New Drug Approvals

Home » FDA 2023 (Page 2)

Category Archives: FDA 2023

DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE

Blog Stats

  • 4,798,920 hits

Flag and hits

Flag Counter

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
Follow New Drug Approvals on WordPress.com

Archives

Categories

Recent Posts

Flag Counter

ORGANIC SPECTROSCOPY

Read all about Organic Spectroscopy on ORGANIC SPECTROSCOPY INTERNATIONAL 

Enter your email address to follow this blog and receive notifications of new posts by email.

Join 37.9K other subscribers
DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO Ph.D

DR ANTHONY MELVIN CRASTO, Born in Mumbai in 1964 and graduated from Mumbai University, Completed his Ph.D from ICT, 1991,Matunga, Mumbai, India, in Organic Chemistry, The thesis topic was Synthesis of Novel Pyrethroid Analogues, Currently he is working with AFRICURE PHARMA, ROW2TECH, NIPER-G, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India as ADVISOR, earlier assignment was with GLENMARK LIFE SCIENCES LTD, as CONSUlTANT, Retired from GLENMARK in Jan2022 Research Centre as Principal Scientist, Process Research (bulk actives) at Mahape, Navi Mumbai, India. Total Industry exp 32 plus yrs, Prior to joining Glenmark, he has worked with major multinationals like Hoechst Marion Roussel, now Sanofi, Searle India Ltd, now RPG lifesciences, etc. He has worked with notable scientists like Dr K Nagarajan, Dr Ralph Stapel, Prof S Seshadri, etc, He did custom synthesis for major multinationals in his career like BASF, Novartis, Sanofi, etc., He has worked in Discovery, Natural products, Bulk drugs, Generics, Intermediates, Fine chemicals, Neutraceuticals, GMP, Scaleups, etc, he is now helping millions, has 9 million plus hits on Google on all Organic chemistry websites. His friends call him Open superstar worlddrugtracker. His New Drug Approvals, Green Chemistry International, All about drugs, Eurekamoments, Organic spectroscopy international, etc in organic chemistry are some most read blogs He has hands on experience in initiation and developing novel routes for drug molecules and implementation them on commercial scale over a 32 PLUS year tenure till date Feb 2023, Around 35 plus products in his career. He has good knowledge of IPM, GMP, Regulatory aspects, he has several International patents published worldwide . He has good proficiency in Technology transfer, Spectroscopy, Stereochemistry, Synthesis, Polymorphism etc., He suffered a paralytic stroke/ Acute Transverse mylitis in Dec 2007 and is 90 %Paralysed, He is bound to a wheelchair, this seems to have injected feul in him to help chemists all around the world, he is more active than before and is pushing boundaries, He has 100 million plus hits on Google, 2.5 lakh plus connections on all networking sites, 100 Lakh plus views on dozen plus blogs, 227 countries, 7 continents, He makes himself available to all, contact him on +91 9323115463, email amcrasto@gmail.com, Twitter, @amcrasto , He lives and will die for his family, 90% paralysis cannot kill his soul., Notably he has 38 lakh plus views on New Drug Approvals Blog in 227 countries......https://newdrugapprovals.wordpress.com/ , He appreciates the help he gets from one and all, Friends, Family, Glenmark, Readers, Wellwishers, Doctors, Drug authorities, His Contacts, Physiotherapist, etc He has total of 32 International and Indian awards

Verified Services

View Full Profile →

Archives

Categories

Flag Counter

Tofersen


all-P-ambo-2′-O-(2-Methoxyethyl)-5-methyl-P-thiocytidylyl-(3’→5′)-2′-O-(2-methoxyethyl)adenylyl-(3’→5′)-2′-O-(2-methoxyethyl)-P-thioguanylyl-(3’→5′)-2′-O-(2-methoxyethyl)guanylyl-(3’→5′)-2′-O-(2-methoxyethyl)-P-thioadenylyl-(3’→5′)-P-thiothymidylyl-(3’→5′)-2′-deoxy-P-thioadenylyl-(3’→5′)-2′-deoxy-5-methyl-P-thiocytidylyl-(3’→5′)-2′-deoxy-P-thioadenylyl-(3’→5′)-P-thiothymidylyl-(3’→5′)-P-thiothymidylyl-(3’→5′)-P-thiothymidylyl-(3’→5′)-2′-deoxy-5-methyl-P-thiocytidylyl-(3’→5′)-P-thiothymidylyl-(3’→5′)-2′-deoxy-P-thioadenylyl-(3’→5′)-2′-O-(2-methoxyethyl)-5-methylcytidylyl-(3’→5′)-2′-O-(2-methoxyethyl)-P-thioadenylyl-(3’→5′)-2′-O-(2-methoxyethyl)guanylyl-(3’→5′)-2′-O-(2-methoxyethyl)-5-methyl-P-thiocytidylyl-(3’→5′)-2′-O-(2-methoxyethyl)-5-methyluridine

C230H317N72O123P19S15  : 7127.86
[2088232-70-4]

Tofersen

CAS 2088232-70-4

FDA APPROVED 4/25/2023, Qalsody

  • Antisense Oligonucleotide Inhibitor Of The Expression Of Superoxide Dismutase 1 Gene
  • DNA, D((2′-O-(2-METHOXYETHYL))M5RC-SP-(2′-O-(2-METHOXYETHYL))RA-(2′-O-(2-METHOXYETHYL))RG-SP-(2′-O-(2-METHOXYETHYL))RG-(2′-O-(2-METHOXYETHYL))RA-SP-T-SP-A-SP-M5C-SP-A-SP-T-SP-T-SP-T-SP-M5C-SP-T-SP-A-SP-(2′-O-(2-METHOXYETHYL))M5RC-(2′-O-(2-METHOXYETHYL))R
  • IONIS SOD1Rx

To treat amyotrophic lateral sclerosis in adults who have a SOD1 gene mutation
Drug Trials Snapshot

A nucleic acid-based drug indicated for the treatment of a specific type of amyotrophic lateral sclerosis.

Tofersen, sold under the brand name Qalsody, is a medication used for the treatment of amyotrophic lateral sclerosis (ALS).[3] Tofersen is an antisense oligonucleotide that targets the production of superoxide dismutase 1, an enzyme whose mutant form is commonly associated with amyotrophic lateral sclerosis. It is administered as an intrathecal injection.[3]

The most common side effects include fatigue, arthralgia (joint pain), increased cerebrospinal (brain and spinal cord) fluid white blood cells, and myalgia (muscle pain).[3]

Tofersen was approved for medical use in the United States in April 2023,[3][6] and in the European Union in May 2024.[4] The US Food and Drug Administration (FDA) considers it to be a first-in-class medication.[7]

Clinical data
Trade namesQalsody
AHFS/Drugs.comMonograph
MedlinePlusa623024
License dataUS DailyMedTofersen
Routes of
administration
Intrathecal
ATC codeN07XX22 (WHO)
Legal status
Legal statusCA℞-only[1]US: ℞-only[2][3]EU: Rx-only[4][5]
Identifiers
CAS Number2088232-70-4
DrugBankDB14782
UNII2NU6F9601K
KEGGD11811
Chemical and physical data
FormulaC230H317N72O123P19S15
Molar mass7127.85 g·mol−1

References

  1. ^ “Register of Innovative Drugs”Health Canada. 3 November 2006. Retrieved 17 April 2025.
  2. ^ “Qalsody- tofersen injection”DailyMed. 25 April 2023. Archived from the original on 8 May 2023. Retrieved 10 June 2023.
  3. Jump up to:a b c d e f g h i j k l “FDA approves treatment of amyotrophic lateral sclerosis associated with a mutation in the SOD1 gene” (Press release). U.S. Food and Drug Administration (FDA). 25 April 2023. Archived from the original on 25 April 2023. Retrieved 25 April 2023. Public Domain This article incorporates text from this source, which is in the public domain.
  4. Jump up to:a b c d “Qalsody EPAR”European Medicines Agency (EMA). 22 February 2024. Retrieved 24 February 2024. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
  5. Jump up to:a b “Qalsody PI”Union Register of medicinal products. 3 June 2024. Retrieved 7 September 2024.
  6. ^ “FDA Grants Accelerated Approval for Qalsody (tofersen) for SOD1-ALS, a Major Scientific Advancement as the First Treatment to Target a Genetic Cause of ALS” (Press release). Biogen. 25 April 2023. Archived from the original on 25 April 2023. Retrieved 25 April 2023 – via GlobeNewswire.
  7. Jump up to:a b New Drug Therapy Approvals 2023 (PDF). U.S. Food and Drug Administration (FDA) (Report). January 2024. Archived from the original on 10 January 2024. Retrieved 9 January 2024.
  8. ^ Liu A (1 May 2019). “Biogen’s antisense ALS drug shows promise in early clinical trial”FierceBiotechArchived from the original on 2 February 2023. Retrieved 25 April 2023.
  9. ^ Langreth R (22 March 2023). “Biogen’s ALS Drug Gets Partial Backing From FDA Panel”Bloomberg News. Retrieved 25 April 2023.
  10. ^ “FDA approves drug which helps to slow progression of rare form of MND”http://www.sheffield.ac.uk. 28 April 2023. Retrieved 16 May 2024.
  11. ^ Berdyński M, Miszta P, Safranow K, Andersen PM, Morita M, Filipek S, et al. (January 2022). “SOD1 mutations associated with amyotrophic lateral sclerosis analysis of variant severity”Scientific Reports12 (1): 103. Bibcode:2022NatSR..12..103Bdoi:10.1038/s41598-021-03891-8PMC 8742055PMID 34996976.
  12. ^ Constantino A (25 April 2023). “FDA grants accelerated approval for Biogen ALS drug that treats rare form of the disease”CNBCArchived from the original on 25 April 2023. Retrieved 25 April 2023.
  13. ^ Constantino A (22 March 2023). “FDA advisors vote against effectiveness of Biogen’s ALS drug for rare and aggressive form of the disease”CNBCArchived from the original on 10 April 2023. Retrieved 25 April 2023.
  14. ^ Robins R (25 April 2023). “F.D.A. Approves Drug for Rare Form of A.L.S.” The New York TimesArchived from the original on 25 April 2023. Retrieved 25 April 2023.
  15. ^ “New treatment for rare motor neuron disease recommended for approval”European Medicines Agency (EMA) (Press release). 23 February 2024. Retrieved 24 February 2024.

////////////tofersen, Qalsody, FDA 2023, APPROVALS 2023, EU 2024, EMA 2024, BIIB 067, BIIB067, IONIS SOD1Rx

Leniolisib


Leniolisib

CAS 1354690-24-6

WeightAverage: 450.466
Monoisotopic: 450.199108558

Chemical FormulaC21H25F3N6O2


  • CDZ-173-NX
  • CDZ173
  • CDZ173-NX


1-[(3S)-3-({6-[6-methoxy-5-(trifluoromethyl)pyridin-3-yl]-5H,6H,7H,8H-pyrido[4,3-d]pyrimidin-4-yl}amino)pyrrolidin-1-yl]propan-1-one

FDA APPROVED Joenja, 3/24/2023, To treat activated phosphoinositide 3-kinase delta syndrome
Drug Trials Snapshot

Leniolisib (INN[3][4]), sold under the brand name Joenja, is a medication used for the treatment of activated phosphoinositide 3-kinase delta syndrome (APDS).[2][5] It is a kinase inhibitor[2][6] that is taken by mouth.[2]

The most common side effects include headachesinusitis, and atopic dermatitis.[5]

Leniolisib was approved for medical use in the United States in March 2023.[5][7][8] It is the first approved medication for the treatment of activated PI3K delta syndrome.[5] The US Food and Drug Administration (FDA) considers it to be a first-in-class medication.[9]

PATENT

US8653092

https://patents.google.com/patent/US8653092B2/en

PATENT

https://patentscope.wipo.int/search/en/WO2012004299

Example 67 was prepared according the general procedure described in scheme 4

Example 67: 1 -{(S)-3-[6-(6-Methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidin-4-ylamino]-pyrrolidin-1-yl}-propan-1-one

To a solution of (S)-3-[6-(6-methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidin-4-ylamino]-pyrrolidine-1 -carboxylic acid tert-butyl ester (intermediate 24) (13.4 g, 27.1 mmol) in CH2CI2 (100 mL), was added TFA (41 .8 mL) and the mixture stirred at rt for 1 h. Concentrated in vacuo and partitioned between 2M NaOH(aq) (300 mL) and CH2CI2 (200 mL). The organic phase was separated and the aqueous phase extracted with CH2CI2 (2 x 200 mL). The organic phases were combined, dried (MgS04) and

evaporated in vacuo to give a brown foam. The foam was dissolved in CH2CI2 (50 mL) and was added simultaneously portionwise with sat.NaHC03(aq) (50 mL) to a vigourously stirring solution of propionyl chloride (2.63 g, 28.5 mmol) in CH2CI2 (50 mL) at rt. The resulting biphasic mixture was stirred at rt for 1 h. Further propionyl chloride (0.566g, 6.12 mmol) was added and continued stirring vigorously for 20 min. The organic layer was separated and the aqueous layer extracted with CH2CI2 (100 mL). The organic layers were combined, dried (MgS04) and concentrated in vacuo to give a brown gum. The gum was stirred in EtOAc (100 mL) and the resulting solid filtered (9.4 g). The mother liquors were concentrated in vacuo and purified by column chromatography through a Biotage® amino silica gel eluting with EtOAc / MeOH, 100/0 to 90/10 to give a yellow foam which was then stirred in EtOAc (20 mL) and the resulting solid filtered (870 mg). Both batches of solids were combined and stirred in refluxing EtOAc (50 mL) for 1 h. Filtered to give 1-{(S)-3-[6-(6-methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidin-4-ylamino]-pyrrolidin-1 -yl}-propan-1 -one as a colourless solid (9.42 g, 76% yield). 1 H NMR (400 MHz, DMSO-d6, 298K) δ ppm 0.95-1.05 (m, 3H) 1 .87-2.32 (m, 4H) 2.77-2.86 (m, 2H) 3.25-3.88 (m, 6H) 3.93 (s, 3H) 3.98 (s, 2H) 4.55-4.80 (m, 1 H) 6.70-6.80 (m, 1 H, N-H) 7.86-7.92 (m, 1 H) 8.27-8.33 (m, 1 H) 8.33-8.37 (m, 1 H) LCMS: [M+H]+=451.0, Rt (6)= 1.49 min.

Alternative synthesis for example 67

A solution of (S)-3-[6-(6-methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidin-4-ylamino]-pyrrolidine-1-carboxylic acid tert-butyl ester (intermediate 24) (29.04 g, 58.73 mmol) in 2-Me-THF (100 mL) was dropwise added into aqueous HCI solution (150 mL, 31 %) over 15 min. The reaction mixture was partitioned between water (300 mL) and isopropyl acetate (100 mL) and the upper organic phase was discarded. The aqueous phase was partitioned between 25% NaOH (aq) (200 g) and 2-Me-THF (200 mL), and the organic phase was collected and dried. Triethylamine (16.32 mL, 1 17.48 mmol) was added into the organic phase followed by dropwise addition of propionyl chloride (6.0 g, 64.6 mmol) at 0 °C. The resulting mixture was stirred at 0 °C for 1 h. The reaction mixture was washed with water (1 10 mL) and the resulting organic phase was concentrated in vacuo to give a brown gum.

The residue was recrystallized with isopropanol and methyl tert-butyl ether to give 1 -{(S)-3- [6-(6-methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidin-4- ylamino]-pyrrolidin-1-yl}-propan-1 -one as a colourless solid (17.2 g, 65% yield).

Crystallization of Example 67 by heating in acetonitrile/water

2.0 g of Example 67 (4.440 mol) were dissolved in 10 mL of acetonitrile and 0.5 mL of water at 75°C. The solution was allowed to cool down to rt within 30 min resulting in a suspension. The mixture was stirred for 16 h at rt. The crystals were collected by filtration. The filter cake was washed 2 times with 1 mL of acetonitrile and afterwards dried for 16 h at 24°C and ca. 10 mbar vacuum. Elementary analysis of the material showed a waterless form.

PAPER

https://pubs.acs.org/doi/10.1021/acsmedchemlett.7b00293

ACS Medicinal Chemistry Letters

Cite this: ACS Med. Chem. Lett. 2017, 8, 9, 975–980

https://doi.org/10.1021/acsmedchemlett.7b00293

The predominant expression of phosphoinositide 3-kinase δ (PI3Kδ) in leukocytes and its critical role in B and T cell functions led to the hypothesis that selective inhibitors of this isoform would have potential as therapeutics for the treatment of allergic and inflammatory disease. Targeting specifically PI3Kδ should avoid potential side effects associated with the ubiquitously expressed PI3Kα and β isoforms. We disclose how morphing the heterocyclic core of previously discovered 4,6-diaryl quinazolines to a significantly less lipophilic 5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine, followed by replacement of one of the phenyl groups with a pyrrolidine-3-amine, led to a compound series with an optimal on-target profile and good ADME properties. A final lipophilicity adjustment led to the discovery of CDZ173 (leniolisib), a potent PI3Kδ selective inhibitor with suitable properties and efficacy for clinical development as an anti-inflammatory therapeutic. In vitro, CDZ173 inhibits a large spectrum of immune cell functions, as demonstrated in B and T cells, neutrophils, monocytes, basophils, plasmocytoid dendritic cells, and mast cells. In vivo, CDZ173 inhibits B cell activation in rats and monkeys in a concentration- and time-dependent manner. After prophylactic or therapeutic dosing, CDZ173 potently inhibited antigen-specific antibody production and reduced disease symptoms in a rat collagen-induced arthritis model. Structurally, CDZ173 differs significantly from the first generation of PI3Kδ and PI3Kγδ-selective clinical compounds. Therefore, CDZ173 could differentiate by a more favorable safety profile. CDZ173 is currently in clinical studies in patients suffering from primary Sjögren’s syndrome and in APDS/PASLI, a disease caused by gain-of-function mutations of PI3Kδ.

Synthesis and full characterization of (S)-1-(3-((6-(6-methoxy-5-(trifluoromethyl)pyridin-3-yl)-
5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin-4-yl)amino)pyrrolidin-1-yl)propan-1-one (3h, CDZ173,
leniolisib)

TFA (41.8 mL) was added to a solution of (S)-3-[6-(6-methoxy-5-trifluoromethyl-pyridin-3-yl)-5,6,7,8-
tetrahydro-pyrido[4,3-d]pyrimidin-4-ylamino]-pyrrolidine-1-carboxylic acid tert-butyl ester (13.4 g,
27.1 mmol) in CH2Cl2 (100 mL), and the mixture was stirred at RT for 1 h. After that time, the mixture
was concentrated under reduced pressure, and the residue was partitioned between NaOH (aqu., 2M,
300 mL) and CH2Cl2 (200 mL). The organic phase was separated, and the aqueous phase was extracted
with CH2Cl2 (2 x 200 mL). The combined organic phases were dried (MgSO4) and concentrated under
reduced pressure. The resulting brown foam was dissolved in CH2Cl2 (50 mL) and added simultaneously with a NaHCO3 solution (aqu., saturated) (50 mL) to a vigorously stirring solution of propionyl chloride (2.63 g, 28.5 mmol) in CH2Cl2 (50 mL) at RT. The resulting biphasic mixture was stirred at RT for
1h. Additional propionyl chloride (0.566 g, 6.12 mmol) was added, and vigorous stirring was continued
for 20 min. The organic layer was separated and the aqueous layer extracted with CH2Cl2 (100 mL). The
combined organic layers were dried (MgSO4) and concentrated under reduced pressure. The resulting
brown gum was stirred in EtOAc (100 mL) and the resulting solid was filtered (9.4 g). The mother liquors were concentrated under reduced pressure and purified by column chromatography through a Biotage®
amino silica gel eluting with EtOAc / MeOH, 100/0 to 90/10. After concentration under reduced
pressure, the resulting yellow foam was stirred in EtOAc (20 mL) and the resulting solid was filtered
(870 mg). Both batches of solids were combined and stirred in refluxing EtOAc (50 mL) for 1h. The
resulting solid was filtered to give the title compound as a colorless solid (9.42 g, 76%). 1H NMR (400
MHz, DMSO-d6, 298K, ca. 1:1 mixture of rotamers) δ ppm 8.35 (m, 1H) 8.30 (m, 1H) 7.89 (m, 1H)
6.80-6.70 (m, 1H, N-H) 4.80-4.55 (m, 1H) 3.93 (s, 3H) 3.98 (s, 2H) 3.88-3.25 (m, 6H) 2.86-2.75 (m,
2H) 2.32-1.87 (m, 4H) 1.05-0.95 (m, 3H); 13C-NMR (150 MHz, DMSO-d6, 298K, ca. 1:1 mixture of
rotamers, data given for cis-isomer): δ ppm 171.3, 158.6, 158.1, 155.5, 153.6, 141.3, 138.0, 125.7,
123.3, 111.1, 109.7, 53.7, 50.8, 49.4, 45.8, 45.8, 44.3, 31.2, 29.7, 26.6, 8.97; LCMS method 1: Rt 1.49
min, calcd for C21H26F3N6O2 [M+H]+
451.2, found 451.0, HRMS (ESI+) calcd for C21H26F3N6O2
[M+H]+ 451.20693, found 451.20642

REF

https://www.researchgate.net/figure/Stereo-divergent-strategy-for-the-synthesis-of-key-intermediates-2-and-3-of-leniolisib_fig6_369490553

REF

https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2024.1337436/full

-Benzyl-4-chloro-5,6,7,8-tetrahydro-pyrido[4,3-d]pyrimidine (compound 1) is coupled with (S)-tert-butyl 3-aminopyrrolidine-1-carboxylate (compound 2) in the presence of triethylamine at 120 °C for 42 h to give compound 3 a 93% yield. The benzyl group is deprotected with 20% palladium hydroxide on carbon and ammonium formate in methanol at 65 °C for 2 h to give compound 4 a 66% yield. Compound 4 is coupled with 5-bromo-2-methoxy-3-(trifluoromethyl)pyridine (compound 5) in the presence of sodium –tert-butoxide, tris(dibenzylideneacetone)dipalladium(0), 2-di-t-butylphosphino-2′-(N,N-dimethylamino)biphenyl in tert-butanol at 100 °C for 5 h to give compound 6 a 74% yield. Deprotection of the Boc group in DCM/TFA, followed by coupling with propionyl chloride in the presence of sodium bicarbonate in DCM at room temperature for 1 h gives the final compound 7 (leniolisib) a 76% yield.

REF

https://www.sciencedirect.com/science/article/abs/pii/S0223523424000047

REF

J. Med. Chem. 2025, 68, 2147−2182

Leniolisib (Joenja). Leniolisib (5), is a twice-daily, orally available selective phosphoinositide 3-kinase-delta
(PI3Kδ) inhibitor developed by Novartis and in-licensed by Pharming Group NV for the treatment of activated phosphoinositide 3-kinase-delta syndrome (APDS). APDS is a primary immunodeficiency caused by mutations in PI3Kδ catalytic (PIK3CD) or regulatory (PIK3R1) subunits. The loss or gain of function of these subunits results in hyperactivity of the PI3Kδ pathway which can result in infections, lymphoprolif
eration, autoimmunity, increased risk of malignant lymphoma and early mortality. 44−46
Current treatment strategies include immunosuppressives such as corticosteroids, antiviral, and antibiotic therapies, stem cell transplantation, and immunoglobulin replacement therapy. However, none of these therapeutic strategies treats the underlying hyperactivity of the PI3Kδpathway. Thus, the approval of leniolisib by the USFDA in March 2023 provided a significant breakthrough therapy for patients 12 years and older.47 48
A concise synthetic route to leniolisib has been disclosed by Novartis,beginning with commercially available tetrahydropyridopyrimidine 5.1 (Scheme 9). An SNAr reaction with amine 5.2 furnished intermediate 5.3 in good yield. Transfer hydrogenation with Pd(OH)2 on carbon to remove the benzyl
group gave free amine 5.4, setting up the system for a Buchwald−Hartwig amination with bromide 5.5 to produce 5.6 in good yield. Protecting group removal and subsequent acylation with ethyl chloroformate provided leniolisib (5) in 76% yield over two steps.

(44) Hoegenauer, K.; Soldermann, N.; Stauffer, F.; Furet, P.;
Graveleau, N.; Smith, A. B.; Hebach, C.; Hollingworth, G. J.; Lewis,
I.; Gutmann, S.; et al. Discovery and pharmacological characterization
of novel quinazoline-based PI3K delta-selective inhibitors. ACS Med.
Chem. Lett. 2016, 7, 762−767.
(45) Hoegenauer, K.; Soldermann, N.; Zécri, F.; Strang, R. S.;
Graveleau, N.; Wolf, R. M.; Cooke, N. G.; Smith, A. B.; Hollingworth,
G. J.; Blanz, J.; et al. Discovery of CDZ173 (Leniolisib), representing a
structurally novel class of PI3K delta-selective inhibitors. ACS Med.
Chem. Lett. 2017, 8, 975−980.

(46) Duggan, S.; Al-Salama, Z. T. Leniolisib: first approval. Drugs
2023, 83, 943−948.
(47) Pharming announces US FDA approval of Joenja® (leniolisib) as
the first and only treatment indicated for APDS. Pharming, March 24, 2023 https://www.pharming.com/news/pharming-announces-us-fda
approval-joenja-leniolisib-first-and-only-treatment-indicated-apds (ac
cessed February 2024).
(48) Fernandes Gomes dos Santos, P. A.; Hogenauer, K.;
Hollingworth, G.; Soldermann, N.; Stowasser, F.; Tufilli, N.; Zecri, F.
Solid forms and salts of tetrahydro-pyrido-pyrimidine derivatives. WO
2013001445 A1, 2013

    Ref

    https://www.sciencedirect.com/science/article/abs/pii/S0223523424000047

    Leniolisib developed by Novartis Pharma AG, was approved on March 24, 2023, making it the first treatment drug for APDS [3]. APDS is an immunodeficiency disorder that primarily occurs due to mutations in the gene responsible for encoding phosphotidylinsitol-3-kinase δ(PI3Kδ). These mutations enhance the function of PI3Kδ, resulting in impaired immune response and heightened vulnerability to infections.
    Leniolisib is capable of inhibiting the hyperactive PI3Kδ enzyme by obstructing the active binding site within the p110δ subunit [54]. Inisolated enzyme assays conducted without cells, the selectivity of PI3K-δ
    was found to be higher compared to PI3Kα(28-fold), PI3K-β (43-fold),and PI3K-γ (257-fold), as well as other enzymes in the kinome [54].Leniolisib demonstrated the ability to decrease phosphoinositide-3-kinase/protein kinase B (pAKT) pathway activity and suppress the growth and activation of B and T cell subsets in cell-based experiments. Leniolisib effectively blocks the signaling pathways responsible for the excessive production of phosphatidylinositol 3,4,5-trisphosphate (PIP3), overactivation of the downstream
    mammalian target of rapamycin (mTOR)/protein kinase B (AKT) pathway, and the imbalanced functioning of B and T cells [55].
    One representative approach of Leniolisib is depicted in Scheme 15 [55]. Pyrrolidine LENI-003 was obtained by nucleophilic substitution of aminopyrrolidine LENI-001 and the 4-Cl of LENI-002 under alkaline conditions, and LENI-004 was obtained by debenzylation of LENI-003 under palladium hydroxide/carbon. LENI-004 and 5-bromo-2-methox y-3-(trifluoromethyl)pyridine (LENI-005) were coupled to obtain LENI-006. LENI-006 was deprotected by TFA and further condensed with acyl chloride to obtain Leniolisib.

    54] V.K. Rao, S. Webster, V. Dalm, A. ˇ Sediv´ a, P.M. van Hagen, S. Holland, S.
    D. Rosenzweig, A.D. Christ, B. Sloth, M. Cabanski, A.D. Joshi, S. de Buck,
    J. Doucet, D. Guerini, C. Kalis, I. Pylvaenaeinen, N. Soldermann, A. Kashyap,
    G. Uzel, M.J. Lenardo, D.D. Patel, C.L. Lucas, C. Burkhart, Effective “activated
    PI3Kδ syndrome”-targeted therapy with the PI3Kδ inhibitor leniolisib, Blood 130
    (2017) 2307–2316.

    [55] K. Hoegenauer, N. Soldermann, F. Z´ ecri, R.S. Strang, N. Graveleau, R.M. Wolf, N.
    G. Cooke, A.B. Smith, G.J. Hollingworth, J. Blanz, S. Gutmann, G. Rummel,
    A. Littlewood-Evans, C. Burkhart, Discovery of CDZ173 (Leniolisib), representing
    a structurally novel class of PI3K delta-selective inhibitors, ACS Med. Chem. Lett.
    8 (2017) 975–980.[55] K. Hoegenauer, N. Soldermann, F. Z´ ecri, R.S. Strang, N. Graveleau, R.M. Wolf, N.
    G. Cooke, A.B. Smith, G.J. Hollingworth, J. Blanz, S. Gutmann, G. Rummel,
    A. Littlewood-Evans, C. Burkhart, Discovery of CDZ173 (Leniolisib), representing
    a structurally novel class of PI3K delta-selective inhibitors, ACS Med. Chem. Lett.
    8 (2017) 975–980.

    ,

    References

    1. ^ “Joenja (Ballia Holdings Pty Ltd)”Therapeutic Goods Administration (TGA). 16 April 2025. Retrieved 3 May 2025.
    2. Jump up to:a b c d e f “Joenja- leniolisib tablet, film coated”DailyMed. 29 March 2023. Retrieved 20 June 2023.
    3. ^ World Health Organization (2016). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 76”. WHO Drug Information30 (3). hdl:10665/331020.
    4. ^ World Health Organization (2017). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 77”. WHO Drug Information31 (1). hdl:10665/330984.
    5. Jump up to:a b c d e f g h i j “FDA approves first treatment for activated phosphoinositide 3-kinase delta syndrome”U.S. Food and Drug Administration (FDA) (Press release). 24 March 2023. Retrieved 24 March 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    6. ^ Duggan S, Al-Salama ZT (July 2023). “Leniolisib: First Approval”Drugs83 (10): 943–948. doi:10.1007/s40265-023-01895-4PMID 37256490S2CID 258989663.
    7. Jump up to:a b “US FDA approves Pharming’s immune disorder drug”. Reuters. Archived from the original on 24 March 2023. Retrieved 24 March 2023.
    8. ^ “Pharming announces US FDA approval of Joenja (leniolisib) as the first and only treatment indicated for APDS” (PDF). Pharming Group N.V. (Press release). 24 March 2023. Retrieved 25 March 2023.
    9. ^ New Drug Therapy Approvals 2023 (PDF). U.S. Food and Drug Administration (FDA) (Report). January 2024. Archived from the original on 10 January 2024. Retrieved 9 January 2024.

    Public Domain This article incorporates text from this source, which is in the public domainBing Chat output modified to create the initial revision of this article. 25 March 2023. – via Microsoft

    Clinical trial number NCT02435173 for “Study of Efficacy of CDZ173 in Patients With APDS/PASLI” at ClinicalTrials.gov

    Clinical data
    Trade namesJoenja
    Other namesCDZ173
    AHFS/Drugs.comMonograph
    MedlinePlusa623016
    License dataUS DailyMedLeniolisib
    Routes of
    administration
    By mouth
    Drug classAntineoplastic
    ATC codeL03AX22 (WHO)
    Legal status
    Legal statusAU: S4 (Prescription only)[1]US: ℞-only[2]
    Identifiers
    CAS Number1354690-24-6as salt: 1354691-97-6
    DrugBankDB16217
    ChemSpider52083264
    UNIIL22772Z9CP
    KEGGD11158as salt: D11159
    ChEMBLChEMBL3643413
    PDB ligand9NQ (PDBeRCSB PDB)
    Chemical and physical data
    FormulaC21H25F3N6O2
    Molar mass450.466 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI

    //////////leniolisib, Joenja, FDA 2023, APPROVALS 2023, CDZ-173-NX, CDZ173, CDZ173-NX

    str1

    AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@gmail.com

    ……

    Rezafungin


    Rezafungin

    CAS 1396640-59-7

    WeightAverage: 1226.411
    Monoisotopic: 1225.602719729

    Chemical FormulaC63H85N8O17

    FDA APPROVED 3/22/2023, Rezzayo, To treat candidemia and invasive candidiasis
    Drug Trials Snapshot

    2-[[(3S,6S,9S,11R,15S,18S,20R,21R,24S,25S,26S)-6-[(1S,2S)-1,2-dihydroxy-2-(4-hydroxyphenyl)ethyl]-11,20,25-trihydroxy-3,15-bis[(1R)-1-hydroxyethyl]-26-methyl-2,5,8,14,17,23-hexaoxo-18-[[4-[4-(4-pentoxyphenyl)phenyl]benzoyl]amino]-1,4,7,13,16,22-hexazatricyclo[22.3.0.09,13]heptacosan-21-yl]oxy]ethyl-trimethylazanium

    • Rezafungin ion
    • Rezafungin cation
    • CD-101
    • SP-3025
    • G013B5478J

    Rezafungin, sold under the brand name Rezzayo (by Melinta Therapeutics), is a medication used for the treatment of invasive candidiasis.[2] It is an echinocandin antifungal[1][4] that acts as a fungal β-glucan synthase inhibitor.[5]

    Rezafungin was approved for medical use in the United States in March 2023,[1][6][5] and in the European Union in December 2023.[2][3]

    CAS No. : 1631754-41-0

    Rezafungin acetate  (Synonyms: Biafungin acetate; CD101 acetate; SP-3025 acetate)

    Rezafungin acetate (Biafungin acetate) is a next-generation, broad-spectrum, and long-lasting echinocandin. Rezafungin acetate shows potent antifungal activity against Candida spp.Aspergillus spp., and Pneumocystis spp..

    SYN

    https://doi.org/10.1021/acs.jmedchem.4c02079
    J. Med. Chem. 2025, 68, 2147−2182

    Rezafungin (Rezzayo). Rezafungin (2) is a secondgeneration echinocandin that was discovered by Seachaid
    Pharmaceuticals and developed by Cidera Therapeutics. The once weekly intravenously administered drug is used to treat candidemia and invasive candidiasis and to prevent invasive fungal diseases in blood and bone marrow transplant patients.23
    Rezafungin was designed to improve the pharmacokinetic properties of the USFDA-approved first-generation echinocandins anidulafungin, caspofungin, and micafungin, enabling less frequent dosing. Mechanistically, echinocandins exert their antifungal activity by inhibiting β-(1→3)-glucan synthase, a
    transmembrane protein complex essential for the synthesis of an important polysaccharide component of the fungal cell wall.
    This noncompetitive inhibition destabilizes the cell wall, leading to osmotic imbalance and fungal cell death.24 Rezafungin was approved by the USFDA in March 2023 for use in patients 18 years and older.25
    An elegant semisynthesis of rezafungin from anidulafungin (2.1) was reported by Cidera Therapeutics that circumvented chemical instability including potential racemization of the
    parent compound (Scheme 3).26,27 The semisynthetic sequence26 begins with boronate formation between the 1,2-diol of 2.1 and 3,4-dimethoxyphenylborane (2.2) utilizing azeotropic distillation, maintaining a constant volume of THF. Addition of a solution of choline chloride, TFA, and TFAA in
    MeCN to the slurry of boronate ester 2.3 gave the choline ether. Selective ether formation at the hemiaminal hydroxyl group occurred due to its increased reactivity compared to the other free hydroxyls in the compound.27 The specific boronate ester used in this sequence was found to be beneficial at minimizing the amount of a diastereomer impurity (at the hemiaminal) formed in the choline conjugation, though the authors of the patent shared that this was unexpected given the remote boronic
    acid from the hemiaminal that participated in conjugation. A 95:5 α:β selectivity of the conjugation was achieved under acidic conditions, and preferential crystallization of the α-isomer while
    maintaining solution equilibrium enabled control of the βisomer to less than 2.0%. Work up of the reaction using ammonium acetate and ammonium hydroxide provided crude
    rezafungin. Ion exchange chromatography was used to remove3,4-dimethyoxyphenyl boronic acid, eluting with ammonium acetate to afford rezafungin (2). Using this synthetic sequence, a purity of 98.49% was reported with only minor amounts of racemization observed (0.77% undesired diastereomer and
    0.51% unwanted epimer at the benzylic center).

    (23) Syed, Y. Y. Rezafungin: first approval. Drugs 2023, 83, 833−840.

    (24) Denning, D. W. Echinocandins: a new class of antifungal. J.
    Antimicrob. Chemother. 2002, 49, 889−891.
    (25) Cidara Therapeutics and Melinta Therapeutics announce FDA
    approval of RezzayoTM (Rezafungin for injection) for the treatment of
    candidemia and invasive candidiasis. Cidera Therapeutics, March 22,

    1. https://www.cidara.com/news/cidara-therapeutics-andmelinta-therapeutics-announce-fda-approval-of-rezzayo-rezafunginfor-injection-for-the-treatment-of-candidemia-and-invasivecandidiasis/ (accessed February 2024).
      (26) Cidara Therapeutics. Synthesis of echinocandin antifungal agent.
      WO 2019241626 A1, 2019.
      (27) Jamison, J. A.; LaGrandeur, L. M.; Rodriguez, M. J.; Turner, W.
      W.; Zeckner, D. J. The synthesis and antifungal activity of nitrogen
      containing hemiaminal ethers of LY303366. J. Antibiot. (Tokyo) 1998,
      51, 239−42

    .

    SYN


    Hughes, D., et al. (2022). Synthesis of echinocandin antifungal agent. (U.S. Patent No. 11,524,980 B2). U.S. Patent and Trademark Office. https://patentimages.storage.googleapis.com/34/d5/c2/1a8cdcfb3fe3db/US11524980.pdf

    https://patentscope.wipo.int/search/en/detail.jsf?docId=US327113930&_cid=P11-MAORN7-73998-1

    Example 9. Synthesis of Compound 1 from the 3,4-dimethoxyphenylboronate Ester of Anidulafungin—Coupling in the Presence of TFAA

          Tetrahydrofuran (700 mL) and anidulafungin (108.44 g) were charged to a 1 L reactor. 3,4-Dimethoxyphenylboronic acid (21.0 g) was then charged and the mixture was stirred at 18-22° C. The reaction mixture was azeodried by distillation of tetrahydrofuran and simultaneous addition of fresh tetrahydrofuran (7.0 L). A constant volume solvent swap to acetonitrile was carried out by addition of acetonitrile (2.1 L) and simultaneous vacuum distillation. After complete turnover to acetonitrile, further distillation was carried out to reduce the volume to 420 mL.
          In a separate vessel, the following were combined with stirring: choline chloride (172 g), acetonitrile (217 mL), trifluoroacetic acid (142 mL), and trifluoroacetic anhydride (8.6 mL). This solution was then added to the slurry containing the anidulafungin boronate ester and the resulting mixture was stirred at 15° C. for 8 hours. The reaction was quenched by charging cooled (T<10° C.) solution of ammonium acetate (4.2 M, 221 mL) to the reactor at once followed by addition of chilled (T<10° C.)) water (221 mL). Then, a cooled (10° C.) solution of ammonium hydroxide (9.0 M, 126.4 mL) was added. The final pH was adjusted to pH 4.0-4.6 by addition of ammonium hydroxide. The crude reaction mixture was diluted with water:acetonitrile (3:1, 6 L) and stored at −20° C.
          Results: compound 1, 76.8%, compound 1 beta-diastereomer, 0.8%.
          A reduction in the level of compound 1 beta-diastereomer has allowed for replacement of the HPLC purification with medium pressure chromatography (MPLC) using a coarser grade of C18 silica (25 to 50 μm). The 3,4-dimethoxyphenyl boronic acid can be separated by ion-exchange capture, eluting with 100 mM ammonium acetate (pH 4.5) in water:acetonitrile 50:50 v:v, which affords salt exchange from trifluoroacetate to acetate.
    Patent NumberPediatric ExtensionApprovedExpires (estimated)
    US10702573No2020-07-072033-03-14US flag
    US9526835No2016-12-272033-03-14US flag
    US8722619No2014-05-132032-03-02US flag
    US11197909No2021-12-142038-07-14US flag
    US11654196No2023-05-232032-03-02US flag
    US11712459No2023-08-012037-03-15US flag
    US11819533No2023-11-212038-07-11US flag

    Medical uses

    In the United States, rezafungin is indicated in adults who have limited or no alternative options for the treatment of candidemia and invasive candidiasis.[1]

    In the European Union, rezafungin is indicated for the treatment of invasive candidiasis in adults.[2]

    Rezafungin, while remaining a hydrophilic compound, exhibits a volume of distribution more than twice that of caspofungin.[7] This pharmacokinetic property has supported its investigation for the treatment of deep-seated Candida infections, including osteomyelitis.[8][9]

    Rezafungin was approved for medical use in the United States in March 2023,[1][10][11] The FDA granted the application for rezafungin orphan drugfast track, and priority review designations.[12]

    In October 2023, the Committee for Medicinal Products for Human Use of the European Medicines Agency adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product Rezzayo, intended for the treatment of invasive candidiasis in adults.[2] The applicant for this medicinal product is Mundipharma GmbH.[2] Rezafungin was approved for medical use in the European Union in December 2023.[3]

    Rezafungin is a member of the family of echinocandins that inhibits 1,3-beta-D-glucan synthase. It is developed by Cidara Therapeutics and approved for the treatment of candidaemia and invasive candidiasis in patients aged >= 18 years who have limited or no alternative treatment options. It is an echinocandin, a quaternary ammonium ion, an antibiotic antifungal drug, an azamacrocycle, a homodetic cyclic peptide and an aromatic ether.

    Brand names

    Rezafungin is the international nonproprietary name.[13]

    Rezafungin is sold under the brand name Rezzayo.[2]

    References

    1. Jump up to:a b c d e “Rezzayo- rezafungin injection, powder, lyophilized, for solution”DailyMed. 8 June 2023. Retrieved 26 December 2023.
    2. Jump up to:a b c d e f g “Rezzayo EPAR”European Medicines Agency (EMA). 12 October 2023. Retrieved 27 December 2023. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
    3. Jump up to:a b c “Rezzayo Product information”Union Register of medicinal products. 22 December 2023. Retrieved 26 December 2023.
    4. ^ Zhao Y, Perlin DS (September 2020). “Review of the Novel Echinocandin Antifungal Rezafungin: Animal Studies and Clinical Data”Journal of Fungi6 (4): 192. doi:10.3390/jof6040192PMC 7712954PMID 32998224.
    5. Jump up to:a b Syed YY (June 2023). “Rezafungin: First Approval”Drugs83 (9): 833–840. doi:10.1007/s40265-023-01891-8PMID 37212966S2CID 258831091.
    6. ^ “Rezzayo approved by FDA amid rapid Candida auris spread”thepharmaletter.com. 23 March 2023.
    7. ^ Albanell-Fernández M (January 2025). “Echinocandins Pharmacokinetics: A Comprehensive Review of Micafungin, Caspofungin, Anidulafungin, and Rezafungin Population Pharmacokinetic Models and Dose Optimization in Special Populations”Clinical Pharmacokinetics64 (1): 27–52. doi:10.1007/s40262-024-01461-5PMC 11762474PMID 39707078.
    8. ^ Grasselli Kmet N, Luzzati R, Monticelli J, Babich S, Conti J, Bella SD (March 2025). “Salvage therapy of complicated Candida albicans spondylodiscitis with Rezafungin”. European Journal of Clinical Microbiology & Infectious Diseasesdoi:10.1007/s10096-025-05117-5PMID 40163284.
    9. ^ Viceconte G, Buonomo AR, Esposito N, Cattaneo L, Somma T, Scirocco MM, et al. (April 2024). “Salvage Therapy with Rezafungin for Candida parapsilosis Spondylodiscitis: A Case Report from Expanded Access Program”Microorganisms12 (5): 903. doi:10.3390/microorganisms12050903PMC 11123963PMID 38792732.
    10. ^ “Novel Drug Approvals for 2023”U.S. Food and Drug Administration (FDA). 22 December 2023. Retrieved 27 December 2023.
    11. ^ “Drug Approval Package: Rezzayo”U.S. Food and Drug Administration (FDA). 18 April 2023. Retrieved 27 December 2023.
    12. ^ New Drug Therapy Approvals 2023 (PDF). U.S. Food and Drug Administration (FDA) (Report). January 2024. Archived from the original on 10 January 2024. Retrieved 9 January 2024.
    13. ^ World Health Organization (2018). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 79”. WHO Drug Information32 (1). hdl:10665/330941.

    Clinical data
    Trade namesRezzayo
    Other namesBiafungin; CD101
    AHFS/Drugs.comMonograph
    MedlinePlusa623021
    License dataUS DailyMedRezafungin
    Routes of
    administration
    Intravenous
    Drug classAntifungal
    ATC codeJ02AX08 (WHO)
    Legal status
    Legal statusUS: ℞-only[1]EU: Rx-only[2][3]
    Pharmacokinetic data
    ExcretionFeces
    Identifiers
    CAS Number1396640-59-7
    PubChem CID78318119
    DrugBankDB16310
    UNIIG013B5478J
    KEGGD11197
    ChEBICHEBI:229680
    Chemical and physical data
    FormulaC63H85N8O17+
    Molar mass1226.412 g·mol−1
    1. Lamoth F: Novel Therapeutic Approaches to Invasive Candidiasis: Considerations for the Clinician. Infect Drug Resist. 2023 Feb 22;16:1087-1097. doi: 10.2147/IDR.S375625. eCollection 2023. [Article]
    2. Miesel L, Lin KY, Ong V: Rezafungin treatment in mouse models of invasive candidiasis and aspergillosis: Insights on the PK/PD pharmacometrics of rezafungin efficacy. Pharmacol Res Perspect. 2019 Nov 20;7(6):e00546. doi: 10.1002/prp2.546. eCollection 2019 Dec. [Article]
    3. Thompson GR 3rd, Soriano A, Cornely OA, Kullberg BJ, Kollef M, Vazquez J, Honore PM, Bassetti M, Pullman J, Chayakulkeeree M, Poromanski I, Dignani C, Das AF, Sandison T, Pappas PG: Rezafungin versus caspofungin for treatment of candidaemia and invasive candidiasis (ReSTORE): a multicentre, double-blind, double-dummy, randomised phase 3 trial. Lancet. 2023 Jan 7;401(10370):49-59. doi: 10.1016/S0140-6736(22)02324-8. Epub 2022 Nov 25. [Article]
    4. Ong V, Wills S, Watson D, Sandison T, Flanagan S: Metabolism, Excretion, and Mass Balance of [(14)C]-Rezafungin in Animals and Humans. Antimicrob Agents Chemother. 2022 Jan 18;66(1):e0139021. doi: 10.1128/AAC.01390-21. Epub 2021 Oct 18. [Article]
    5. FDA Approved Drug Products: REZZAYO (rezafungin) injection for intravenous use (March 2023) [Link]
    6. Globe News Wire: Cidara Therapeutics and Melinta Therapeutics Announce FDA Approval of REZZAYO (rezafungin for injection) for the Treatment of Candidemia and Invasive Candidiasis [Link]
    7. EMA Summary of Product Characteristics: REZZAYO (rezafungin) solution for infusion [Link]

    //////////Rezafungin, Rezzayo, APROVALS 2023, FDA 2023, Rezafungin ion, Rezafungin cation, CD 101, SP 3025, G013B5478J

    Omaveloxolone


    Omaveloxolone

    CAS
    1474034-05-3

    N-[(4aS,6aR,6bS,8aR,12aS,14aR,14bS)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl-10,14-dioxo-1,2,3,4,4a,5,6,6a,6b,7,8,8a,9,10,12a,14,14a,14b-octadecahydropicen-4a-yl]-2,2-difluoropropanamide

    N-[(4aS,6aR,6bS,8aR,12aS,14aR,14bS)-11-cyano-2,2,6a,6b,9,9,12a-heptamethyl-10,14-dioxo-1,3,4,5,6,7,8,8a,14a,14b-decahydropicen-4a-yl]-2,2-difluoropropanamide

    FDA 2023, 2/28/2023, To treat Friedrich’s ataxia
    Drug Trials Snapshot

    WeightAverage: 554.723
    Monoisotopic: 554.331999611

    Chemical FormulaC33H44F2N2O3

    • RTA 408
    • RTA-408
    • OriginatorDartmouth College; University of Texas M. D. Anderson Cancer Center
    • DeveloperBiogen
    • ClassAnalgesics; Anti-inflammatories; Antineoplastics; Eye disorder therapies; Neuroprotectants; Small molecules; Triterpenes
    • Mechanism of ActionNF-E2-related factor 2 stimulants
    • Orphan Drug StatusYes – Friedreich’s ataxia; Malignant melanoma
    • MarketedFriedreich’s ataxia
    • Phase IIMitochondrial disorders; Ocular inflammation; Ocular pain
    • Phase I/IIMalignant melanoma
    • PreclinicalBrain disorders
    • DiscontinuedDuchenne muscular dystrophy; Non-small cell lung cancer; Radiation-induced skin damage
    • 08 Apr 2025Biogen completes a phase I pharmacokinetics trial (In volunteers) in USA (PO) (NCT06612879)
    • 17 Mar 2025Registered for Friedreich’s ataxia (In adolescents, In adults) in Canada (PO)
    • 18 Oct 2024Biogen initiates enrolment in a phase I pharmacokinetics trial (In volunteers) in USA (PO) (NCT06612879)

    Omaveloxolone, sold under the brand name Skyclarys, is a medication used for the treatment of Friedreich’s ataxia.[2][5] It is taken by mouth.[2]

    The most common side effects include an increase in alanine transaminase and an increase of aspartate aminotransferase, which can be signs of liver damage, headache, nausea, abdominal pain, fatigue, diarrhea and musculoskeletal pain.[5]

    Omaveloxolone was approved for medical use in the United States in February 2023,[2][5][6][7][8] and in the European Union in February 2024.[3] The US Food and Drug Administration (FDA) considers it to be a first-in-class medication.[9]

    SYNTHESIS

    PATENT
    Sheikh, AY et al. (2018). Bardoxolonmethyl-2,2-difluoropropionamide derivatives, polymorphe forms and procedures for use thereof. DK/EP 2989114 T3. Danish Patent and Trademark Office. Available at https://patentimages.storage.googleapis.com/51/87/43/97d0fb3e69ee73/DK2989114T3.pdf

    https://patentscope.wipo.int/search/en/detail.jsf?docId=EP159939262&_cid=P21-MAKI10-93498-1

    [0164]  Reagents and conditions: (a) (PhO) 2PON 3 (DPPA), triethylamine, toluene, 0 °C for 5 minutes, then ambient temperature overnight, ∼94%; (b) benzene, 80 °C for 2 hours; (c) HCl, CH 3CN, ambient temperature for 1 hour; (d) CH 3CF 2CO 2H, dicyclohexylcarbodiimide, 4-(dimethylamino)pyridine, CH 2Cl 2, ambient temperature overnight, 73% from RTA 401 (4 steps).

    [0165]Compound 1: RTA 401 (20.0 g, 40.6 mmol), triethylamine (17.0 mL, 122.0 mmol), and toluene (400 mL) were added into a reactor and cooled to 0 °C with stirring. Diphenyl phosphoryl azide (DPPA) (13.2 mL, 61.0 mmol) was added with stirring at 0 °C over 5 minutes, and the mixture was continually stirred at room temperature overnight (HPLC-MS check shows no RTA 401 left). The reaction mixture was directly loaded on a silica gel column and purified by column chromatography (silica gel, 0% to 5% ethyl acetate in CH 2Cl 2) to give compound 1 (19.7 g, ∼94%, partially converted into compound 2) as a white foam.

    [0166]Compound 2: Compound 1 (19.7 g, ∼38.1 mmol) and benzene (250 mL) were added into a reactor and heated to 80 °C with stirring for 2 hours (HPLC-MS check shows no compound 1 left). The reaction mixture was concentrated at reduced pressure to afford crude compound 2 as a solid residue, which was used for the next step without purification.

    [0167]Compound 3: Crude compound 2 (≤38.1 mmol) and CH 3CN (200 mL) were added into a reactor and cooled to 0 °C with stirring. HCl (12 N, 90 mL) was added at 0 °C over 1 minute, and the mixture was continually stirred at room temperature for 1 hour (HPLC-MS check shows no compound 2 left). The reaction mixture was cooled to 0 °C and 10% NaOH (∼500 mL) was added with stirring. Then, saturated NaHCO 3 (1 L) was added with stirring. The aqueous phase was extracted by ethyl acetate (2×500 mL). The combined organic phase was washed by H 2O (200 mL), saturated NaCl (200 mL), dried over Na 2SO 4, and concentrated to afford crude compound 3 (16.62 g) as a light yellow foam, which was used for the next step without purification.

    [0168]RTA 408: Crude amine 3 (16.62 g, 35.9 mmol), CH 3CF 2CO 2H (4.7388 g, 43.1 mmol), and CH 2Cl 2 (360 mL) were added into a reactor with stirring at room temperature. Then, dicyclohexylcarbodiimide (DCC) (11.129 g, 53.9 mmol) and 4-(dimethylamino)pyridine (DMAP) (1.65 g, 13.64 mmol) were added and the mixture was continually stirred at room temperature overnight (HPLC-MS check shows no compound 3 left). The reaction mixture was filtered to remove solid by-products, and the filtrate was directly loaded on a silica gel column and purified by column chromatography (silica gel, 0% to 20% ethyl acetate in hexanes) twice to give compound RTA 408 (16.347 g, 73% from RTA 401 over 4 steps) as a white foam: 1H NMR (400 MHz, CD 3Cl) δ ppm 8.04 (s, 1H), 6.00 (s, 1H), 5.94 (s, br, 1H), 3.01 (d, 1H, J = 4.8 Hz), 2.75-2.82 (m, 1H), 1.92-2.18 (m, 4H), 1.69-1.85 (m, 7H), 1.53-1.64 (m, 1H), 1.60 (s, 3H), 1.50 (s, 3H), 1.42 (s, 3H), 1.11-1.38 (m, 3H), 1.27 (s, 3H), 1.18 (s, 3H), 1.06 (s, 3H), 1.04 (s, 3H), 0.92 (s, 3H); m/z 555 (M+1).

    SYNTHESIS
    J. Med. Chem. 2025, 68, 2147−2182

    Omaveloxolone (Skyclarys). Omaveloxolone (6) was approved in February 2023 for the treatment of Friedreich’s Ataxia (FRDA), a genetic, neurodegenerative disease. Patients with FRDA have lowered activity of the frataxin gene (FXN), attributed to an expansion of a guanine-adenine-adenine (GAA)
    triplet. The resulting decrease in frataxin limits the production of iron−sulfur clusters, leading to accumulation of iron in the mitochondria and oxidative stress which in turn leads to cell damageanddeath.49
    Omaveloxoloneactivates the nuclear factor erythroid 2-related factor 2 (Nrf2), an important pathway in
    oxidative stress. It acts by preventing ubiquitination and subsequent degradation of Nrf2, keeping levels high enough to counteract the oxidative stress associated with FRDA. 50
    Omaveloxolone was developed by Reata Pharmaceuticals (which was acquired by Biogen in September 2023) and was granted orphan drug, fast track, priority review, and rare pediatric disease designations. 51Omaveloxolone (6) is a semisynthetic triterpenoid based on the oleanolic acid scaffold.52
    advanced intermediate 6.1,The synthesis started from the53also known as CDDO orbardoxolone, which has individually been investigated fortherapeutic benefits from Nrf2 activation (Scheme 10).
    Treatment of acid 6.1 with DPPA produced the azide, and subsequent heating in benzene generated isocyanate 6.2 via aCurtius rearrangement. Hydrolysis with aqueous acid generated amine 6.3, and an amidation with 2,2-difluoropropanoic acid produced omaveloxolone (6). A yield of 73% over the sequence was reported, and intermediates were used crude with no purification between steps.

    (49) Ghanekar, S. D.; Miller, W. W.; Meyer, C. J.; Fenelon, K. J.;
    Lacdao, A.; Zesiewicz, T. A. Orphan drugs in development for the
    treatment of Friedreich’s ataxia: focus on omaveloxolone. Degener.
    Neurol. Neuromuscular Dis. 2019, 9, 103−107.
    (50) Abeti, R.; Baccaro, A.; Esteras, N.; Giunti, P. Novel Nrf2-inducer
    prevents mitochondrial defects and oxidative stress in Friedreich’s
    ataxia models. Front. Cell. Neurosci. 2018, 12, 188.
    (51) Lee,A.Omaveloxolone:first approval. Drugs 2023, 83, 725−729.
    (52) Anderson, E.; Decker, A.; Liu, X. Synthesis, pharmaceutical use,
    and characterization of crystalline forms of 2,2-difluoropropionamide
    derivatives of bardoxolone methyl. WO 2013163344, 2013.
    (53) Honda, T.; Rounds, B. V.; Gribble, G. W.; Suh, N.; Wang, Y.;
    Sporn, M. B. Design and synthesis of 2-cyano-3,12-dioxoolean-1,9
    dien-28-oic acid, a novel and highly active inhibitor of nitric oxide
    production in mouse macrophages. Bioorg. Med. Chem. Lett. 1998, 8,
    2711−2714.

    SYN

    European Journal of Medicinal Chemistry 265 (2024) 116124

    Omaveloxolone (Skyclarys)
    Omaveloxolone was granted FDA approval on February 28, 2023, to treat Friedrich’s ataxia in individuals aged 16 and older [2]. Omaveloxolone possesses antioxidant and anti-inflammatory properties, making it a semi-synthetic triterpenoid compound. It has the ability to function as a stimulator of nuclear factor-erythroid 2 related factor 2(Nrf2), a transcription factor that reduces oxidative stress. In individuals
    suffering from FA, a genetic disorder characterized by mitochondrial dysfunction, the Nrf2 pathway is compromised, leading to a decrease in Nrf2 activity. Hence, Omaveloxolone, an Nrf2 activator, can be
    employed as a therapeutic option for the management of these in dividuals [23].The process route of Omaveloxolone is described below in Scheme 724]. The substitution reaction of carboxylic acid OMAV-001 with diphenylphosphoryl azide (DPPA) gave the acyl azide OMAV-002,which underwent Curtius-rearrangement under heating conditions to produce isocyanate OMAV-003. The amine OMAV-004 was obtained under acidic conditions. OMAV-004 was condensed with 2,2-difluoro propionic acid to obtain the final product Omaveloxolone.

    [23] B.L. Probst, I. Trevino, L. McCauley, R. Bumeister, I. Dulubova, W.C. Wigley, D.
    A. Ferguson, RTA 408, A novel synthetic triterpenoid with broad anticancer and
    anti-inflammatory activity, PLoS One 10 (2015) e0122942.
    [24] E. Anderson, A. Decker, X. Liu Synthesis, Pharmaceutical Use, and
    Characterization of Crystalline Forms of 2,2-difluoropropionamide Derivatives of
    Bardoxolone Methyl, 2013. WO2013163344.

    .

    Medical uses

    Omaveloxolone is indicated for the treatment of Friedreich’s ataxia.[2][5]

    Friedreich’s ataxia causes progressive damage to the spinal cord, peripheral nerves, and the brain, resulting in uncoordinated muscle movement, poor balance, difficulty walking, changes in speech and swallowing, and a shortened lifespan.[5] The condition can also cause heart disease.[5] This disease tends to develop in children and teenagers and gradually worsens over time.[5]

    Although rare, Friedreich’s ataxia is the most common form of hereditary ataxia in the United States, affecting about one in every 50,000 people.[5]

    Mechanism of action

    The mechanism of action of omaveloxolone and its related compounds has been demonstrated to be through a combination of activation of the antioxidative transcription factor Nrf2 and inhibition of the pro-inflammatory transcription factor NF-κB.[10]

    Nrf2 transcriptionally regulates multiple genes that play both direct and indirect roles in producing antioxidative potential and the production of cellular energy (i.e., adenosine triphosphate or ATP) within the mitochondria. Consequently, unlike exogenously administered antioxidants (e.g.vitamin E or Coenzyme Q10), which provide a specific and finite antioxidative potential, omaveloxolone, through Nrf2, broadly activates intracellular and mitochondrial antioxidative pathways, in addition to pathways that may directly increase mitochondrial biogenesis (such as PGC1α) and bioenergetics.[11]

    History

    Omaveloxolone is a second generation member of the synthetic oleanane triterpenoid compounds and in clinical development by Reata PharmaceuticalsPreclinical studies have demonstrated that omaveloxolone possesses antioxidative and anti-inflammatory activities[10][12] and the ability to improve mitochondrial bioenergetics.[11] Omaveloxolone is under clinical investigation for a variety of indications, including Friedreich’s ataxiamitochondrial myopathiesimmunooncology, and prevention of corneal endothelial cell loss following cataract surgery.

    The efficacy and safety of omaveloxolone was evaluated in a 48-week randomized, placebo-controlled, and double-blind study [Study 1 (NCT02255435)] and an open-label extension.[5] Study 1 enrolled 103 individuals with Friedreich’s ataxia who received placebo (52 individuals) or omaveloxolone 150 mg (51 individuals) for 48 weeks.[5] Of the research participants, 53% were male, 97% were white, and the mean age was 24 years at study entry.[5] Nine (18%) patients were younger than age 18.[5] The primary objective was to evaluate the change in the modified Friedreich’s Ataxia Rating Scale (mFARS) score compared to placebo at week 48.[5] The mFARS is a clinical assessment that measures disease progression, namely swallowing and speech (bulbar), upper limb coordination, lower limb coordination, and upright stability.[5] Individuals receiving omaveloxolone performed better on the mFARS than people receiving placebo.[5]

    The US Food and Drug Administration (FDA) granted the application for omaveloxolone orphan drugfast trackpriority review, and rare pediatric disease designations.[5][9]

    Society and culture

    Omaveloxolone was approved for medical use in the United States in February 2023.[2][5]

    In December 2023, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency adopted a positive opinion, recommending the granting of a marketing authorization for the medicinal product Skyclarys, intended for the treatment of Friedreich’s ataxia.[3] The applicant for this medicinal product is Reata Ireland Limited.[3] Omaveloxolone was approved for medical use in the European Union in February 2024.[3][4]

    References

    1. ^ “Register of Innovative Drugs”Health Canada. 3 November 2006. Retrieved 17 April 2025.
    2. Jump up to:a b c d e f “Skyclarys- omaveloxolone capsule”DailyMed. 12 May 2023. Archived from the original on 1 July 2023. Retrieved 16 December 2023.
    3. Jump up to:a b c d e “Skyclarys EPAR”European Medicines Agency (EMA). 14 December 2023. Archived from the original on 15 December 2023. Retrieved 16 December 2023. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.
    4. Jump up to:a b “Skyclarys product information”Union Register of medicinal products. 12 February 2024. Retrieved 19 February 2024.
    5. Jump up to:a b c d e f g h i j k l m n o p q “FDA approves first treatment for Friedreich’s ataxia”U.S. Food and Drug Administration (FDA). 28 February 2023. Archived from the original on 1 March 2023. Retrieved 28 February 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    6. ^ “Reata Pharmaceuticals Announces FDA Approval of Skyclarys (Omavaloxolone), the First and Only Drug Indicated for Patients with Friedreich’s Ataxia”Reata Pharmaceuticals Inc. (Press release). 28 February 2023. Archived from the original on 1 March 2023. Retrieved 28 February 2023.
    7. ^ Lee A (June 2023). “Omaveloxolone: First Approval”Drugs83 (8): 725–729. doi:10.1007/s40265-023-01874-9PMID 37155124S2CID 258567442Archived from the original on 9 December 2023. Retrieved 16 December 2023.
    8. ^ Subramony SH, Lynch DL (May 2023). “A Milestone in the Treatment of Ataxias: Approval of Omaveloxolone for Friedreich Ataxia”. Cerebellum23 (2): 775–777. doi:10.1007/s12311-023-01568-8PMID 37219716S2CID 258843532.
    9. Jump up to:a b New Drug Therapy Approvals 2023 (PDF). U.S. Food and Drug Administration (FDA) (Report). January 2024. Archived from the original on 10 January 2024. Retrieved 9 January 2024.
    10. Jump up to:a b Reisman SA, Lee CY, Meyer CJ, Proksch JW, Ward KW (July 2014). “Topical application of the synthetic triterpenoid RTA 408 activates Nrf2 and induces cytoprotective genes in rat skin”. Archives of Dermatological Research306 (5): 447–454. doi:10.1007/s00403-013-1433-7PMID 24362512S2CID 25733020.
    11. Jump up to:a b Neymotin A, Calingasan NY, Wille E, Naseri N, Petri S, Damiano M, et al. (July 2011). “Neuroprotective effect of Nrf2/ARE activators, CDDO ethylamide and CDDO trifluoroethylamide, in a mouse model of amyotrophic lateral sclerosis”Free Radical Biology & Medicine51 (1): 88–96. doi:10.1016/j.freeradbiomed.2011.03.027PMC 3109235PMID 21457778.
    12. ^ Reisman SA, Lee CY, Meyer CJ, Proksch JW, Sonis ST, Ward KW (May 2014). “Topical application of the synthetic triterpenoid RTA 408 protects mice from radiation-induced dermatitis”Radiation Research181 (5): 512–520. Bibcode:2014RadR..181..512Rdoi:10.1667/RR13578.1PMID 24720753S2CID 23906747.

    Clinical trial number NCT02255435 for “RTA 408 Capsules in Patients With Friedreich’s Ataxia – MOXIe” at ClinicalTrials.gov

    Clinical data
    Trade namesSkyclarys
    Other namesRTA 408
    AHFS/Drugs.comMonograph
    License dataUS DailyMedOmaveloxolone
    Routes of
    administration
    By mouth
    ATC codeN07XX25 (WHO)
    Legal status
    Legal statusCA℞-only[1]US: ℞-only[2]EU: Rx-only[3][4]
    Identifiers
    showIUPAC name
    CAS Number1474034-05-3 
    PubChem CID71811910
    IUPHAR/BPS7573
    DrugBankDB12513
    ChemSpider34980948 
    UNIIG69Z98951Q
    KEGGD10964
    ChEBICHEBI:229661
    CompTox Dashboard (EPA)DTXSID101138251 
    Chemical and physical data
    FormulaC33H44F2N2O3
    Molar mass554.723 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI
    1. Zesiewicz TA, Hancock J, Ghanekar SD, Kuo SH, Dohse CA, Vega J: Emerging therapies in Friedreich’s Ataxia. Expert Rev Neurother. 2020 Dec;20(12):1215-1228. doi: 10.1080/14737175.2020.1821654. Epub 2020 Sep 21. [Article]
    2. Jiang Z, Qi G, Lu W, Wang H, Li D, Chen W, Ding L, Yang X, Yuan H, Zeng Q: Omaveloxolone inhibits IL-1beta-induced chondrocyte apoptosis through the Nrf2/ARE and NF-kappaB signalling pathways in vitro and attenuates osteoarthritis in vivo. Front Pharmacol. 2022 Sep 27;13:952950. doi: 10.3389/fphar.2022.952950. eCollection 2022. [Article]
    3. Shekh-Ahmad T, Eckel R, Dayalan Naidu S, Higgins M, Yamamoto M, Dinkova-Kostova AT, Kovac S, Abramov AY, Walker MC: KEAP1 inhibition is neuroprotective and suppresses the development of epilepsy. Brain. 2018 May 1;141(5):1390-1403. doi: 10.1093/brain/awy071. [Article]
    4. Probst BL, Trevino I, McCauley L, Bumeister R, Dulubova I, Wigley WC, Ferguson DA: RTA 408, A Novel Synthetic Triterpenoid with Broad Anticancer and Anti-Inflammatory Activity. PLoS One. 2015 Apr 21;10(4):e0122942. doi: 10.1371/journal.pone.0122942. eCollection 2015. [Article]
    5. Lynch DR, Farmer J, Hauser L, Blair IA, Wang QQ, Mesaros C, Snyder N, Boesch S, Chin M, Delatycki MB, Giunti P, Goldsberry A, Hoyle C, McBride MG, Nachbauer W, O’Grady M, Perlman S, Subramony SH, Wilmot GR, Zesiewicz T, Meyer C: Safety, pharmacodynamics, and potential benefit of omaveloxolone in Friedreich ataxia. Ann Clin Transl Neurol. 2018 Nov 10;6(1):15-26. doi: 10.1002/acn3.660. eCollection 2019 Jan. [Article]
    6. Zighan M, Arkadir D, Douiev L, Keller G, Miller C, Saada A: Variable effects of omaveloxolone (RTA408) on primary fibroblasts with mitochondrial defects. Front Mol Biosci. 2022 Aug 12;9:890653. doi: 10.3389/fmolb.2022.890653. eCollection 2022. [Article]
    7. FDA Approved Drug Products: SKYCLARYS (omaveloxolone) capsules for oral use (February 2023) [Link]
    8. EMA Approved Drug Products: Skyclarys (omaveloxolone) Oral Capsules [Link]
    9. Health Canada Approved Drug Products: SKYCLARYS (Omaveloxolone) Capsules For Oral Use [Link]

    ///////////Omaveloxolone, Skyclarys, Friedrich’s ataxia, FDA 2023, APPROVALS 2023, RTA 408, RTA-408, omaveloxolona, RTA 408, 63415, PP415, orphan drugfast trackpriority review, rare pediatric disease

    str1

    AS ON JUNE2025 4.45 LAKHS VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@gmail.com

    ……

    Elacestrant 


    Elacestrant 

    (6R)-6-[2-[ethyl-[[4-[2-(ethylamino)ethyl]phenyl]methyl]amino]-4-methoxyphenyl]-5,6,7,8-tetrahydronaphthalen-2-ol

    (6R)-6-{2-[ethyl({4-[2-(ethylamino)ethyl]phenyl}methyl)amino]-4-methoxyphenyl}-5,6,7,8-tetrahydronaphthalen-2-ol

    FDA 1/27/2023, Orserdu

    WeightAverage: 458.646
    Monoisotopic: 458.293328472

    Chemical FormulaC30H38N2O2

    To treat estrogen receptor-positive, human epidermal growth factor receptor 2-negative, ESR1-mutated, advanced or metastatic breast cancer with disease progression following at least one line of endocrine therapy
    Drug Trials Snapshot

    Elacestrant, sold under the brand name Orserdu, is a selective estrogen receptor degrader (SERD) used in the treatment of breast cancer.[1][4] It is taken by mouth.[1][4]

    Elacestrant is an antiestrogen that acts as an antagonist of estrogen receptors, which are the biological targets of endogenous estrogens like estradiol.[1] The most common side effects of elacestrant include body pain, nausea and vomiting, increased serum lipids, elevated liver enzymes, fatigue, decreased hemoglobin, raised creatinine, decreased appetite, diarrhea, headache, constipation, abdominal pain, and hot flashes.[2]

    Elacestrant was approved for medical use in the United States in January 2023,[1][2][5][6] and in the European Union in September 2023.[3][7]

    PATENTS


    Cruskie MP, et al. (2019). Polymorphic forms of RAD1901-2HCl (U.S. Patent No. 10,385,008 B2). U.S. Patent and Trademark Office. https://patentimages.storage.googleapis.com/42/82/b6/e9fcbbbd08054e/US10385008.pdf

    Patent NumberPediatric ExtensionApprovedExpires (estimated)
    US10071066No2018-09-112034-10-10US flag
    US10385008No2019-08-202038-01-05US flag
    US10420734No2019-09-242034-10-10US flag
    US10745343No2020-08-182038-01-05US flag
    US11779552No2023-10-102034-10-10US flag
    US11819480No2023-11-212036-11-29US flag
    US7612114No2009-11-032026-08-18US flag
    US8399520No2013-03-192023-12-25US flag

    PATENT

    https://patents.google.com/patent/US10385008B2/en

    Medical uses

    Elacestrant is indicated for the treatment of postmenopausal women or adult men with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative, ESR1mutatedadvanced or metastatic breast cancer with disease progression following at least one other line of endocrine therapy.[2][4]

    Pharmacology

    Pharmacodynamics

    Elacestrant is an antiestrogen that acts as an antagonist of estrogen receptors, specifically targeting the estrogen receptor alpha (ERα), which is the biological target of endogenous estrogens like estradiol.[1] Additionally, elacestrant is a selective estrogen receptor degrader (SERD), meaning it induces the degradation of ERα.[1][8]

    Pharmacokinetics

    Elacestrant has an oral bioavailability of approximately 10%.[1] Its plasma protein binding exceeds 99% and remains independent of concentration.[1] Elacestrant is metabolized in the liver, primarily by the cytochrome P450 enzyme CYP3A4 and to a lesser extent by CYP2A6 and CYP2C9.[1] The elimination half-life of elacestrant is 30 to 50 hours.[1] It is excreted primarily in feces (82%) and to a lesser extent in urine (7.5%).[1]

    History

    The efficacy of elacestrant was evaluated in the EMERALD trial, which was a randomized, open-label, active-controlled, multicenter study involving 478 postmenopausal women and men with ER-positive, HER2-negative advanced or metastatic breast cancer. Among them, 228 participants had ESR1 mutations. Eligible participants had experienced disease progression on one or two prior lines of endocrine therapy, including one line with a CDK4/6 inhibitor, and could have received up to one prior line of chemotherapy in the advanced or metastatic setting.[2]

    Participants were randomly assigned in a 1:1 ratio to receive either elacestrant 345 mg orally once daily or investigator’s choice of endocrine therapy. The choices for the control arm included fulvestrant, or an aromatase inhibitor. Randomization was stratified based on whether the ESR1 mutation was detected or not, prior treatment with fulvestrant, and presence of visceral metastasis.[2]

    The FDA granted the application for elacestrant priority review and fast track designations.[2]

    Research

    It is a nonsteroidal combined selective estrogen receptor modulator (SERM) and selective estrogen receptor degrader (SERD) (described as a “SERM/SERD hybrid (SSH)”) that was discovered by Eisai and is under development by Radius Health and Takeda for the treatment estrogen receptor (ER)-positive advanced breast cancer.[9] Elacestrant has dose-dependent, tissue-selective estrogenic and antiestrogenic activities, with biphasic weak partial agonist activity at the ER at low doses and antagonist activity at higher doses.[10] It shows agonistic activity on bone and antagonistic activity on breast and uterine tissues.[11] Unlike the SERD fulvestrant, elacestrant is able to readily cross the blood-brain-barrier into the central nervous system, where it can target breast cancer metastases in the brain,[10][11] and is orally bioavailable and does not require intramuscular injection.[10][11]

    References

    1. Jump up to:a b c d e f g h i j k l m n o p q “Orserdu- elacestrant tablet, film coated”DailyMed. 8 February 2023. Archived from the original on 11 February 2023. Retrieved 11 February 2023.
    2. Jump up to:a b c d e f g “FDA approves elacestrant for ER-positive, HER2-negative, ESR1-mutated advanced or metastatic breast cancer”U.S. Food and Drug Administration (FDA). 27 January 2023. Archived from the original on 2 February 2023. Retrieved 1 February 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    3. Jump up to:a b “Orserdu Product information”Union Register of medicinal products. 18 September 2023. Retrieved 1 October 2023.
    4. Jump up to:a b c d “Orserdu EPAR”European Medicines Agency (EMA). 9 October 2023. Retrieved 9 October 2023.
    5. ^ https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2023/217639Orig1s000ltr.pdf Archived 2023-02-02 at the Wayback Machine Public Domain This article incorporates text from this source, which is in the public domain.
    6. ^ “Stemline Therapeutics Inc., a wholly owned subsidiary of Menarini Group, Receives Approval from U.S. FDA for Orserdu (elacestrant) as the First and Only Treatment Specifically Indicated for Patients with ESR1 Mutations in ER+, HER2- Advanced or Metastatic Breast Cancer”Radius (Press release). 31 January 2023. Archived from the original on 2 February 2023. Retrieved 1 February 2023.
    7. ^ “EC approves Menarini Group’s Orserdu for advanced or metastatic breast cancer”PMLive. 21 September 2023. Retrieved 22 September 2023.
    8. ^ Lloyd MR, Wander SA, Hamilton E, Razavi P, Bardia A (2022). “Next-generation selective estrogen receptor degraders and other novel endocrine therapies for management of metastatic hormone receptor-positive breast cancer: current and emerging role”Therapeutic Advances in Medical Oncology14: 17588359221113694. doi:10.1177/17588359221113694PMC 9340905PMID 35923930.
    9. ^ Clinical trial number NCT03778931 for “Phase 3 Trial of Elacestrant vs. Standard of Care for the Treatment of Patients With ER+/HER2- Advanced Breast Cancer” at ClinicalTrials.gov
    10. Jump up to:a b c Wardell SE, Nelson ER, Chao CA, Alley HM, McDonnell DP (October 2015). “Evaluation of the pharmacological activities of RAD1901, a selective estrogen receptor degrader”Endocrine-Related Cancer22 (5): 713–724. doi:10.1530/ERC-15-0287PMC 4545300PMID 26162914.
    11. Jump up to:a b c Garner F, Shomali M, Paquin D, Lyttle CR, Hattersley G (October 2015). “RAD1901: a novel, orally bioavailable selective estrogen receptor degrader that demonstrates antitumor activity in breast cancer xenograft models”Anti-Cancer Drugs26 (9): 948–956. doi:10.1097/CAD.0000000000000271PMC 4560273PMID 26164151.
    Clinical data
    Pronunciation/ˌɛləˈsɛstrənt/
    EL-ə-SES-trənt
    Trade namesOrserdu
    Other namesRAD-1901; ER-306323
    License dataUS DailyMedElacestrant
    Routes of
    administration
    By mouth
    ATC codeL02BA04 (WHO)
    Legal status
    Legal statusUS: ℞-only[1][2]EU: Rx-only[3][4]
    Pharmacokinetic data
    Bioavailability~10%[1]
    Protein binding>99%[1]
    MetabolismLiver (major: CYP3A4, minor: CYP2A6CYP2C9)[1]
    Elimination half-life30–50 hours[1]
    ExcretionFeces (82%), urine (7.5%)[1]
    Identifiers
    showIUPAC name
    CAS Number722533-56-4
    PubChem CID23642301
    DrugBankDB06374
    ChemSpider57583807
    UNIIFM6A2627A8
    KEGGD11671
    ChEMBLChEMBL4297509
    PDB ligandI0V (PDBeRCSB PDB)
    CompTox Dashboard (EPA)DTXSID901045846 
    ECHA InfoCard100.312.890 
    Chemical and physical data
    FormulaC30H38N2O2
    Molar mass458.646 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI

    /////////Elacestrant, Orserdu, FDA 2023, APPROVALS 2023, FM6A2627A8, WHO 10247, ER 306323, RAD 1901, RAD1901

    Pirtobrutinib


    Pirtobrutinib

    • CAS 2101700-15-4
    • JAYPIRCA
    • RXC-005
    • LY3527727
    • LOXO-305
    • WHO 11681
    • WeightAverage: 479.436
    • Monoisotopic: 479.158052208
    • Chemical FormulaC22H21F4N5O3

    5-amino-3-[4-[[(5-fluoro-2-methoxybenzoyl)amino]methyl]phenyl]-1-[(2S)-1,1,1-trifluoropropan-2-yl]pyrazole-4-carboxamide

    FDA 2023, 1/27/2023, Jaypirca

    To treat relapsed or refractory mantle cell lymphoma in adults who have had at least two lines of systemic therapy, including a BTK inhibitor
    Drug Trials Snapshot

    Pirtobrutinib, sold under the brand name Jaypirca, is an anticancer medication that is used to treat mantle cell lymphoma.[1][2][4] It inhibits B cell lymphocyte proliferation and survival by binding and inhibiting Bruton’s tyrosine kinase (BTK).[5] It is taken by mouth.[1]

    The most common adverse reactions include fatigue, musculoskeletal pain, diarrhea, edema, dyspnea, pneumonia, and bruising.[4][6] The most common adverse reactions when used to treat chronic lymphocytic leukemia or small lymphocytic leukemia include fatigue, bruising, cough, musculoskeletal pain, COVID-19, diarrhea, pneumonia, abdominal pain, dyspnea, hemorrhage, edema, nausea, pyrexia, and headache.[7]

    Pirtobrutinib was approved for medical use in the United States in January 2023,[4][8][9][10] and in the European Union in November 2023.[2]

    PATENTS

    Guisot, N. (2017). Compounds useful as kinase inhibitors (WO 2017/103611 A1). World Intellectual Property Organization. https://patentimages.storage.googleapis.com/d7/16/21/9300e49071a21a/WO2017103611A1.pdf

    https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017103611&_cid=P10-MAG7OA-80884-1

    [00381] Example 120: 5-amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]-1- (2,2,2-trifluoro-1 -methyl-ethyl)pyrazole-4-carboxamide

    N-[(2,2,2-Trifluoro-1-methyl-ethylidene)aminolbenzamide

    General procedure S, benzhydrazide (49.9 mmol) and 1,1,1- trifluoroacetone (74.9 mmol) gave, after washing, the titled compound as a white solid. UPLC-MS (ES + , Short acidic): 1.45 min, m/z 230.9 [M+H] +

    -Amino-3-[4-[[(5-fluoro-2-methoxy-benzoyl)amino]methyl]phenyl]-1-(2,2,2-trifluoro-1-methyl-ethyl)pyrazole-4-carboxamide

    General procedure M, N-[[4-[5-amino-4-cyano-1-(2,2,2-trifluoro-1-methyl-ethyl)pyrazol-3-yl]phenyl]methyl]-5-fluoro-2-methoxy-benzamide (0.83 mmol) gave, after purification, the titled compound (0.42 mmol) as a white solid. UPLC-MS (ES + , Short acidic): 1.55 min, m/z 480.1 [M+H] + . UPLC-MS (ES + , Long acidic): 3.57 min, m/z 480.1 [M+H] + . 1 H NMR (400 MHz, DMSO-d 6 , δ): 8.84 (t, J = 6.1 Hz, 1H), 7.52 (dd, J = 9.2, 3.3 Hz, 1H), 7.48-7.41 (m, 4H), 7.37-7.32 (m, 1H), 7.19 (dd, J = 9.1, 4.3 Hz, 1H), 6.67 (s, 2H), 5.35-5.24 (m, 1H), 4.56 (d, J = 6.0 Hz, 2H), 3.90 (s, 3H), 1.62 (d, J = 6.9 Hz, 3H).

    MORE

    Medical uses

    In the United States, pirtobrutinib is indicated to treat relapsed or refractory mantle cell lymphoma after at least two lines of systemic therapy, including a Bruton’s tyrosine kinase (BTK) inhibitor.[1][11] In December 2023, the US Food and Drug Administration (FDA) expanded the indication for pirtobrutinib to include the treatment of adults with chronic lymphocytic leukemia or small lymphocytic leukemia.[7][12]

    In the European Union, pirtobrutinib is indicated for the treatment of mantle cell lymphoma.[2]

    Mechanism of action

    B cells are white cells of the lymphocyte subtype that produce antibodies, but when some of them grow uncontrollably they can be a cause of cancer. A key enzyme in B cell stimulation and survival is BTK, and pirtobrutinib inhibits BTK in a way that is different from the prototypical BTK inhibitor ibrutinib by binding in a different way that avoids a genetic change (mutation at active site cysteine residue C481 in BTK) that can make some tumors less responsive to ibrutinib.[5]

    History

    Pirtobrutinib is manufactured by Eli Lilly and Company and was approved by the US Food and Drug Administration in January 2023, for the treatment of mantle cell lymphoma that has become refractory to other BTK inhibitors.[13]

    Efficacy was evaluated in BRUIN (NCT03740529), an open-label, multicenter, single-arm trial of pirtobrutinib monotherapy that included 120 participants with mantle cell lymphoma previously treated with a Bruton’s tyrosine kinase (BTK) inhibitor.[4] Participants had a median of three prior lines of therapy, with 93% having two or more prior lines.[4] The most common prior Bruton’s tyrosine kinase inhibitors received were ibrutinib (67%), acalabrutinib (30%), and zanubrutinib (8%); 83% had discontinued their last Bruton’s tyrosine kinase inhibitor due to refractory or progressive disease.[4] The trial was conducted at 49 sites in 10 countries in the United States, Europe, Australia, and Asia.[6] The same trial was used to assess safety and efficacy.[6]

    Efficacy was evaluated in BRUIN (NCT03740529], an open-label, international, single-arm, multicohort trial that included 108 participants with chronic lymphocytic leukemia or small lymphocytic lymphoma previously treated with at least two prior lines of therapy, including a Bruton’s tyrosine kinase (BTK) inhibitor and a B-cell lymphoma-2 (BCL-2) inhibitor.[7] Participants received a median of five prior lines of therapy (range: 2 to 11).[7] Seventy-seven percent of participants discontinued the last BTK inhibitor for refractory or progressive disease.[7] Pirtobrutinib was administered orally at 200 mg once daily and was continued until disease progression or unacceptable toxicity.[7]

    Society and culture

    In April 2023, the Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency (EMA) adopted a positive opinion, recommending the granting of a conditional marketing authorization for the medicinal product Jaypirca, intended for the treatment of relapsed or refractory mantle cell lymphoma (MCL).[14] The applicant for this medicinal product is Eli Lilly Nederland B.V.[14] Pirtobrutinib was approved for medical use in the European Union in November 2023.[2]

    References

    1. Jump up to:a b c d “Jaypirca- pirtobrutinib tablet, coated”DailyMed. 27 January 2023. Archived from the original on 11 February 2023. Retrieved 11 February 2023.
    2. Jump up to:a b c d e “Jaypirca EPAR”European Medicines Agency (EMA). 20 November 2023. Archived from the original on 22 November 2023. Retrieved 22 November 2023.
    3. ^ “Jaypirca Product information”Union Register of medicinal products. 31 October 2023. Archived from the original on 22 November 2023. Retrieved 22 November 2023.
    4. Jump up to:a b c d e f “FDA grants accelerated approval to pirtobrutinib for relapsed or refractory mantle cell lymphoma”. FDA. 27 January 2023. Archived from the original on 28 January 2023. Retrieved 28 January 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    5. Jump up to:a b Aslan B, Kismali G, Iles LR, Manyam GC, Ayres ML, Chen LS, et al. (May 2022). “Pirtobrutinib inhibits wild-type and mutant Bruton’s tyrosine kinase-mediated signaling in chronic lymphocytic leukemia”Blood Cancer Journal12 (5): 80. doi:10.1038/s41408-022-00675-9PMC 9123190PMID 35595730.
    6. Jump up to:a b c “Drug Trials Snapshots: Jaypirca”U.S. Food and Drug Administration (FDA). 27 January 2023. Retrieved 13 May 2024.
    7. Jump up to:a b c d e f “FDA grants accelerated approval to pirtobrutinib for chronic lymphocytic leukemia and small lymphocytic lymphoma”U.S. Food and Drug Administration (FDA). 1 December 2023. Archived from the original on 3 December 2023. Retrieved 3 December 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    8. ^ “U.S. FDA Approves Jaypirca (pirtobrutinib), the First and Only Non-Covalent (Reversible) BTK Inhibitor, for Adult Patients with Relapsed or Refractory Mantle Cell Lymphoma After at Least Two Lines of Systemic Therapy, Including a BTK Inhibitor” (Press release). Eli Lilly. 27 January 2023. Archived from the original on 30 January 2023. Retrieved 31 January 2023 – via PR Newswire.
    9. ^ Keam SJ (April 2023). “Pirtobrutinib: First Approval”Drugs83 (6): 547–553. doi:10.1007/s40265-023-01860-1PMID 37004673S2CID 257912433Archived from the original on 19 November 2023. Retrieved 19 November 2023.
    10. ^ Telaraja D, Kasamon YL, Collazo JS, Leong R, Wang K, Li P, et al. (August 2023). “FDA Approval Summary: Pirtobrutinib for Relapsed or Refractory Mantle Cell Lymphoma”Clinical Cancer Research30 (1): OF1 – OF6. doi:10.1158/1078-0432.CCR-23-1272PMC 10841293PMID 37624619S2CID 265965744.
    11. ^ De SK (October 2023). “Pirtobrutinib: First Non-covalent Tyrosine Kinase Inhibitor for Treating Relapsed or Refractory Mantle Cell Lymphoma in Adults”. Current Medicinal Chemistry31doi:10.2174/0109298673251030231004052822PMID 37818564S2CID 263828536.
    12. ^ “Jaypirca (pirtobrutinib) Now Approved by U.S. FDA for the Treatment of Adult Patients with Chronic Lymphocytic Leukemia or Small Lymphocytic Lymphoma Who Have Received at Least Two Lines of Therapy, Including a BTK Inhibitor and a BCL-2 Inhibitor” (Press release). Eli Lilly. 1 December 2023. Archived from the original on 3 December 2023. Retrieved 3 December 2023 – via PR Newswire.
    13. ^ “FDA approves Eli Lilly’s drug for rare blood cancer”Reuters. 27 January 2023. Archived from the original on 28 January 2023.
    14. Jump up to:a b “Jaypirca: Pending EC decision”European Medicines Agency. 26 April 2023. Archived from the original on 26 April 2023. Retrieved 27 April 2023. Text was copied from this source which is copyright European Medicines Agency. Reproduction is authorized provided the source is acknowledged.

    Further reading

    Clinical data
    Trade namesJaypirca
    Other namesLOXO-305
    AHFS/Drugs.comMonograph
    MedlinePlusa623012
    License dataUS DailyMedPirtobrutinib
    Routes of
    administration
    By mouth
    Drug classProtein kinase inhibitor
    ATC codeL01EL05 (WHO)
    Legal status
    Legal statusUS: ℞-only[1]EU: Rx-only[2][3]
    Identifiers
    showIUPAC name
    CAS Number2101700-15-4
    PubChem CID129269915
    DrugBankDB17472
    ChemSpider114875989
    UNIIJNA39I7ZVB
    KEGGD12050
    ChEBICHEBI:229212
    ChEMBLChEMBL4650485
    PDB ligandY7W (PDBeRCSB PDB)
    Chemical and physical data
    FormulaC22H21F4N5O3
    Molar mass479.436 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI
    1. Jensen JL, Mato AR, Pena C, Roeker LE, Coombs CC: The potential of pirtobrutinib in multiple B-cell malignancies. Ther Adv Hematol. 2022 Jun 16;13:20406207221101697. doi: 10.1177/20406207221101697. eCollection 2022. [Article]
    2. Aslan B, Kismali G, Iles LR, Manyam GC, Ayres ML, Chen LS, Gagea M, Bertilaccio MTS, Wierda WG, Gandhi V: Pirtobrutinib inhibits wild-type and mutant Bruton’s tyrosine kinase-mediated signaling in chronic lymphocytic leukemia. Blood Cancer J. 2022 May 20;12(5):80. doi: 10.1038/s41408-022-00675-9. [Article]
    3. Alu A, Lei H, Han X, Wei Y, Wei X: BTK inhibitors in the treatment of hematological malignancies and inflammatory diseases: mechanisms and clinical studies. J Hematol Oncol. 2022 Oct 1;15(1):138. doi: 10.1186/s13045-022-01353-w. [Article]
    4. Mato AR, Shah NN, Jurczak W, Cheah CY, Pagel JM, Woyach JA, Fakhri B, Eyre TA, Lamanna N, Patel MR, Alencar A, Lech-Maranda E, Wierda WG, Coombs CC, Gerson JN, Ghia P, Le Gouill S, Lewis DJ, Sundaram S, Cohen JB, Flinn IW, Tam CS, Barve MA, Kuss B, Taylor J, Abdel-Wahab O, Schuster SJ, Palomba ML, Lewis KL, Roeker LE, Davids MS, Tan XN, Fenske TS, Wallin J, Tsai DE, Ku NC, Zhu E, Chen J, Yin M, Nair B, Ebata K, Marella N, Brown JR, Wang M: Pirtobrutinib in relapsed or refractory B-cell malignancies (BRUIN): a phase 1/2 study. Lancet. 2021 Mar 6;397(10277):892-901. doi: 10.1016/S0140-6736(21)00224-5. [Article]
    5. Wang E, Mi X, Thompson MC, Montoya S, Notti RQ, Afaghani J, Durham BH, Penson A, Witkowski MT, Lu SX, Bourcier J, Hogg SJ, Erickson C, Cui D, Cho H, Singer M, Totiger TM, Chaudhry S, Geyer M, Alencar A, Linley AJ, Palomba ML, Coombs CC, Park JH, Zelenetz A, Roeker L, Rosendahl M, Tsai DE, Ebata K, Brandhuber B, Hyman DM, Aifantis I, Mato A, Taylor J, Abdel-Wahab O: Mechanisms of Resistance to Noncovalent Bruton’s Tyrosine Kinase Inhibitors. N Engl J Med. 2022 Feb 24;386(8):735-743. doi: 10.1056/NEJMoa2114110. [Article]
    6. FDA Approved Drug Products: JAYPIRCA (pirtobrutinib) tablets for oral use [Link]
    7. BioSpace: U.S. FDA Approves Jaypirca (pirtobrutinib), the First and Only Non-Covalent (Reversible) BTK Inhibitor, for Adult Patients with Relapsed or Refractory Mantle Cell Lymphoma After at Least Two Lines of Systemic Therapy, Including a BTK Inhibitor [Link]

    //////////////Jaypirca, FDA 2023, APPROVALS 2023, Pirtobrutinib, RXC-005, LY3527727, LOXO-305, LOXO 305, WHO 11681

    Capivasertib


    Capivasertib.png

    Capivasertib

    C21H25ClN6O2

     428.915

    • 1143532-39-1

    AZD 5363

    4-amino-N-[(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide

    (S)-4-AMINO-N-(1-(4-CHLOROPHENYL)-3-HYDROXYPROPYL)-1-(7H-PYRROLO[2,3-D]PYRIMIDIN-4-YL)PIPERIDINE-4-CARBOXAMIDE

    FDA APPROVED 11/16/2023, To treat breast cancer that meets certain disease criteria, Truqap

    Capivasertib, sold under the brand name Truqap, is an anti-cancer medication used for the treatment of breast cancer.[1][2]

    The most common adverse reactions include diarrhea, cutaneous adverse reactions, increased random glucose, decreased lymphocytes, decreased hemoglobin, increased fasting glucose, nausea, fatigue, decreased leukocytes, increased triglycerides, decreased neutrophils, increased creatinine, vomiting, and stomatitis.[3]

    In November 2023, capivasertib was approved in the United States for people with hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer when used in combination with fulvestrant.[3][4][5]

    Capivasertib is a novel pyrrolopyrimidine derivative, and an orally available inhibitor of the serine/threonine protein kinase AKT (protein kinase B) with potential antineoplastic activity. Capivasertib binds to and inhibits all AKT isoforms. Inhibition of AKT prevents the phosphorylation of AKT substrates that mediate cellular processes, such as cell division, apoptosis, and glucose and fatty acid metabolism. A wide range of solid and hematological malignancies show dysregulated PI3K/AKT/mTOR signaling due to mutations in multiple signaling components. By targeting AKT, the key node in the PIK3/AKT signaling network, this agent may be used as monotherapy or combination therapy for a variety of human cancers.

    Medical uses

    Capivasertib, used in combination with fulvestrant (Faslodex), is indicated for adults with hormone receptor-positive, human epidermal growth factor receptor 2-negative locally advanced or metastatic breast cancer with one or more PIK3CA/AKT1/PTEN-alterations, as detected by an FDA-approved test, following progression on at least one endocrine-based regimen in the metastatic setting or recurrence on or within twelve months of completing adjuvant therapy.[1][3]

    History

    Efficacy was evaluated in CAPItello-291 (NCT04305496), a randomized, double-blind, placebo-controlled, multicenter trial in 708 participants with locally advanced or metastatic HR-positive, HER2-negative breast cancer, of which 289 participants had tumors with PIK3CA/AKT1/PTEN-alterations.[3] All participants were required to have progression on aromatase inhibitor-based treatment.[3] Participants could have received up to two prior lines of endocrine therapy and up to one line of chemotherapy for locally advanced or metastatic disease.[3]

    PATENT

    US10654855,

    EXAMPLE 9: (S)-4-AMINO-N-(1-(4-CHLOROPHENYL)-3-HYDROXYPROPYL)-1-(7H-PYRROLO[2,3-D]PYRIMIDIN-4-YL)PIPERIDINE-4-CARBOXAMIDE (E9)


    (MOL)(CDX)
          HCl (4M in Dioxane) (3.00 mL, 12.00 mmol) was added to (S)-tert-butyl 4-(1-(4-chlorophenyl)-3-hydroxypropylcarbamoyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidin-4-ylcarbamate (Intermediate 22) (1.27 g, 2.40 mmol) in dichloromethane (20 mL). The resulting suspension was stirred at 20° C. for 16 hours. The reaction mixture was filtered through a PTFE filter cup and the crude solid was purified by preparative HPLC (Waters XTerra C18 column, 5 μm silica, 19 mm diameter, 100 mm length), using decreasingly polar mixtures of water (containing 1% TFA) and MeCN as eluents. Fractions containing the desired compound were purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 7M NH 3/MeOH and pure fractions were evaporated to dryness to afford (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (0.200 g, 19.4%) as a white solid. 1H NMR (399.9 MHz, DMSO-d6) δ 1.45 (2H, d), 1.86 (1H, d), 1.90-1.93 (1H, m), 2.19 (2H, s), 3.38 (2H, q), 3.51-3.58 (2H, m), 4.35-4.38 (2H, m), 4.53 (1H, t), 4.88 (1H, d), 6.58 (1H, t), 7.16 (1H, t), 7.32-7.38 (4H, m), 8.12 (1H, s), 8.43 (1H, d), 11.63 (1H, s), m/z (ESI+) (M+H)+=429; HPLC tR=1.46 min.

    EXAMPLE 9 ALTERNATIVE ROUTE 1: (S)-4-AMINO-N-(1-(4-CHLOROPHENYL)-3-HYDROXYPROPYL)-1-(7H-PYRROLO[2,3-D]PYRIMIDIN-4-YL)PIPERIDINE-4-CARBOXAMIDE


    (MOL)(CDX)
          N-Ethyldiisopropylamine (1.676 ml, 9.62 mmol) was added to (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)piperidine-4-carboxamide (Intermediate 49) (1 g, 3.21 mmol) and 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (0.493 g, 3.21 mmol) in butan-1-ol (15 ml). The resulting solution was stirred at 60° C. for 18 hours. The reaction mixture was diluted with EtOAc (50 mL), and washed sequentially with water (25 mL) and saturated brine (25 mL). The organic layer was dried over MgSO 4, filtered and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 0 to 6% MeOH with ammonia in DCM. Pure fractions were evaporated to dryness to afford (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (842 mg) as a white foam. (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide was stirred in ethyl acetate (7 mL) for 18 hours. The solid was collected by filtration, washed with a small amount of ethyl acetate and vacuum oven dried at 55° C. for 18 hours to afford (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (0.585 g, 42.5%) as a white solid.
          m/z (ES+) (M+H)+=429; HPLC tR=1.60 min.
          1H NMR (400.13 MHz, DMSO-d 6) δ 1.39-1.47 (2H, m), 1.80-2.02 (4H, m), 2.17 (2H, s), 3.35-3.40 (2H, m), 3.50-3.59 (2H, m), 4.34-4.41 (2H, m), 4.53 (1H, t), 4.88 (1H, d), 6.57 (1H, m), 7.14-7.16 (1H, m), 7.31-7.37 (4H, m), 8.12 (1H, s), 8.42 (1H, d), 11.62 (1H, s)

    EXAMPLE 9 ALTERNATIVE ROUTE 2: (S)-4-AMINO-N-(1-(4-CHLOROPHENYL)-3-HYDROXYPROPYL)-1-(7H-PYRROLO[2,3-D]PYRIMIDIN-4-YL)PIPERIDINE-4-CARBOXAMIDE


    (MOL)(CDX)
          (S)-3-Amino-3-(4-chlorophenyl)propan-1-ol (Intermediate 47) (2.055 g, 11.07 mmol) was added in one portion to 4-(tert-butoxycarbonylamino)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxylic acid (Intermediate 1) (4 g, 11.07 mmol) and DIPEA (5.80 ml, 33.20 mmol) in DMA (40 ml). HATU (4.63 g, 12.18 mmol) was added and the resulting solution was stirred at 20° C. for 24 hours. The reaction mixture was evaporated to dryness then diluted with EtOAc (300 mL), and washed sequentially with water (50 mL) and saturated brine (50 mL). The organic layer was dried over MgSO 4, filtered and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 2 to 6% MeOH with ammonia in DCM. Pure fractions were evaporated to dryness and triturated with dioxane (40 ml) to afford (S)-tert-butyl 4-(1-(4-chlorophenyl)-3-hydroxypropylcarbamoyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidin-4-ylcarbamate (Intermediate 22) (4.82 g, 82%) as a white solid. (S)-tert-butyl 4-(1-(4-chlorophenyl)-3-hydroxypropylcarbamoyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidin-4-ylcarbamate (Intermediate 22) (4.82 g, 82%) was suspended in dioxane (40.0 ml) and 4M hydrogen chloride in dioxane (7.69 ml, 221.36 mmol) added. The reaction was stirred at ambient temperature for 2 hours. The crude product was purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 3.5M NH 3/MeOH and pure fractions were evaporated to dryness. The crude product was purified by preparative HPLC (Waters XBridge Prep C18 OBD column, 5 μm silica, 19 mm diameter, 100 mm length), using decreasingly polar mixtures of water (containing 1% NH 3) and MeCN as eluents. Fractions containing the desired compound were evaporated to dryness to afford (S)-4-amino-N-(1-(4-chlorophenyl)-3-hydroxypropyl)-1-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine-4-carboxamide (1.200 g, 25.3%) as a white solid.
          m/z (ES+) (M+H)+=429; HPLC tR=1.67 min.
    1H NMR matches previous.
    str1
    Flag Counter

    AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@amcrasto

    //////////

    Clinical data
    Trade namesTruqap
    Other namesAZD-5363
    AHFS/Drugs.comTruqap
    License dataUS DailyMedCapivasertib
    Routes of
    administration
    By mouth
    Drug classThreonine kinase inhibitor
    ATC codeL01EX27 (WHO)
    Legal status
    Legal statusUS: ℞-only[1]
    Identifiers
    showIUPAC name
    CAS Number1143532-39-1
    PubChem CID25227436
    DrugBankDB12218
    ChemSpider28189073
    UNIIWFR23M21IE
    KEGGD11371
    ChEMBLChEMBL2325741
    PDB ligand0XZ (PDBeRCSB PDB)
    CompTox Dashboard (EPA)DTXSID40150710 
    ECHA InfoCard100.208.066 
    Chemical and physical data
    FormulaC21H25ClN6O2
    Molar mass428.92 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI

    References

    1. Jump up to:a b c “Truqap- capivasertib tablet, film coated”DailyMed. 16 November 2023. Archived from the original on 20 November 2023. Retrieved 20 November 2023.
    2. ^ Turner NC, Oliveira M, Howell SJ, Dalenc F, Cortes J, Gomez Moreno HL, et al. (June 2023). “Capivasertib in Hormone Receptor–Positive Advanced Breast Cancer”. New England Journal of Medicine388 (22): 2058–2070. doi:10.1056/NEJMoa2214131PMID 37256976S2CID 259002400.
    3. Jump up to:a b c d e f “FDA approves capivasertib with fulvestrant for breast cancer”U.S. Food and Drug Administration. 16 November 2023. Archived from the original on 17 November 2023. Retrieved 17 November 2023. Public Domain This article incorporates text from this source, which is in the public domain.
    4. ^ “Oncology (Cancer) / Hematologic Malignancies Approval Notifications”U.S. Food and Drug Administration. 16 November 2023. Archived from the original on 17 November 2023. Retrieved 17 November 2023.
    5. ^ “Truqap (capivasertib) plus Faslodex approved in the US for patients with advanced HR-positive breast cancer”AstraZeneca (Press release). 17 November 2023. Archived from the original on 17 November 2023. Retrieved 17 November 2023.

    External links

    • Clinical trial number NCT04305496 for “Capivasertib+Fulvestrant vs Placebo+Fulvestrant as Treatment for Locally Advanced (Inoperable) or Metastatic HR+/HER2- Breast Cancer (CAPItello-291)” at ClinicalTrials.gov

    ///////Capivasertib, Truqap, FDA 2023, APPROVALS 2023, AZD 5363

    NC1(CCN(CC1)C1=C2C=CNC2=NC=N1)C(=O)N[C@@H](CCO)C1=CC=C(Cl)C=C1

    Eplontersen


    Eplontersen

    AKCEA-TTR-LRx

    • ION-682884 FREE ACID
    • ISIS-682884 FREE ACID

    UNII0GRZ0F5XJ6

    CAS number1637600-16-8

    STR1

    Eplontersen, FDA APP, 12/21/2023, To treat polyneuropathy of hereditary transthyretin-mediated amyloidosis, Wainua

    AKCEA-TTR-LRx is under investigation in clinical trial NCT04136184 (Neuro-ttransform: A Study to Evaluate the Efficacy and Safety of Akcea-ttr-lrx in Participants With Hereditary Transthyretin-mediated Amyloid Polyneuropathy).

    Eplontersen, sold under the brand name Wainua, is a medication used for the treatment of transthyretin-mediated amyloidosis.[1] It is a transthyretin-directed antisense oligonucleotide.[1] It was developed to treat hereditary transthyretin amyloidosis by Ionis Pharmaceuticals and AstraZeneca.[2][3][4][5]

    It was approved for medical use in the United States in December 2023.[6][7][8]

    Medical uses

    Eplontersen is indicated for the treatment of the polyneuropathy of hereditary transthyretin-mediated amyloidosis in adults.[1]

    Society and culture

    Names

    Eplontersen is the international nonproprietary name.[9]

    //////////////

    str1
    Flag Counter

    AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@amcrasto

    //////////

    Clinical data
    Trade namesWainua
    Other namesAKCEA-TTR-LRx
    AHFS/Drugs.comEplontersen
    License dataUS DailyMedEplontersen
    Routes of
    administration
    Subcutaneous
    ATC codeN07XX21 (WHO)
    Legal status
    Legal statusUS: ℞-only[1]
    Identifiers
    CAS Number1637600-16-8
    DrugBankDB16199
    UNII0GRZ0F5XJ6

    References

    1. Jump up to:a b c d https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/217388s000lbl.pdf
    2. ^ “Ionis announces FDA acceptance of New Drug Application for eplontersen for the treatment of hereditary transthyretin-mediated amyloid polyneuropathy (ATTRv-PN)” (Press release). Ionis Pharmaceuticals. 7 March 2023. Archived from the original on 26 September 2023. Retrieved 21 December 2023 – via PR Newswire.
    3. ^ Coelho, Teresa; Waddington Cruz, Márcia; Chao, Chi-Chao; Parman, Yeşim; Wixner, Jonas; Weiler, Markus; et al. (February 2023). “Characteristics of Patients with Hereditary Transthyretin Amyloidosis-Polyneuropathy (ATTRv-PN) in NEURO-TTRansform, an Open-label Phase 3 Study of Eplontersen”Neurology and Therapy12 (1): 267–287. doi:10.1007/s40120-022-00414-zPMC 9837340PMID 36525140.
    4. ^ Coelho, Teresa; Marques, Wilson; Dasgupta, Noel R.; Chao, Chi-Chao; Parman, Yeşim; França, Marcondes Cavalcante; et al. (October 2023). “Eplontersen for Hereditary Transthyretin Amyloidosis With Polyneuropathy”. The Journal of the American Medical Association330 (15): 1448–1458. doi:10.1001/jama.2023.18688PMC 10540057. PMID 37768671.
    5. ^ Diep, John K.; Yu, Rosie Z.; Viney, Nicholas J.; Schneider, Eugene; Guo, Shuling; Henry, Scott; et al. (December 2022). “Population pharmacokinetic/pharmacodynamic modelling of eplontersen, an antisense oligonucleotide in development for transthyretin amyloidosis”. British Journal of Clinical Pharmacology88 (12): 5389–5398. doi:10.1111/bcp.15468PMID 35869634S2CID 250989659.
    6. ^ “Eplontersen: FDA-Approved Drugs”U.S. Food and Drug Administration (FDA). Retrieved 21 December 2023.
    7. ^ “Wainua (eplontersen) granted regulatory approval in the U.S. for the treatment of adults with polyneuropathy of hereditary transthyretin-mediated amyloidosis”Ionis Pharmaceuticals, Inc. (Press release). 21 December 2023. Retrieved 22 December 2023.
    8. ^ “Wainua (eplontersen) granted first-ever regulatory approval in the US for the treatment of adults with polyneuropathy of hereditary transthyretin-mediated amyloidosis”AstraZeneca US (Press release). 22 December 2023. Retrieved 22 December 2023.
    9. ^ World Health Organization (2021). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 85”. WHO Drug Information35 (1). hdl:10665/340684.

    External links

    • Clinical trial number NCT04136184 for “NEURO-TTRansform: A Study to Evaluate the Efficacy and Safety of Eplontersen (Formerly Known as ION-682884, IONIS-TTR-LRx and AKCEA-TTR-LRx) in Participants With Hereditary Transthyretin-Mediated Amyloid Polyneuropathy” at ClinicalTrials.gov
    • Clinical trial number NCT01737398 for “Efficacy and Safety of Inotersen in Familial Amyloid Polyneuropathy” at ClinicalTrials.gov

    ///////////Eplontersen, Wainua, FDA 2023, APPROVALS 2023, ION-682884 FREE ACID, ISIS-682884 FREE ACID

    Iptacopan


    Iptacopan

    1644670-37-0

    422.525, C25H30N2O4

    • 4-((2S,4S)-4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl) benzoic acid
    • BENZOIC ACID, 4-((2S,4S)-4-ETHOXY-1-((5-METHOXY-7-METHYL-1H-INDOL-4-YL)METHYL)-2-PIPERIDINYL)-
    • Iptacopan
    • LNP 023
    • LNP-023
    • LNP023
    • NVP-LNP023
    • NVP-LNP023-NX

    Fda approved, To treat paroxysmal nocturnal hemoglobinuria, 12/5/2023, Fabhalta ‘CHINA 2024

    Iptacopan is a small-molecule factor B inhibitor previously investigated as a potential treatment for the rare blood disease paroxysmal nocturnal hemoglobinuria (PNH) by inhibiting the complement factor B.1 Factor B is a positive regulator of the alternative complement pathway, where it activates C3 convertase and subsequently C5 convertase.2 This is of particular importance to PNH, where one of the disease hallmarks is the mutation of the PIGA gene. Due to this mutation, all progeny erythrocytes will lack the glycosyl phosphatidylinositol–anchored proteins that normally anchor 2 membrane proteins, CD55 and CD59, that protect blood cells against the alternative complement pathway.3 Additionally, iptacopan has the benefit of targeting factor B, which only affect the alternative complement pathway, leaving the classic and lectin pathway untouched for the body to still mount adequate immune responses against pathogens.2

    On December 6th, 2023, Iptacopan under the brand name Fabhalta was approved by the FDA for the treatment of adults with PNH. This approval was based on favorable results obtained from the phase III APPL-PNH and APPOINT-PNH studies, where 82.3% and 77.5% of patients experienced a sustained hemoglobin improvement without transfusions respectively.5

    Iptacopan , sold under the brand name Fabhalta, is a medication used for the treatment of paroxysmal nocturnal hemoglobinuria.[1] It is a complement factor B inhibitor that was developed by Novartis.[1] It is taken by mouth.[1]

    Iptacopan was approved by the US Food and Drug Administration (FDA) for the treatment of adults with paroxysmal nocturnal hemoglobinuria in December 2023.[2][3]

    Medical uses

    Iptacopan is indicated for the treatment of adults with paroxysmal nocturnal hemoglobinuria.[1][4]

    Side effects

    The FDA label for iptacopan contains a black box warning for the risk of serious and life-threatening infections caused by encapsulated bacteria, including Streptococcus pneumoniaeNeisseria meningitidis, and Haemophilus influenzae type B.[1]

    Research

    In a clinical study with twelve participants, iptacopan as a single drug led to the normalization of hemolytic markers in most patients, and no serious adverse events occurred during the 12-week study.[5][6]

    Iptacopan is also investigated as a drug in other complement-mediated diseases, like age-related macular degeneration and some types of glomerulopathies.[7]


    PATENT

    US9682968

    https://patents.google.com/patent/US9682968B2/en

    Example-26Example-26a4-((2S,4S)-(4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl))benzoic acid ((+) as TFA Salt)

    Figure US09682968-20170620-C00315

    A mixture of methyl 4-((2S,4S)-4-ethoxy-1-((5-methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl)benzoate, Intermediate 6-2b peak-1 (tr=1.9 min), (84 mg, 0.192 mmol) and LiOH in H2O (1 mL, 1 mmol) in THF (1 mL)/MeOH (2 mL) was stirred at room temperature for 16 h, and then concentrated. The resulting residue was purified by RP-HPLC (HC-A) to afford the title compound. Absolute stereochemistry was determined by comparison with enantiopure synthesis in Example-26c. 1H NMR (TFA salt, 400 MHz, D2O) δ 8.12 (d, J=8.19 Hz, 2H), 7.66 (br. d, J=8.20 Hz, 2H), 7.35 (d, J=3.06 Hz, 1H), 6.67 (s, 1H), 6.25 (d, J=3.06 Hz, 1H), 4.65 (dd, J=4.28, 11.49 Hz, 1H), 4.04 (d, J=13.00 Hz, 1H), 3.87-3.98 (m, 2H), 3.53-3.69 (m, 5H), 3.38-3.50 (m, 1H), 3.20-3.35 (m, 1H), 2.40 (s, 3H), 2.17-2.33 (m, 2H), 2.08 (br. d, J=15.70 Hz, 1H), 1.82-1.99 (m, 1H), 1.28 (t, J=7.03 Hz, 3H); HRMS calcd. for C26H31N2O(M+H)423.2284, found 423.2263.

    PATENT

    Example 1

          Intermediate 1:

          To a 3 L three-necked flask were successively added tetrahydrofuran (150 mL) and 4-bromoxynil (50 g). Isopropylmagnesium chloride lithium chloride coordination complex (1.3 M, 210 mL) was slowly added to the reaction system under nitrogen atmosphere. After the reaction was carried out at room temperature for 2 h, the reaction system was diluted with anhydrous tetrahydrofuran (500 mL) for dilution. The reaction system was cooled to −5° C., and 4-methoxypyridine (25 mL) was added, followed by slowly dropwise addition of benzyl chloroformate (35 mL) (the system temperature was maintained below 0° C.). After the dropwise addition was completed, the reaction system was successively reacted at 0° C. for 2 h, and then warmed to room temperature and reacted at that temperature for 16 h. After the reaction was completed, hydrochloric acid solution (6 M, 150 mL) was added. The mixture was stirred at room temperature for half an hour, added with water (1000 mL) for dilution, and extracted twice with ethyl acetate (500 mL). The extract phase was washed with saturated brine (50 mL), then dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated, and the resulting crude product was separated and purified by a silica gel column (petroleum ether:ethyl acetate=3:1 to 1:1) to give intermediate 1 (23 g, yield: 23%). MS m/z (ESI): 333.0 [M+H].
          Intermediate 2:

          To a 500 mL single-neck flask were successively added intermediate 1 (28 g), zinc powder (55 g) and acetic acid (200 mL). The reaction mixture was heated to 100° C. and reacted at that temperature for 16 h. After the reaction was completed, the reaction mixture was filtered. The filtrate was added with water (500 mL) for dilution and extracted with ethyl acetate (500 mL). The extract phase was washed twice with saturated aqueous sodium bicarbonate solution (500 mL), washed once with saturated brine (100 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give intermediate 2 (26 g, yield: 73%). MS m/z (ESI): 334.8 [M+H].
          Intermediate 3:

          To a 1 L single-neck flask were successively added tetrahydrofuran (100 mL), ethanol (100 mL) and intermediate 2 (26 g), and sodium borohydride (2 g) was added in batches. The mixture was reacted at room temperature for 2 h. After the reaction was completed, the system was cooled to 0° C., and saturated aqueous ammonium chloride solution (100 mL) was added until the temperature did not increase any more. Water (300 mL) was added for dilution, followed by extraction with ethyl acetate (200 mL×2). The extract phase was washed with saturated brine (500 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give intermediate 3 (25 g, yield: 76%). MS m/z (ESI): 336.9 [M+H].
          Intermediate 4:

          Dichloromethane (200 mL) was added to a 500 mL single-neck flask, and then intermediate 3 (25 g), imidazole (6.6 g) and tert-butyldiphenylchlorosilane (25 g) were successively added. The mixture was reacted at room temperature for 2 h. After the reaction was completed, water (500 mL) was added for dilution, followed by the extraction with dichloromethane (200 mL). The extract phase was washed with water (50 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was separated and purified by a silica gel column (petroleum ether:ethyl acetate=10:1) to give intermediate 4 (5.7 g, yield: 13%, R f=0.55; isomer R f=0.50). MS m/z (ESI): 597.0 [M+23].
          Intermediate 5:

          To a 250 mL single-neck flask were successively added a solution of tetrabutylammonium fluoride in tetrahydrofuran (1 M, 30 mL) and intermediate 4 (5 g). The mixture was reacted at room temperature for 2 h. After the reaction was completed, water (100 mL) was added for dilution, followed by the extraction with ethyl acetate (50 mL×3). The extract phase was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure. The residue was separated and purified by a silica gel column (petroleum ether:ethyl acetate=3:1 to 0:1) to give a racemic intermediate. The intermediate was subjected to SFC chiral resolution (apparatus: SFC Thar prep 80; column: CHIRALPAK AD-H, 250 mm×20 mm, 5 m; modifier: 35% methanol (0.2% aqueous ammonia); column temperature: 40° C.; column pressure: 60 bar; wavelength: 214/254 nm; flow rate: 40 g/min; Rt=4.78 min) to give intermediate 5 (1.2 g, yield: 41%). MS m/z (ESI): 358.8 [M+23].
          Intermediate 6:

          To a 100 mL single-neck flask were successively added N,N-dimethylformamide (15 mL) as a solvent, intermediate 5 (1.2 g) and iodoethane (1.1 g). After the reaction system was cooled to 0° C., sodium hydrogen (60%, 243 mg) was added. Then the system was warmed to room temperature and reacted at that temperature for 2 h. After the reaction was completed, water (30 mL) was added for dilution, followed by the extraction with ethyl acetate (50 mL). The extract phase was washed with saturated brine (10 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give intermediate 6 (1.2 g, yield: 83%). MS m/z (ESI): 386.9 [M+23].
          Intermediate 7:

          To a 100 mL single-neck flask were successively added methanol (10 mL), water (10 mL), concentrated sulfuric acid (10 mL) and intermediate 6 (1.2 g). The mixture was heated to 80° C. and reacted at that temperature for 48 h. After the reaction was completed, the reaction mixture was concentrated to remove methanol. The residue was made neutral with saturated aqueous sodium hydroxide solution and extracted three times with ethyl acetate (10 mL). The extract phase was washed with saturated brine (5 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give intermediate 7 (850 mg, yield: 81%). MS m/z (ESI): 264.1 [M+H]. 1H NMR (400 MHz, CDCl 3) δ 8.01 (d, J=8.3 Hz, 2H), 7.49 (d, J=8.3 Hz, 2H), 4.13 (dd, J=11.7, 2.4 Hz, 1H), 3.92 (s, 3H), 3.82-3.70 (m, 1H), 3.62-3.47 (m, 2H), 3.27-3.10 (m, 1H), 3.02-2.88 (m, 1H), 2.07-1.97 (m, 1H), 1.95-1.85 (m, 1H), 1.82-1.62 (m, 2H), 1.27 (t, J=7.0 Hz, 3H).
          Intermediate 8:

          To a 250 mL single-neck flask were successively added dichloromethane (50 mL), 5-methoxy-7-methyl-1H-indole (3 g), BOC anhydride (5.68 g), 4-dimethylaminopyridine (227 mg) and triethylamine (2.26 g). The mixture was reacted at room temperature for 16 h. After the reaction was completed, the reaction mixture was quenched by adding saturated ammonium chloride solution (5 mL) and extracted three times with dichloromethane (20 mL). The combined organic phases were washed with water (5 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether:ethyl acetate=10:1) to give intermediate 8 (4.6 g, yield: 94%). MS m/z (ESI): 262.0 [M+H].
          Intermediate 9:

          To a 250 mL single-neck flask were successively added dichloromethane (80 mL), N-methylformanilide (3.8 g) and oxalyl chloride (3.6 g). The mixture was stirred at room temperature for 3 h. Then the reaction temperature was lowered to −14° C., and intermediate 8 (2.5 g) was added. The reaction system was naturally warmed to room temperature and stirred for 1 h. After the reaction was completed, the reaction liquid was poured into ice water and extracted three times with dichloromethane (100 mL). The combined extract phases were washed twice with water (10 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated. The residue was separated and purified by a silica gel column (petroleum ether:ethyl acetate=20:1) to give intermediate 9 (1.3 g, yield: 47%). MS m/z (ESI): 290.0 [M+H]. 1H NMR (400 MHz, CDCl 3) δ 10.65 (s, 1H), 7.65 (d, J=3.4 Hz, 1H), 7.49 (d, J=3.4 Hz, 1H), 6.76 (s, 1H), 3.98 (s, 3H), 2.70 (s, 3H), 1.65 (s, 9H).
          Intermediate 10:

          To a 50 mL three-necked flask were successively added 1,2-dichloroethane (5 mL), intermediate 7 (127 mg) and intermediate 9 (130 mg). The mixture was reacted at room temperature for 18 h. Then sodium triacetoxyborohydride (438.72 mg) was added, and the system was successively reacted at room temperature for 18 h. After the reaction was completed, dichloromethane (10 mL) was added for dilution, followed by a wash with 10 mL of water. The organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated. The residue was separated and purified by a silica gel column (methanol:dichloromethane=1:10) to give intermediate 10 (50 mg, yield: 14.58%). MS m/z (ESI): 437.3 [M+H], RT=1.142 min.
          Intermediate 11:

          To a 50 mL three-necked flask were successively added tetrahydrofuran (0.5 mL), methanol (0.5 mL), water (0.5 mL), sodium hydroxide (44 mg) and intermediate 10 (50 mg). The mixture was reacted at room temperature for 18 h. After the reaction was completed, the reaction liquid was directly concentrated under reduced pressure and lyophilized to give intermediate 11 (50 mg, yield: 92%). MS m/z (ESI): 423.1 [M+H].

    PAPER

    https://pubs.acs.org/doi/abs/10.1021/acs.jmedchem.9b01870

    The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.

    Abstract Image
    STR1

    a Reagents and conditions: (a) i PrMgCl·LiCl, Cbz-Cl, THF; (b) Zn, AcOH; (c) LiBH4, THF; (d) TBDPS-Cl, imidazole, DMF; (e) separation of diastereomers by flash chromatography; (f) TBAF, THF; (g) NaH, EtI, DMF; (h) Ba(OH)2, i PrOH, H2O; (i) K2CO3, MeI, DMF; (j) H2, Pd/C, MeOH; (k) (±)-50, DIPEA, DMA; (l) K2CO3, MeOH; then TMS-diazomethane, toluene, MeOH; (m) chiral SFC; (n) LiOH, H2O, MeOH, THF; (o) (2S,4S)-50, NaBH(OAc)3, DCE.

    4-((2S,4S)-(4-Ethoxy-1-((5-methoxy-7-methyl-1H-indol-4- yl)methyl)piperidin-2-yl))benzoic Acid (41, LNP023). Step 1: tert-Butyl 4-(((2S,4S)-4-Ethoxy-2-(4-(methoxycarbonyl)phenyl)- piperidin-1-yl)methyl)-5-methoxy-7-methyl-1H-indole-1-carboxylate (58). To a solution of tert-butyl 4-formyl-5-methoxy-7-methyl1H-indole-1-carboxylate (57) (1.5 g, 5.18 mmol) and methyl 4- ((2S,4S)-4-ethoxypiperidin-2-yl)benzoate ((2S,4S)-50) (1.185 g, 4.50 mmol) in DCE (20 mL) was added NaBH(OAc)3 (3 g, 14.1 mmol), and this was stirred at rt for 21.5h. Additional tert-butyl 4-formyl-5- methoxy-7-methyl-1H-indole-1-carboxylate (57) (500 mg, 1.90 mmol) was added, and this was stirred for 20 h. The reaction was diluted with EtOAc, washed successively with 5% aqueous NaHCO3, H2O, and brine, dried over Na2SO4, filtered, and concentrated to provide the title compound (2.415 g, quant) which was used without further purification. MS (ESI+) m/z 537.4 (M + H). The absolutestereochemistry was ultimately determined via cocrystallization of 41 with the catalytic domain of FB. Step 2: 4-((2S,4S)-(4-Ethoxy-1-((5-methoxy-7-methyl-1H-indol-4- yl)methyl)piperidin-2-yl))benzoic Acid (41, LNP023). To a solution of tert-butyl 4-(((2S,4S)-4-ethoxy-2-(4-(methoxycarbonyl)phenyl)- piperidin-1-yl)methyl)-5-methoxy-7-methyl-1H-indole-1-carboxylate (58) (2.415 g, 4.50 mmol) in THF (10 mL) and MeOH (20 mL) was added 1 M LiOH in H2O (15 mL, 15 mmol), and this was stirred at 70 °C for 8 h. The reaction was cooled to rt, diluted with H2O, half saturated aqueous KHSO4 and citric acid, saturated with sodium chloride, then extracted with 9:1 DCM/TFE, dried with Na2SO4, filtered, and concentrated. RP-HPLC-B purification provided the title compound (730 mg, 38% for 2 steps). 1 H NMR (400 MHz, D2O) δ 7.96 (d, J = 8.0 Hz, 2H), 7.58 (d, J = 8.1 Hz, 2H), 7.30 (d, J = 3.2 Hz, 1H), 6.66 (s, 1H), 6.20 (s, 1H), 4.62−4.47 (m, 1H), 4.06 (d, J = 13.2 Hz, 1H), 3.97−3.76 (m, 2H), 3.66−3.48 (m, 5H), 3.43−3.29 (m, 1H), 3.26−3.15 (m, 1H), 2.35 (s, 3H), 2.31−2.11 (m, 2H), 2.00 (d, J = 15.4 Hz, 1H), 1.93−1.74 (m, 1H), 1.25−1.07 (m, 3H). HRMS calcd for C25H31N2O4 (M + H)+ 423.2284, found 423.2263. 4-((2S,4S)-(4-Ethoxy-1-((5-methoxy-7-methyl-1H-indol-4- yl)methyl)piperidin-2-yl))benzoic Acid Hydrochloride (41· HCl). To a solution of 41 (620 mg, 1.47 mmol) in H2O (10 mL) and acetonitrile (3 mL) was added 5 M aqueous HCl (0.5 mL, 2.5 mmol). The mixture was then lyophilized, and the resulting solid was suspended in i PrOH and heated to 70 °C. The mixture turned into a solution after 1.5 h and was then cooled to rt with stirring. After about 5 h, the mixture turned into a suspension and the solid was collected by filtration and dried under high vacuum at 50 °C to provide the title compound as the hydrochloride salt (450 mg, 65%). 1 H NMR (400 MHz, methanol-d4) δ 10.73 (s, 1H), 8.23 (d, J = 8.2 Hz, 2H), 7.74 (d, J = 8.3 Hz, 2H), 7.36−7.31 (m, 1H), 6.77 (s, 1H), 6.42−6.31 (m, 1H), 4.40−4.19 (m, 2H), 3.87−3.80 (m, 1H), 3.76 (s, 3H), 3.68− 3.50 (m, 4H), 3.45−3.38 (m, 1H), 2.51 (s, 3H), 2.30−2.18 (m, 2H), 2.13−1.89 (m, 2H), 1.31 (t, J = 7.0 Hz, 3H). MS (ESI+) m/z 423.3 (M + H).

    SYN
    European Journal of Medicinal Chemistry 291 (2025) 117643

    Iptacopan (Fabhalta®), a first-in-class oral therapeutic agent discovered by Novartis, specifically targets the complement Factor B protein within the alternative complement system. NMPA granted
    marketing authorization in 2024, indicated for complement inhibitor-naïve adult patients diagnosed with paroxysmal nocturnal hemoglobinuria (PNH) [75]. By competitively binding to the catalytic domain of
    Factor B, the drug effectively blocks C3 convertase assembly, thereby suppressing downstream cleavage of C3 into its active fragments. This dual inhibitory action addresses both intravascular erythrocyte
    destruction and extravascular hemolytic processes characteristic of PNHpathogenesis [76]. Clinical validation emerged from the multinational APPOINT-PNH study (ClinicalTrials.gov identifier NCT04820530), where treatment-naïve participants exhibited sustained hemoglobin
    stabilization (≥12 g/dL) in 79.6 % of cases, achieving transfusion in dependence over 24 weeks. Secondary endpoints revealed significant improvements in fatigue scores and health-related quality metrics [77]. Safety monitoring identified encapsulated bacterial infection as critical risks, necessitating mandatory vaccination ≥2 weeks pre-treatment. Common treatment-emergent adverse events comprised transient gastrointestinal disturbances (nausea 18.3 %, diarrhea 14.7 %) and mild
    cephalgia (22.1 %), with resolution typically occurring within 4 weeks [78].
    The synthetic pathway of Iptacopan, delineated in Scheme 18, initiates with nucleophilic substitution between Ipta-001 and Ipta-002, followed by Grignard coupling yielding Ipta-003 [79]. This intermedi
    ate undergoes NaBH4-mediated reduction and TMSCl-induced silanization to afford Ipta-004. Acid-catalyzed TMS deprotection (HCl/MeOH) delivers Ipta-005, which progresses through sequential alkylation (methyl iodide/K2CO3 catalytic hydrogenation (H)/Pd–C), transesterification (EtONa), and  to construct Ipta-006. Condensation with Ipta-007 and subsequent reduction forms Ipta-008. Strategic TFA-mediated Boc cleavage in DCM followed by HCl-induced salt formation in dioxane ultimately furnishes Iptacopan hydrochloride.

    75-79

    [75] Iptacopan, Drugs and Lactation Database (Lactmed®), National Institute of Child
    Health and Human Development, Bethesda (MD), 2006.
    [76] J.H. Jang, L. Wong, B.S. Ko, S.S. Yoon, K. Li, I. Baltcheva, P.K. Nidamarthy,
    R. Chawla, G. Junge, E.S. Yap, Iptacopan monotherapy in patients with paroxysmal
    nocturnal hemoglobinuria: a 2-cohort open-label proof-of-concept study, Blood
    Adv 6 (2022) 4450–4460.
    [77] A.M. Risitano, C. de Castro, B. Han, A.G. Kulasekararaj, J.P. Maciejewski,
    P. Scheinberg, Y. Ueda, S. Vallow, G. Bermann, M. Dahlke, R. Kumar, R. Peffault de
    Latour, Patient-reported improvements in patients with PNH treated with
    iptacopan from two phase 3 studies, Blood Adv 9 (2025) 1816–1826.
    [78] C.M. de Castro, B.J. Patel, Iptacopan for the treatment of paroxysmal nocturnal
    hemoglobinuria, Expert Opin Pharmacother 25 (2024) 2331–2339.
    [79] N. Mainolfi, T. Ehara, R.G. Karki, K. Anderson, A. Mac Sweeney, S.M. Liao, U.
    A. Argikar, K. Jendza, C. Zhang, J. Powers, D.W. Klosowski, M. Crowley,
    T. Kawanami, J. Ding, M. April, C. Forster, M. Serrano-Wu, M. Capparelli,
    R. Ramqaj, C. Solovay, F. Cumin, T.M. Smith, L. Ferrara, W. Lee, D. Long,
    M. Prentiss, A. De Erkenez, L. Yang, F. Liu, H. Sellner, F. Sirockin, E. Valeur,
    P. Erbel, D. Ostermeier, P. Ramage, B. Gerhartz, A. Schubart, S. Flohr, N. Gradoux,
    R. Feifel, B. Vogg, C. Wiesmann, J. Maibaum, J. Eder, R. Sedrani, R.A. Harrison,
    M. Mogi, B.D. Jaffee, C.M. Adams, Discovery of 4-((2S,4S)-4-Ethoxy-1-((5-
    methoxy-7-methyl-1H-indol-4-yl)methyl)piperidin-2-yl)benzoic acid (LNP023), a
    factor B inhibitor specifically designed to be applicable to treating a diverse array
    of complement mediated diseases, J. Med. Chem. 63 (2020) 5697–5722.

    .

    //////////////

    str1
    Flag Counter

    AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@amcrasto

    ///////////

    Clinical data
    Trade namesFabhalta
    Other namesLNP023
    AHFS/Drugs.comFabhalta
    License dataUS DailyMedIptacopan
    Routes of
    administration
    By mouth
    Drug classComplement factor B inhibitor
    ATC codeNone
    Legal status
    Legal statusUS: ℞-only[1]
    Identifiers
    CAS Number1644670-37-0
    PubChem CID90467622
    DrugBankDB16200
    ChemSpider75533872
    UNII8E05T07Z6W
    KEGGD12251D12252
    ChEMBLChEMBL4594448
    PDB ligandJGQ (PDBeRCSB PDB)
    Chemical and physical data
    FormulaC25H30N2O4
    Molar mass422.525 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI

    References

    1. Jump up to:a b c d e f “Fabhalta- iptacopan capsule”DailyMed. 5 December 2023. Archived from the original on 10 December 2023. Retrieved 10 December 2023.
    2. ^ “Novartis receives FDA approval for Fabhalta (iptacopan), offering superior hemoglobin improvement in the absence of transfusions as the first oral monotherapy for adults with PNH”Novartis (Press release). Archived from the original on 12 December 2023. Retrieved 6 December 2023.
    3. ^ “Novel Drug Approvals for 2023”U.S. Food and Drug Administration (FDA). 6 December 2023. Archived from the original on 21 January 2023. Retrieved 10 December 2023.
    4. ^ https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2023/218276Orig1s000ltr.pdf Archived 10 December 2023 at the Wayback Machine Public Domain This article incorporates text from this source, which is in the public domain.
    5. ^ Jang JH, Wong L, Ko BS, Yoon SS, Li K, Baltcheva I, et al. (August 2022). “Iptacopan monotherapy in patients with paroxysmal nocturnal hemoglobinuria: a 2-cohort open-label proof-of-concept study”Blood Advances6 (15): 4450–4460. doi:10.1182/bloodadvances.2022006960PMC 9636331PMID 35561315.
    6. ^ “Novartis Phase III APPOINT-PNH trial shows investigational oral monotherapy iptacopan improves hemoglobin to near-normal levels, leading to transfusion independence in all treatment-naïve PNH patients”Novartis (Press release). Archived from the original on 12 December 2023. Retrieved 6 September 2023.
    7. ^ Schubart A, Anderson K, Mainolfi N, Sellner H, Ehara T, Adams CM, et al. (April 2019). “Small-molecule factor B inhibitor for the treatment of complement-mediated diseases”Proceedings of the National Academy of Sciences of the United States of America116 (16): 7926–7931. Bibcode:2019PNAS..116.7926Sdoi:10.1073/pnas.1820892116PMC 6475383PMID 30926668.

    External links

    • Clinical trial number NCT04558918 for “Study of Efficacy and Safety of Twice Daily Oral LNP023 in Adult PNH Patients With Residual Anemia Despite Anti-C5 Antibody Treatment (APPLY-PNH)” at ClinicalTrials.gov
    • Clinical trial number NCT04820530 for “Study of Efficacy and Safety of Twice Daily Oral Iptacopan (LNP023) in Adult PNH Patients Who Are Naive to Complement Inhibitor Therapy (APPOINT-PNH)” at ClinicalTrials.gov

    ///////Iptacopan, fda 2023,  approvals, 2023,  paroxysmal nocturnal hemoglobinuria, 12/5/2023, Fabhalta , LNP 023, LNP-023, LNP023, NVP-LNP023, NVP-LNP023-NX

    NEW DRUG APPROVALS

    ONE TIME

    $10.00

    Etrasimod


    Etrasimod

    • APD334
    • C26H26F3NO3
    • 457.493

    1206123-37-6
    2-[(3R)-7-{[4-cyclopentyl-3-(trifluoromethyl)phenyl]methoxy}-1H,2H,3H,4H-cyclopenta[b]indol-3-yl]acetic acid

    img
    Etrasimod arginineMXE5EMA09L1206123-97-8GVPVVOSNDUAUKM-BPGOJFKZSA-N

    Name: Etrasimod arginine
    CAS#: 1206123-97-8 (arginine)
    Chemical Formula: C32H40F3N5O5
    Exact Mass: 631.30
    Molecular Weight: 631.700

    FDA APPROVED, To treat moderately to severely active ulcerative colitis in adults,

    10/12/2023
    Velsipity

    Etrasimod, sold under the brand name Velsipity, is a medication that is used for the treatment of ulcerative colitis (UC).[1] It is a selective sphingosine-1-phosphate (S1P) receptor modulator that modifies the activity of the immune system.[1] It is taken by mouth.[1]

    Etrasimod was discovered by Arena Pharmaceuticals, with subsequent development by Pfizer.[2]

    Etrasimod is a synthetic next-generation selective Sphingosine 1-phosphate (S1P) receptor modulator that targets the S1P1,4,5 with no detectable activity on S1P2 and S1P3 receptors. S1P receptors are membrane-derived lysophospholipid signaling molecules that are involved in the sequestration of circulating peripheral lymphocytes in lymph nodes.1 Therefore, S1P receptor modulators like etrasimod were investigated in treating immune-mediated diseases like ulcerative colitis where a high level of inflammatory T cells is present in the gastrointestinal tract, thus causing diffuse mucosal inflammation.1 In fact, it has been observed that antigen-activated T cells within peripheral lymphoid organs can transiently downregulate S1P receptor levels to facilitate immune cells trafficking into the intestinal mucosa.2

    Etrasimod was approved on October 13, 2023, by the FDA under the brand name VELSIPITY for the treatment of adults with moderately to severely active ulcerative colitis. This approval was based on favorable results obtained from Pfizer’s Elevate UC Phase III registrational program, consisting of the Elevate UC 52 and Elevate UC 12 clinical trials, that investigates the efficacy of a 2-mg daily dose regimen of etrasimod, with a 32% and 26% remission rate observed in UC 52 and UC 12 trials respectively.4

    Medical uses

    Etrasimod is used for the treatment of moderate to severe ulcerative colitis.[1]

    Mechanism of action

    It works by causing T cells to become trapped in the lymph nodes, preventing them from entering the bloodstream, from where they would travel to other tissues in the body and mediate inflammation.[3][4][5][6][7][8]

    Society and culture

    Legal status

    Velsipity was approved by the US Food and Drug Administration (FDA) in October 2023.[1][9][10]

    Names

    Etrasimod is the international nonproprietary name.[11]

    SYN

    ACS Med. Chem. Lett.2014, 5, 12, 1313–1317

    Publication Date:November 4, 2014

    https://doi.org/10.1021/ml500389m

    APD334 was discovered as part of our internal effort to identify potent, centrally available, functional antagonists of the S1P1 receptor for use as next generation therapeutics for treating multiple sclerosis (MS) and other autoimmune diseases. APD334 is a potent functional antagonist of S1P1 and has a favorable PK/PD profile, producing robust lymphocyte lowering at relatively low plasma concentrations in several preclinical species. This new agent was efficacious in a mouse experimental autoimmune encephalomyelitis (EAE) model of MS and a rat collagen induced arthritis (CIA) model and was found to have appreciable central exposure.

    Abstract Image

    APD334 is the second eluting enantiomer (most retained) with a retention time of 48.4 minutes. 1H NMR (400 MHz, DMSO-d6) δ ppm 1.54-1.75 (m, 4H), 1.79-1.92 (m, 2H), 1.95-2.16 (m, 3H), 2.39 (dd, J = 16.0, 8.8 Hz, 1H), 2.61-2.83 (m, 4H), 3.23-3.34 (m, 1H), 3.45-3.56 (m, 1H), 5.14 (s, 2H), 6.74 (dd, J = 8.7, 2.4 Hz, 1H), 6.92 (d, J = 2.3 Hz, 1H), 7.24 (d, J = 8.8 Hz, 1H), 7.64 (d, J = 8.1 Hz, 1H), 7.72 (d, J = 8.6 Hz, 1H), 7.74 (s, 1H), 10.50 (s, 1H), 12.24 (bs, 1H). 13C APT NMR (100 MHz, DMSO-d6): δ up (C, CH2): 23.1, 25.5, 35.5, 35.6, 68.6, 117.0, 124.7 (q, J = 273 Hz), 124.2, 126.8 (q, J = 28 Hz), 128.7, 136.1, 136.2, 144.6, 147.0, 151.9, 173.4; down (CH, CH3): 35.0, 40.5, 102.1, 110.0, 112.4, 124.1 (q, J = 5.7 Hz), 128.4, 131.7. 19F NMR (400 MHz, DMSO-d6) δ ppm -57.4. LCMS (ESI+): calcd for C26H27F3NO3+ [M+H] 458.19; found, 458.4. HRMS (ESI-): calcd for C26H25F3NO3- [M-H] 456.1792; found, 456.1776.

    str1
    Flag Counter

    AS ON DEC2021 3,491,869 VIEWS ON BLOG WORLDREACH AVAILABLEFOR YOUR ADVERTISEMENT

    wdt-16

    join me on Linkedin

    Anthony Melvin Crasto Ph.D – India | LinkedIn

    join me on Researchgate

    RESEARCHGATE

    This image has an empty alt attribute; its file name is research.jpg

    join me on Facebook

    Anthony Melvin Crasto Dr. | Facebook

    join me on twitter

    Anthony Melvin Crasto Dr. | twitter

    +919321316780 call whatsaapp

    EMAIL. amcrasto@amcrasto

    ///////////

    Skeletal formula of etrasimod
    Clinical data
    Trade namesVelsipity
    Other namesAPD334, APD-334
    License dataUS DailyMedEtrasimod
    Routes of
    administration
    By mouth
    Drug classSphingosine-1-phosphate receptor modulator
    ATC codeNone
    Legal status
    Legal statusUS: ℞-only[1]
    Pharmacokinetic data
    Protein binding97.9%[medical citation needed]
    MetabolismLiver (CYP2C82C93A4)[medical citation needed]
    Elimination half-life30 hours[medical citation needed]
    ExcretionFeces (82%), kidneys (5%)[medical citation needed]
    Identifiers
    showIUPAC name
    CAS Number1206123-37-6as arginine: 1206123-97-8
    PubChem CID44623998
    DrugBankDB14766as arginine: DBSALT003430
    ChemSpider52084233as arginine: 57643656
    UNII6WH8495MMHas arginine: MXE5EMA09L
    KEGGD10930as arginine: D10931
    ChEMBLChEMBL3358920
    Chemical and physical data
    FormulaC26H26F3NO3
    Molar mass457.493 g·mol−1
    3D model (JSmol)Interactive image
    showSMILES
    showInChI

    References

    1. Jump up to:a b c d e f Pfizer (12 October 2023). “Velsipity (etrasimod) tablets, for oral use” (PDF). U.S. Food and Drug Administration (FDA). Retrieved 18 October 2023.
    2. ^ Bayer M (2 May 2023). “Pfizer tosses newly acquired meds out of the Arena”Fierce Biotech. Retrieved 13 October 2023.
    3. ^ Atreya R, Neurath MF (April 2023). “The sphingosine-1-phosphate receptor agonist etrasimod in ulcerative colitis”. Lancet401 (10383): 1132–1133. doi:10.1016/S0140-6736(23)00228-3PMID 36871570.
    4. ^ Sandborn WJ, Vermeire S, Peyrin-Biroulet L, Dubinsky MC, Panes J, Yarur A, et al. (April 2023). “Etrasimod as induction and maintenance therapy for ulcerative colitis (ELEVATE): two randomised, double-blind, placebo-controlled, phase 3 studies”. Lancet401 (10383): 1159–1171. doi:10.1016/S0140-6736(23)00061-2PMID 36871574.
    5. ^ Dal Buono A, Gabbiadini R, Alfarone L, Solitano V, Repici A, Vetrano S, et al. (July 2022). “Sphingosine 1-Phosphate Modulation in Inflammatory Bowel Diseases: Keeping Lymphocytes Out of the Intestine”Biomedicines10 (7). doi:10.3390/biomedicines10071735PMC 9313037PMID 35885040.
    6. ^ Argollo M, Furfaro F, Gilardi D, Roda G, Allocca M, Peyrin-Biroulet L, et al. (April 2020). “Modulation of sphingosine-1-phosphate in ulcerative colitis”. Expert Opin Biol Ther20 (4): 413–420. doi:10.1080/14712598.2020.1732919PMID 32093531.
    7. ^ Al-Shamma H, Lehmann-Bruinsma K, Carroll C, Solomon M, Komori HK, Peyrin-Biroulet L, et al. (June 2019). “The Selective Sphingosine 1-Phosphate Receptor Modulator Etrasimod Regulates Lymphocyte Trafficking and Alleviates Experimental Colitis”. J Pharmacol Exp Ther369 (3): 311–317. doi:10.1124/jpet.118.254268PMID 30872391.
    8. ^ Peyrin-Biroulet L, Christopher R, Behan D, Lassen C (May 2017). “Modulation of sphingosine-1-phosphate in inflammatory bowel disease”. Autoimmun Rev16 (5): 495–503. doi:10.1016/j.autrev.2017.03.007PMID 28279838.
    9. ^ Brooks M (13 October 2023). “FDA Approves New Drug for Ulcerative Colitis”Medscape. Retrieved 13 October 2023.
    10. ^ https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2023/216956Orig1s000ltr.pdf
    11. ^ World Health Organization (2017). “International nonproprietary names for pharmaceutical substances (INN): recommended INN: list 78”. WHO Drug Information31 (3). hdl:10665/330961.

    /////////Etrasimod, APD334, Velsipity, FDA 2023, APPROVALS 2023

    wdt

    NEW DRUG APPROVALS

    ONE TIME

    $10.00