric hydroxide, ammonium hydroxide or organic amines such as N-methylglucamine, choline, arginine and the like. For other examples of pharmaceutically acceptable salts, reference can be made to Gould (1986, Int J Pharm 33: 201-217).
According to a further aspect of the invention, there is a provided a pharmaceutical composition comprising the compound or mixture of the invention as described herein and a pharmaceutically or therapeutically acceptable excipient or carrier.
The term “pharmaceutically or therapeutically acceptable excipient or carrier” refers to a solid or liquid filler, diluent or encapsulating substance which does not interfere with the effectiveness or the biological activity of the active ingredients and which is not toxic to the host, which may be either humans or animals, to which it is administered. Depending upon the particular route of administration, a variety of pharmaceutically acceptable carriers such as those well known in the art may be used. Non-limiting examples include sugars, starches, cellulose and its derivatives, malt, gelatin, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-free water.
All suitable modes of administration are contemplated according to the invention. For example, administration of the medicament may be via oral, subcutaneous, direct intravenous, slow intravenous infusion, continuous intravenous infusion, intravenous or epidural patient controlled analgesia (PCA and PCEA), intramuscular, intrathecal, epidural, intracistemal, intraperitoneal, transdermal, topical, transmucosal, buccal, sublingual, transmucosal, inhalation, intranasal, intra-atricular, intranasal, rectal or ocular routes. The medicament may be formulated in discrete dosage units and can be prepared by any of the methods well known in the art of pharmacy.
All suitable pharmaceutical dosage forms are contemplated. Administration of the medicament may for example be in the form of oral solutions and suspensions, tablets, capsules, lozenges, effervescent tablets, transmucosal films, suppositories, buccal products, oral mucoretentive products, topical creams, ointments, gels, films and patches, transdermal patches, abuse deterrent and abuse resistant formulations, sterile solutions suspensions and depots for parenteral use, and the like, administered as immediate release, sustained release, delayed release, controlled release, extended release and the like.
Another aspect of the invention is the use of the compound or mixture of the invention as defined herein in the manufacture of a medicament for the treatment of a disease or disorder.
A further aspect of the invention is the compound or mixture of the invention for use as an inducer of Z A1AT secretion.
Further provided is the compound or mixture of the invention as defined herein for use in the treatment of a disease or disorder.
The invention also encompasses a method of treating a disease or disorder, comprising the step of administering the compound or mixture or the pharmaceutical composition of the invention as defined herein to a patient in need thereof.
The invention further encompasses the use of a compound or mixture of the invention as an inducer of Z A1AT secretion. The use may be in the treatment of a disease or disorder. Additionally or alternatively, the use may be in vitro , for example in an in vitro assay.
A disease or disorder suitable for treatment according to the relevant aspects of the invention is one which is characterised by low plasma levels of A1AT, for example AATD.
The invention also provides the use of a racemic compound of formula (I) in the manufacture of a medicament for the treatment of a disease or disorder, wherein the disease or disorder is AATD.
Also provided is the use of a racemic compound of formula (I) as an inducer of Z A1AT secretion. The use of a numerical range in this description is intended unambiguously to include within the scope of the invention all individual integers within the range and all the combinations of upper and lower limit numbers within the broadest scope of the given range.
As used herein, the term “comprising” is to be read as meaning both comprising and consisting of. Consequently, where the invention relates to a “pharmaceutical composition comprising as active ingredient” a compound, this terminology is intended to cover both compositions in which other active ingredients may be present and also compositions which consist only of one active ingredient as defined.
Unless otherwise defined, all the technical and scientific terms used here have the same meaning as that usually understood by an ordinary specialist in the field to which this invention belongs. Similarly, all the publications, patent applications, all the patents and all other references mentioned here are incorporated by way of reference in their entirety (where legally permissible).
Particular non-limiting examples of the present invention will now be described.
Experimental
General Method
Compounds of formula 1 were prepared using the following synthetic procedure.
Figure imgf000008_0001
Scheme 1
In Scheme 1 above, Ri , n and m have the same meanings as set out herein.
The carboxylic acid (1 equivalent), potassium hydroxide (1 equivalent) and potassium carbonate (2 equivalents) were added to water and stirred. The sulfonyl chloride (1 equivalent) was added and the reaction was stirred at room temperature for 3 hours. The reaction was cooled to 0°C and acidified with 2M hydrochloric acid to give a white precipitate. This precipitate was dried and triturated with n-pentane to give the compound of formula (I).
Synthesis of specific examples
Example 1: (R)-l-((3-chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3- carboxylic acid
(S)- l-((3-chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid was prepared using the following synthesis procedure.
Figure imgf000008_0002
(S)-Piperidine-3-carboxylic acid (7.7 mmol), potassium hydroxide (7.7 mmol) and potassium carbonate (15.4 mmol) were added to water (20ml) and stirred. 3-Chloro-2- (trifluoromethyl)benzenesulfonyl chloride (7.7 mmol) was added and the reaction was stirred at room temperature for 3 hours. The reaction was cooled to 0°C and acidified with 2M hydrochloric acid to give a white precipitate. This precipitate was dried and triturated with n-pentane to give (S)-l-((3-chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3- carboxylic acid.
Example 2: (R)- 1 -((3 -chloro-2-(trifluoromethyl)phenyl) sulfonyl)piperidine-3 - carboxylic acid
(R)-l-((3-Chloro-2-(trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid was prepared in the same manner as (S)-l-((3-chloro-2-
(trifluoromethyl)phenyl)sulfonyl)piperidine-3-carboxylic acid, but using (R)-piperidine- 3-carboxylic acid.
Example 3
The following compounds were also prepared using the general method as set out above.
Figure imgf000010_0001

Claims

Claims
1. A compound of formula (I):
Figure imgf000011_0001
wherein
• Ri , R2, R3 and R4 are independently selected from chloride, fluoride, trifluoromethyl, -O-methyl and hydrogen,
• m and n are independently 1, 2 or 3, but the combination where both n and m are 1 is excluded,
• when the combination of m and n results in a chiral centre, optionally both enantiomers and the racemic mixture are included, and the compound is
Figure imgf000012_0001
2. A mixture of the two enantiomers of any compound according to claim 1, wherein the mixture is either racemic or has one enantiomer in excess of the other enantiomer.
3. The compound or mixture according to any preceding claim in a pharmaceutically acceptable salt form.
4. A pharmaceutical composition comprising a compound or mixture according to any one of claims 1 to 3 and a pharmaceutically or therapeutically acceptable excipient or carrier.
5. Use of a compound or mixture according to any one of claims 1 to 3 in the manufacture of a medicament for the treatment of a disease or disorder.
6. A compound or mixture according to any one of claims 1 to 3 for use in the treatment of a disease or disorder.
7. A compound or mixture according to any one of claims 1 to 3 for use as an inducer of Z A1AT secretion.
8. A method of treating a disease or disorder, comprising the step of administering a compound or mixture according to any one of claims 1 to 3, or a pharmaceutical composition according to claim 4, to a patient in need thereof.
9. Use of a compound or mixture according to any one of claims 1 to 3 in the treatment of a disease or disorder.
10. The use according to claim 9 as an inducer of Z A1AT secretion.
11. The use according to either of claim 9 or claim 10 wherein the use is in vitro.
12. The use according to claim 5, the compound or mixture for use according to claim 6, the method of treatment according to claim 8, or the use according to any one of claims 9 to 11, wherein the disease or disorder is AATD.
15. Use of a racemic compound of formula (I) as defined in any of claims 1 to 3 in the manufacture of a medicament for the treatment of a disease or disorder, wherein the disease or disorder is AATD.
16. Use of a racemic compound of formula (I) as defined in any of claims 1 to 3 as an inducer of Z A1AT secretion.
PCT/GB2022/051503 2021-06-15 2022-06-15 Compounds and their use for the treatment of alpha1-antitrypsin deficiency WO2022263817A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB2108542.8A GB202108542D0 (en) 2021-06-15 2021-06-15 Compounds and their use for the treatment of alpha1-antitrypsin deficiency
GB2108542.8 2021-06-15

Publications (1)

Publication Number Publication Date
WO2022263817A1 true WO2022263817A1 (en) 2022-12-22

Family

ID=76954608

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2022/051503 WO2022263817A1 (en) 2021-06-15 2022-06-15 Compounds and their use for the treatment of alpha1-antitrypsin deficiency

Country Status (2)

Country Link
GB (1) GB202108542D0 (en)
WO (1) WO2022263817A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005113542A2 (en) * 2004-05-20 2005-12-01 Elan Pharmaceuticals, Inc. N-cyclic sulfonamido inhibitors of gamma secretase
EP2471363A1 (en) * 2010-12-30 2012-07-04 Bayer CropScience AG Use of aryl-, heteroaryl- and benzylsulfonamide carboxylic acids, -carboxylic acid esters, -carboxylic acid amides and -carbonitriles and/or its salts for increasing stress tolerance in plants
WO2020120992A1 (en) * 2018-12-14 2020-06-18 Z Factor Limited COMPOUNDS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY
WO2021067584A1 (en) * 2019-10-02 2021-04-08 Vertex Pharmaceuticals Incorporated Methods of treatment for alpha-1 antitrypsin deficiency
WO2021116703A1 (en) * 2019-12-13 2021-06-17 Z Factor Limited COMPOUNDS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005113542A2 (en) * 2004-05-20 2005-12-01 Elan Pharmaceuticals, Inc. N-cyclic sulfonamido inhibitors of gamma secretase
EP2471363A1 (en) * 2010-12-30 2012-07-04 Bayer CropScience AG Use of aryl-, heteroaryl- and benzylsulfonamide carboxylic acids, -carboxylic acid esters, -carboxylic acid amides and -carbonitriles and/or its salts for increasing stress tolerance in plants
WO2020120992A1 (en) * 2018-12-14 2020-06-18 Z Factor Limited COMPOUNDS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY
WO2021067584A1 (en) * 2019-10-02 2021-04-08 Vertex Pharmaceuticals Incorporated Methods of treatment for alpha-1 antitrypsin deficiency
WO2021116703A1 (en) * 2019-12-13 2021-06-17 Z Factor Limited COMPOUNDS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 21 November 2016 (2016-11-21), XP002807232, Database accession no. 2035392-14-2 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 24 November 2016 (2016-11-24), XP002807231, Database accession no. 2036545-67-0 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 28 November 2016 (2016-11-28), XP002807230, Database accession no. 2039706-22-2 *
GOULD, INT J PHARM, vol. 33, 1986, pages 201 - 217

Also Published As

Publication number Publication date
GB202108542D0 (en) 2021-07-28

Similar Documents

Publication Publication Date Title
JP5255456B2 (en) Pharmaceutical composition containing pyrazine derivative and method of using pyrazine derivative in combination
US20230089087A1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
US20230102488A1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
US20230129181A1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
EP4072675B1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
WO2022263817A1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
WO2022263819A1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
WO2022263820A1 (en) COMPOUNDS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY
EP4073068B1 (en) Compounds and their use for the treatment of alpha1-antitrypsin deficiency
WO2022263816A1 (en) COMPOSITIONS AND THEIR USE FOR THE TREATMENT OF α1-ANTITRYPSIN DEFICIENCY
WO2021255436A1 (en) 4-((6-oxopyrimidin-1(6h)-yl)methyl)benzoic acid as an inducer of z a1 at secretion for treating aatd

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: 22736328

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE