New Drug Approvals

Home » Antineoplastic

Category Archives: Antineoplastic

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

Blog Stats

  • 4,808,911 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

Beroterkib


Beroterkib

CAS 2095719-92-7

MF C29H31ClFN5O5 MW584.0 g/mol

(2R)-2-(6-{5-chloro-2-[(oxan-4-yl)amino]pyrimidin-4-yl}-1,3-dihydro-2H-1-oxoisoindol-2-yl) -N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]propanamide

(2R)-2-[5-[5-chloro-2-(oxan-4-ylamino)pyrimidin-4-yl]-3-oxo-1H-isoindol-2-yl]-N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]propanamide

(alphaR)-6-[5-chloro-2-[(tetrahydro-2H-pyran-4-yl)amino]-4-pyrimidinyl]-N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]-1,3-dihydro-alpha-methyl-1-oxo-2H-isoindole-2-acetamide

(2R)-2-[5-[5-chloro-2-(oxan-4-ylamino)pyrimidin-4-yl]-3-oxo-1H-isoindol-2-yl]-N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]propanamide
extracellular signal-regulated kinases (ERK) inhibitor, antineoplastic, ASTX029, ASTX 029, 14FDK6ISC9, Beroterkib anhydrous, AT 35029

Beroterkib Anhydrous is the anhydrous form of beroterkib, an orally bioavailable inhibitor of the extracellular signal-regulated kinases (ERK) 1 and 2, with potential antineoplastic activity. Upon administration, beroterkib specifically binds to and inhibits both ERK 1 and 2, thereby preventing the activation of mitogen-activated protein kinase (MAPK)/ERK-mediated signal transduction pathways. This results in the inhibition of ERK-dependent tumor cell proliferation and survival. The MAPK/ERK pathway is often upregulated in a variety of tumor cell types and plays a key role in the proliferation, differentiation and survival of tumor cells.

  • Study of ASTX029 in Subjects With Advanced Solid TumorsCTID: NCT03520075Phase: Phase 1/Phase 2Status: CompletedDate: 2025-07-03
  • Phase I/II Study of a Combination of Decitabine and Cedazuridine (ASTX727) and ASTX029, an ERK Inhibitor, for Patients With RAS Pathway Mutant Myelodysplastic Syndromes and Myelodysplastic/Myeloproliferative NeoplasmsCTID: NCT06284460Phase: Phase 1/Phase 2Status: WithdrawnDate: 2024-10-24
  • A Phase 1 Study to Evaluate the Effect of Food on Pharmacokinetics of ASTX029CTID: NCT04466514Phase: Phase 1Status: CompletedDate: 2024-08-02

PAT

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017068412&_cid=P21-MK4TZX-17603-1

Example 685: (2R)-2-(6-{5-chloro-2-[(oxan-4-yl)amino]pyrimidin-4-yl}-1-oxo-2,3-dihydro- 1H-isoindol-2-yl)-N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]propanamide

A stirred solution of (R)-2-(6-(5-chloro-2-((oxan-4-yl)amino)pyrimidin-4-yl)-1-oxoisoindolin-2- yl)propanoic acid (70 mg, 0.168 mmol), (S)-2-amino-2-(3-fluoro-5-methoxyphenyl)ethanol, HCl (41 mg, 0.185 mmol) and triethylamine (0.094 ml, 0.672 mmol) in DMF (1 ml) was treated with TBTU (65 mg, 0.202 mmol) and stirred at room temperature overnight. The mixture was diluted with ethyl acetate (20 ml), was washed successively with 1M KHSO4 (10 ml), NaHCO3 (10 ml), brine (2x 10 ml) and then water (4x 10 ml), was dried (MgSO4) and evaporated. The residue was purified by chromatography (SiO2, 12 g column, 0- 5% EtOOH in EtOAc) to give a glass, which was triturated with ether (2 ml) to give a solid. The solid was collected by filtration, washed with ether (2x 1 ml) and dried under vacuum at 50°C overnight to give the titlecompound (64.3 mg, 64.3 %) as a cream solid. 1H NMR (DMSO, 400 MHz) δ 8.56 (1H, d), 8.44 (1H, s), 8.07 ‒ 8.00 (1H, m), 7.97 (1H, dd), 7.74 (1H, d), 7.61 (1H, s), 6.76 ‒ 6.64 (3H, m), 4.99 (1H, q), 4.91 (1H, t), 4.86 ‒ 4.70 (2H, m), 4.60 (1H, d), 4.00 ‒ 3.80 (3H, m), 3.76 (3H, s), 3.60 ‒ 3.47 (2H, m), 3.40 ‒ 3.33 (2H, m), 1.84 (2H, d), 1.59 ‒ 1.39 (5H, m). ). LCMS: [M+H]+ = 584.

SYN

US10457669,

https://patentscope.wipo.int/search/en/detail.jsf?docId=US237389744&_cid=P21-MK4U5F-21416-1

Example 685: (2R)-2-(6-{5-chloro-2-[(oxan-4-yl)amino]pyrimidin-4-yl}-1-oxo-2,3-dihydro-1H-isoindol-2-yl)-N-[(1S)-1-(3-fluoro-5-methoxyphenyl)-2-hydroxyethyl]propanamide

      
      A stirred solution of (R)-2-(6-(5-chloro-2-((oxan-4-yl)amino)pyrimidin-4-yl)-1-oxoisoindolin-2-yl)propanoic acid (70 mg, 0.168 mmol), (S)-2-amino-2-(3-fluoro-5-methoxyphenyl)ethanol, HCl (41 mg, 0.185 mmol) and triethylamine (0.094 ml, 0.672 mmol) in DMF (1 ml) was treated with TBTU (65 mg, 0.202 mmol) and stirred at room temperature overnight. The mixture was diluted with ethyl acetate (20 ml), was washed successively with 1M KHSO (10 ml), NaHCO (10 ml), brine (2×10 ml) and then water (4×10 ml), was dried (MgSO 4) and evaporated. The residue was purified by chromatography (SiO 2, 12 g column, 0-5% EtOOH in EtOAc) to give a glass, which was triturated with ether (2 ml) to give a solid. The solid was collected by filtration, washed with ether (2×1 ml) and dried under vacuum at 50° C. overnight to give the title compound (64.3 mg, 64.3%) as a cream solid. 1H NMR (DMSO, 400 MHz) δ 8.56 (1H, d), 8.44 (1H, s), 8.07-8.00 (1H, m), 7.97 (1H, dd), 7.74 (1H, d), 7.61 (1H, s), 6.76-6.64 (3H, m), 4.99 (1H, q), 4.91 (1H, t), 4.86-4.70 (2H, m), 4.60 (1H, d), 4.00-3.80 (3H, m), 3.76 (3H, s), 3.60-3.47 (2H, m), 3.40-3.33 (2H, m), 1.84 (2H, d), 1.59-1.39 (5H, m).). LCMS: [M+H] +=584.

SYN

US10457669,

PAT

str1

AS ON OCT2025 4.511 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

……

REF

//////////////Beroterkib, extracellular signal-regulated kinases (ERK) inhibitor, antineoplastic, ASTX029, ASTX 029, 14FDK6ISC9, Beroterkib anhydrous, AT 35029

Atirmociclib


Atirmociclib

CAS 2380321-51-5

MF C22H27ClFN5O3,
463.9 g/mol

(3S,4R)-4-[[5-chloro-4-[7-fluoro-2-(2-hydroxypropan-2-yl)-3-propan-2-ylbenzimidazol-5-yl]pyrimidin-2-yl]amino]oxan-3-ol

(3S,4R)-4-({5-chloro-4-[4-fluoro-2-(2-hydroxypropan-2-yl)-1-(propan2-yl)-1H-1,3-benzimidazol-6-yl]pyrimidin-2-yl}amino)oxan-3-ol

 1,5-anhydro-3-({5-chloro-4-[4-fluoro-2-(2-hydroxpropan-2-yl)-1-(propan-2-yl)-1H-benzimidazol-6-yl]pyrimidin-2-yl}amino)-2,3-dideoxy-D-threo-pentitol

D-threo-Pentitol, 1,5-anhydro-3-[[5-chloro-4-[4-fluoro-2-(1-hydroxy-1-methylethyl)-1-(1-methylethyl)-1H-benzimidazol-6-yl]-2-pyrimidinyl]amino]-2,3-dideoxy-
cyclin-dependent kinase (CDK) inhibitor, antineoplastic, PF 07220060, S743GOJ5LJ, CDK4/6-IN-6

Atirmociclib is an orally bioavailable inhibitor of cyclin-dependent kinase 4 (CDK4), with potential antineoplastic activity. Upon administration, atirmociclib selectively inhibits CDK4, which inhibits the phosphorylation of retinoblastoma protein (Rb) early in the G1 phase, prevents CDK-mediated G1-S-phase transition and leads to cell cycle arrest. This suppresses DNA replication and inhibits tumor cell proliferation. CDK4, a serine/threonine kinase, is upregulated in many tumor cell types and plays a key role in the regulation of both cell cycle progression from the G1-phase into the S-phase and tumor cell proliferation.

Atirmociclib (development code PF-07220060) is an investigational orally bioavailable and CDK4-specific inhibitor being developed by Pfizer for the treatment of various solid tumors, particularly hormone receptor-positive, HER2-negative breast cancer.[1][2] The safety and efficacy of atirmociclib have not been established, as it remains in clinical development as of September 2025.[3][4][5]

SYN

https://pubs.acs.org/doi/10.1021/acs.jmedchem.5c02137

PAT

https://patentscope.wipo.int/search/en/detail.jsf?docId=US275481329&_cid=P22-MK0K3I-13424-1

Example A94 (Scheme A-15): Preparation of 1,5-anhydro-3-({5-chloro-4-[4-fluoro-2-(2-hydroxypropan-2-yl)-1-(propan-2-yl)-1H-benzimidazol-6-yl]pyrimidin-2-yl}amino)-2,3-dideoxy-D-threo-pentitol

Step 8: Synthesis of 1,5-anhydro-3-({5-chloro-4-[4-fluoro-2-(2-hydroxypropan-2-yl)-1-(propan-2-yl)-1H-benzimidazol-6-yl]pyrimidin-2-yl}amino)-2,3-dideoxy-D-threo-pentitol (Example A94)

      A 2 L three-neck round bottom flask was charged with 2-[6-(2,5-dichloropyrimidin-4-yl)-4-fluoro-1-(propan-2-yl)-1H-benzimidazol-2-yl]propan-2-ol (A-23) (112 g, 292 mmol), 3-amino-1,5-anhydro-2,3-dideoxy-D-threo-pentitol hydrochloride (51.6 g, 336 mmol), and MeCN (1.1 L). DIPEA (132 g, 1.02 mol, 178 mL) was added at room temperature. The reaction mixture was heated to 80° C. (internal temperature) and stirred at the same temperature for 40 h to provide a brown solution. LCMS analysis showed remaining starting material. Additional 3-amino-1,5-anhydro-2,3-dideoxy-D-threo-pentitol hydrochloride (6.73 g, 43.8 mmol) was added at 80° C. (internal temperature) and the reaction was stirred at 80° C. (internal temperature) for an additional 10 h. The reaction mixture was cooled to room temperature and concentrated under vacuum. The residue was taken up in 1:1 EtOAc/H 2O (1.5 L). Some solids were precipitated. EtOH (100 mL) was added. The organic layer was collected and the aqueous layer was extracted with EtOAc (2×500 mL). The combined organic layers were washed with H 2O (2×300 mL), dried over Na 2SO 4, and filtered. To the filtrate was added sulfhydryl silica gel (Accela, 8 g, 0.7-1.4 mmol/g). The resulting mixture was stirred at room temperature for 1 h and then filtered through a pad of Celite. Treatment with sulfhydryl silica gel was repeated in identical fashion and the filtrate was concentrated to dryness. The crude residue was slurried in MeCN (500 mL) at room temperature for 16 h. The suspension was filtered and the filter cake was washed with MeCN (2×100 mL). The filter cake was slurried again with MeCN (300 mL) at room temperature for 6 h. The mixture was filtered and the filter cake was washed with MeCN (2×100 mL). The filter cake was collected and dried under vacuum and then dried in a drying oven (45° C. for 20 h, 50° C. for 64 h) to provide 1,5-anhydro-3-({5-chloro-4-[4-fluoro-2-(2-hydroxpropan-2-yl)-1-(propan-2-yl)-1H-benzimidazol-6-yl]pyrimidin-2-yl}amino)-2,3-dideoxy-D-threo-pentitol (Example A94) (90 g, 66% yield) as a white solid. 1H NMR (400 MHz, 80° C., DMSO-d 6) δ 8.38 (s, 1H), 8.00 (s, 1H), 7.43 (d, J=11.8 Hz, 1H), 7.13 (d, J=7.5 Hz, 1H), 5.80 (hept, J=7.0 Hz, 1H), 5.56 (s, 1H), 4.71 (d, J=5.3 Hz, 1H), 3.91-3.79 (m, 3H), 3.61-3.52 (m, 1H), 3.41-3.31 (m, 1H), 3.12-3.07 (m, 1H), 2.09-2.00 (m, 1H), 1.70 (s, 6H), 1.67-1.52 (m, 7H); 19F NMR (377 MHz, CDCl 3) δ −127.2; m/z (ESI+) for (C 2227ClFN 63), 464.2 (M+H) +; [α] D 22=−12.6 (c=0.2, MeOH).

PAT

https://patentscope.wipo.int/search/en/detail.jsf?docId=US275481329&_cid=P22-MK0KHW-23947-1

PAT

str1

AS ON OCT2025 4.511 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

……

Mechanism of action

Atirmociclib is designed as a CDK4-specific inhibitor, distinguishing it from dual CDK4/6 inhibitors currently approved for cancer treatment.[6] The drug targets cyclin-dependent kinase 4, which plays a role in cell cycle regulation.[1][7][8]

Atirmociclib functions as a selective inhibitor of the CDK4/cyclin D complex, which plays a crucial role in cell cycle regulation.[4] The drug works by targeting the CDK4 kinase, rendering the retinoblastoma (Rb)/E2F transcription system inactive, which ultimately leads to cell cycle arrest in the G1 phase.[4] This mechanism is particularly effective in tumors that have lost Rb cell cycle-suppressive function, a common feature in various solid tumors.[5]

The selective nature of atirmociclib represents a significant advancement over existing dual CDK4/6 inhibitors.[6] By specifically targeting CDK4 while limiting CDK6 inhibition, atirmociclib is designed to maintain antitumor efficacy while potentially reducing dose-limiting hematologic toxicities, particularly neutropenia, which is believed to be primarily driven by CDK6 inhibition.[9]

Clinical development

Atirmociclib is currently being evaluated in clinical trials for the treatment of advanced solid tumors.[1] Clinical studies are ongoing with estimated completion dates extending to 2027–2028, reflecting the early stage of development for this investigational compound.[1]

Preclinical research published in Cancer Cell in March 2025 reported atirmociclib as a next-generation CDK4-selective inhibitor with enhanced anti-tumor activity and reduced predicted toxicity compared to FDA-approved dual CDK4/6 inhibitors, though these findings require validation in clinical studies.[6]

Preclinical studies

Preclinical research has demonstrated that atirmociclib exhibits enhanced anti-tumor activity compared to FDA-approved dual CDK4/6 inhibitors while showing reduced predicted toxicity.[6] Studies have shown that CDK4-selective inhibition can provide improved preclinical anti-tumor efficacy and safety profiles compared to dual CDK4/6 inhibition strategies.[10]

The preclinical development program has explored combination approaches with various therapeutic modalities, including endocrine therapy, CDK2 inhibition, HER2 antibodies, and immune checkpoint inhibitors.[6] These combination strategies are designed to counter resistance mechanisms to CDK4 inhibition and expand the potential therapeutic applications of cell cycle targeting therapy.[6]

Clinical trials

Atirmociclib has entered clinical development as part of Pfizer’s extensive oncology pipeline.[11] The clinical program is evaluating atirmociclib both as a single agent and in combination with other therapeutic approaches, particularly focusing on patients with hormone receptor-positive, HER2-negative breast cancer.[9][12][13][14][15][16][17]

Early clinical studies have included heavily pretreated patient populations, including those who have previously received CDK4/6 inhibitor therapy.[9] This approach allows for the evaluation of atirmociclib’s potential to overcome resistance to existing CDK4/6 inhibitors and provide therapeutic benefit in patients with limited treatment options.[9]

Safety profile and toxicity

One of the key differentiating features of atirmociclib is its potential for improved safety profile compared to existing dual CDK4/6 inhibitors.[6] The selective targeting of CDK4 while limiting CDK6 inhibition is specifically designed to reduce neutropenia, the most common dose-limiting toxicity associated with current CDK4/6 inhibitors.[18]

The rationale for this approach is based on preclinical evidence suggesting that neutropenia is primarily driven by CDK6 inhibition rather than CDK4 inhibition.[18] By selectively targeting CDK4, atirmociclib aims to maintain therapeutic efficacy while potentially allowing for higher or more sustained dosing without the dose-limiting hematologic toxicities that can compromise treatment outcomes with existing agents.[18]

Regulatory status

As of September 2025, atirmociclib remains an investigational drug that has not received approval from the FDA or other regulatory agencies.[5] The compound is part of Pfizer’s oncology development pipeline.[5]

References

  1.  Pfizer (2 February 2025). A Phase 1/2A Study Evaluating the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Anti-Tumor Activity of Pf-07220060 as a Single Agent and as Part of Combination Therapy in Participants With Advanced Solid Tumors (Report). clinicaltrials.gov.
  2.  Shapiro GI (March 2017). “The evolving role of cyclin-dependent kinase inhibitors in cancer management”. Clinical Advances in Hematology & Oncology15 (3): 174–177. PMID 28398270.
  3.  “CDK4 inhibitor PF-07220060”http://www.cancer.gov. 2 February 2011. Retrieved 3 September 2025.
  4.  “Pfizer Pipeline”Pfizer.
  5.  “Atirmociclib PF-07220060”Pfizer Oncology Development. Retrieved 3 September 2025.
  6.  Chang J, Lu J, Liu Q, Xiang T, Zhang S, Yi Y, et al. (March 2025). “Single-cell multi-stage spatial evolutional map of esophageal carcinogenesis”. Cancer Cell43 (3): 380–397.e7. doi:10.1016/j.ccell.2025.02.009PMID 40068596.
  7.  Topacio BR, Zatulovskiy E, Cristea S, Xie S, Tambo CS, Rubin SM, et al. (May 2019). “Cyclin D-Cdk4,6 Drives Cell-Cycle Progression via the Retinoblastoma Protein’s C-Terminal Helix”Molecular Cell74 (4): 758–770.e4. doi:10.1016/j.molcel.2019.03.020PMC 6800134PMID 30982746.
  8.  Helsten T, Kato S, Schwaederle M, Tomson BN, Buys TP, Elkin SK, et al. (July 2016). “Cell-Cycle Gene Alterations in 4,864 Tumors Analyzed by Next-Generation Sequencing: Implications for Targeted Therapeutics”. Molecular Cancer Therapeutics15 (7): 1682–1690. doi:10.1158/1535-7163.MCT-16-0071PMID 27196769.
  9.  “ESMO 2024 – combos could be the way forward for CDK2”ApexOnco. 15 September 2024.
  10.  Palmer CL, Boras B, Pascual B, Li N, Li D, Garza S, et al. (March 2025). “CDK4 selective inhibition improves preclinical anti-tumor efficacy and safety”Cancer Cell43 (3): 464–481.e14. doi:10.1016/j.ccell.2025.02.006PMID 40068598.
  11.  “Pfizer Highlights Diverse Oncology Portfolio and Combination Approaches at ESMO 2024”Pfizer. 2024.
  12.  Pfizer (12 August 2025). A Phase 1/2a Dose Escalation and Expansion Study to Evaluate Safety, Tolerability, Pharmacokinetic, Pharmacodynamic, and Anti-Tumor Activity of Pf-07248144 in Participants With Advanced or Metastatic Solid Tumors (Report). clinicaltrials.gov.
  13.  Pfizer (2 July 2025). An Interventional Safety and Efficacy Phase 1/2, Open-Label Study to Investigate Tolerability, Pk, and Antitumor Activity of Vepdegestrant (Arv-47/Pf-07850327), an Oral Proteolysis Targeting Chimera, in Combination With Pf-07220060 in Participants Aged 18 Years and Older With Er+/her2- Advanced or Metastatic Breast Cancer (Report). clinicaltrials.gov.
  14.  Pfizer (14 November 2024). A Phase 1/2, Open-Label, Multicenter, Dose Escalation and Dose Expansion Study to Evaluate the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Antitumor Activity of PF-07220060 in Combination With Pf-07104091 Plus Endocrine Therapy in Participants With Advanced Solid Tumors (Report). clinicaltrials.gov.
  15.  Pfizer (17 June 2025). (FOURLIGHT-3) (Report). clinicaltrials.gov.
  16.  Pfizer (13 March 2025). An Interventional, Open-Label, Randomized, Multicenter Phase 3 Study of PF-07220060 Plus Letrozole Compared to cdk4/6 Inhibitor Plus Letrozole in Participants Over 18 Years of Age With Hormone Receptor (Hr)-Positive, her2-Negative Advanced/Metastatic Breast Cancer Who Have Not Received Any Prior Systemic Anticancer Treatment for Advanced/Metastatic Disease (FOURLIGHT-1) (Report). clinicaltrials.gov.
  17.  Pfizer (15 November 2024). An Interventional, Open-Label, Randomized, Multicenter, Phase 2 Study of Pf-07220060 Plus Letrozole Compared to Letrozole Alone in Postmenopausal Women 18 Years or Older With Hormone Receptor-Positive, her2-Negative Breast Cancer in the Neoadjuvant Setting (Report). clinicaltrials.gov.
  18.  “Pfizer dials down its atirmociclib ambitions”ApexOnco. 1 May 2025.
Identifiers
IUPAC name
CAS Number2380321-51-5
PubChem CID146219790
ChemSpider115009592
UNIIS743GOJ5LJ
KEGGD12834
ChEMBLChEMBL5187755
Chemical and physical data
FormulaC22H27ClFN5O3
Molar mass463.94 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

///////////Atirmociclib, cyclin-dependent kinase (CDK) inhibitor, antineoplastic, PF 07220060, S743GOJ5LJ, CDK4/6-IN-6

Asaretoclax


Asaretoclax

CAS 2363074-01-3

MF C47H57F2N7O7S, MW 902.1 g/mol

4-[4-[[2-[3-(difluoromethyl)-1-bicyclo[1.1.1]pentanyl]-4,4-dimethylcyclohexen-1-yl]methyl]piperazin-1-yl]-N-[4-[(4-hydroxy-4-methylcyclohexyl)methylamino]-3-nitrophenyl]sulfonyl-2-(1H-pyrrolo[2,3-b]pyridin-5-yloxy)benzamide

2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)-N-((4-((((1r,4r)-4-hydroxy-4-methylcyclohexyl)methyl)amino)-3-nitrophenyl)sulfonyl)benzamide

B-cell lymphoma 2 (Bcl-2) inhibitor, antineoplastic, GY6FD5FXA3, HY 159817, ABT 263

Asaretoclax is an orally bioavailable inhibitor of the anti-apoptotic protein B-cell lymphoma 2 (Bcl-2), with potential pro-apoptotic and antineoplastic activities. Upon oral administration, asaretoclax targets, binds to and inhibits the activity of Bcl-2. This restores apoptotic processes in tumor cells. Bcl-2 is overexpressed in many cancers and plays an important role in the negative regulation of apoptosis; its expression is associated with increased drug resistance and tumor cell survival.

SYN

https://patentscope.wipo.int/search/en/detail.jsf?docId=US309776623&_cid=P21-MJZ42N-73938-1

Example 34

2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1l-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)-N-((4-((((1r,4r)-4-hydroxy-4-methylcyclohexyl)methyl)amino)-3-nitrophenyl)sulfonyl)benzamide

Intermediate 18

Intermediate 18

4-((((1r,4r)-4-hydroxy-4-methylcyclohexyl)methyl)amino)-3-nitrobenzenesulfonamide

 Intermediate 18 was prepared following a procedure described in WO2014/165044A1. LC/MS (ESI) m/z 344.1 [M+H] +.

Intermediate 30

Intermediate 30

2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoic Acid

Step 1: Methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((2-(3-(difluoromethyl)bicyclo[1.1.1]pentan-1-yl)-4,4-dimethylcyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate (Intermediate 30-1) was prepared following the procedure described in Step 1, Route C for Intermediate 28 using Intermediate 24 in place of Intermediate 22. LCMS (ESI) m/z 591.2 [M+H] +.
      Step 2: Intermediate 30 was prepared following the procedure described in Step 5, Route B for Intermediate 26 using Intermediate 30-1 in place of methyl 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxy)-4-(4-((4,4-dimethyl-2-(3-methylbicyclo[1.1.1]pentan-1-yl)cyclohex-1-en-1-yl)methyl)piperazin-1-yl)benzoate. LCMS (ESI) m/z 577.5[M+H] +.

Example 34 was prepared following General Procedure A using Intermediate 30 and Intermediate 18. 1H NMR (400 MHz, DMSO-d 6) δ 11.70 (s, 1H), 11.40 (br s, 1H), 8.59-8.49 (m, 2H), 8.04 (d, J=2.0 Hz, 1H), 7.78 (d, J=8.8 Hz, 1H), 7.53-7.48 (m, 3H), 7.06 (d, J=9.2 Hz, 1H), 6.72 (d, J=7.2 Hz, 1H), 6.38 (s, 1H), 6.25 (s, 1H), 5.99 (t, J=56.8 Hz, 1H), 4.25 (s, 1H), 3.33-3.25 (m, 2H), 3.18-3.05 (m, 4H), 2.97 (s, 2H), 2.40-2.28 (m, 4H), 2.05-1.95 (m, 2H), 1.94 (s, 6H), 1.71-1.59 (m, 5H), 1.58-1.49 (m, 2H), 1.39-1.28 (m, 2H), 1.27-1.20 (m, 2H), 1.18-1.09 (m, 2H), 1.10 (s, 3H), 0.83 (s, 6H); LC/MS (ESI) m/z 902.6 [M+H] +.

SYN

PAT

https://patentscope.wipo.int/search/en/detail.jsf?docId=US384526484&_cid=P21-MJZ3XL-69589-1

PAT

Benzamide compounds

Publication Number: US-2021009543-A1

Priority Date: 2018-01-10

str1

AS ON OCT2025 4.511 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

……

/////////Asaretoclax, B-cell lymphoma 2 (Bcl-2) inhibitor, antineoplastic, GY6FD5FXA3, HY 159817, ABT 263

Zomiradomide


Zomiradomide

CAS 2655656-99-6

MF C45H48F3N7O6S MW871.97

  • N-[2-[4-[[6-[2-[[2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindol-4-yl]amino]ethyl]-2-azaspiro[3.3]heptan-2-yl]methyl]cyclohexyl]-5-(2-hydroxypropan-2-yl)-1,3-benzothiazol-6-yl]-6-(trifluoromethyl)pyridine-2-carboxamide
  • N-[2-[trans-4-[[6-[2-[[2-(2,6-Dioxo-3-piperidinyl)-2,3-dihydro-1,3-dioxo-1H-isoindol-4-yl]amino]ethyl]-2-azaspiro[3.3]hept-2-yl]methyl]cyclohexyl]-5-(1-hydroxy-1-methylethyl)-6-benzothiazolyl]-6-(trifluoromethyl)-2-pyridinecarboxamide

antineoplastic, IRAK degrader-1, AQ5UXV5646

Zomiradomide is an orally active PROTAC degrader for IRAK4 (DC50=6 nM), thereby inhibiting the NF-κB signaling pathway. Zomiradomide acts also as a molecular glue, recruiting Ikaros and Aiolos, and mediating their degradation (DC50 for Ikaros is 1 nM), thereby activating the type I IFN signaling pathway.

Zomiradomide is a small molecule protein degrader of interleukin-1 receptor-associated kinase 4 (IRAK4) and the immunomodulatory imide drug (IMiD) substrates Ikaros (IKZF1) and Aiolos (IKZF3), with potential immunomodulating and antineoplastic activities. Upon administration, zomiradomide modulates the E3 (ubiquitin) ligase and targets IRAK4, Ikaros and Aiolos for ubiquitination. This induces proteasome-mediated degradation of IRAK4, Ikaros and Aiolos. The degradation of IRAK4 inhibits IRAK4-mediated signaling and prevents the activation of IRAK4-mediated nuclear factor-kappa B (NF-kB) signaling and decreases the expression of inflammatory cytokines and certain pro-survival factors. This inhibits the proliferation of IRAK4-overactivated tumor cells, which are found in cells harboring MYD88 activating mutations or those with overactivated toll-like receptor (TLR) pathways. The degradation of the transcription factors Ikaros and Aiolos leads to a downregulation of other proteins, including interferon regulatory factor 4 (IRF4), which upregulates type I interferon signaling and further inhibits NF-kB activation. This leads to apoptosis and the inhibition of tumor cell proliferation. IRAK4, a serine/threonine-protein kinase that plays a key role in both the TLR and IL-1R signaling pathways, is activated though the adaptor protein MYD88 and links the TLR and IL-1R signaling pathway to the NF-kB pathway.

SYN

WO2022027058

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022027058&_cid=P20-MJGJKA-81687-1

Example 1. Synthesis of N-[2-[4-[[6-[2-[[2-(2,6-dioxo-3-piperidyl)-1,3-dioxo-isoindolin-4-yl]amino]ethyl]-2- azaspiro[3.3]heptan-2-yl]methyl]cyclohexyl]-5-(1-hydroxy-1-methyl-ethyl)-1,3-benzothiazol-6-yl]-6-(trifluoromethyl)pyridine-2-carboxamide (Compound A)

[00349] To a solution of 4-[2-(2-azaspiro[3.3]heptan-6-yl)ethylamino]-2-(2,6-dioxo-3-piperidyl)isoindoline -1,3-dione (75.8 mg, 148 umol, TFA salt, Intermediate ATH) in THF (2 mL) was added TEA (15.0 mg, 148 umol), then the mixture stirred at 25 °C for 10 min. Next, HOAc (8.92 mg, 148 umol) and N-[2-(4-formylcyclohexyl)-5-(1-hydroxy-1-methyl-ethyl)-1,3-benzothiazol-6-yl]-6-(trifluoromethyl)pyridine-2-carboxamide (73.0 mg, 148 umol, Intermediate BAX) were added to the mixture and the mixture was stirred at 25 °C for 20 minutes, then NaBH(OAc)3 (62.9 mg, 297 umol) was added to the mixture at 0 °C. The reaction mixture was stirred at 0-25 °C for 2 hours. On completion, the reaction mixture was quenched with H2O (1 mL) and concentrated in vacuo. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18150*25*10 um; mobile phase: [water(0.225%FA)-ACN]; B%: 31%-58%, 9 min) to give the title compound (59.1 mg, 41% yield) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 12.54 (s, 1H), 11.09 (s, 1H), 9.06 (s, 1H), 8.49 – 8.44 (m, 1H), 8.38 (t, J = 8.0 Hz, 1H), 8.19 (d, J = 8.0 Hz, 1H), 7.88 (s, 1H), 7.58 (t, J = 8.0 Hz, 1H), 7.10 – 6.99 (m, 2H), 6.47 (t, J = 5.6 Hz, 1H), 6.07 (s, 1H), 5.05 (dd, J = 5.6, 12.8 Hz, 1H), 3.54 – 3.47 (m, 2H), 3.25 – 3.18 (m, 4H), 3.06 – 2.99 (m, 1H), 2.93 – 2.83 (m, 1H), 2.63 – 2.56 (m, 1H), 2.54 (s, 3H), 2.30 – 2.21 (m, 2H), 2.30 – 2.21 (m, 3H), 2.06 – 1.99 (m, 1H), 1.88 – 1.77 (m, 4H), 1.68 – 1.61 (m, 8H), 1.58 – 1.49 (m, 2H), 1.45 – 1.36 (m, 1H), 1.15 – 1.02 (m, 2H); LC-MS (ESI+) m/z 872.2 (M+H)+.

PAT

str1

AS ON OCT2025 4.511 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

……


[1]. Weiss Matthew M. Preparation of benzothiazole derivatives as IRAK degraders and uses thereof. World Intellectual Property Organization, WO2021127190 A1. 2021-06-24.[2]. Zeng S, et al. Current advances and development strategies of orally bioavailable PROTACs. Eur J Med Chem. 2023 Dec 5;261:115793.  [Content Brief][3]. Weiss MM, et al., Discovery of KT-413, a Targeted Protein Degrader of IRAK4 and IMiD Substrates Targeting MYD88 Mutant Diffuse Large B-Cell Lymphoma. J Med Chem. 2024 Jul 11;67(13):10548-10566.  [Content Brief]

/////////zomiradomide, antineoplastic, IRAK degrader-1, AQ5UXV5646

Zemirciclib


Zemirciclib

CAS 2057509-72-3

MF C22H28ClN5O2, 429.9 g/mol

(1S,3R)-3-acetamido-N-[5-chloro-4-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)pyridin-2-
yl]cyclohexane-1-carboxamide

(1S,3R)-3-acetamido-N-[5-chloro-4-(5,5-dimethyl-4,6-dihydropyrrolo[1,2-b]pyrazol-3-yl)pyridin-2-yl]cyclohexane-1-carboxamide

(1S,3R)-3-acetamido-N-(5-chloro-4-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)pyridin-2-yl)cyclohexanecarboxamide

cyclin-dependent kinase inhibitor, antineoplastic, AZD 4573, UNII-E5XSP3X68B

Zemirciclib is a selective, short-acting inhibitor of the serine/threonine cyclin-dependent kinase 9 (CDK9), the catalytic subunit of the RNA polymerase II (RNA Pol II) elongation factor positive transcription elongation factor b (PTEF-b; PTEFb), with potential antineoplastic activity. Upon intravenous administration, zemirciclib binds to and blocks the phosphorylation and kinase activity of CDK9, thereby preventing PTEFb-mediated activation of RNA Pol II, leading to the inhibition of gene transcription of various anti-apoptotic proteins. This induces cell cycle arrest and apoptosis, and leads to a reduction in tumor cell proliferation. CDK9 regulates elongation of transcription through phosphorylation of RNA polymerase II at serine 2 (p-Ser2-RNAPII). It is upregulated in various tumor cell types and plays a key role in the regulation of Pol II-mediated transcription of anti-apoptotic proteins. Tumor cells are dependent on anti-apoptotic proteins for their survival.

AZD-4573 is a small molecule drug with a maximum clinical trial phase of II and has 1 investigational indication.

  • AZD4573 in Novel Combinations With Anti-cancer Agents in Patients With Advanced Blood CancerCTID: NCT04630756Phase: Phase 1/Phase 2Status: CompletedDate: 2025-04-09
  • AZD4573 as Monotherapy or in Combinations With Anti-cancer Agents in Patients With r/r PTCL or r/r cHLCTID: NCT05140382Phase: Phase 2Status: CompletedDate: 2024-08-28
  • Study to Assess Safety, Tolerability, Pharmacokinetics and Antitumor Activity of AZD4573 in Relapsed/Refractory Haematological MalignanciesCTID: NCT03263637Phase: Phase 1Status: CompletedDate: 2021-10-22

SYN

SYN

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2017001354&_cid=P22-MJC84G-87476-1

Example 14: (1S,3R)-3-acetamido-N-(5-chloro-4-(5,5-dimethyl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)pyridin-2-yl)cyclohexanecarboxamide

PAT

str1

AS ON OCT2025 4.511 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

……

/////////Zemirciclib, cyclin-dependent kinase inhibitor, antineoplastic, AZD 4573, UNII-E5XSP3X68B

Zelenirstat


Zelenirstat

CAS 1215011-08-7

MF C24H30Cl2N6O2S, 537.5 g/mol

2,6-dichloro-N-[1,5-dimethyl-3-(2-methylpropyl)-1Hpyrazol-4-yl]-4-[2-(piperazin-1-yl)pyridin-4-yl]benzene-1-sulfonamide
N-myristoyltransferase inhibitor, antineoplastic, PCLX 001, DDD86481, CCI 002, DDD 86481

Zelenirstat (PCLX-001) is an investigational, oral small-molecule drug that inhibits N-myristoyltransferases (NMTs), enzymes crucial for adding fatty acids to proteins, a process vital for cell signaling and membrane attachment. Developed by Pacylex Pharmaceuticals, it’s being tested for various cancers, showing promise in hematologic cancers like AML and lymphomas, as well as solid tumors, by disrupting cancer cell survival and growth, with early trials indicating good safety and potential efficacy. 

How it works:

  • Targets NMT enzymes: Zelenirstat blocks NMT1 and NMT2, preventing myristoylation (adding a fatty acid) to proteins.
  • Disrupts cancer cell processes: This inhibition interferes with essential cell signaling and stability, especially in cancer cells where NMT expression is altered, leading to cell death (apoptosis).
  • Affects mitochondrial function: It also disrupts mitochondrial complex I and oxidative phosphorylation, vital for leukemia stem cell survival, notes Pacylex Pharmaceuticals. 

Development & Status:

  • Orphan Drug Status: Granted for Acute Myeloid Leukemia (AML).
  • Clinical Trials: A Phase 1 trial demonstrated good safety and early signs of activity in patients with advanced solid tumors and lymphomas, leading to further development.
  • New Drug Class: It represents a novel approach to cancer treatment, distinct from many existing therapies. 

Potential Applications:

  • Acute Myeloid Leukemia (AML)
  • B-cell Lymphomas (like Diffuse Large B-Cell Lymphoma)
  • Colorectal Carcinoma
  • Other cancers, including breast, lung, bladder, and pancreatic cancers, show sensitivity in preclinical models. 

Zelenirstat, also known as PCLX-001, is an investigational new drug that is being evaluated for the treatment of cancer and as an antiviral agent. It is a small molecule inhibitor targets both N-myristoyltransferase 1 (NMT1) and N-myristoyltransferase 2 (NMT2) proteins, which are responsible for myristoylation. Its dual mechanism of action disrupts both cell signaling and energy production in cancer cells.

Zelenirstat is a strong pan-N myristoyl transferase inhibitor, which prevents addition of myristic acid into penultimate glycine of protein with myristoylation signal, and initially has been introduced as anti-tumor drug.[1][2][3] It has completed phase I clinical trial and is going through escalation phase.[4] Its prototype DDD85646 as well as other NMT inhibitors such as IMP-1088 have strong antiviral activities against viruses that required myristoylated proteins to complete their life cycle, including hemorrhagic viruses, such as lassa and argentinian virus, and pox viruses, such as vaccinia and monkeypox.[5][6]

Zelenirstat is an orally bioavailable inhibitor of the enzyme N-myristoyl transferase (NMT), with potential antineoplastic activity. Upon oral administration, zelenirstat targets and binds to NMT, especially NMT type 2 (NMT2). This prevents NMT-mediated signaling and myristoylation. This inhibits proliferation of certain cancer cells in which NMT expression is lost. Zelenirstat also inhibits B-cell receptor (BCR) signaling and reduces the levels of Src-family tyrosine kinases (SFKs). NMTs mediate myristoylation, a key process by which the fatty acid myristate is added to proteins and allows proteins to interact with cell membranes and become part of the cell signaling system. NMT expression is lost in numerous cancers, such as blood cancer cells, thereby making these cells more sensitive to zelenirstat compared to normal cells. The loss of NMT expression may promote tumorigenesis.

Mechanism of action

Zelenirstat acts by inhibiting NMT I and II enzymes, which are required to complete the myristoylation of proteins. Without myristoylation, these proteins are targeted for proteasomal degradation.[7]

PCLX-001 is a first-in-kind N-Myristoyltransferase (NMT) inhibitor being developed by [Pacylex Pharmaceuticals](https://pacylex.com). Current studies have shown that PCLX-001 works differently than other known cancer drugs and has high activity and positive results in breast, lung, bladder and pancreas cancers.

  • Study of PCLX-001 in R/R Advanced Solid Malignancies and B-cell LymphomaCTID: NCT04836195Phase: Phase 1Status: CompletedDate: 2025-04-17
  • Study of Oral PCLX-001 in R/R Acute Myeloid LeukemiaCTID: NCT06613217Phase: Phase 1Status: RecruitingDate: 2025-03-10

REF

SYN

US9156811B2

DDD 86481

https://patentscope.wipo.int/search/en/detail.jsf?docId=US73438944&_cid=P12-MJAUPA-00022-1

INTERMEDIATE 23A

4-Bromo-2,6-dichloro-N-(3-isobutyl-1,5-dimethyl-1H-pyrazol-4-yl)-benzenesulfonamide

      Prepared from 4-bromo-2,6-dichlorobenzenesulfonyl chloride (0.68 g, 2.1 mmol) and 4-amino-1,5-dimethyl-3-isobutyl-1H-pyrazole (0.35 g, 2.1 mmol) in pyridine (5 ml) according to the method of intermediate 1, to give the title compound as a white solid (120 mg, 0.26 mmol, 12%). δH (D-6 DMSO, 300K) 7.65 (2H, s), 6.54 (1H, s), 3.70 (3H, s), 2.17 (3H, s), 1.96 (2H, d J 7.9 Hz), 1.74 (1H, m), 0.78 (6H, d J 6.6 Hz). m/z (ES +, 70V) 456.0 (MH +).

EXAMPLE DDD86481

2,6-Dichloro-N-(3-isobutyl-1,5-dimethyl-1H-pyrazol-4-yl)-4-(2-piperazin-1-yl-pyridin-4-yl)-benzenesulfonamide

      Prepared from the sulphonamide of intermediate 23A (115 mg, 0.25 mmol), 2-(1-piperazinyl)pyridine-4-boronic acid pinacol ester (80 mg, 0.28 mmol), tribasic potassium phosphate (60 mg, 0.28 mmol), and Pd(PPh 3(30 mg, 0.026 mmol) in DMF (1.6 ml) and water (0.4 ml), according to the method of intermediate 11, to give the title compound as a yellow solid (73 mg, 0.14 mmol, 54%). δH (D-6 DMSO, 300K) 8.20, (1H, d J 5.2 Hz), 8.06 (2H, s), 7.15 (1H, s), 7.02 (1H, d J 5.2 Hz), 3.60 (3H, s), 3.52 (4H, m), 2.80 (4H, m), 1.98 (3H, s), 1.92 (2H, d J 7.3 Hz), 1.70 (1H, m), 0.70 (6H, d J 6.6 Hz). m/z (ES +, 70V) 537.2 (MH +).

SYN

WO-2010026365

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2010026365&_cid=P12-MJAUOO-99381-1

PAT

N-myristoyl transferase inhibitors

Publication Number: WO-2010026365-A1

Priority Date: 2008-09-02

str1

AS ON OCT2025 4.511 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

……

References

  1.  Gamma JM, Liu Q, Beauchamp E, Iyer A, Yap MC, Zak Z, et al. (January 2025). “Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signaling and Oxidative Phosphorylation, Killing Acute Myeloid Leukemia Cells”Molecular Cancer Therapeutics24 (1): 69–80. doi:10.1158/1535-7163.MCT-24-0307PMC 11694064PMID 39382188.
  2.  Sangha R, Jamal R, Spratlin J, Kuruvilla J, Sehn LH, Beauchamp E, et al. (August 2024). “A first-in-human phase I trial of daily oral zelenirstat, a N-myristoyltransferase inhibitor, in patients with advanced solid tumors and relapsed/refractory B-cell lymphomas”Investigational New Drugs42 (4): 386–393. doi:10.1007/s10637-024-01448-wPMC 11327210PMID 38837078.
  3.  Sangha RS, Jamal R, Spratlin J, Kuruvilla J, Sehn LH, Weickert M, et al. (June 2024). “Final results of a first-in-human phase I dose escalation trial of daily oral zelenirstat, a n-myristoyltransferase inhibitor, in patients with advanced solid tumors and relapsed/refractory B-cell lymphomas”. Journal of Clinical Oncology42 (16_suppl): 3082. doi:10.1200/JCO.2024.42.16_suppl.3082ISSN 0732-183X.
  4.  Spratlin JL, Sangha RS, Jamal R, Beauchamp E, Berthiaume LG, Mackey JR (20 January 2024). “A first-in-human, open-label, phase I trial of daily oral zelenirstat, an NMT inhibitor, in patients with relapsed/refractory advanced cancer including gastrointestinal cancers”Journal of Clinical Oncology42 (3_suppl): 129–129. doi:10.1200/jco.2024.42.3_suppl.129. Retrieved 19 January 2025.
  5.  Witwit H, Betancourt CA, Cubitt B, Khafaji R, Kowalski H, Jackson N, et al. (August 2024). “Cellular N-Myristoyl Transferases Are Required for Mammarenavirus Multiplication”Viruses16 (9): 1362. doi:10.3390/v16091362PMC 11436053PMID 39339839.
  6.  Witwit H, Cubitt B, Khafaji R, Castro EM, Goicoechea M, Lorenzo MM, et al. (January 2025). “Repurposing Drugs for Synergistic Combination Therapies to Counteract Monkeypox Virus Tecovirimat Resistance”Viruses17 (1): 92. doi:10.3390/v17010092ISSN 1999-4915PMC 11769280.
  7.  Witwit H, Betancourt CA, Cubitt B, Khafaji R, Kowalski H, Jackson N, et al. (August 2024). “Cellular N-Myristoyl Transferases Are Required for Mammarenavirus Multiplication”Viruses16 (9): 1362. doi:10.3390/v16091362PMC 11436053PMID 39339839.
Clinical data
Other namesPCLX-001
Identifiers
IUPAC name
CAS Number1215011-08-7
PubChem CID58561243
DrugBankDB15567
ChemSpider35034199
UNII5HY8BYC3Q6
ChEMBLChEMBL3357685
Chemical and physical data
FormulaC24H30Cl2N6O2S
Molar mass537.50 g·mol−1
3D model (JSmol)Interactive image
SMILES
InChI

////////zelenirstat, N-myristoyltransferase inhibitor, antineoplastic, PCLX 001, DDD86481, CCI 002, DDD 86481

Zamzetoclax


Zamzetoclax

CAS 2388470-64-0

MF C38H46ClN5O6S MW736.32

N-[(3′R,4S,6′R,7′S,8′E,11′S)-7-chloro-7′-methoxy-11′-methyl-13′,15′-dioxospiro[2,3-dihydro-1H-naphthalene-4,22′-20-oxa-13λ6-thia-1,14-diazatetracyclo[14.7.2.03,6.019,24]pentacosa-8,13,16(25),17,19(24)-pentaene]-13′-yl]-3-methoxy-1-methylpyrazole-4-carboxamide

B-cell lymphoma 2 (Bcl-2) inhibitor, antineoplastic, RRS8GZU2UN

Zamzetoclax (compound 1) is a potential Mcl-1 inhibitor.

REFDiscovery of an Oral, Beyond-Rule-of-Five Mcl-1 Protein–Protein Interaction Modulator with the Potential of Treating Hematological Malignancies

Publication Name: Journal of Medicinal Chemistry

Publication Date: 2023-04-28

PMID: 37114951

DOI: 10.1021/acs.jmedchem.2c01953

SYN

WO 2019/222112

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2019222112&_cid=P12-MJ7Z15-98773-1

Example 154

[0447] Example 154 was synthesized in the same manner as Example 18 using 3-methoxy-1-methyl-1H-pyrazole-4-carboxylic acid and Example 109. Example 109 (620 mg, 1.04 mmol) was dissolved in dichloromethane (12 mL). 3-Methoxy-1-methyl-1H-pyrazole-4-carboxylic acid (324 mg, 2.08 mmol, 2 equiv.) and N-(3-dimethylaminopropyl)-N¢-ethylcarbodiimide hydrochloride (400 mg, 2.08 mmol, 2 equiv.) were added. The reaction mixture was stirred for 5 minutes at room temperature before DMAP (253 mg, 2.08 mmol, 2 equiv.) was added in a single portion. The reaction mixture was stirred overnight at room temperature and the progress of the reaction was monitored by LCMS. Upon completion, the reaction mixture was concentrated under reduced pressure, and the residue was purified by Gilson reverse phase prep HPLC (60-100% ACN/H2O with 0.1% TFA) to give Example 154.1H NMR (400 MHz, methanol-d4) d 8.07 (s, 1H), 7.76 (d, J = 8.6 Hz, 1H), 7.34 (d, J = 8.2 Hz, 1H), 7.22– 7.10 (m, 3H), 6.92 (d, J = 8.2 Hz, 1H), 6.20– 6.05 (m, 1H), 5.63 (dd, J = 15.5, 8.0 Hz, 1H), 4.10 (d, J = 12.0 Hz, 1H), 4.06 (s, 4H), 3.91– 3.83 (m, 1H), 3.82 (s, 3H), 3.79 (s, 1H), 3.72 (d, J = 14.4 Hz, 1H), 3.38 (d, J = 14.5 Hz, 1H), 3.30 (s, 3H), 3.09 (dd, J = 15.1, 10.0 Hz, 1H), 2.89– 2.72 (m, 2H), 2.51 (d, J = 26.7 Hz, 2H), 2.24 (dd, J = 10.9, 6.0 Hz, 2H), 2.12 (d, J = 13.7 Hz, 1H), 2.02– 1.70 (m, 4H), 1.54– 1.40 (m, 1H), 1.14 (d, J = 6.1 Hz, 3H). LCMS-ESI+ (m/z): calcd for C38H46ClN5O6S: 735.28; found: 735.94.

SYN

https://patentscope.wipo.int/search/en/detail.jsf;jsessionid=33B420439B8C0CFAAEE83F88092DF1B6.wapp1nC?docId=US413449521&_cid=P12-MJ7YW7-95600-1

WO 2019/222112 discloses novel 3′,4,4′,5-tetrahydro-2H,2′H-spiro[benzo[b][1,4]oxazepine-3,1′-naphthalene] derivatives that are active against MCL-1. For example, Compound 1 (below) has been shown to be an effective MCL-1 inhibitor

SYN

US10703733,

SYN

WO2023215404

PAT

str1

AS ON OCT2025 4.511 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

……

[1]. Salts and polymorphs of certain MCL-1 inhibitors. World Intellectual Property Organization, WO2023215404 A1 2023-11-09.

///////////zamzetoclax, B-cell lymphoma 2 (Bcl-2) inhibitor, antineoplastic, RRS8GZU2UN

Vilzemetkib


Vilzemetkib

CAS 1363402-44-1

MF C36H36F2N4O5 MW 642.7 g/mol

1-N‘-[4-[7-[[1-(cyclopentylamino)cyclopropyl]methoxy]-6-methoxyquinolin-4-yl]oxy-3-fluorophenyl]-1-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide

hepatocyte growth factor receptor inhibitor, antineoplastic, AL 2846, FJ4Y6XP24Y

Vilzemetkib (also known as AL2846) is an investigational, orally active small-molecule drug that acts as a potent inhibitor of the c-Met receptor tyrosine kinase, a protein often overexpressed in cancers, aiming to block tumor growth, survival, and spread by disrupting key cellular signals. It’s being studied in clinical trials, often in combination with other agents like TQB2450 (a PD-L1 inhibitor), for advanced cancers such as esophageal and liver cancer, showing promise in immunotherapy-resistant patients. 

How it Works:

  • Targets c-Met: Vilzemetkib binds to the c-Met protein, preventing its phosphorylation (activation).
  • Blocks Signaling: This action disrupts downstream pathways crucial for cancer cell proliferation, survival, invasion, metastasis, and new blood vessel formation (angiogenesis). 

Development & Use:

  • Developer: Developed by Advenchen Laboratories.
  • Status: Investigational drug, currently in clinical trials.
  • Research Focus: Studied for cancers like esophageal squamous cell carcinoma (ESCC) and hepatocellular carcinoma (HCC). 

Key Information:

  • Chemical Name: 1,1-Cyclopropanedicarboxamide, N-[4-[[7-[[1-(cyclopentylamino)cyclopropyl]methoxy]-6-methoxy-4-quinolinyl]oxy]-3-fluorophenyl]-N′-(4-fluorophenyl)-.
  • Purpose: Potential anti-cancer (antineoplastic) activity. 
  • OriginatorAdvenchen Laboratories
  • DeveloperAdvenchen Laboratories; Chia Tai Tianqing Pharmaceutical Group
  • ClassAntineoplastics; Small molecules
  • Mechanism of ActionReceptor protein-tyrosine kinase antagonists
  • Phase IIINon-small cell lung cancer; Thyroid cancer
  • Phase IILung cancer; Ovarian cancer
  • Phase I/IIColorectal cancer; Neurofibromatosis 1; Pancreatic cancer
  • No development reportedSolid tumours
  • 28 Oct 2025No recent reports of development identified for phase-I development in Solid-tumours(Combination therapy, In the elderly, Late-stage disease, Second-line therapy or greater, In adults) in China (PO, Capsule)
  • 10 Oct 2025700363489: CTP push: KDM and HE updated
  • 26 Aug 2025Chemical structure information added.

Vilzemetkib is an orally bioavailable small molecule inhibitor of the oncoprotein c-Met (hepatocyte growth factor receptor; HGFR), with potential antineoplastic activity. Upon oral administration vilzemetkib targets and binds to the c-Met protein, prevents c-Met phosphorylation and disrupts c-Met-dependent signal transduction pathways. This may induce cell death in tumor cells overexpressing c-Met protein or expressing constitutively activated c-Met protein. c-Met protein is overexpressed or mutated in many tumor cell types and plays key roles in tumor cell proliferation, survival, invasion, metastasis, and tumor angiogenesis.

SYN

[US20120123126]

https://patentscope.wipo.int/search/en/detail.jsf?docId=US73570351&_cid=P20-MJ6JF6-22611-1

EXAMPLE 6

N-(4-(7-((1-(cyclopentylamino)cyclopropyl)methoxy)-6-methoxyquinolin-4-yloxy)-3-fluoro-phenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide

      The title compound was prepared by similar manner to Example 3, by using cyclopentanone instead of tetrahydro-4H-pyran-4-one. Mass: (M+1), 643

SYN

WO-2022268158-A1

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2022268158&_cid=P20-MJ6JJ7-25153-1

WO2012034055 discloses N-(4-((7-((1-(cyclopentylamino)cyclopropyl)methoxy)-6-methoxyquinolone-4-yl)oxy)-3-fluorophenyl)-N-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide (hereinafter referred to as compound (I)) as a c-Met kinase inhibitor and its use in inhibiting tyrosine kinase activity. Compound (I) is a novel class of compounds with excellent pharmacological properties, capable of inhibiting the activity of various protein tyrosine kinases, such as c-Met, VEGFr, EGFr, c-kit, PDGF, FGF, SRC, Ron, Tie2, etc. This disclosure relates to the treatment of neurofibromatosis type I with compound (I).

SYN

WO-2012034055-A2

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2012034055&_cid=P20-MJ6JLU-26634-1

Example 6

N-(4-(7-((1-(cyclopentylamino)cyclopropyl)methoxy)-6-methoxyquinolin-4-yloxy)-3-fluoro-phenyl)-N-(4-fluorophenyl)cyclopropane- 1,1-dicarboxamide

The title compound was prepared by similar manner to Example 3, by using cyclopentanone instead of tetrahydro-4H-pyran-4-one. Mass: (M + 1), 643

PAT

str1

AS ON OCT2025 4.511 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

……

////////Vilzemetkib, hepatocyte growth factor receptor inhibitor, antineoplastic, AL 2846, FJ4Y6XP24Y

Tigozertinib


Tigozertinib

CAS 2660250-10-0

MF C28H37FN6O3S MW 556.7

3-Isoquinolinamine, N-[2-[(3S,4R)-3-fluoro-4-methoxy-1-piperidinyl]-4-pyrimidinyl]-5-(1-methylethyl)-8-[(2R,3S)-2-methyl-3- [(methylsulfonyl)methyl]-1-azetidinyl]-

N-{2-[(3S,4R)-3-fluoro-4-methoxypiperidin-1-yl]pyrimidin-4-yl}-8-{(2R,3S)-3-[(methanesulfonyl)methyl]-2-methylazetidin-1-yl}-5-(propan-2-yl)isoquinolin-3-amine

N-[2-[(3S,4R)-3-fluoro-4-methoxypiperidin-1-yl]pyrimidin-4-yl]-8-[(2R,3S)-2-methyl-3-(methylsulfonylmethyl)azetidin-1-yl]-5-propan-2-ylisoquinolin-3-amine

N-(2-((3S,4R)-3-fluoro-4-methoxypiperidin-l-yl)pyrimidin-4-yl)-5-isopropyl-8-(3-(methylsulfonylmethyl)azetidin-l-yl)isoquinolin-3-amine

epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, antineoplastic, PA4PTH5HL9, BLU 945


Tigozertinib (BLU-945) is currently under investigation in clinical trial NCT04862780 (Phase 1/​2 Study Targeting EGFR Resistance Mechanisms in NSCLC) for the treatment of NSCLC.

Tigozertinib is a fourth-generation, orally bioavailable, mutant-selective, epidermal growth factor receptor (EGFR) inhibitor, with potential antineoplastic activity. Upon oral administration, tigozertinib targets, binds to and inhibits the activity of EGFR with C797S triple mutations including ex19del/T790M/C797S and L858R/T790M/C797S, thereby preventing EGFR-mediated signaling. This may both induce cell death and inhibit tumor growth in EGFR-overexpressing tumor cells. EGFR, a receptor tyrosine kinase mutated in many tumor cell types, plays a key role in tumor cell proliferation and tumor vascularization. BLU-945 inhibits mutated forms of EGFR with C797S mutation, which prevents covalent bond formation with third-generation EGFR inhibitors leading to drug resistance. BLU-945 may have enhanced anti-tumor effects in tumors with C797S-mediated resistance when compared to other EGFR tyrosine kinase inhibitors.Tigozertinib is a fourth-generation, orally bioavailable, mutant-selective, epidermal growth factor receptor (EGFR) inhibitor, with potential antineoplastic activity. Upon oral administration, tigozertinib targets, binds to and inhibits the activity of EGFR with C797S triple mutations including ex19del/T790M/C797S and L858R/T790M/C797S, thereby preventing EGFR-mediated signaling. This may both induce cell death and inhibit tumor growth in EGFR-overexpressing tumor cells. EGFR, a receptor tyrosine kinase mutated in many tumor cell types, plays a key role in tumor cell proliferation and tumor vascularization. BLU-945 inhibits mutated forms of EGFR with C797S mutation, which prevents covalent bond formation with third-generation EGFR inhibitors leading to drug resistance. BLU-945 may have enhanced anti-tumor effects in tumors with C797S-mediated resistance when compared to other EGFR tyrosine kinase inhibitors.

  • First-in-Human, Phase 1b/2a Trial of a Multipeptide Therapeutic Vaccine in Patients With Progressive GlioblastomaCTID: NCT04116658Phase: Phase 1/Phase 2Status: CompletedDate: 2025-11-28
  • (SYMPHONY) Phase 1/2 Study Targeting EGFR Resistance Mechanisms in NSCLCCTID: NCT04862780Phase: Phase 1Status: TerminatedDate: 2025-02-10
  • A Novel Therapeutic Vaccine (EO2401) in Metastatic Adrenocortical Carcinoma, or Malignant Pheochromocytoma/ParagangliomaCTID: NCT04187404Phase: Phase 1/Phase 2Status: TerminatedDate: 2024-11-12

REF

SYN

WO2021133809

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021133809&_cid=P11-MJ29N8-15768-1

SYN

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2021133809&_cid=P11-MJ292P-02302-1

Example 5, Compound 117: Synthesis of (3S,4R)-3-fluoro-l-(4-(5-isopropyl-8-((2R,3S)-2-methyl-3-(methylsulfonylmethyl)azetidin-l-yl)isoquinolin-3-ylamino)pyrimidin-2-yl)-4-methylpiperidin-4-ol

To a solution of 3-chloro-8-[(2R,3S)-3-(methanesulfonylmethyl)-2-methylazetidin-l-yl]-5-(propan-2-yl)isoquinoline(28 g,76.3mmol, from step 1 of Example 3), (3R,4S)-l-(4-aminopyrimidin-2-yl)-3-fluoro-4-methylpiperidin-4-ol(17.2g,76.3mmol, peak 1 from Example B12), CS2CO3 (49.8 g, 152 mmol),C-phos (4.27 g, 9.15mmol, 2-dicyclohexylphosphino-2’,6’-bis(N,N-dimethylamino)biphenyl) and Pd2(dba)3 (3.94 g, 3.81 mmol) in dioxane (400 mL) was heated to 100 °C for 16 h under N2 atmosphere. The mixture reaction was filtered and the filtrate was concentration under vacuum. The residue was applied onto a silica gel column with EA/PE (2: 1) to give product 28.8 g (67%) as a light-yellow solid.

OTHERS

PAT

str1

AS ON OCT2025 4.511 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

……

///////////Tigozertinib, antineoplastic, PA4PTH5HL9, BLU 945

Tambiciclib


Tambiciclib

CAS 2247481-08-7

MF C25H35ClN6O2S, 519.10

4-[[[4-[5-chloro-2-[[4-[[(2R)-1-methoxypropan-2-yl]amino]cyclohexyl]amino]-4-pyridinyl]-1,3-thiazol-2-yl]amino]methyl]oxane-4-carbonitrile

2H-PYRAN-4-CARBONITRILE, 4-(((4-(5-CHLORO-2-((TRANS-4-(((1R)-2-METHOXY-1-METHYLETHYL)AMINO)CYCLOHEXYL)AMINO)-4-PYRIDINYL)-2-THIAZOLYL)AMINO)METHYL)TETRAHYDRO-

cyclin-dependent kinase inhibitor, antineoplastic, GFH 009, JSH 009, XDZ7VK8CXC, Orphan Drug , Acute myeloid leukaemia, Peripheral T-cell lymphoma

Tambiciclib (GFH009, JSH-009) is an orally active, highly potent and selective CDK9 inhibitor (IC50 = 1 nM), demonstrating >200-fold selectivity over other CDKs, >100-fold selectivity over DYRK1A/B, and excellent selectivity over 468 kinases/mutants. Tambiciclib demonstrates potent in vitro and in vivo antileukemic efficacy in acute myeloid leukemia (AML) mouse models by inhibiting RNA Pol II phosphorylation, downregulating MCL1 and MYC, and inducing apoptosis. Tambiciclib can be used for AML research.

Tambiciclib is a selective inhibitor of the serine/threonine cyclin-dependent kinase 9 (CDK9), the catalytic subunit of the RNA polymerase II (RNA Pol II) elongation factor positive transcription elongation factor b (PTEF-b; PTEFb), with potential antineoplastic activity. Upon administration, tambiciclib targets, binds to and blocks the phosphorylation and kinase activity of CDK9, thereby preventing PTEFb-mediated activation of RNA Pol II, leading to the inhibition of gene transcription of various anti-apoptotic proteins. This induces cell cycle arrest and apoptosis and prevents tumor cell proliferation. CDK9 regulates elongation of transcription through phosphorylation of RNA Pol II at serine 2 (p-Ser2-RNAPII). It is upregulated in various tumor cell types and plays a key role in the regulation of Pol II-mediated transcription of anti-apoptotic proteins. Tumor cells are dependent on anti-apoptotic proteins for their survival.

  • OriginatorGenFleet Therapeutics
  • DeveloperGenFleet Therapeutics; Sellas Life Sciences Group
  • ClassAntineoplastics; Small molecules
  • Mechanism of ActionCyclin-dependent kinase 9 inhibitors
  • Orphan Drug StatusYes – Acute myeloid leukaemia; Peripheral T-cell lymphoma
  • Phase IIAcute myeloid leukaemia
  • Phase I/IIDiffuse large B cell lymphoma; Haematological malignancies; Peripheral T-cell lymphoma
  • Phase ISolid tumours
  • PreclinicalColorectal cancer; T-cell prolymphocytic leukaemia
  • 13 Oct 2025Preclinical trials in T-cell prolymphocytic leukaemia (Combination therapy) in USA (Parenteral)
  • 13 Oct 2025Preclinical trials in T-cell prolymphocytic leukaemia (Monotherapy) in USA (Parenteral)
  • 13 Oct 2025Pharmacodynamics data from preclinical studies in T-cell prolymphocytic leukaemia released by SELLAS Life Sciences

CLINICAL

  • A Study of GFH009 in Combination With Zanubrutinib in Subjects With Relapsed or Refractory DLBCLCTID: NCT06375733Phase: Phase 1/Phase 2Status: RecruitingDate: 2025-08-12
  • A Study of GFH009 Monotherapy in Patients with Relapsed or Refractory Peripheral T-cell Lymphoma (PTCL)CTID: NCT05934513Phase: Phase 1/Phase 2Status: RecruitingDate: 2024-12-13

Discovery of 4-(((4-(5-chloro-2-(((1s,4s)-4-((2-methoxyethyl)amino)cyclohexyl)amino)pyridin-4-yl)thiazol-2-yl)amino)methyl)tetrahydro-2H-pyran-4-carbonitrile (JSH-150) as a novel highly selective and potent CDK9 kinase inhibitor

Publication Name: European Journal of Medicinal Chemistry

Publication Date: 2018-10-05

PMID: 30253346

DOI: 10.1016/j.ejmech.2018.09.025

SYN

WO-2020244612-A1

SYN

WO-2024239512-A1

SYN

https://patentscope.wipo.int/search/en/detail.jsf?docId=WO2018192273&_cid=P12-MJ18VV-17351-1

Example 1: Synthesis of 4-(((4-(5-chloro-2-(((1R,4r)-4-(((R)-1-methoxypropyl-2-yl)amino) cyclohexyl)amino)pyridin-4-yl)thiazolyl)amino)methyl)tetrahydro-2H-pyran-4- carboxynitrile

Step 1: Synthesis of 5-chloro-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborhecyclopentan-2-yl)pyridine 

[0102]5-Chloro-2-fluoropyridine-4-boronic acid (0.7 g, 4.46 mmol) and pinacol (0.63 g, 5.35 mmol) were added to 50 mL of toluene, and the mixture was refluxed at 120 °C overnight. TLC showed a small amount of starting material remaining. The reaction mixture was cooled to room temperature and concentrated, then dried by an oil pump to give 0.92 g of a white solid compound, 5-chloro-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborhexacyclopentan-2-yl)pyridine, yield 80%, MS (ESI): m/z 258.1 (M+H) + . 

[0103]Step 2: Synthesis of (S)-1-methoxypropyl-2-yl-4-toluenesulfonyl ester 

[0104]60% sodium hydride (NaH) (6.52 g, 283 mmol) was added to anhydrous tetrahydrofuran (THF) (200 mL). The mixture was cooled to 0 °C in an ice bath under nitrogen protection, and (S)-(+)-1-methoxy-2-propanol (21 g, 233 mmol) was added dropwise. After the addition was complete, the mixture was brought to room temperature and stirred for 1.5 hours. The reaction mixture was then cooled back to 0 °C, and a tetrahydrofuran (THF) solution of p-toluenesulfonyl chloride (45.3 g, 283 mmol) (200 mL) was added dropwise. After the addition was complete, the mixture was stirred overnight at room temperature. TLC showed that the starting material had reacted completely. The reaction mixture was diluted with ethyl acetate (500 mL), and the reaction was quenched by adding water (500 mL) dropwise while cooling in an ice bath. The mixture was separated, and the aqueous phase was extracted once more with ethyl acetate (200 mL). The combined organic phases were washed with water (200 mL) and then with saturated brine (200 mL). The crude product was dried with anhydrous sodium sulfate, filtered, and concentrated to obtain 43 g of a pale yellow oily substance. Column separation (petroleum ether/ethyl acetate = 5/1) yielded 37 g of (S)-1-methoxypropyl-2-yl-4-toluenesulfonyl ester, a pale yellow oily substance, with a yield of 65.1%. MS (ESI): m/z 245.1 (M+H) + . 

[0105]Step 3: Synthesis of (1r,4R)-N 

1 -((R)-1-methoxypropyl-2-yl)cyclohexane-1,4-diamine 

[0106](S)-1-methoxypropyl-2-yl 4-toluenesulfonyl ester (5 g, 20.5 mmol) and trans-1,4-cyclohexanediamine (5.84 g, 51.2 mmol) were added to 50 mL of acetonitrile and heated to 90 °C overnight. The reaction was monitored by TLC until complete. After cooling, the reaction solution was filtered, the filtrate was concentrated, and the residue was dissolved in dichloromethane and separated by silica gel stirring column (dichloromethane/methanol = 10/1) to give 2.5 g of the pale yellow liquid compound (1r,4R)-N 

1 -((R)-1-methoxypropyl-2-yl)cyclohexane-1,4-diamine, yield 65%, MS (ESI): m/z 187.3 (M+H) + . 

[0107]Step 4: Synthesis of tert-butyl 5-bromothiazol-2-ylcarbamate 

[0108]105 g (403 mmol) of 5-bromothiazol-2-amine hydrobromide was suspended in 500 mL of tetrahydrofuran. Dimethylaminopyridine (2.41 g, 20 mmol) was added, resulting in a white turbidity. A tetrahydrofuran solution of di-tert-butyl dicarbonate (105.6 g, 484.6 mmol) was slowly added dropwise, and the reaction was allowed to proceed at room temperature for two days. The reaction solution was concentrated and dissolved in 300 mL of dichloromethane. The solution was mixed with silica gel and separated by column chromatography (petroleum ether/ethyl acetate = 10/1-6/1 gradient elution) to give 45 g of off-white solid, yield 40%. MS (ESI): m/z 278.98 (M+H) + . 

[0109]Step 5: Synthesis of tert-butyl 4-bromothiazol-2-ylcarbamate 

[0110]A 200 mL solution of diisopropylamine (64 mL, 446 mmol) in tetrahydrofuran was added to a dry three-necked flask. Under nitrogen protection, the mixture was cooled to 0 °C, and n-butyllithium (2.5 M, 173 mL, 431.7 mmol) was added dropwise. The reaction was allowed to proceed for 1 hour after the addition was complete. Then, a 400 mL solution of 5-bromothiazol-2-ylcarbamate in tetrahydrofuran was added dropwise at 0 °C. The reaction was allowed to proceed for 2 hours after the addition was complete. TLC showed that the reaction was complete. At 0℃, ice water (5 mL) was slowly added dropwise to quench the reaction. After stirring for 30 minutes, saturated ammonium chloride (500 mL) aqueous solution was added. The mixture was separated, and the aqueous layer was extracted with dichloromethane (2 × 300 mL). The organic layers were combined, washed with saturated brine, dried with anhydrous sodium sulfate, filtered, concentrated, and recrystallized from petroleum ether:ethyl acetate = 30:1. 31 g of tert-butyl 4-bromothiazol-2-ylcarbamate was obtained as a white solid, yield 77.5%. MS (ESI): m/z 278.98 (M+H) + . 

[0111]Step Six: Synthesis of Methyl 4-cyano-tetrahydro-2H-pyran-4-carbonate 

[0112]Methyl cyanoacetate (39.1 g, 395.3 mmol) and 2,2-dibromoethyl ether (100 g, 434.8 mmol) were added to 600 mL of dimethylformamide, followed by DBU (90 g, 593 mmol). The mixture was heated to 85 °C and reacted for 3 hours. TLC showed that the starting material reacted completely. The solid was filtered off, washed with ethyl acetate (2 × 300 mL), and the mother liquor was concentrated to obtain a brown oily substance. The oil was distilled under reduced pressure at an internal temperature of 65-70 °C, and the fraction collected was a colorless liquid. Crystallization was observed to give 42 g of a white solid, 4-cyano-tetrahydro-2H-pyran-4-carbonate. Yield: 62.8%, MS (ESI): m/z 178.2 (M+H) + . 

[0113]Step 7: Synthesis of 4-(hydroxymethyl)-tetrahydro-2H-pyran-4-carboxynitrile 

[0114]4-Cyano-tetrahydro-2H-pyran-4-carbonate methyl ester (42 g, 248.4 mmol) was dissolved in 400 mL of ethylene glycol dimethyl ether and 40 mL of methanol. The mixture was cooled to 0 °C in an ice bath, and sodium borohydride (11.1 g, 149 mmol) was added in portions. After the addition was complete, the mixture was allowed to rise to room temperature and stirred for 16 hours. The reaction was completed by TLC. The reaction solution was concentrated, and methanol was added to quench excess sodium borohydride. The solution was then concentrated again. Column chromatography (petroleum ether/ethyl acetate = 5/1) yielded 28 g of 4-(hydroxymethyl)-tetrahydro-2H-pyran-4-carboxynitrile, a pale yellow oil, yield: 79.5%, MS (ESI): m/z 142.1 (M+H) + . 

[0115]Step 8: Synthesis of tert-butyl (4-bromothiazolyl)((4-cyanotetrahydro-2H-pyran-4-yl)methyl)carbamate 

[0116]4-(hydroxymethyl)-tetrahydro-2H-pyran-4-carboxynitrile, 4-bromothiazol-2-ylcarbamate tert-butyl ester, and triphenylphosphine were added to anhydrous tetrahydrofuran (THF) and cooled to 0°C. Diisopropyl azodicarbonate (DIAD) was added dropwise. The mixture was stirred at room temperature for 10 minutes, then heated to 40°C overnight. The reaction solution was concentrated, and the residue was dissolved in dichloromethane. The solution was mixed with silica gel and separated by column chromatography (petroleum ether/ethyl acetate = 50/1, 30/1, 20/1) to obtain (4-bromothiazol-2-yl)((4-cyanotetrahydro-2H-pyran-4-yl)methyl)carbamate tert-butyl ester, a white solid of 365 mg, yield 50%. MS (ESI): m/z 402.1 (M+H) + . 

[0117]Step Nine: Synthesis of tert-butyl (4-(5-chloro-2-fluoropyridin-4-yl)thiazolyl)((4-cyano-tetrahydro-2H-pyran-4-yl)methyl)carbamate 

[0118]5-Chloro-2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborhexacyclopentan-2-yl)pyridine and sodium carbonate were added to a mixture of dimethyl ether/H₂O 

/ dioxane. The system was purged with nitrogen twice. Then, tert-butyl (4-bromothiazolyl)((4-cyanotetrahydro-2H-pyran-4-yl)methyl)carbamate and tetraphenylphosphine palladium Pd(pph 3 ) 

4 were added . The system was purged with nitrogen three times. The temperature was then raised to 70°C and the reaction was carried out for 6 hours. TLC showed that only half of the starting material remained. Heating was then stopped and the reaction was terminated. The reaction solution was cooled to room temperature, ethyl acetate and methanol were added, and the mixture was filtered. The filter cake was washed with ethyl acetate, the filtrate was concentrated, and the residue was dissolved in dichloromethane. The residue was washed with saturated brine, separated, and the organic phase was dried over anhydrous sodium sulfate. The mixture was filtered, and silica gel was added for mixing. The sample was separated by column chromatography (petroleum ether/ethyl acetate = 30/1) to give 3.2 g of (4-(5-chloro-2-fluoropyridin-4-yl)thiazolyl)((4-cyano-tetrahydro-2H-pyran-4-yl)methyl)carbamate, a white foamy solid, with a yield of 55%. MS (ESI): m/z 453.1 (M+H) + . 

[0119]Step 10: Synthesis of 4-(((4-(5-chloro-2-(((1R,4r)-4-(((R)-1-methoxypropyl-2-yl)amino)cyclohexyl)amino)pyridin-4-yl)thiazolyl)amino)methyl)tetrahydro-2H-pyran-4-carboxynitrile 

[0120]The tert-butyl carbamate (4-(5-chloro-2-fluoropyridin-4-yl)thiazolyl)((4-cyano-tetrahydro-2H-pyran-4-yl)methyl)carbamate (3.2 g, 7.1 mmol) and (1r,4R)-N 

1 -((R)-1-methoxypropyl-2-yl)cyclohexane-1,4-diamine (3.9 g, 21.2 mmol) and diisopropylethylamine (DIPEA) were added to 30 mL of dimethyl sulfoxide. Under nitrogen protection, the mixture was heated to 100-110 °C and reacted for two days. The reaction was monitored by TLC and LCMS. The starting material (4-(5-chloro-2-fluoropyridin-4-yl)thiazolyl)((4-cyano-tetrahydro-2H-pyran-4-yl)methyl)carbamate tert-butyl ester had completely disappeared, with some BOC-free intermediate remaining. The reaction was stopped, and the reaction solution was cooled and diluted with ethyl acetate (60 mL). Water (150 mL) was added under ice bath. The mixture was separated, and the aqueous layer was extracted again with ethyl acetate (2 × 50 mL). The organic layers were combined, washed with saturated brine (100 mL), dried with anhydrous sodium sulfate, filtered, and concentrated to obtain a crude product of yellowish-brown oil. Column separation (acetonitrile/water/trifluoroacetic acid = 80/20/0.001) yielded 700 mg of 4-(((4-(5-chloro-2-(((1R,4r)-4-(((R)-1-methoxypropyl-2-yl)amino)cyclohexyl)amino)pyridin-4-yl)thiazolyl)amino)methyl)tetrahydro-2H-pyran-4-carboxynitrile, a pale yellow solid. Yield: 19.1%. ¹H NMR (400 MHz, CDCl₃ 

) )δ8.06(s,1H),7.38(s,1H),6.97(s,1H),5.92(brs,1H),4.45(d,J=8.0Hz,1H),4.02(dd,J 1=2.8Hz, J2=12Hz,2H),3.71-3.74(m,4H),3.54-3.56(m,1H),3.35(s,3H),3.21-3.25(m,2 H),3.00-3.05(m,1H),2.50-2.60(m,1H),2.15(d,J=9.6Hz,2H),2.04-2.07(m,1H),1.95(d ,J=12.8Hz,3H),1.74-1.82(m,3H),1.10-1.30(m,4H),1.00(d,J=.4Hz,3H),MS(ESI):m/z 519.3(M+H) + .

SYN

https://patentscope.wipo.int/search/en/detail.jsf?docId=US376039987&_cid=P12-MJ18R0-12787-1

PAT

str1

AS ON OCT2025 4.511 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

……

//Tambiciclib, cyclin-dependent kinase inhibitor, antineoplastic, GFH 009, JSH 009, XDZ7VK8CXC, Orphan Drug , Acute myeloid leukaemia, Peripheral T-cell lymphoma