AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

ND 2158

 Uncategorized  Comments Off on ND 2158
Apr 102016
 

(2S)-2-hydroxy-3-[(3R)-12-{[(1r,4r)-4-(morpholin-4-yl)cyclohexyl]oxy}-7-thia-9,11-diazatricyclo[6.4.0.0²,⁶]dodeca-1(12),2(6),8,10-tetraen-3-yl]propanamide

S)-2-hydroxy-3-((R)-4-(((lr,4R)-4-morpholinocyclohexyl)oxy)-6,7-dihydro-5H-cyclopenta [4,5] thieno [2,3-d] pyrimidin-5-yl)propanamide

 CAS 1388896-07-8
C22 H30 N4 O4 S
5H-​Cyclopenta[4,​5]​thieno[2,​3-​d]​pyrimidine-​5-​propanamide, 6,​7-​dihydro-​α-​hydroxy-​4-​[[trans-​4-​(4-​morpholinyl)​cyclohexyl]​oxy]​-​, (αS,​5R)​-
Molecular Weight446.56

STR3

ND 2158

IRAK4, 446.2

C22H30N4O4S

Company Nimbus Therapeutics LLC
Description IL-1 receptor-associated kinase 4 (IRAK4) inhibitor
Molecular Target Interleukin-1 receptor-associated kinase 4 (IRAK4)
Mechanism of Action Interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor
Therapeutic Modality Small molecule

ND-2158 is a potent and selective experimental inhibitor of IRAK4 described in patent WO2013106535 [2] and in a poster presented at the American College of Rheumatology meeting in 2012 (Abstract #1062 in Supplement: Abstracts of the American College of Rheumatology & Association of Rheumatology Health Professionals, Annual Scientific Meeting, November 9-4, 2012 Washington DC, Volume 64, Issue S10, Page S1-S1216).

 

PATENT

WO2013106535

http://www.google.com/patents/WO2013106535A1?cl=en

Figure imgf000085_0001

Figure imgf000086_0001

 

Scheme II

 

Example 88: (S)-l-((R)-4-(((lr,4R)-4-morpholinocyclohexyl)oxy)-6,7-dihydro- 5H-cyclopenta[4,5]thieno[2,3-d]pyrimidin-5-yl)butan-2-ol (1-64) and Example 89: (R)-l- ((R)-4-(((lr,4R)-4-morpholinocyclohexyl)oxy)-6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-

Synthesis of compound 88.1. Note: For the preparation of the starting material compound 29.2, please see Example 29. A solution of

yl)cyclohexyl]oxy]-7-thia-9,l l-diazatricyclo[6.4.0.0[2,6]]dodeca-l(8),2(6),9,l l-tetraen-3- yl]ethan-l-ol (190 mg, 0.47 mmol, 1.00 equiv) in 10 mL of dichloromethane was added Dess- Martin periodinane at 0 °C in a water/ice bath under nitrogen. The resulting mixture was stirred for 2 h at room temperature. After completion of the reaction, the mixture was then diluted with saturated aqueous sodium bicarbonate and extracted with 3 x 30 mL of ethyl acetate. The combined organic layers were dried over sodium sulfate and concentrated under vacuum. The residue was applied onto a silica gel column with ethyl acetate/petroleum ether (1 :5 to 1 : 1) to afford 2-[(3Λ)-12-[[4-^ο 1ιο1ϊη-4-γ1)ογο1ο1ιβχγ1]οχγ]-7-ωΕ-9,11- diazatricyclo[6.4.0.0[2,6]]dodeca-l(8),2(6),9,l l-tetraen-3-yl]acetaldehyde (130 mg, 69%) as a colorless oil. MS (ES): m/z 402 [M+H]+.

Synthesis of Compound 1-64 and Compound 1-65. A solution of [(3i?)-12-[[4- (moφholin-4-yl)cyclohexyl]oxy]-7-thia-9,l l-diazatricyclo[6.4.0.0[2,6]]dodeca-l(8),2(6),9,l l- tetraen-3-yl]acetaldehyde (130 mg, 0.32 mmol, 1.00 equiv) in 5 mL of anhydrous THF was added bromo(ethyl)magnesium (1 M in THF, 0.62 mL, 2.0 equiv) dropwise at 0 °C under nitrogen. The resulting solution was stirred for 4 h at room temperature and then quenched by the addition of saturated aqueous NH4CI and extracted with 3 x 50 mL of DCM/i-PrOH (3:1). The combined organic layers was dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product (150 mg) was purified by preparative HPLC under the following conditions (SHIMADZU): column: SunFire Prep C18, 19*150 mm 5um; mobile phase: water with 0.05% NH4CO3 and CH3CN (6.0% CH3CN up to 54.0% in 25 min); UV detection at 254/220 nm to afford (S)-l-((R)-4-(((lr,4R)-4-moφholinocyclohexyl)oxy)-6,7-dihydro-5H- cyclopenta[4,5]thieno[2,3-d]pyrimidin-5-yl)butan-2-ol (11.8 mg) and (R)-l-((R)-4-(((lr,4R)-4- mo holinocyclohexyl)oxy)-6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidin-5-yl)butan- 2-ol (23.9 mg) as white solids.

Example 88 (1-64): MS: 432 (M+H)+. ¾ NMR (300 MHz, CDC13) S 8.47 (s, 2H), 5.24-5.20 (m, 1H), 3.75-3.58 (m, 5H), 3.06-2.93 (m, 2H), 2.70-2.61 (m, 4H), 2.28-1.98 (m, 3H), 1.59-1.41 (m, 10H), 1.28-1.23 (m, 2H),0.95-0.85 (m, 3H).

Example 89 (1-65): MS: 432 (M+H)+. ¾ NMR (300 MHz, CDC13) S 8.47 (s, 2H), 5.25 (m, 1H), 3.71-3.39 (m, 6H), 3.04-2.90 (m, 2H), 2.67-2.55 (m, 5H), 2.34-2.22 (m, 4H), 2.01- 1.81 (m, 3H), 1.64-1.39 (m, 7H), 0.94-0.92 (m, 3H).

WATCH OUT SYNTHESIS COMING…………

 

PATENT

WO 2014011906

https://www.google.co.in/patents/WO2014011906A2?cl=en

 

PATENT

WO-2014194242

https://www.google.com/patents/WO2014194242A2?cl=en

 

Example 49: Synthesis of Intermediate 49.1.

Image loading...

step 1 step 2

35.1 49.1 49.2 Image loading...

step 3 49 3

] Intermediate 49.3 was prepared from 35.1 in a manner analogous to the synthesis of 36.3. Isolated 150 mg of a white solid in 57% overall yield. MS (ES): m/z 402 [M+H]+.

Example 50: Synthesis of Intermediate 50.4.

Image loading...

49.3 50.1 50.2

Image loading...

50.3 50.4

Intermediate 50.4 was prepared from 49.3 in a manner analogous to the synthesis of 1-25, except that HCl/MeOH rather than TBAF/THF was used in the second step. Isolated 124 mg of a white solid in 48% overall yield. MS (ES): m/z 447 [M+H]+. 1H NMR (400 MHz, CDCls): δ 8.46 (s, 1H), 5.28-5.25 (m, 1H), 4.17-4.06 (m, 51H), 3.74-3.72 (m, 5H), 3.37-2.98 (m, 2H), 2.72-2.28 (m, 10H), 2.11-2.08 (m, 2H), 1.79-1.46 (m, 5H).

Example 51: Synthesis of (S)-2-hydroxy-3-((R)-4-(((lr,4R)-4- morpholinocyclohexyl)oxy)-6,7-dihydro-5H-cyclopenta [4,5] thieno [2,3-d] pyrimidin-5- yl)propanamide (1-34) and Example 52: Synthesis of (R)-2-hydroxy-3-((R)-4-(((lr,4R)-4- morpholinocyclohexyl)oxy)-6,7-dihydro-5H-cyclopenta [4,5] thieno [2,3-d] pyrimidin-5- yl)propanamide (1-44)

Image loading...

The racemic 50.4 (1.6 g, 96.5% purity) was separated by Chiral-HPLC with the following conditions (Gilson G x 281): column: Chiralpak AD-H, 2*25 cm Chiral-P(AD-H); mobile phase: phase A: hex (O. P/oDEA) (HPLC grade), phase B: IPA (HPLC grade), gradient: 30% B in 9 min; flow rate: 20 mL/min; UV detection at 220/254 nm. The former fractions (tR = 4.75 min) were collected and evaporated under reduced pressure and lyophilized overnight to afford 1-44 (520 mg) with 100% ee as a white solid. And the latter fractions (tR = 5.82 min) were handled as the former fractions to give the desired 1-34 (510 mg) with 99.6%> ee as a white solid. The ee values of the two isomers were determined by the chiral-HPLC with the following conditions (SHIMADZU-SPD-20A): column: Chiralpak AD-H, 0.46*25 cm, 5um (DAICEL); mobile phase: hex (0.1% TEA): IPA = 85:15; UV detection at 254 nm. Flow rate: 1.0 mL/min. tR (1-44) = 7.939 min and tR (1-34) = 11.918 min.

[00431] Analytical data for 1-44: MS: (ES, m/z) 447 [M+H]+. 1H NMR (400 MHz, CD3OD+CDCI3): δ 8.47 (s, 1H), 5.32-5.22 (m, 1H), 4.08 (dd, 1H), 4.89-4.62 (m, 5H), 3.20-3.10 (m, 1H), 3.05-2.95 (m, 1H), 2.75-2.55 (m, 5H), 2.44-2.38 (m, 2H), 2.34-2.28 (m, 3H), 2.10 (d, 2H), 1.82-1.62 (m, 3H), 1.58-1.40 (m, 2H).

Analytical data for 1-34: MS: (ES, m/z) 447 [M+H]+. 1H NMR (400 MHz, CDC13): δ 8.46 (s, 1H), 5.32-5.22 (m, 1H), 4.15 (t, 1H), 3.73 (t, 4H), 3.59 (td, 1H), 3.19-3.08 (m, 1H), 3.02- 2.92 (m, 1H), 2.78-2.70 (m, 1H), 2.69-2.60 (m, 4H), 2.58-2.20 (m, 5H), 2.10 (d, 2H), 1.75-1.63 (m, 3H), 1.53-1.40 (m, 2H).

 

Paper

http://pubs.acs.org/doi/abs/10.1021/jm5016044

Recent Advances in the Discovery of Small Molecule Inhibitors of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4) as a Therapeutic Target for Inflammation and Oncology Disorders

Miniperspective

Nimbus Discovery, 25 First Street, Suite 404, Cambridge, Massachusetts 02141, United States
Schrödinger Inc., 120 West Forty-Fifth Street, New York, New York 10036, United States
J. Med. Chem., 2015, 58 (1), pp 96–110
DOI: 10.1021/jm5016044
Abstract Image

IRAK4, a serine/threonine kinase, plays a key role in both inflammation and oncology diseases. Herein, we summarize the compelling biology surrounding the IRAK4 signaling node in disease, review key structural features of IRAK4 including selectivity challenges, and describe efforts to discover clinically viable IRAK4 inhibitors. Finally, a view of knowledge gained and remaining challenges is provided.

STR3

  1. 78 Romero, D. L.; Robinson, S.; Wessel, M. D.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO201401902, January 16, 2014.

  2. 79.

    Harriman, G. C.; Romero, D. L.; Masse, C. E.; Robinson, S.; Wessel, M. D.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO2014011911A2, January 16, 2014.

  3. 80.

    Harriman, G. C.; Wester, R. T.; Romero, D. L.; Masse, C. E.; Robinson, R.; Greenwood, J. R. IRAK Inhibitors and Uses Thereof. WO2014011906A2, January 16, 2014

 

Patent ID Date Patent Title
US2013231328 2013-09-05 IRAK INHIBITORS AND USES THEREOF

PATENT

STR3

 

WO 2014194242

WO 2013106535

WO 2012097013

US20070155777 * Feb 21, 2007 Jul 5, 2007 Amgen, Inc. Antiinflammation agents
US20100041676 * Feb 18, 2010 Hirst Gavin C Kinase inhibitors
US20100143341 * Jun 21, 2006 Jun 10, 2010 Develogen Aktiengesellschaft Thienopyrimidines for pharmaceutical compositions
US20120015962 * Jan 19, 2012 Nidhi Arora PYRAZOLO[1,5a]PYRIMIDINE DERIVATIVES AS IRAK4 MODULATORS
US20120283238 * Nov 8, 2012 Nimbus Iris, Inc. Irak inhibitors and uses thereof
References
1. Chaudhary D, Robinson S, Romero DL. (2015)
Recent Advances in the Discovery of Small Molecule Inhibitors of Interleukin-1 Receptor-Associated Kinase 4 (IRAK4) as a Therapeutic Target for Inflammation and Oncology Disorders.
J. Med. Chem.58 (1): 96-110. [PMID:25479567]
2. Harriman GC, Wester RT, Romero DL, Robinson S, Shelley M, Wessel MD, Greenwood JR, Masse CE, Kapeller-Libermann R. (2013)
Irak inhibitors and uses thereof.
Patent number: WO2013106535. Assignee: Nimbus Iris, Inc.. Priority date: 18/07/2013. Publication date: 10/01/2012.

http://nimbustx.com/sites/default/files/uploads/posters/irak4_nimbus_acr_poster_2012_small.pdf

///////ND 2158, IRAK4, ND-2158, NIMBUS, 1388896-07-8

NC(=O)C(CC1CCc2c1c1c(ncnc1s2)OC1CCC(CC1)N1CCOCC1)O

C1CC(CCC1N2CCOCC2)OC3=C4C5=C(CCC5CC(C(=O)N)O)SC4=NC=N3

Share

Curis and Aurigene’s AUPM 170, CA 170

 Uncategorized  Comments Off on Curis and Aurigene’s AUPM 170, CA 170
Apr 082016
 

Curis, Inc.

 

1,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds of formula (I):

ONE EXAMPLE

 

OR

1,3,4-oxadiazole and 1 ,3,4-thiadiazole compounds of formula (I):

STR3

EXAMPLES

STR3

PREDICTED AUPM 170, CA 170, AUPM-170, CA-170

STR3

Synthesis coming………….

WATCH THIS SPACE

 

Aurigene Discovery Technologies Limited INNOVATOR

Curis with the option to exclusively license Aurigene’s orally-available small molecule antagonist of programmed death ligand-1 (PD-L1) in the immuno-oncology field

Addressing immune checkpoint pathways is a well validated strategy to treat human cancers and the ability to target PD-1/PD-L1 and other immune checkpoints with orally available small molecule drugs has the potential to be a distinct and major advancement for patients.

Through its collaboration with Aurigene, Curis is now engaged in the discovery and development of the first ever orally bioavailable, small molecule antagonists that target immune checkpoint receptor-ligand interactions, including PD-1/PD-L1 interactions.  In the first half of 2016, Curis expects to file an IND application with the U.S. FDA to initiate clinical testing of CA-170, the first small molecule immune checkpoint antagonist targeting PD-L1 and VISTA.  The multi-year collaboration with Aurigene is focused on generation of small molecule antagonists targeting additional checkpoint receptor-ligand interactions and Curis expects to advance additional drug candidates for clinical testing in the coming years. The next immuno-oncology program in the collaboration is currently targeting the immune checkpoints PD-L1 and TIM3.

In November 2015, preclinical data were reported. Data demonstrated tha the drug rescued and sustained activation of T cells functions in culture. CA-170 resulted in anti-tumor activity in multiple syngeneic tumor models including melanoma and colon cancer. Similar data were presented at the 2015 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference in Boston, MA

By August 2015, preclinical data had been reported. Preliminary data demonstrated that in in vitro studies, small molecule PD-L1 antagonists induced effective T cell proliferation and IFN-gamma production by T cells that were specifically suppressed by PD-L1 in culture. The compounds were found to have effects similar to anti-PD1 antibodies in in vivo tumor models

 

(Oral Small Molecule PD-L1/VISTAAntagonist)

Certain human cancers express a ligand on their cell surface referred to as Programmed-death Ligand 1, or PD-L1, which binds to its cognate receptor, Programmed-death 1, or PD-1, present on the surface of the immune system’s T cells.  Cell surface interactions between tumor cells and T cells through PD-L1/PD-1 molecules result in T cell inactivation and hence the inability of the body to mount an effective immune response against the tumor.  It has been previously shown that modulation of the PD-1 mediated inhibition of T cells by either anti-PD1 antibodies or anti-PD-L1 antibodies can lead to activation of T cells that result in the observed anti-tumor effects in the tumor tissues.  Therapeutic monoclonal antibodies targeting the PD-1/PD-L1 interactions have now been approved by the U.S. FDA for the treatment of certain cancers, and multiple therapeutic monoclonal antibodies targeting PD-1 or PD-L1 are currently in development.

In addition to PD-1/PD-L1 immune regulators, there are several other checkpoint molecules that are involved in the modulation of immune responses to tumor cells1.  One such regulator is V-domain Ig suppressor of T-cell activation or VISTA that shares structural homology with PD-L1 and is also a potent suppressor of T cell functions.  However, the expression of VISTA is different from that of PD-L1, and appears to be limited to the hematopoietic compartment in tissues such as spleen, lymph nodes and blood as well as in myeloid hematopoietic cells within the tumor microenvironment.  Recent animal studies have demonstrated that combined targeting/ blockade of PD-1/PD-L1 interactions and VISTA result in improved anti-tumor responses in certain tumor models, highlighting their distinct and non-redundant functions in regulating the immune response to tumors2.

As part of the collaboration with Aurigene, in October 2015 Curis licensed a first-in-class oral, small molecule antagonist designated as CA-170 that selectively targets PD-L1 and VISTA, both of which function as negative checkpoint regulators of immune activation.  CA-170 was selected from the broad PD-1 pathway antagonist program that the companies have been engaged in since the collaboration was established in January 2015.  Preclinical data demonstrate that CA-170 can induce effective proliferation and IFN-γ (Interferon-gamma) production (a cytokine that is produced by activated T cells and is a marker of T cell activation) by T cells that are specifically suppressed by PD-L1 or VISTA in culture.  In addition, CA-170 also appears to have anti-tumor effects similar to anti-PD-1 or anti-VISTA antibodies in multiple in vivo tumor models and appears to have a good in vivo safety profile.  Curis expects to file an IND and initiate clinical testing of CA-170 in patients with advanced tumors during the first half of 2016.

Curis, Inc.

Jan 21, 2015

Curis and Aurigene Announce Collaboration, License and Option Agreement to Discover, Develop and Commercialize Small Molecule Antagonists for Immuno-Oncology and Precision Oncology Targets

— Agreement Provides Curis with Option to Exclusively License Aurigene’s Antagonists for Immuno-Oncology, Including an Antagonist of PD-L1 and Selected Precision Oncology Targets, Including an IRAK4 Kinase Inhibitor —

— Investigational New Drug (IND) Application Filings for Both Initial Collaboration Programs Expected this Year —

— Curis to issue 17.1M shares of its Common Stock as Up-front Consideration —

— Management to Host Conference Call Today at 8:00 a.m. EST —

LEXINGTON, Mass. and BANGALORE, India, Jan. 21, 2015 (GLOBE NEWSWIRE) — Curis, Inc. (Nasdaq:CRIS), a biotechnology company focused on the development and commercialization of innovative drug candidates for the treatment of human cancers, and Aurigene Discovery Technologies Limited, a specialized, discovery stage biotechnology company developing novel therapies to treat cancer and inflammatory diseases, today announced that they have entered into an exclusive collaboration agreement focused on immuno-oncology and selected precision oncology targets. The collaboration provides for inclusion of multiple programs, with Curis having the option to exclusively license compounds once a development candidate is nominated within each respective program. The partnership draws from each company’s respective areas of expertise, with Aurigene having the responsibility for conducting all discovery and preclinical activities, including IND-enabling studies and providing Phase 1 clinical trial supply, and Curis having responsibility for all clinical development, regulatory and commercialization efforts worldwide, excluding India and Russia, for each program for which it exercises an option to obtain a license.

The first two programs under the collaboration are an orally-available small molecule antagonist of programmed death ligand-1 (PD-L1) in the immuno-oncology field and an orally-available small molecule inhibitor of Interleukin-1 receptor-associated kinase 4 (IRAK4) in the precision oncology field. Curis expects to exercise its option to obtain exclusive licenses to both programs and file IND applications for a development candidate from each in 2015.

“We are thrilled to partner with Aurigene in seeking to discover, develop and commercialize small molecule drug candidates generated from Aurigene’s novel technology and we believe that this collaboration represents a true transformation for Curis that positions the company for continued growth in the development and eventual commercialization of cancer drugs,” said Ali Fattaey, Ph.D., President and Chief Executive Officer of Curis. “The multi-year nature of our collaboration means that the parties have the potential to generate a steady pipeline of novel drug candidates in the coming years. Addressing immune checkpoint pathways is now a well validated strategy to treat human cancers and the ability to target PD-1/PD-L1 and other immune checkpoints with orally available small molecule drugs has the potential to be a distinct and major advancement for patients. Recent studies have also shown that alterations of the MYD88 gene lead to dysregulation of its downstream target IRAK4 in a number of hematologic malignancies, including Waldenström’s Macroglobulinemia and a subset of diffuse large B-cell lymphomas, making IRAK4 an attractive target for the treatment of these cancers. We look forward to advancing these programs into clinical development later this year.”

Dr. Fattaey continued, “Aurigene has a long and well-established track record of generating targeted small molecule drug candidates with bio-pharmaceutical collaborators and we have significantly expanded our drug development capabilities as we advance our proprietary drug candidates in currently ongoing clinical studies. We believe that we are well-positioned to advance compounds from this collaboration into clinical development.”

CSN Murthy, Chief Executive Officer of Aurigene, said, “We are excited to enter into this exclusive collaboration with Curis under which we intend to discover and develop a number of drug candidates from our chemistry innovations in the most exciting fields of cancer therapy. This unique collaboration is an opportunity for Aurigene to participate in advancing our discoveries into clinical development and beyond, and mutually align interests as provided for in our agreement.  Our scientists at Aurigene have established a novel strategy to address immune checkpoint targets using small molecule chemical approaches, and have discovered a number of candidates that modulate these checkpoint pathways, including PD-1/PD-L1. We have established a large panel of preclinical tumor models in immunocompetent mice and can show significant in vivo anti-tumor activity using our small molecule PD-L1 antagonists.  We are also in the late stages of selecting a candidate that is a potent and selective inhibitor of the IRAK4 kinase, demonstrating excellent in vivo activity in preclinical tumor models.”

In connection with the transaction, Curis has issued to Aurigene approximately 17.1 million shares of its common stock, or 19.9% of its outstanding common stock immediately prior to the transaction, in partial consideration for the rights granted to Curis under the collaboration agreement. The shares issued to Aurigene are subject to a lock-up agreement until January 18, 2017, with a portion of the shares being released from the lock-up in four equal bi-annual installments between now and that date.

The agreement provides that the parties will collaborate exclusively in immuno-oncology for an initial period of approximately two years, with the option for Curis to extend the broad immuno-oncology exclusivity.

In addition Curis has agreed to make payments to Aurigene as follows:

  • for the first two programs: up to $52.5 million per program, including $42.5 million per program for approval and commercial milestones, plus specified approval milestone payments for additional indications, if any;
  • for the third and fourth programs: up to $50 million per program, including $42.5 million per program for  approval and commercial milestones, plus specified approval milestone payments for additional indications, if any; and
  • for any program thereafter: up to $140.5 million per program, including $87.5 million per program in approval and commercial milestones, plus specified approval milestone payments for additional indications, if any.

Curis has agreed to pay Aurigene royalties on any net sales ranging from high single digits to 10% in territories where it successfully commercializes products and will also share in amounts that it receives from sublicensees depending upon the stage of development of the respective molecule.
About Immune Checkpoint  Modulation and Programmed Death 1 Pathway

Modulation of immune checkpoint pathways has emerged as a highly promising therapeutic approach in a wide range of human cancers. Immune checkpoints are critical for the maintenance of self-tolerance as well as for the protection of tissues from excessive immune response generated during infections. However, cancer cells have the ability to modulate certain immune checkpoint pathways as a mechanism to evade the immune system. Certain immune checkpoint receptors or ligands are expressed by various cancer cells, targeting of which may be an effective strategy for generating anti-tumor activity. Some immune-checkpoint modulators, such as programmed death 1 (PD-1) protein, specifically regulate immune cell effector functions within tissues. One of the mechanisms by which tumor cells block anti-tumor immune responses in the tumor microenvironment is by upregulating ligands for PD-1, such as PD-L1. Hence, targeting of PD-1 and/or PD-L1 has been shown to lead to the generation of effective anti-tumor responses.
About Curis, Inc.

Curis is a biotechnology company focused on the development and commercialization of novel drug candidates for the treatment of human cancers. Curis’ pipeline of drug candidates includes CUDC-907, a dual HDAC and PI3K inhibitor, CUDC-427, a small molecule antagonist of IAP proteins, and Debio 0932, an oral HSP90 inhibitor. Curis is also engaged in a collaboration with Genentech, a member of the Roche Group, under which Genentech and Roche are developing and commercializing Erivedge®, the first and only FDA-approved medicine for the treatment of advanced basal cell carcinoma. For more information, visit Curis’ website at www.curis.com.

About Aurigene

Aurigene is a specialized, discovery stage biotechnology company, developing novel and best-in-class therapies to treat cancer and inflammatory diseases. Aurigene’s Programmed Death pathway program is the first of several immune checkpoint programs that are at different stages of discovery and preclinical development. Aurigene has partnered with several large- and mid-pharma companies in the United States and Europe and has delivered multiple clinical compounds through these partnerships. With over 500 scientists, Aurigene has collaborated with 6 of the top 10 pharma companies. Aurigene is an independent, wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (NYSE:RDY). For more information, please visit Aurigene’s website at http://aurigene.com/.

 

POSTER

STR3

STR3

 

STR3

WO2011161699, WO2012/168944, WO2013144704 and WO2013132317 report peptides or peptidomimetic compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

PATENT

WO 2015033299

Example 5: Synthesis of

The compound was synthesised using similar procedure as depicted in Example 4 (compound 4) using D-amino acids are linked up in reverse order. Boc-D-Thr(‘Bu)-OH was used in place of Boc-Ser(‘Bu)-OH, Fmoc-D-Asn(trt)-OH in place of Fmoc-Asn(trt)-OH and H-D-Ser(‘Bu)-0’Bu was used in place of H-Thr^Bu^O’Bu to yield 0.3 g crude material of the title compound. The cmde solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.3 (M+H)+. HPLC: tR = 13.58 min.

Example 8: Synthesis of

The compound was synthesised using similar procedure as depicted in Example 2 (compound 2) using Fmoc-Glu(0’Bu)-OH instead of Fmoc-Asn(Trt)-OH to get 0.4 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 362.1 (M+H)+. HPLC: tR = 13.27 min.

 

PATENT

WO2015033301

Example 3: Synthesis of compound 3

Step 3a:

3a

Lawesson’s reagent (2.85 g, 7.03 mmol) was added to a solution of compound 2e (4 g, 4.68 mmol) in THF (40 mL) and stirred at 75°C for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced

pressure and the obtained residue was partitioned between ice water and ethyl acetate. The organic layer was washed with NaHCC>3 solution followed brine solution. The organic layer was dried over Na2S04, filtered and evaporated under reduced pressure to get residue which was further purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in hexane) to afford 2.7 g of compound 3a (Yield: 67.66%). LCMS: 852.3 (M+H)+,

Step 3

3a 3b

Fmoc group on compound 3a was deprotected by adding diethylamine (3.8 mL) to the solution of compound 3a (1 g, 1.17 mmol) in CH2CI2 (3.8 mL). The reaction mixture was stirred at room temperature for 30 min. The resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by neutral alumina column chromatography (eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to attain 0.62 g of compound 3b. LCMS: 630.5 (M+H)+.

Step 3c

To a solution of compound 3b (0.6 g) in CH2CI2 (7.5 mL), trifluoroacetic acid (2.5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h. The resulting solution was concentrated in vacuum to get 0.13 g of compound 3 which was purified by preparative HPLC method described under experimental conditions. LCMS: 232.3 (M+H)+.

Example 1: Synthesis of compound 1

Step la:

Potassium carbonate (7.9 g, 57.39 mmol) and Methyl iodide (1.3 mL, 21.04 mmol) were added to a solution of compound la (5.0 g, 19.13 mmol) in DMF (35 mL) and stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2S04 and evaporated under reduced pressure to get 5.0 g of compound lb (Yield: 96.1%). LCMS: 176.1 (M-Boc)+.

Step lb:

Hydrazine hydrate (7.2 mL) was added to a solution of compound lb (5.0 g, 18.16 mmol) in methanol (30 mL) and stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure, the residue obtained was partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2S04 and evaporated under reduced pressure to get 4.0 g of compound lc (Yield: 80.0%). LCMS: 276.3 (M+H)+. Step lc:

NMM (0.67 ml, 6.52 mmol) was slowly added to a stirred solution of lc (1.2 g, 4.35 mmol), Id (1.43 g, 4.35 mmol), HOBt (0.7 g, 5.22 mmol) and EDC.HC1 (0.99 g, 5.22 mmol) in DMF (15 mL) at 0°C. The reaction mixture was stirred at room temperature for 12 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice and the solid precipitated was filtered and dried under vacuum to obtain 2.0 g of pure product le (Yield: 83.3%). LCMS: 591.5 (M+Na)+.

St

1 e

1f

To a stirred solution of le (1.5 g, 2.63 mmol) in dry THF (15.0 mL) and DMF (5.0 mL) triphenylphosphine (1.38 g, 5.27 mmol) and iodine (1.33 g, 5.27 mmol) were added at 0°C. After the iodine was completely dissolved, Et3N (1.52 mL, 10.54 mmol) was added to this reaction mixture at ice cold temperature. Reaction mixture was allowed to attain room temperature and stirred for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water and extracted with ethyl acetate. Organic layer was washed with saturated sodium thiosulphate and brine solution.

The separated Organic layer was dried over Na2SC>4 and evaporated under reduced pressure to get residue, which was further purified by silica gel column chromatography (eluent: 30% ethyl acetate in hexane) to afford 0.8 g of compound If (Yield: 55%). LCMS: 551.3 (M+H)+.

Step le:

1f i g

Fmoc group was deprotected by the addition of diethylamine (20.0 mL) to a solution of compound If (0.8 g, 1.45 mmol) in CH2CI2 (20.0 mL) at 0°C. The reaction was stirred at room temperature for 2 h. The resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by neutral alumina column chromatography (eluent: 2% methanol in chloroform) to afford 0.38 g of compound lg (Yield: 80.0%): LCMS: 329.4 (M+H)+.

Step If:

ig 1 i

Compound lg (0.38 g, 1.16 mmol), TEA (0.33 mL, 2.32 mmol) dissolved in DMF (10 mL) were added drop wise to a solution of lh (0.55 g, 1.39 mmol) at 0°C for urea bond formation and the mixture was stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water, the solid precipitated was filtered and dried under vacuum to get crude compound, which was further purified by silica gel column chromatography (eluent: 0-35% ethyl acetate in hexane) to get 0.4 g of product li (Yield: 59.7%). LCMS: 586.4 (M+H)+.

Step lg:

BocHN’ IJ, H LT Y~™

1

To a solution of compound li (0.4 g, 0.68 mmol) in CH2CI2 (5 m L), trifluoro acetic acid (5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h to remove the acid sensitive protecting groups. The resulting solution was concentrated under nitrogen and the solid material was purified by preparative HPLC method as described under experimental conditions (Yield: 0.05 g). LCMS: 318.0 (M+H)+; HPLC: tR= 10.96 min.

Synthesis of compound lh (N02-C6H4-OCO-Thr(tBu)- 0¾u):

To a solution of 4-nitrophenylchloroformate (4.79 g, 23.77 mmol) in DCM (25.0 mL) was added a solution of H-Thr(tBu)-OtBu (5.0 g, 21.61 mmol) TEA (6.2 mL, 43.22 mmol) in CH2CI2 (25 mL) slowly at 0°C and allowed to stir for 30 min. The completion of the reaction was confirmed by TLC analysis. After completion of reaction it was diluted with DCM and washed with 1.0 M of citric acid followed by 1.0 M sodium carbonate solution. The organic layer was dried over Na2S04 and evaporated under reduced pressure to afford crude compound 1 h, which was further purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in hexane) to get 3.0 g of product lh. jH NMR (CDCI3, 400 MHz): £1.17 (s, 9H), 1 .28 (d, 3H), .50 (s, 9H), 4.11 (m, 1 H), 4.28 (m, 1H , 5.89 (d, 1H), 7.37 (d, 2H), 8.26 (d, 2H).

Pottayil Sasikumar

Pottayil Sasikumar

Ph D
Associate Research Director
Bengaluru · Medicinal Chemistry

 

Murali Ramachandra

Murali Ramachandra

PhD
Senior Vice President
Aurigene Discovery Technologie…, Bengaluru · Preclinical Biology
Sudarshan N.S

Scientist at Aurigene Discovery Technologies Limited

Nagaraj Gowda

Nagaraj Gowda

Group lead-immunology, Aurigene Discovery Technologies Ltd.

 Susanta Samajdar

Research Director at Aurigene Discovery Technologies

Brahma Reddy V, Thomas Antony, Murali Ramachandra, Venkateshwar Rao G, Wesley Roy Balasubramanian, Kishore Narayanan, Samiulla DS, Aravind AB, and Shekar Chelur.

REFERENCES

US20150073024

WO2011161699A2 27 Jun 2011 29 Dec 2011 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2012168944A1 21 Dec 2011 13 Dec 2012 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
WO2013132317A1 4 Mar 2013 12 Sep 2013 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
WO2013144704A1 28 Mar 2013 3 Oct 2013 Aurigene Discovery Technologies Limited Immunomodulating cyclic compounds from the bc loop of human pd1

http://www.curis.com/pipeline/immuno-oncology/pd-l1-antagonist

http://www.curis.com/images/stories/pdfs/posters/Aurigene_PD-L1_VISTA_AACR-NCI-EORTC_2015.pdf

////////Curis and Aurigene,  AUPM 170, CA 170, AUPM-170, CA-170, PD-L1, VISTA antagonist

Share

Curis and Aurigene’s CA 4948, AU 4948

 Uncategorized  Comments Off on Curis and Aurigene’s CA 4948, AU 4948
Apr 082016
 

Curis, Inc.

 

STR3

CHEMBL3353198.png

Example 13 WO2015104688

6-(6-aminopyridin-3-yl)-N-(2-morpholin-4-yl-1,3-benzothiazol-6-yl)pyridine-2-carboxamide

Molecular Formula: C22H20N6O2S
Molecular Weight: 432.4982 g/mol
1428335-77-6
[2,​3′-​Bipyridine]​-​6-​carboxamide, 6′-​amino-​N-​[2-​(4-​morpholinyl)​-​6-​benzothiazolyl]​-

PROBABLE STRUCTURE

Example 1 ……..6′-amino-N-(2-morpholinooxazolo[4,5-b]pyridin-6-yl)-[2,3′-bipyridine]-6-carboxamideWO2015104688

STR3

Compound-6:  6′-amino-N-(5-(cyclopropyIamino)-2-morpholinobenzo [d]oxazoI-6-yl)-[2,3′-bipyridine]-6-carboxamide.WO2013042137

PROBABLE CA 4948, AU 4948,AU-4948, CA-4948

STRUCTURE AND SYNTHESIS COMING……..

Company Aurigene Discovery Technologies Ltd.
Description Oral IL-1 receptor-associated kinase 4 (IRAK4) inhibitor
Molecular Target Interleukin-1 receptor-associated kinase 4 (IRAK4)
Mechanism of Action
Therapeutic Modality Small molecule
Latest Stage of Development Preclinical
Standard Indication B cell lymphoma
Indication Details Treat diffuse large B cell lymphoma (DLBCL)
Regulatory Designation
Partner Curis Inc.

Interleukin-1 Receptor Associated Kinase-4 (IRAK-4) is a serine/threonine protein kinase belonging to tyrosine like kinase (TLK) family. IRAK-4 is one of the important signalling components downstream of IL-1/Toll family of receptors (IL-1R, IL-18R, IL-33R, Toll-like receptors). Recent studies have reported occurrence of oncogenic mutations in MYD88 in 30% of ABC diffuse large B cell lymphomas (ABC DLBCL) and 90% of Waldenstrom’s macroglobulinemia (WM). Most of ABC DLBCLs have a single amino acid substitution of proline for the leucine at position 265 (L265P) in the TIR domain of MYD88 protein resulting in constitutive activation of IRAK-4. Thus, IRAK4 is an attractive therapeutic target for the treatment of B-cell lymphomas with activating MYD88 L265P mutation. We have designed, synthesized and tested small molecule IRAK-4 inhibitors based on hits originating from Aurigene’ s compound library. These novel compounds were profiled for IRAK4 kinase inhibition, anti-proliferative activity, kinase selectivity, and drug-like properties. Furthermore, selected compounds were tested in a proliferation assay and pIRAK1 mechanistic assay using ABC-DLBCL cell lines with activating MYD88 L265P mutation, OCI-lLy10 and OCI-lLy3. We have identified a series of novel bicyclic heterocycles as potent inhibitors of IRAK-4. Aurigene Lead compound exhibited potent inhibitory activity for IRAK-4 with an IC50 of 3nM in biochemical assay. Aurigene Lead compound inhibited pIRAK1 levels, and proliferation of OCI-Ly3 and OCI-Ly10 cells with an IC501of 132nM and 52nM respectively. To the best of our knowledge, Aurigene Lead compound represents the most potent IRAK4 inhibitor reported for target modulation and anti-proliferative activity in DLBCL cell lines with activating MYD88 L265P mutation. Aurigene Lead compound has good oral pharmacokinetic profile in mice and has demonstrated excellent pharmacodynamic effect in an in vivo LPS induced TNF-α model with an ED50 of 3.8 mg/Kg in mice. Preliminary in vitro tox studies indicated clean safety profile. Demonstration of efficacy in OCI-lLy10 mouse tumor model is ongoing. In summary, a series of potent IRAK-4 inhibitors belonging to 3 different chemical series have been discovered and are being evaluated for treatment of B-cell lymphomas.

Curis with the option to exclusively license Aurigene’s orally-available small molecule inhibitor of Interleukin-1 receptor-associated kinase 4 (IRAK4) in the precision oncology field. Curis expects to exercise its option to obtain exclusive licenses to both programs and file IND applications for a development candidate from each in 2015.

Recent studies have also shown that alterations of the MYD88 gene lead to dysregulation of its downstream target IRAK4 in a number of hematologic malignancies, including Waldenström’s Macroglobulinemia and a subset of diffuse large B-cell lymphomas, making IRAK4 an attractive target for the treatment of these cancers.

Curis, Inc.

Jan 21, 2015

Curis and Aurigene Announce Collaboration, License and Option Agreement to Discover, Develop and Commercialize Small Molecule Antagonists for Immuno-Oncology and Precision Oncology Targets

— Agreement Provides Curis with Option to Exclusively License Aurigene’s Antagonists for Immuno-Oncology, Including an Antagonist of PD-L1 and Selected Precision Oncology Targets, Including an IRAK4 Kinase Inhibitor —

— Investigational New Drug (IND) Application Filings for Both Initial Collaboration Programs Expected this Year —

— Curis to issue 17.1M shares of its Common Stock as Up-front Consideration —

— Management to Host Conference Call Today at 8:00 a.m. EST —

LEXINGTON, Mass. and BANGALORE, India, Jan. 21, 2015 (GLOBE NEWSWIRE) — Curis, Inc. (Nasdaq:CRIS), a biotechnology company focused on the development and commercialization of innovative drug candidates for the treatment of human cancers, and Aurigene Discovery Technologies Limited, a specialized, discovery stage biotechnology company developing novel therapies to treat cancer and inflammatory diseases, today announced that they have entered into an exclusive collaboration agreement focused on immuno-oncology and selected precision oncology targets. The collaboration provides for inclusion of multiple programs, with Curis having the option to exclusively license compounds once a development candidate is nominated within each respective program. The partnership draws from each company’s respective areas of expertise, with Aurigene having the responsibility for conducting all discovery and preclinical activities, including IND-enabling studies and providing Phase 1 clinical trial supply, and Curis having responsibility for all clinical development, regulatory and commercialization efforts worldwide, excluding India and Russia, for each program for which it exercises an option to obtain a license.

The first two programs under the collaboration are an orally-available small molecule antagonist of programmed death ligand-1 (PD-L1) in the immuno-oncology field and an orally-available small molecule inhibitor of Interleukin-1 receptor-associated kinase 4 (IRAK4) in the precision oncology field. Curis expects to exercise its option to obtain exclusive licenses to both programs and file IND applications for a development candidate from each in 2015.

“We are thrilled to partner with Aurigene in seeking to discover, develop and commercialize small molecule drug candidates generated from Aurigene’s novel technology and we believe that this collaboration represents a true transformation for Curis that positions the company for continued growth in the development and eventual commercialization of cancer drugs,” said Ali Fattaey, Ph.D., President and Chief Executive Officer of Curis. “The multi-year nature of our collaboration means that the parties have the potential to generate a steady pipeline of novel drug candidates in the coming years. Addressing immune checkpoint pathways is now a well validated strategy to treat human cancers and the ability to target PD-1/PD-L1 and other immune checkpoints with orally available small molecule drugs has the potential to be a distinct and major advancement for patients. Recent studies have also shown that alterations of the MYD88 gene lead to dysregulation of its downstream target IRAK4 in a number of hematologic malignancies, including Waldenström’s Macroglobulinemia and a subset of diffuse large B-cell lymphomas, making IRAK4 an attractive target for the treatment of these cancers. We look forward to advancing these programs into clinical development later this year.”

Dr. Fattaey continued, “Aurigene has a long and well-established track record of generating targeted small molecule drug candidates with bio-pharmaceutical collaborators and we have significantly expanded our drug development capabilities as we advance our proprietary drug candidates in currently ongoing clinical studies. We believe that we are well-positioned to advance compounds from this collaboration into clinical development.”

CSN Murthy, Chief Executive Officer of Aurigene, said, “We are excited to enter into this exclusive collaboration with Curis under which we intend to discover and develop a number of drug candidates from our chemistry innovations in the most exciting fields of cancer therapy. This unique collaboration is an opportunity for Aurigene to participate in advancing our discoveries into clinical development and beyond, and mutually align interests as provided for in our agreement.  Our scientists at Aurigene have established a novel strategy to address immune checkpoint targets using small molecule chemical approaches, and have discovered a number of candidates that modulate these checkpoint pathways, including PD-1/PD-L1. We have established a large panel of preclinical tumor models in immunocompetent mice and can show significant in vivo anti-tumor activity using our small molecule PD-L1 antagonists.  We are also in the late stages of selecting a candidate that is a potent and selective inhibitor of the IRAK4 kinase, demonstrating excellent in vivo activity in preclinical tumor models.”

In connection with the transaction, Curis has issued to Aurigene approximately 17.1 million shares of its common stock, or 19.9% of its outstanding common stock immediately prior to the transaction, in partial consideration for the rights granted to Curis under the collaboration agreement. The shares issued to Aurigene are subject to a lock-up agreement until January 18, 2017, with a portion of the shares being released from the lock-up in four equal bi-annual installments between now and that date.

The agreement provides that the parties will collaborate exclusively in immuno-oncology for an initial period of approximately two years, with the option for Curis to extend the broad immuno-oncology exclusivity.

In addition Curis has agreed to make payments to Aurigene as follows:

  • for the first two programs: up to $52.5 million per program, including $42.5 million per program for approval and commercial milestones, plus specified approval milestone payments for additional indications, if any;
  • for the third and fourth programs: up to $50 million per program, including $42.5 million per program for  approval and commercial milestones, plus specified approval milestone payments for additional indications, if any; and
  • for any program thereafter: up to $140.5 million per program, including $87.5 million per program in approval and commercial milestones, plus specified approval milestone payments for additional indications, if any.

Curis has agreed to pay Aurigene royalties on any net sales ranging from high single digits to 10% in territories where it successfully commercializes products and will also share in amounts that it receives from sublicensees depending upon the stage of development of the respective molecule.

About IRAK4:

Interleukin-1 receptor-associated kinase 4, or IRAK4 is a signaling kinase that becomes inappropriately activated in certain cancers including activated B cell-diffuse large B cell lymphoma (ABC-DLBCL), an aggressive form of lymphoma with poor prognosis. There appears to be a mechanistic link with IRAK4 in ABC-DLBCL where these tumors from approximately 35% of patients harbor oncogenic mutations in the MYD88 gene, which encodes an adaptor protein that interacts directly with IRAK4. MYD88 mutations appear to constitutively activate the IRAK4 kinase complex, driving pro-survival pathways in ABC-DLBCL disease. Oncogenic MYD88 mutations have also been identified in other cancers, including in over 90% of patients with Waldenström’s Macroglobulinemia as well as in a subset of patients with chronic lymphocytic leukemia (CLL).

About Curis, Inc.

Curis is a biotechnology company focused on the development and commercialization of novel drug candidates for the treatment of human cancers. Curis’ pipeline of drug candidates includes CUDC-907, a dual HDAC and PI3K inhibitor, CUDC-427, a small molecule antagonist of IAP proteins, and Debio 0932, an oral HSP90 inhibitor. Curis is also engaged in a collaboration with Genentech, a member of the Roche Group, under which Genentech and Roche are developing and commercializing Erivedge®, the first and only FDA-approved medicine for the treatment of advanced basal cell carcinoma. For more information, visit Curis’ website at www.curis.com.

About Aurigene

Aurigene is a specialized, discovery stage biotechnology company, developing novel and best-in-class therapies to treat cancer and inflammatory diseases. Aurigene’s Programmed Death pathway program is the first of several immune checkpoint programs that are at different stages of discovery and preclinical development. Aurigene has partnered with several large- and mid-pharma companies in the United States and Europe and has delivered multiple clinical compounds through these partnerships. With over 500 scientists, Aurigene has collaborated with 6 of the top 10 pharma companies. Aurigene is an independent, wholly owned subsidiary of Dr. Reddy’s Laboratories Ltd. (NYSE:RDY). For more information, please visit Aurigene’s website at http://aurigene.com/.

Small Molecule IRAK4 Kinase Inhibitor)

Innate immune responses mediated through Toll-like receptors or certain interleukin receptors are important mediators of the body’s initial defense against foreign antigens, while their dysregulation is associated with certain inflammatory conditions.  Toll-like receptor and interleukin receptor signaling through the adaptor protein MYD88, results in the assembly and activation of IRAK4, initiating a signaling cascade that induces cytokine and survival factor expression mediated by the transcription factor NFκB. More recently, components of this pathway are recognized to be genetically altered and have important roles in specific human cancers.  Toll-like receptor and interleukin receptor signaling through the adaptor protein MYD88, results in the assembly and activation of IRAK4, initiating a signaling cascade that induces cytokine and survival factor expression mediated by the transcription factor NFκB.  MYD88 gene mutations are shown to occur in approximately 30% of Activated B-Cell (ABC) subtype of diffuse large B-cell lymphomas (DLBCL)1,2 and in over 90% of the B-cell malignancy Waldenstrom’s macroglobulinemia.3  Due to IRAK4’s central role in these signaling pathways, it is considered an attractive target for generation of therapeutics to treat these B-cell malignancies as well as certain inflammatory diseases.

As part of the collaboration with Aurigene, in October 2015 we exercised our option to exclusively license a program of orally-available, small molecule inhibitors of IRAK4 kinase, including the development candidate, CA-4948.  Curis expects to file an IND and initiate clinical testing of CA-4948 in patients with advanced hematologic cancers during the second half of 2016.

1Nature. 2011; 470(7332):115–1192Immunology and Cell Biology. 2011; 89(6):659–6603N Engl J Med. 30, 2012; 367(9):826–833

CLIP

In November 2015, preclinical data were presented at the 2015 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference in Boston, MA

Aurigene Collaboration (IRAK4 Inhibitor):

In October 2015, Curis exercised its option to exclusively license a program of orally available small molecule inhibitors of IRAK4 kinase, a serine/threonine kinase involved in innate immune responses as well as in certain hematologic cancers. The Company has since designated the development candidate as CA-4948 and expects to file an IND application for this molecule during 2016.

In November 2015, Curis’ collaborator Aurigene presented preclinical data from the IRAK4 program at the 2015 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference in Boston, MA. This presentation included data from chemically distinct series of small molecule compounds with potent IRAK4 inhibitory activity in biochemical assays as well as in in vivo preclinical models, including MYD88 mutant DLBCL xenograft tumor models as well as a model of inflammatory disease.

CLIP

In April 2014, preclinical data presented at the CHI’s Ninth Drug Discovery Chemistry Conference in San Diego, CA, showed the compounds in vivo to have activity down to 10 mg/kg .

 

CLIP

10:50 Novel IRAK4 Inhibitors for Oncology and Inflammation
Susanta SamajdarSusanta Samajdar, Ph.D., Research Director, Medicinal Chemistry, Aurigene Discovery Technologies Limited
This presentation will discuss the discovery and optimization of hit series, some preliminary in vivo data, combination therapy strategy, present focus and further advancements.
CLIP

April 24-25 2014
Drug Discovery Chemistry – CHI’s Ninth Annual Conference: Fifth Annual Kinase inhibitor Chemistry, San Diego, CA, USA

Novel IRAK4 inhibitors

Susanta Samajdar from Aurigene Discovery Technologies presented the discovery of new IRAK4 (IL-1 receptor-associated kinase 4) inhibitors. Research began with a HTS campaign using two types of libraries: rationally designed novel scaffolds by hopping and morphing of known IRAK4 inhibitors and novel scaffolds identified by virtual screening of drug-like commercial library. A benzoxazol series was identified and crystallography was used to help their design. Lead optimization culminated in the identification of very potent compounds (AU-2807 and AU-2202) in cell assay (inflammation pathway and oncology pathway, respectively). The compounds were also active against Flt3 and KDR. Some PD in vivo data using LPS and TNFalpha release were presented in which the compound showed activity down to 10 mg/kg: no other in vivo model data were disclosed, but it was mentioned that studies in the CIA (collagen induced arthritis) model was ongoing. Dr Samajdar answered to three questions, one related to IRAK1 selectivity (the answer was that the compound is fully selective against IRAK1 and IRAK2). It was also mentioned that the compounds have a PBB higher than 98%. And the last question was related to the synergetic effect with BTK inhibitor in activated B-cell like diffuse large B-cell lymphoma, and this effect was observed with these compounds.

Susanta Samajdar

Research Director at Aurigene Discovery Technologies

 

 

PATENT

http://www.google.com/patents/WO2013042137A1?cl=en

Compound-6: Synthesis of 6′-amino-N-(5-(cyclopropyIamino)-2-morpholinobenzo [d]oxazoI-6-yl)-[2,3′-bipyridine]-6-carboxamide.

Step_l^N-cyclopropyl-2-morpholino-6-nitrobenzo[d]oxazol-5-amine.

N-cyclopropyl-2-moφholino-6-nitrobenzo[d]oxazol-5-amine(0.7g,70%) was prepared from 5-fluoro-2-mo holino-6-nitrobenzo[d]oxazole(lg,Intermediate-2) by treating with cyciopropanamine in sealed tube at 100°C for 8-14h. The progress of the reaction was monitored by TLC. After the reaction was completed, it was extracted with water (15ml) and dichioromethane (2x 15ml). The organic layer was collected, washed with brine, dried over sodium sulfate and concentrated under reduced pressure to get the crude. MS (ES) m/e 305(M+1, 50%).

Steg2:6-bromo-N-(5-(cyclopropylamino)-2-morpholinobenzo[d]oxazol-6-yl)

picolinamide.

Step Π and ii):The process of these steps are adopted from step 2 and step 3 of compound- 1.

Step3:6′-amino-N-(5-(cvclopropvlamino)-2-morpholinobenzord]oxazol-6-yl)-r2,3′- bipyridine]-6-carboxamide.

(i) N-(4-methoxybenzyl)-5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)pyridin

Na2C03, Pd(dppf)Cl2, ACN, H20, 80-100°C, 8-14h; TFA, 60-70°C, 8-14h.

6′-amino-N-(5-(cyclopropylamino)-2-mo holinobenzo[d]oxazol-6-yl)-[2,3′-bipyridine]-6- carboxamide (0.03g,61%) was prepared from 6-bromo-N-(5-(cyclopropyIamino)-2- moφholinobenzo[d]o azoI-6-yl)picolinamide(0.07g, step-3) by following the same process used in step-1 and 2 of compound-3.

Ή NMR (400 MHz, DMSO-< ):6 1 1.63 (s, IH), 8.90 (s, IH), 8.61 (s, IH), 8.55 (s, IH), 8.37- 8.03 (m, 2H), 7.39 (s, IH), 6.80-6.62 (s, IH), 3.80-3.59 (m, 15H), 2.88-2.64 (m, 2H). MS (ESI): 472 (M+l , 60%).

 

 

PATENT

WO2015104688

Example 13

6′-amino-N-(2-morphol ne]-6-carboxamide

Step-1: Synthesis of 6-chloro thiazolo[4,5-c]pyridine-2(3H)-thione

Using the same reaction conditions as described in step 1 of example 1, 4,6-dichloropyridin-3-amine (1.3 g, 7 mmol) was cyclised using potassium ethyl xanthate (2.55 g, 15 mmol) in DMF (25mL) at 150°C for 8h to afford the title compound (1.3 g, 86.6 %) as a light brown solid.

1HNMR (400 MHz, DMSO-d6): δ 14.2-14.0 (b, 1H), 8.274 (s, 1H), 7.931 (s, 1H); LCMS: 100%, m/z = 201.3 (M+l)+.

Step-2: Synthesis of 4-(6-chloro thiazolo[4,5-c]pyridin-2-yl) morpholine

To a suspension of 6-chlorothiazolo[4,5-c]pyridine-2(3H)-thione (0.3 g, 1.16 mmol) in

DCM (4 mL), oxalyl chloride (0.2 mL, 2.38 mmol) and DMF (1.5 mL) were added at 0°C. The resulting mixture was slowly allowed to warm to room temperature and stirred there for 1 h. The reaction mixture was again cooled to 0°C and triethyl amine (0.66 mL, 4.76 mmol) and morpholine (0.13 mL, 1.75 mmol) were added. The reaction mixture was stirred at RT for 1 h and quenched with water and extracted with ethyl acetate. The combined organic layers were washed with water, brine, dried over sodium sulphate and concentrated under reduced pressure. The crude material was purified by column chromatography (EtOAc/n-hexanes 3:7) to afford the title compound (0.14 g, 39.6 %) as a light brown solid.

1H NMR (400 MHz, DMSO-d6): δ 8.47 (s, 1H), 8.04 (s, 1H), 3.74-3.72 (m, 4H), 3.61-3.59 (m, 4H); LCMS: m/z = 256.1 (M+l)+.

Step-3: Synthesis of 6′-amino-/V-(2-morpholino thiazolo [4,5-c]pyridin-6-yl)-[2,3′-bipyridine]-6-carboxamide

Using the same reaction conditions as described in step 4 of example 12, 4-(6-chlorothiazolo[4,5-c] pyridin-2-yl) morpholine (0.081 g, 0.32 mmol), was coupled with tert-butyl (6-carbamoyl-[2,3′-bipyridin]-6′-yl)carbamate (intermediate 2) (0.1 g, 0.32 mmol) using cesium carbonate (0.21 g, 0.64 mmol), XantPhos (0.028g, 0.047mmol) and Pd2(dba)3 (0.015 mg, 0.015 mmol) in toluene : dioxane (2:2mL) to get the crude product. The resultant crude was purified by 60-120 silica gel column chromatography using 2% methanol in DCM as eluent. Further the resultant crude was purified by prep HPLC to afford title compound (0.01 g, 6 %) as an off-white solid.

1H NMR (400 MHz, DMSO-d6): δ 10.65 (s, 1H), 8.88 (d, 1H), 8.85 (dd, 1H), 8.71 (s, 1H), 8.55 (s, 1H), 8.22-8.13 (m, 4 H), 7.09 (d, 1H), 3.73 (t, 4H), 3.58 (t, 4H). LCMS: 100%, m/z = 434.2 (M+l)+.

 

Example 11

(S)-2-(2-methylpyridin-4-yl)-N-(2-morpholino-5-(pyrrolidin-3-ylamino)oxazolo[4,5-b]pyridin-6-yl)oxazole-4-carboxamide

Step l:Preparation of (S)-tert-butyl 3-((2-morpholino-6-nitrooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine- 1 -carboxylate

A solution of 5-chloro-2-morpholino-6-nitrooxazolo[4,5-b]pyridine (300mg, 1.0563 mmol) (S)-tert-butyl 3 -aminopyrrolidine- 1 -carboxylate (237mg, 1.267 mmol) and potassium carbonate (292mg, 2.112 mmol) in DMF (2mL) was heated at 100°C for 2h. Reaction was quenched with ice water and filtered the solid. The resultant crude was purified by 60-120 silica gel column chromatography using 1 % methanol in DCM as eluent to obtain the title compound (350mg, 76.25%). LCMS: m/z: 435.4 (M+l)+.

Step 2:Preparation of (S)-tert-butyl 3-((6-amino-2-morpholinooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine- 1 -carboxylate

Using the same reaction conditions as described in step 5 of example 1, (S)-tert-butyl 3- ((2-morpholino-6-nitrooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine-l -carboxylate (350mg, 0.806 mmol) was reduced with zinc dust (422mg, 6.451 mmol) and ammonium chloride (691mg, 12.903 mmol) in THF/methanol/H20 (10mL/2mL/lmL) to get the title compound (240mg, 71.8%). LCMS: m/z: 405.2 (M+l)+.

Step 3:Preparation of (S)-tert-butyl 3-((6-(2-(2-methylpyridin-4-yl)oxazole-4-carboxamido)-2-morpholinooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine-l-carboxylate

Using the same reaction conditions as described in step 6 of example 1, (S)-tert-butyl 3-((6-amino-2-morpholinooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine-l -carboxylate (115mg, 0.284 mmol), was coupled with 2-(2-methylpyridin-4-yl)oxazole-4-carboxylic acid (70mg, 0.341 mmol) using EDCI.HCl (82mg, 0.426 mmol), HOBt (58mg, 0.426 mmol), DIPEA (0.199mL, 1.138 mmol) in DMF (2mL) to afford the title compound (lOOmg, 59.52%). LCMS: m/z: 591.4 (M+l)+.

Step 4: Preparation of (S)-2-(2-methylpyridin-4-yl)-N-(2-morpholino-5-(pyrrolidin-3-ylamino)oxazolo[4,5-b]pyridin-6-yl)oxazole-4-carboxamide

Using the same reaction conditions as described in step 8 of example 1, (S)-tert-butyl 3- ((6-(2-(2-methylpyridin-4-yl)oxazole-4-carboxamido)-2-morpholinooxazolo[4,5-b]pyridin-5-yl)amino)pyrrolidine-l -carboxylate (lOOmg, 0.169 mmol) was deprotected using methanolic HC1 (5mL) to get the crude product. This was then purified by prep HPLC to get the title compound (9mg, 10.84%).

1HNMR (CDCI3, 400MHz): δ 9.91 (s, 1H), 8.78 (s, 1H), 8.74-8.73 (d, 1H), 8.45 (s, 1H), 7.82 (s, 1H), 7.76-7.74 (d, 1H), 4.50 (s, 1H), 4.04-4.03 (d, 4H), 3.30-3.00 (m, 7H), 2.70 (s, 3H), 2.40-1.80 (m, 4H), 1.00-0.08 (m, 1H). LCMS: 100%, m/z = 491.3 (M+l)+.

 

REFERENCES

http://www.curis.com/images/stories/pdfs/posters/Aurigene_IRAK4_AACR-NCI-EORTC_2015.pdf

http://www.curis.com/images/stories/pdfs/posters/Aurigene_IRAK4_AACR_20150421.pdf

1Nature. 2011; 470(7332):115–119

2Immunology and Cell Biology. 2011; 89(6):659–660

3N Engl J Med. 30, 2012; 367(9):826–833

April 2014, preclinical data presented at the CHI’s Ninth Drug Discovery Chemistry Conference in San Diego, CA

November 2015, preclinical data were presented at the 2015 AACR-NCI-EORTC Molecular Targets and Cancer Therapeutics Conference in Boston, MA

http://pubs.acs.org/doi/abs/10.1021/jm5016044

http://cancerres.aacrjournals.org/content/75/15_Supplement/3646

2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3646. doi:10.1158/1538-7445.AM2015-3646

 

////////IRAK4 Kinase Inhibitor, Curis,  Aurigene,  CA 4948, AU 4948, CA-4948, AU-4948, 1428335-77-6

c21ccc(cc1sc(n2)N3CCOCC3)NC(c4nc(ccc4)c5ccc(nc5)N)=O

Share

DS 2330 by Daiichi Sankyo

 phase 1  Comments Off on DS 2330 by Daiichi Sankyo
Apr 072016
 

 

str1

DS 2330

 

 

4-[2-(4-{[2-({3-[(trans-4-carboxy-cyclohexyl)(ethyl)sulfocarbamoyl]benzoyl}amino)-5-(piperidin-1-yl)benzoyl]amino}phenyl)ethyl]benzoic acid,

4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate

CAS 1634680-81-1
C43 H48 N4 O8 S, 780.9
Benzoic acid, 4-​[2-​[4-​[[2-​[[3-​[[(trans-​4-​carboxycyclohexyl)​ethylamino]​sulfonyl]​benzoyl]​amino]​-​5-​(1-​piperidinyl)​benzoyl]​amino]​phenyl]​ethyl]​-
CIS CAS 1634681-85-8
DISODIUM SALT 1634681-00-7
  • OriginatorDaiichi Sankyo Inc
  • ClassHyperphosphataemia therapies

useful for treating hyperphosphatemia, DS-2330, a phosphorous lowering agent, being developed by Daiichi Sankyo, for treating hyperphosphatemia in chronic kidney disease. In April 2016, DS-2330 was reported to be in phase 1 clinical development.

  • Phase IHyperphosphataemia
  • 31 Oct 2015Phase-I clinical trials in Hyperphosphataemia in USA (unspecified route)

 

str1

 

SEE  WO2015108038,

PATENT

WO2014175317

http://www.google.com/patents/EP2990400A1?cl=en

 

str1

PATENT

WO-2016047613

he problem is to provide a pharmaceutical for the prevention or treatment of hyperphosphatemia. The solution is a salt of a compound including formula (I), or a crystal of a hydrate thereof.

 

 

 

 

 

 

(Example 1)
disodium 4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl ) benzoyl] amino} phenyl) ethyl] benzoic acid trihydrate
Disodium 4- [2- (4 – { [2 – ({3 – [(trans-4-carboxylatocyclohexyl) (ethyl) sulfamoyl] benzoyl} amino) – 5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate trihydrate
of α crystal

 

[Formula 7] crystal of disodium salt trihydrate of (α crystal)

 

(1)
4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] 1 mol / L NaOH aqueous solution to benzoic acid (1.2 g) (3.1 mL) was added and dissolved completely. After stirring at room temperature for 1 day was added acetonitrile (60 mL), at 40 ° C.
and stirred for further 1 day. The precipitated solid was collected by filtration, and 3 hours drying under reduced pressure at room temperature to give the title compound 1.1 g (85%).
(2)
 4- [2- (4 – {[2 – ({3 – [(trans-4-carboxy-cyclohexyl) (ethyl) sulfur carbamoyl] benzoyl} amino) -5- (piperidin-1-yl) benzoyl] amino} phenyl) ethyl] benzoate (40.0 g)
in water (46.4 mL), 1-PrOH (72 mL), 4 mol / L NaOH aqueous solution (25.54 mL) was added, then filtered after stirring insolubles at room temperature, water / 1-PrOH: was washed with (3 7, 80 mL). The filtrate was heated up to 40 ℃, 1-PrOH the (160 mL) was added, and further seed crystal (α crystals, 0.2g) was added. Then the temperature was raised to 50 ℃, 1-PrOH (96 ml) was added, and the mixture was stirred overnight.Thereafter, 1-PrOH (480 ml) was added and after overnight stirring, was collected by filtration the precipitated solid was cooled to room temperature.Thereafter, and vacuum dried overnight at 40 ° C., to give the title compound 39.4 g (96%).

 

REFERENCES

http://www.daiichisankyo.com/media_investors/investor_relations/ir_calendar/files/005280/Presentation%20Material.pdf

////////////DS 2330, DS-2330, DAIICHI SANKYO, phase 1

O=C(O)[C@@H]1CC[C@H](CC1)N(CC)S(=O)(=O)c2cccc(c2)C(=O)Nc5ccc(cc5C(=O)Nc4ccc(CCc3ccc(cc3)C(=O)O)cc4)N6CCCCC6

OR

O=C(O)[C@@H]1CC[C@H](CC1)N(CC)S(=O)(=O)c2cccc(c2)C(=O)Nc5ccc(cc5C(=O)Nc4ccc(CCc3ccc(cc3)C(=O)O)cc4)N6CCCCC6

 

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

Join me on Linkedin

View Anthony Melvin Crasto Ph.D's profile on LinkedIn

Join me on Facebook FACEBOOK

Join me on twitterFollow amcrasto on Twitter
Join me on google plus Googleplus

 amcrasto@gmail.com

Share

ICH Q3D implemented in the European Pharmacopoeia: Revision of Two General Monographs with Regard to Elemental Impurities

 regulatory  Comments Off on ICH Q3D implemented in the European Pharmacopoeia: Revision of Two General Monographs with Regard to Elemental Impurities
Apr 072016
 

 

ICH Q3D implemented in the European Pharmacopoeia: Revision of Two General Monographs with Regard to Elemental Impurities

Two general monographs of the European Pharmacopoeia have been revised and published for comment in the newest “Pharmeuropa” edition. Read more about what you will have to consider in future with regard to the control of elemental impurities in pharmaceutical preparations, APIs and excipients.

see

http://www.gmp-compliance.org/enews_05296_ICH-Q3D-implemented-in-the-European-Pharmacopoeia-Revision-of-Two-General-Monographs-with-Regard-to-Elemental-Impurities_15499,15332,S-AYL_n.html

In a press release dated 30 November 2015, the EDQM announced the revision of two general pharmacopoeial monographs: “Substances for pharmaceutical use” (2034) and “Pharmaceutical preparations” (2619). The decision was taken during the 153rd session of the European Pharmacopoeia Commission; the Commission follows its strategy for implementing the ICH Guideline Q3D “Guideline for Elemental Impurities” in the European Pharmacopoeia. A section “Elemental Impurities” has been added to both monographs which emphasizes that the provisions laid down in General Chapter 5.20 of the Pharmacopoeia (identical in wording with ICH Q3D) apply to the limits of metallic impurities and to their control. For pharmaceutical preparations and substances for pharmaceutical use outside the scope of Chapter 5.20 (e.g. unlicensed patient-specific preparations, herbal products, radiopharmaceuticals, etc.), the manufacturer is obliged to perform a risk assessment with regard to the limits of those impurities and – if necessary – to use validated analytical procedures for their determination. The principles to be applied for such a risk assessment arise from a press release from the EDQM dated 7 August 2015: it is expected that the provisions defined on the Guidelines ICH Q3D or ICH Q9 are followed. Lastly, according to this press release, the control strategy of elemental impurities as well as the substantial demonstration of suitability of the analytical methods used in the marketing authorisation dossier remains in the responsibility of the manufacturer.

The definition of “Substances for pharmaceutical use” in the monograph 2034 states: “Substances for pharmaceutical use are any organic or inorganic substances or excipients for the production of medicinal products for human or veterinary use. … Substances for pharmaceutical use may be used as such or as starting materials for subsequent formulation to prepare medicinal products. Depending on the formulation, certain substances may be used either as active substances or as excipients.”

The drafts of the two revised general monographs are accessible for free in the Journal “Pharmeuropa“, Edition 28/2. You only need to register and log in with your password. The comment deadline for both monograph drafts ends on 30 June 2016.

 

 

 

 

 

 

 

 

str1

str1

 

str1

 

str1

 

 

Other reading

https://www.thermoscientific.com/content/dam/tfs/ATG/CMD/cmd-documents/ref/third/USP-Primer-eBook.pdf

https://www.aaps.org/uploadedFiles/Content/Sections_and_Groups/Regional_Discussion_Groups/Southern_California_Pharmaceutical/SCPDG%20Impurities_Olsen%20Jan%20event.pdf

http://www.ich.org/fileadmin/Public_Web_Site/ICH_ Products/Guidelines/Quality/Q3D/Q3D_Step_4.pdf

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q3D/Q3D_IWG_Final_Concept_Paper_ October_21_2014.pdf

http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Quality/Q3D/Q3D_IWG_Final_Business_Plan_ October_21_2014.pdf

///////ICH Q3D,  implemented,  European Pharmacopoeia, Revision, Two General Monographs,  Elemental Impurities

Share

The new Annex 16 “Certification by a Qualified Person and Batch Release” will become effective as of 15 April 2016.

 regulatory  Comments Off on The new Annex 16 “Certification by a Qualified Person and Batch Release” will become effective as of 15 April 2016.
Apr 072016
 

 

The new Annex 16 is coming into Force

The new Annex 16 “Certification by a Qualified Person and Batch Release” will become effective as of 15 April 2016. The contents will reflect the coming state of expectations regarding the batch release.

see

http://www.gmp-compliance.org/enews_05188_The-new-Annex-16-is-coming-into-Force_15099,15432,Z-QAMPP_n.html

The new Annex 16 “Certification by a Qualified Person and Batch Release” will become effective as of 15 April 2016.

It is centrally pointed out that the main duty of a Qualified Person (QP) is the certification of batches. In this context, the QP must personally ensure that the responsibilities listed under Chapter 1.6 are fulfilled. Chapter 1.7 lists many other responsibilities to be guaranteed by the QP. However the related activities can be delegated and the QP can rely on the respective quality management systems. Yet, the “QP should have on-going assurance that this reliance is well founded” (1.7). The 21 responsibilites listed include amongst others:

  • The starting materials used comply with the requirements and the supply chain is known and under control.
  • The necessary audits have been carried out and the audit reports are available.
  • The manufacturing processes and testing methods are validated and in accordance with the marketing authorisation.
  • Changes have been assessed and completed accordingly.

In this context, it is important to mention that the Annex clearly highlights that the overall responsibility (safety, quality and efficacy) for a medicinal product lies with the marketing authorization holder (MAH). “However, the QP is responsible for ensuring that each individual batch has been manufactured and checked (…) in accordance with the requirements of the marketing authorisation (MA) and with Good Manufacturing Practice (GMP)” (see general principles).

In cases where the QP has to rely on the functioning QM system of another site, the QP must ensure that a documented review and permission of audit reports by third parties is available.

Another important section clarifies the role of the QP with regard to deviations and includes a few elements from EMA’s position paper on QP Discretion (published in February 2006 and updated in January 2008). Chapter 3 of the new Annex describes the “handling of unexpected deviations”. A batch with an unexpected deviation concerning the manufacturing process may be certified if the result of a risk analysis performed shows that “the potential impact of the deviation on quality, safety or efficacy of the batch(es) concerned and conclusion that the impact is negligible.” In cases where a deviation concerns specification defined in the marketing authorisation as essential for the release (OOS; out of specification), the QP will still have no scope left.

During the consultation phase, interest groups have expressed their concerns with regard to the sampling of imported products. Now, the new Annex 16 makes clear that “samples may either be taken after arrival in the EU, or be taken at the manufacturing site in the third in accordance with a technically justified approach which is documented within the company’s quality system. (…) Any samples taken outside the EU should be shipped under equivalent transport conditions as the batch that they represent.”

Regarding any requirements on import, the new Annex 16 “Certification by a Qualified Person and Batch Release” has been kept relatively short. Those requirements will probably be set in the new Annex 21.

//////new Annex 16,  Certification by a Qualified Person and Batch Release,  15 April 2016, Qualified Person (QP),  certification of batches

Share

GDC-0084

 phase 1, Uncategorized  Comments Off on GDC-0084
Apr 062016
 

 

GDC-0084
CAS#: 1382979-44-3
Chemical Formula: C18H22N8O2
Exact Mass: 382.1866

Synonym: RG7666; RG-7666; RG 7666; GDC-0084; GDC0084; GDC 0084.

IUPAC/Chemical Name: 5-(6,6-dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine

Company Roche
Description Phosphoinositide 3-kinase (PI3K) inhibitor
Molecular Target Phosphoinositide 3-kinase (PI3K)
Mechanism of Action Phosphoinositide 3-kinase (PI3K) inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Phase I
Standard Indication Brain cancer
Indication Details Treat progressive or recurrent high-grade glioma
Regulatory Designation
Partner Genentech Inc.
  • Originator Genentech
  • Class Antineoplastics; Small molecules
  • Mechanism of Action 1 Phosphatidylinositol 3 kinase inhibitors
  • 28 Jan 2015 Discontinued – Phase-I for Glioma in Spain (unspecified route)
  • 28 Jan 2015 Discontinued – Phase-I for Glioma in USA (unspecified route)
  • 01 Jan 2015 Genentech completes a phase I trial in Glioma in USA and Spain (NCT01547546)

GDC-0084, also known as RG7666, is a phosphatidylinositol 3-kinase (PI3K) inhibitor with potential antineoplastic activity. PI3K inhibitor GDC-0084 specifically inhibits PI3K in the PI3K/AKT kinase (or protein kinase B) signaling pathway, thereby inhibiting the activation of the PI3K signaling pathway. This may result in the inhibition of both cell growth and survival in susceptible tumor cell populations. Activation of the PI3K signaling pathway is frequently associated with tumorigenesis.

 

str1

http://pubs.acs.org/doi/pdf/10.1021/acsmedchemlett.6b00005

 

 

Abstract Image

An improved, efficient process with a significantly reduced process mass intensity (PMI) led to the multikilogram synthesis of a brain penetrant PI3K inhibitor GDC-0084. Highlights of the synthesis include a phase transfer catalyzed annulation in water, an efficient Suzuki-Miyaura cross-coupling of a chloropyrimidine with an arylboronic acid using a low palladium catalyst loading, and the development of a controlled crystallization to provide the API. The process delivered GDC-0084 with low levels of both impurities and residual metals.

Development of an Efficient, Safe, and Environmentally Friendly Process for the Manufacture of GDC-0084

Small Molecule Process Chemistry, Small Molecule Analytical Chemistry, Genentech, Inc., A Member of the Roche Group, 1 DNA Way, South San Francisco, California 94080, United States
Org. Process Res. Dev., Article ASAP
DOI: 10.1021/acs.oprd.6b00011
Publication Date (Web): March 11, 2016
Copyright © 2016 American Chemical Society

//////GDC-0084

NC1=NC=C(C2=NC(N3CCOCC3)=C4N=C(C(C)(C)OCC5)N5C4=N2)C=N1

str1

str1

5-(6,6-Dimethyl-4-morpholino-8,9-dihydro-6H-[1,4]oxazino[4,3-e]purin-2-yl)pyrimidin-2-amine GDC-0084 

mp 211 °C; 1H NMR (500 MHz, DMSO-d6) δ 9.09 (s, 2H), 7.03 (s, 2H), 4.32–4.17 (m, 4H), 4.17–4.04 (m, 4H), 3.84–3.65 (m, 4H), 1.58 (s, 6H); 13C NMR (125 MHz, DMSO-d6) δ 163.8, 157.6, 154.2, 152.5, 151.3, 151.0, 120.3, 117.3, 73.7, 66.2, 57.8, 45.2, 41.5, 27.3. HRMS [M + H]+calcd for C18H22N8O2 383.1938; found 383.1945.

  1. The Discovery of Clinical Development Candidate GDC-0084, a Brain Penetrant Inhibitor of Class I Phosphoinositide 3-Kinases (PI3K) and mTOR.

    HeffronT.NdubakuC.SalphatiL.AlickeB.CheongJ.;DrobnickJ.EdgarK.GouldS.LeeL.LesnickJ.LewisC.NonomiyaJ.Pangj.PliseE.Sideris,S.WallinJ.WangL.ZhangX.OliveroA. ACS Med. Chem. Lett. 2016, , DOI: 10.1021/acsmedchemlett.6b00005

  2. 3.

    (a) Purine Derivatives Useful as PI3 Kinase Inhibitors. GoldsmithP.HancoxT. C.HudsonA.PeggN. A.KulagowskiJ. J.NadinA. J.PriceS. PCT Int. Appl. WO 2009053716 A1 Apr 30, 2009.

    (b) Preparation of Purine Derivatives with PI3K Inhibitory Activity and Methods of Use Thereof. CastanedoG.Chuckowree,I.FolkesA.SutherlinD. P.WanN. C. PCT Int. Appl. WO 2009146406 A1 Dec 3, 2009

Share

Continuous-Flow Process for the Synthesis of m-Nitrothioanisole

 Uncategorized  Comments Off on Continuous-Flow Process for the Synthesis of m-Nitrothioanisole
Apr 062016
 

Abstract Image

A continuous-flow process for the preparation of m-nitrothioanisole has been set up. The starting material m-nitroaniline was diazotized to give diazonium chloride, followed by azo-coupling with sodium thiomethoxide to give 1-(methylthio)-2-(3-nitrophenyl)diazene, then dediazoniated to gain m-nitrothioanisole in high yield. The continuous-flow process minimized accumulation of the energetic intermediate diazonium salt and has a better capacity for adapting large-scale production. A solvent was introduced in the azo-coupling section to create a biphasic flow system. Side products were inhibited eminently in this flow process.

Continuous-Flow Process for the Synthesis of m-Nitrothioanisole

Zhiqun Yu, Xiaoxuan Xie, Hei Dong, Jiming Liu, and Weike Su*

National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, P. R. China

Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, P. R. China

Org. Process Res. Dev., Article ASAP

DOI: 10.1021/acs.oprd.6b00023

Publication Date (Web): March 24, 2016

Copyright © 2016 American Chemical Society

*Tel.: (+86)57188320899. E-mail: pharmlab@zjut.edu.cn.

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.6b00023

////////

Share

BMS 919373

 phase 2, Uncategorized  Comments Off on BMS 919373
Apr 062016
 

str1

.

Bethany Halford on Twitter: “BMS-919373, from $BMS for …https://twitter.com/beth_halford/status/634105343719682048

Aug 19, 2015 – BMS919373, from $BMS for atrial fibrillation #ACSBoston MEDI 1st disclosures @bmsnews pic.twitter.com/y3D4Yv2U7M.

BMS 919373

 CAS 1272353-82-8
C25 H20 N6 O2 S, 468.53
3-​Pyridinesulfonamide, 5-​[5-​phenyl-​4-​[(2-​pyridinylmethyl)​amino]​-​2-​quinazolinyl]​-
5-[5-phenyl-4-[[(pyridin-2-yl)methyl]amino]quinazolin-2-yl]pyridine-3-sulfonamide
  • Phase IIParoxysmal atrial fibrillation
  • Phase IAcute coronary syndromes; Atrial fibrillation
  •  CAS HCL SALT 1272356-77-0
Company Bristol-Myers Squibb Co.
Description IKur antagonist
Molecular Target Potassium channel Kv1.5 (KCNA5)
Mechanism of Action Potassium channel Kv1.5 (KCNA5) inhibitor
Therapeutic Modality Small molecule
Latest Stage of Development Phase I
Standard Indication Fibrillation
Indication Details Treat atrial fibrillation

Synthesis

str1

 

 

 

str1

 

 

PATENT

WO 2011028741

http://www.google.co.in/patents/WO2011028741A1?cl=en

EXAMPLE 7

5-(5-Phenyl-4-(pyridin-2-ylmethylamino)quinazolin-2-yl)pyridine-3-sulfonamide

Figure imgf000216_0001

Step 1. Preparatio -Bromopyridine-3 -sulfonamide

Figure imgf000216_0002

See also U.S. Publication Nos. 2006/217387 and 2006/375834, and J. Org. Chem., 54:389 (1989). A mixture of pyridine-3 -sulfonic acid (10.3 g, 64.8 mmol), phosphorous pentachloride (20.82 g, 100 mmol) and phosphorous oxychloride (10 mL, 109 mmol) was heated to reflux where it stirred for 4h. At the conclusion of this period, the reaction mixture was allowed to cool to room temperature. Once at the prescribed temperature, the reaction mixture was evaporated to dryness under reduced pressure to yield a residue. The residue was treated with bromine (6.00 mL, 1 16 mmol) and then heated to reflux where it stirred for 14h. After this time, the reaction mixture was cooled to 0 °C and then a saturated solution of NH4OH in ¾0 (40 mL) was slowly added. The resulting mixture was allowed to warm to room temperature where it stirred for 30 min. The reaction mixture was then filtered and the filter cake was washed with hexane to afford 5 -bromopyridine-3 -sulfonamide (6.0 g) as an off- white solid. The product was used without further purification. LCMS Method Q: retention time 0.75 min; [M+l] = 237.0.

Step 2. Preparation of pyridine-3-sulfonamide-5-ylboronic acid pinacol ester

Figure imgf000217_0001

See also WO2008/150827 Al and WO2008/144463. A mixture of 5- bromopyridine-3 -sulfonamide (1.5 g, 6.33 mmol), bis(pinacolato)diboron (2.41 g, 9.5 mmol) and potassium acetate (1.86 g, 19.0 mmol) in 1,4-dioxane (15 mL) was degassed with nitrogen for 15 min then (l, l’-bis(diphenylphosphino)- ferrocene)palladium (II) chloride dichloromethane complex (232 mg, 0.317 mmol) was added and the resulting mixture was degassed again with nitrogen for 10 min. At the conclusion of this period, the reaction mixture was heated in a microwave at 120 °C for 45 min. After this time, the reaction mixture was filtered through CELITE® and the filtrate was concentrated under reduced pressure to provide pyridine-3- sulfonamide-5-ylboronic acid pinacol ester (740 mg) as a brown solid. The product was used without further purification. XH NMR (400 MHz, DMSO-d6) δ (ppm): 8.83 (s, 1H), 8.80 (s, 1H), 8.26 (s, 1H), 7.56-7.74 (bs, 2H), 1.17 (s, 12H).

Step 3. Example 7

Figure imgf000217_0002

To a solution of 2-chloro-5-phenyl-N-(pyridin-2-ylmethyl)quinazolin-4- amine (150 mg, 0.43 mmol) in 1,4-dioxane (6 mL) and ¾0 (1 mL) under nitrogen was added pyridine-3-sulfonamide-5-ylboronic acid pinacol ester (185 mg, 0.65 mmol), and potassium carbonate (119 mg, 0.86 mmol). Upon completion of addition, the mixture was degassed with nitrogen for 15 minutes and then (1, 1′- bis(diphenylphosphino)ferrocene)palladium (II) chloride dichloromethane complex (31 mg, 0.043 mmol) was added. The resulting mixture was again degassed with nitrogen for 10 min. After this time, the mixture was heated to 90 °C where it stirred for 16h. At the conclusion of this period, the reaction mixture was allowed to cool to room temperature. Once at the prescribed temperature, the reaction mixture was quenched by the addition of water and then transferred to a separation funnel. The aqueous layer was extracted with ethyl acetate. The combined organic portions were washed with water and saturated NaCl, dried over Na2S04, filtered and concentrated under reduced pressure. The resulting concentrate was purified by preparative TLC using 5% methanol in dichloromethane to afford Example 7 (50 mg) as a brown solid. ‘H NMR (400 MHz, DMSO-d6) δ (ppm): 9.81 (s, 1H), 9.17 (s, 1H), 9.09 (s, 1H), 8.24 (d, J= 4.4 Hz, 1H), 7.94 (d, J=7.2 Hz, 1H), 7.86 (t, J= 7.6 Hz, 1Η),7.75-7.72 (t, J= 7.6 Hz, 3H), 7.59-7.51 (m, 5H), 7.34 (d, J=7.2 Hz, 2H), 7.24 (t, J=6.4 Hz, 1H), 6.98 (t, J= 3.2 Hz, 1H), 4.77 (d, J= 4.0 Hz, 2H). LCMS Method Q: retention time 1.39 min; [M+l] = 469.0. HPLC Method B: purity 98.1%, retention time = 8.74 min. [00120] Alternatively, Example 7 can be synthesized as follows:

Step 1. Preparation of 5-Bromo-pyridine-3-sulfonyl chloride

Figure imgf000218_0001

PC15 (2.95 Kg, 14.16 moles) and POCl3 (2.45 Kg, 15.98 moles) were added into pyridine-3 -sulfonic acid (1.5 Kg, 9.42 mol) in 10 L RB flask equipped with mechanical stirrer under inert atmosphere. The reaction mass was heated to 120- 125°C where it stirred for 18 h. After this time, the reaction progress was monitored by HPLC, which indicated the reaction was complete. Excess POCI3 was removed under vacuum to give a residue. The residue was cooled to ambient temperature and bromine (1.2 Kg, 7.5 moles) was added. Upon completion of addition, the resulting mixture was heated to 120-125°C where it stirred for 5 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated the reaction was complete. The reaction mixture was cooled to ambient temperature and then poured into ice-water (10 L), and the resulting mixture was extracted with DCM (10.5 Lx2). The DCM extracts were combined and the solvent was removed under vacuum to yield crude product (1.8 Kg, 74.4% yield).

Step 2. Preparation of 5-bromo-N-tert-butylpyridine-3 -sulfonamide

Figure imgf000219_0001

Crude 5 -bromopyridine-3-sulfonyl chloride from step 1 above was dissolved in THF (14 L, 8 vol) and then transferred to a 20 L RB flask equipped with mechanical stirrer under inert atmosphere. The solution was cooled to 0-5°C and tert- butyl amine (1.95 Kg, 26.66 moles) was added at 0-5°C. Upon completion of addition, the reaction mixture was warmed to ambient temperature where it stirred for 2 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated that the reaction was complete. The solvent was evaporated under vacuum to give a thick residue. The residue was dissolved in ethyl acetate (18 L, 12 vol). The organic layer was separated, washed with water (9 L, 5 vol) and then concentrated under vacuum to yield a residue. Hexanes (9 L, 5 vol) were added to the residue and the product precipitated out and was collected by filtration to yield a free flowing yellow solid (1.5 Kg, 54.28% overall yield). ¾ NMR (DMSO-D6, 400 MHz, δ ppm); 8.99 (d, J = 2Hz, 1H), 8.81 (d, J= 2 Hz, 1H), 8.29 (t, J= 2Hz, 1H). [M++l] = 293. Step 3. Preparation of 5-bromo-N-tert-butylpyridine-3 -sulfonamide

Figure imgf000220_0001

5 -Bromo-N-tert-butylpyridine-3 -sulfonamide (1.5 Kg, 5.11 moles) was dissolved in dimethylformamide (7.5 L, 5 vol) and the solution was added to a 20 L glass-lined reactor equipped with mechanical stirrer. The solution was degassed with nitrogen for 30 min. After this time, potassium ferrocyanide trihydrate (867 g, 2.05 moles), sodium carbonate (1.08 Kg, 10.189 moles), copper (I) iodide (73.2 g, 0.374 moles) and dichloro-bis (triphenylphosphine) palladium (II) (71.6 g, 0.102 moles) were added. Upon completion of addition, the reaction mixture was heated to 120- 125°C where it stirred for 4 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated the reaction was complete. The reaction mixture was cooled to ambient temperature and then filtered through a celite bed. Water (18 L, 12 vol) was added into the filtrate and the resulting mixture was extracted with ethyl acetate (7.5L*2). The organic layers were combined, washed with water and then concentrated to yield a thick residue. Hexanes (7.5 L, 5 vol) were added to the residue. The product precipitated out and was collected by filtration to yield a free flowing yellow solid (1.0 Kg, 82.8% yield, 89% purity by HPLC). ¾ NMR (DMSO-D6, 400 MHz, δ ppm); 9.21 – 9.24 (d,d J= 7.2Hz, 3.2Hz, 2H), 8.70-8.71(m,lH), 7.98 (s, lH). [M++l] = 239.2.

Step 4. Preparation of 3-aminobiphenyl-2-carbonitrile

Figure imgf000220_0002

2-Amino-6-bromo-benzonitrile (1.0 Kg, 5.07 moles) and toluene (10 L, 10 vol) were added to a 20 L glass-lined reactor equipped with mechanical stirrer under inert atmosphere. Potassium acetate (996 g, 10.16 moles) and phenylboronic acid (866, 7.10 moles) were added into the solution and the solution was degassed with nitrogen for 30 min. After this time, dichloro-bis (triphenylphosphine) palladium (II) (17.8 g, 0.025 moles) was added to the reaction mixture at ambient temperature. The mixture was heated to 110°C, where it stirred for 17 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated the reaction was completed. The reaction mixture was filtered through a celite bed. The filtrate was transferred back to the reactor and concentrated hydrochloric acid (-35%, 2 L, 2 vol) was charged to the reactor at ambient temperature. The HCl salt of the title compound precipitated out from the reaction and was collected by filtration. The HCl salt was transferred into the 20 L reactor and then made basic with 10% NaOH solution (pH 8-9). The resulting product was extracted with ethyl acetate (10 L, 10 vol). The ethyl acetate layer was washed with water (5 L, 5 vol) and then the solvent was evaporated under vacuum to give a residue. Hexanes (5 L, 5 vol) were added to the residue at 35-40°C, and the resulting slurry was cooled to ambient temperature. Once at the prescribed temperature, the product was collected by filtration to provide a pale yellow solid (802 g, 81.4%, 99% by HPLC). XH NMR (DMSO-D6, 400 MHz, δ ppm); 7.43-7.52 (m, 5H), 7.33-7.37 (m, 1H), 6.83 (d, J=8Hz, 1H), 6.62 (d, J=8Hz, 1H), 6.1 (s, 2H). ES-MS: [M++l] = 194.23.

Step 5. Preparation of 5-(4-amino-5-phenylquinazolin-2-yl)-N-tert-butylpyridine-3-

Figure imgf000221_0001

3-Aminobiphenyl-2-carbonitrile (1028 g, 5.30 moles), 5-bromo-N-tert- butylpyridine-3 -sulfonamide (1440 g, 5.55 moles) and 1,4-dioxane (10 L, 10 vol) were added to a 20 L glass-lined reactor equipped with mechanical stirrer. Sodium tert-butoxide (1.275 Kg 12.870 moles) was added to the solution portion-wise at 20- 30°C. Upon completion of addition, the reaction mixture was heated to reflux where it stirred for 2 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated the reaction was complete. The reaction mixture was cooled to 30-35°C and then poured into water (40 L, 40 vol). The resulting mixture was extracted with DCM (20 L*2). The DCM layers were combined, washed with water (10 L, 10 vol) and then dried over sodium sulfate. The solvent was evaporated under vacuum to give a residue. Isopropyl alcohol (1.2 L, 1.2 vol) was added to the residue at 40°C. The resulting precipitate slurry was cooled to 10-15°C and then stirred for 2 h. After this time, the precipitate was collected by filtration and dried at 50°C for 16 h to yield the product (1.9 Kg, 82.9% yield, 99% purity by HPLC). Ή NMR (DMSO-D6, 400 MHz, δ ppm); 9.72 (s, 1H), 9.11 (s, 2H), 7.83-7.94 (m, 4H), 7.49-7.60 (m, 5H), 7.31 (d,d /=6.8Hz,1.2Hz, 1H). ES-MS: [M++l] = 433.53.

Step 6. Preparation of N-tert-butyl-5-(5-phenyl-4-(pyridin-2-ylmethylamino) quinazolin-2-yl) pyridine-3 -sulfonamide

Figure imgf000222_0001

2-(Chloromethyl) pyridine hydrochloride (564 g, 3.44 moles) and dimethyl acetamide (7L, 7 vol) were added to a 20 L RB flask- 1 equipped with mechanical stirrer under inert atmosphere. The resulting solution was cooled to 0- 5°C and triethylamine (346.3, 3.44 moles) was added at 0-5°C. 5-(4-Amino-5- phenylquinazolin-2-yl)-N-tert-butylpyridine-3-sulfonamide (1.0 Kg. 2.306 moles) and dimethylacetamide (4 L, 4 vol) were added to a separate 20 L RB flask-2 equipped with mechanical stirrer under inert atmosphere. This solution was cooled to 0-5°C and sodium tert-butoxide (884 g, 9.24 moles) was added at 0-5°C. The resulting solution was stirred to affect dissolution and then transferred to the RB flask- 1 at 0- 5°C. Upon completion of addition, the reaction mixture was stirred at 0-5°C for 2 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated that the reaction was complete. The reaction mass was poured into water (60 L, 60 vol) with stirring. The crude product was collected by filtration and dried at 60°C for 12 h. After this time, the dried material was dissolved in THF (20 L, 20 vol). Upon dissolution, 6M HC1 in isopropyl alcohol (1 L, 1 vol) was added at 20-25°C. The crude HCL salt of the product was obtained a pale-yellow free flow solid (920 g, 71% yield, 93% purity by HPLC). The crude HC1 salt (1.345 Kg, 2.56moles), methanol (6.7 L, 5 vol) and dichloromethane (13.5 L, 10 vol) were added to a 20 L glass-lined reactor equipped with mechanical stirrer. The slurry was stirred for 20-30 min at 30°C. After this time, the solvent was distilled to 4 vol with respect to input under vacuum. The resulting slurry was cooled to 20-25°C, where stirred for 2 h. At the conclusion of this period, the slurry was filtered and dried at 50°C for 6 h to yield the product (1.1 Kg, 82% yield, 98% purity by HPLC). XH NMR (DMSO- D6, 400 MHz, δ ppm); 9.72 (s, 1H), 9.10-9.14 (m, 2H), 8.39 (s, 1H), 7.92-8.03 (m, 4H), 7.56-7.58 (m, 5H), 7.43-7.49 (m, 3H), 7.1 (bs, 1H), 4.88 (s, 2H), 1.17 (2, 9H).

Step 7. Example 7

Figure imgf000223_0001

N-tert-butyl-5-(5-phenyl-4-(pyridin-2-ylmethylamino) quinazolin-2-yl) pyridine-3 -sulfonamide (1.0 Kg, 1.9 moles) and concentrated hydrochloric acid (7 L, 7 vol) were added to a 20 L glass-lined reactor equipped with mechanical stirrer. The reaction mixture was heated to 90-100°C where it stirred for 1 h. At the conclusion of this period, the reaction progress was monitored by HPLC, which indicated the reaction was complete. The reaction mixture was cooled to 5-10°C and the pH was adjusted to 1.7 to 2.0 using 12% aqueous sodium hydroxide solution. Once at the prescribed pH, the crude HC1 salt of the product was collected by filtration. The HC1 salt filter cake and ethanol (5 L, 5 vol) were added to 10 L glass-lined reactor equipped with a mechanical stirrer. The resulting mixture was made basic to pH 7-8 at 20-25°C using triethyl amine (2.25 Kg, 22.23 moles). Once at the prescribed pH, the basic mixture was stirred for 2 h. After this time, the free base of product was filtered and washed with water (10 L, 10 vol) followed by ethanol (2L, 2 vol). The resulting product was dried at 50-55°C for 8 h to yield Example 7 (644 g, 72% yield, 99.9% purity by HPLC).

XH NMR (DMSO-D6, 400 MHz, δ ppm); 9.81 (d, J=2.0Hz, 1H), 9.18 (t, J=2Hz, 1H), 9.1 1 (d, J=2Hz, 1H), 8.23 (d, J=4.4Hz, 1H), 7.92-7.94 (m, 1H), 7.83-7.87 (m, 1H), 7.78 (s, 2H), 7.70-7.72 (m, 1H), 7.50-7.59 (m, 5H), 7.31-7.34 (m, 2H), 7.22-7.25 (m, 1H), 6.95 (t, J=4Hz, 1H), 4.76 (d, J=4Hz, 2H). ES-MS: [M++l] = 469.

 

/////////atrial fibrillation, Potassium channel Kv1.5 (KCNA5) inhibitor, IKur antagonist, Bristol-Myers Squibb Co., BMS 919373, BMS-919373, PHASE 2

NS(=O)(=O)c1cc(cnc1)c4nc2cccc(c2c(NCc3ccccn3)n4)c5ccccc5

Share

BTI-320 (formerly PAZ320), Soluble mannan polysaccharides from Boston Therapeutics for the treatment of type 2 diabetes in combination with oral agents or insulin

 phase 2, Uncategorized  Comments Off on BTI-320 (formerly PAZ320), Soluble mannan polysaccharides from Boston Therapeutics for the treatment of type 2 diabetes in combination with oral agents or insulin
Apr 062016
 

CAM00001-1

BTI-320 (formerly PAZ320)

PAZ 320

Non-insulin dependent diabetes

Alpha-glucosidase inhibitor; Hydrolase inhibitor; Sucrose alpha-glucosidase inhibitor

Composition of chemically purified (fractionation) soluble mannan polysaccharides from legume’s seeds

BTI-320 is in phase II clinical development at Boston Therapeutics for the treatment of type 2 diabetes in combination with oral agents or insulin, and also for the treatment of high-risk patients with pre-diabetes. A chewable tablet formulation is being developed. The product is already available as dietary supplement.

Company Boston Therapeutics Inc.
Description Chewable polysaccharide that inhibits alpha glucosidase
Molecular Target
Mechanism of Action Alpha glucosidase inhibitor
Therapeutic Modality Macromolecule: Polysaccharide
Latest Stage of Development Phase II
Standard Indication Diabetes
Indication Details Treat Type II diabetes

 

 

PATENT

http://www.google.co.in/patents/WO2012061675A1?cl=en

A composition of chemically purified soluble mannans from legumes’ seeds (e.g. Ceratonia siliqua, Cæsalpinia spinosa Trigonelle foenum-graecum, and Cyamopsis tetragonolobus) and their use in the assembly of palatable dietary supplements is disclosed herein. The fractionation process provides high-quality physiologically soluble, chemically modified and purified homogeneous size polysaccharide fibers, devoid of natural impurities, for example proteins, alkaloids, glycoalkaloids, and/or environmental impurities including heavy metals, agricultural residues and microbial toxins. This process provides hypoallergenic dietary fibers devoid of any potential allergens, cytotoxins, and gastrointestinal toxins. A sequential process for assembly of the soluble fibers with plurality of molecular weights to create a time controlled dissolution of the functional high and low molecular weight fibers for improving solubility and palatability with improved dietary performance in the oral and gastro-intestinal system is also disclosed herein.

Fig. 1 illustrates a block flow diagram of an embodiment of a method for recovering purified mannan polysaccharides;

Fig. 2 illustrates a chemical structure of a mannan polysaccharide;

CAM00001-1

Fig. 3 illustrates a block flow diagram of an embodiment of a method for recovering high molecular weight (HMW) purified mannan polysaccharides;

Fig. 4 illustrates a block flow diagram of an embodiment of a method for recovering low molecular weight (LMW) purified mannan polysaccharides;

 

REFERENCES

https://clinicaltrials.gov/show/NCT02060916

https://clinicaltrials.gov/show/NCT02358668

BTI-320, a nonsystemic novel drug to control glucose uptake into the bloodstream, functions as a competitive inhibitor of sugar hydrolyzing enzymes
75th Annu Meet Sci Sess Am Diabetes Assoc (ADA) (June 5-9, Boston) 2015, Abst 974-P

Boston Therapeutics’ Hong Kong Affiliate Advance Pharmaceutical’s BTI-320 Clinical Trial Reaches Mid-Point by Enrolling 30 Patients at the Chinese University of Hong Kong
Boston Therapeutics Press Release 2015, July 08

Insight into the molecular mechanism of action of BTI320, a non-systemic novel drug to control serum glucose levels in individuals with diabetes50th Annu Meet Eur Assoc Study Diabetes (EASD) (September 15-19, Vienna) 2014, Abst 545

////BTI-320, PAZ320, PHASE 2, BTI 320, PAZ 320, Macromolecule,  Polysaccharide, Non-insulin dependent diabetes, Alpha-glucosidase inhibitor,  Hydrolase inhibitor,  Sucrose alpha-glucosidase inhibitor, phase II clinical development,  Boston Therapeutics, Soluble mannan polysaccharides

Composition of chemically purified (fractionation) soluble mannan polysaccharides from legume’s seeds

POLYMER OF BELOW

CAS 9036-88-8, 51395-96-1

refractive index : 78.5 ° (C=1.4, H2O)

Ailes;MANNAN;K-41K1;D-Mannan;NSC 174478;NSC 174479;NSC 174481;NSC 307194;NSC 174477;NSC 174473

ChemSpider 2D Image | Mannosan | C6H10O5

D-Mannan C41H60O31S5 (cas 9036-88-8) Molecular Structure

Chemical name: 1,6-Anhydro-β-D-mannopyranose
Synonyms: 1,6-Anhydro-D-mannose; 1,6-Anhydromannose; Mannosan; NSC 226600;
CAS Number: 14168-65-1
Possible CAS #: NA
Molecular form.: C₆H₁₀O₅
Appearance: White to Pale Beige Solid
Melting Point: 182-184°C
Mol. Weight: 162.14

Summary:
Mannans are major constitutents of hemicelluloses in plant tissue and are polymers composed of β(1→4)-linked mannose and glucose residues. Some contain galactopyranosyl side chains (see a galactomannan).

Slightly galactosylated mannans (4% galactose), considered as linear β(1→4)-D-mannans, have been isolated from the seed endosperm of vegetable ivory nut ( Phytelephas macrocarpa) and date ( Phoenix dactylifera) .

str1

Glycan icon:

 

a mannan compound structure

 

Child Classes: a 1,6-α-D-mannan backbone (0), a galactoglucomannan (0), a galactomannan (0), a glucomannan (0), a mannan oligosaccharide (1)

SMILES: C(O)C4(C(O[R1])C(O)C(O)C(OC3(C(O)C(O)C(OC2(C(O)C(O)C(OC1(C(O)C(O)C(O[R2])OC(CO)1))OC(CO)2))OC(CO)3))O4)

CAS:9036-88-8,

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: