AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

EMA/ FDA Mutual Recognition Agreement on drug facility inspections moving forward

 regulatory  Comments Off on EMA/ FDA Mutual Recognition Agreement on drug facility inspections moving forward
Nov 222016
 

 

Image result for signing animations

 

Image result for FDA

 

Image result for EMA

EMA/ FDA Mutual Recognition Agreement moving forward
A possible agreement between the EMA and the US FDA on mutual recognition agreement on drug facility inspections could already be signed in January 2017.

http://www.gmp-compliance.org/enews_05650_EMA–FDA-Mutual-Recognition-Agreement-moving-forward_15642,15660,15656,Z-QAMPP_n.html

A possible agreement between the European Medicines Agency EMA and the US Food and Drug Administration FDA on mutual recognition of drug facility inspections could already be signed in January 2017. This is noted in a report of the EU Commission: “The state-of-play and the organisation of the evaluation of the US and the EU GMP inspectorates were discussed. In light of the progress achieved, the conclusion of a mutual recognition agreement of Good Manufacturing Practices (GMPs) inspections by January 2017 is under consideration.”

But, according to the Commission, some issues are still not resolved – like, for example, the exchange of confidential information and the inclusion of veterinary products in the scope of the text.

The “Report of the 15th Round of Negotations for the Transatlantic Trade and Invesment Partnership” summaries the 15th round of negotiations for the Transatlantic Trade and Investment Partnership (TTIP) from 3rd to 7th October 2016 in New York.

////////EMA, FDA,  Mutual Recognition Agreement, drug facility inspections

Share

New Warning Letter of the FDA with the Focus on “Data Integrity”

 regulatory  Comments Off on New Warning Letter of the FDA with the Focus on “Data Integrity”
Sep 092016
 

The FDA has set the focus of its inspections on data integrity for quite some time already. The most recent Warning Letter addressed to a Chinese API manufacturer dated August 2016 clearly concentrates on the topic data integrity. Please find out more about the current FDA Warning Letter in this News.

http://www.gmp-compliance.org/enews_05557_New-Warning-Letter-of-the-FDA-with-the-Focus-on-%22Data-Integrity%22_15488,15844,Z-QCM_n.html

Again, the focus of FDA’s Warning Letter for the Chinese API manufacturer Zhejiang Medicine Co. Ltd. dated 4th August 2016 is on the lack of data integrity. Among other things, records of activities were made not at the time when they have been performed. Moreover, original data have been deleted. A number of alarming findings were discovered in the course of the FDA inspection in June 2015.

The FDA is now expecting concrete measures (“Data Integrity Remediation”) from the company. For this, the FDA expressly recommended to retain qualified, external consultants. Among the measures to be taken:

A – A comprehensive investigation of the extent of incorrect data
1. An extensive plan for the execution of the investigation
2. Interviews of current and former employees to clarify the root cause of incorrect data
3. An assessment of the extent of data integrity deficits.
4. A comprehensive retrospective assessment of the performance of analytical testing.

B – A current risk assessment of the possible effects of the deficiencies identified on the quality of the medicinal products, up to the risk to patients!

C – A management strategy for the implementation of CAPA plans.

All in all, there were great concerns about the authenticity and reliability of the data produced in that company.

To find out more access the complete Warning Letter for Zhejiang Medicine Co. Ltd.

Image result for Zhejiang Medicine Co. Ltd,

//////////Zhejiang Medicine Co. Ltd, Warning Letter,  FDA, Data Integrity

Share

FDA published generic user fee for 2017: for ANDA, DMF, and for Facility (API, FDF)

 regulatory  Comments Off on FDA published generic user fee for 2017: for ANDA, DMF, and for Facility (API, FDF)
Aug 032016
 

 

.

 

placeholder+image

http://www.raps.org/Regulatory-Focus/News/2016/07/26/25394/FDA-Lowers-ANDA-Fee-Rates-for-2017/

Generic drugmakers submitting abbreviated new drug applications (ANDAs) and prior approval supplements (PAS) will see their US Food and Drug Administration (FDA) fee rates drop in 2017, though all other rates, including those for drug master files (DMF) and facility fees will increase when compared to 2016.

For FY 2017, the generic drug fee rates are: ANDA ($70,480, down from $76,030 in 2016), PAS ($35,240, down from $38,020 in 2016), DMF ($51,140, up from $42,170 in 2016), domestic active pharmaceutical ingredient (API) facility ($44,234, up from $40,867 in 2016), foreign API facility ($59,234, up from $55,867 in 2016), domestic finished dose formulation (FDF) facility ($258,646, up from $243,905), and foreign FDF facility ($273,646, up from $258,905 in 2016).

The new fees are effective 1 October 2016 and will remain in effect through 30 September 2017.

FDA explained the increases and decreases in fees, noting that for ANDA and PAS fees, the agency is expecting an increase in the number of submissions estimated to be submitted in FY 2017 when compared to 2016. For 2017, the agency estimates that approximately 891 new original ANDAs and 439 PASs will be submitted and incur filing fees.

Fees for DMFs will increase, meanwhile, because of an expected decrease in the number of submissions estimated to be submitted in 2017 (FDA is estimating 379 fee-paying DMFs for 2017), when compared to the estimated submissions from 2016.

And all facility fees will increase in 2017 when compared to the previous year because of a decrease in the number of facilities that self-identified (the total number of FDF facilities identified through self-identification was 675, of which 255 were domestic facilities and 420 foreign facilities; while the total number of API facilities self-identified was 789, of which 101 were domestic facilities and 688 were foreign facilities), FDA said.

How FDA Calculates the Fees

In order to calculate the ANDA fee, FDA estimated the number of full application equivalents (FAEs) that will be submitted in FY 2017, which is done by assuming ANDAs count as one FAE and PASs (supplements) count as one-half of an FAE, since the fee for a PAS is one half of the fee for an ANDA.

The Generic Drug User Fee Act (GDUFA) also requires that 75% of the fee paid for an ANDA or PAS filing be refunded if either application is refused due to issues other than a failure to pay the fees.

And since this is the last year of this iteration of GDUFA (the next version is still in the works), the agency is allowed to further increase the fee revenues and fees established if such an adjustment is necessary to provide for not more than three months of operating reserves for the first three months of FY 2018, though FDA estimates that the GDUFA program will have carryover balances for such activities in excess of three months of such operating reserves, so FDA will not be performing a final year adjustment.

To pay the fees, companies must complete a Generic Drug User Fee Cover Sheet, available at http://www.fda.gov/gdufa and generate a user fee identification (ID) number. Payment must be made in US currency drawn on a US bank by electronic check, check, bank draft, US postal money order or wire transfer.

Federal Register Notice

See more at: http://www.raps.org/Regulatory-Focus/News/2016/07/26/25394/FDA-Lowers-ANDA-Fee-Rates-for-2017/#sthash.FNo99XHR.dpuf

 

 

/////////////FDA,  generic user fee,  2017, ANDA, DMF,  Facility, API, FDF

Share

FDA issues new Draft Guidance on Elemental Impurities

 regulatory  Comments Off on FDA issues new Draft Guidance on Elemental Impurities
Jul 072016
 

The recently issued FDA Guideline on Elemental Impurities as a draft describes the procedure for controlling elemental impurities for medicinal products with and without official USP monograph. Read in what cases the FDA expects the fulfilment of the requirements of the Guideline ICH Q3D respectively of the general USP Chapter <232> und <233>.

see

http://www.gmp-compliance.org/enews_05465_FDA-issues-new-Draft-Guidance-on-Elemental-Impurities_15332,S-AYL_n.html

The ICH Q3D “Guideline for Elemental Impurities” was issued in December 2014 and recommended for adoption in the regulations portfolio of the ICH regions Europe, USA and Japan according to the ICH step-by-step procedure (Step 5). With the publication of the “ICH guideline Q3D on elemental impurities” (EMA/CHMP/ICH/353369/2013) in August 2015 the European Medicines Agency (EMA) implemented this step and determined June 2016 (for medicinal products to be newly approved) and December 2017 (for already approved medicinal products) as the dates for the Guideline to come into effect. The FDA took over the ICH Q3D Guideline in September 2015.

On 30 June 2016 the FDA Guidance for Industry “Elemental Impurities in Drug Products” was issued as a draft and is now open for comments for a period of 60 days.

The requirements of the Guidance apply to

  • New compendial and noncompendial NDA or ANDA drug products
  • Drug products not approved under an NDA or ANDA – as, e.g., compendial and noncompendial nonprescription OTC products.

Compendial medicinal products are generally supposed to fulfil the requirements defined in the general USP Chapters <232> und <233>. However, in the following cases the provisions of ICH Q3D have to be met:

  • For noncompendial drug products,
  • For metallic impurities listed only in ICH Q3D but not in the general USP Chapters <232> and <233>.

Correspondingly these provisions do also apply for changes to approved medicinal products, made with the goal to fulfil the requirements of the chapters <232> and <233> respectively of ICH Q3D. For compendial medicinal products the result of the change must be the compliance with <232> and <233>, noncompendial products have to comply with the provisions of ICH Q3D.

The FDA generally considers these kind of changes as low risk with regard to negative effects on identity, strength, quality, purity or potency. For that reason they are not subject to the CBE change procedure and can be reported to the FDA as part of the annual report.

The general USP Chapter <232> only comprises the PDE values of 15 elements, while ICH Q3D covers 24 elements. Otherwise both chapters were adapted to ICH Q3D and issued in the second supplementary volume of USP 38-NF 33 on 1 December 2015. However, both chapters can only be applied to compendial products starting on 1 January 2018 – the date mentioned in the General Notices 5.60.30 “Elemental Impurities in USP Drug Products and Dietary Supplements”. This is nearly the date (December 2017) determined for the application of ICH Q3D respectively the European Guideline (EMA/CHMP/ICH 353369/2013).

///////////FDA, Draft Guidance, Elemental Impurities

Share

FDA publishes Technical Guide on Quality Metrics

 regulatory  Comments Off on FDA publishes Technical Guide on Quality Metrics
Jun 302016
 

 

The FDA has published a supplementing Guide on Quality Metrics. This is a very unusual step as the contents of the guide are planned to be integrated into the Guideline on Quality Metrics which hasn’t been finalised yet. Read more about the Technical Quality Metrics Guide.

see http://www.gmp-compliance.org/enews_05437_FDA-publishes-Technical-Guide-on-Quality-Metrics_15515,S-QSB_n.html

The FDA has published a supplementing Guide on Quality Metrics. This is a very unusual step as the contents of the guide are planned to be integrated into the Guideline on Quality Metrics which hasn’t been finalised yet.

The so-called FDA Quality Metrics Technical Conformance Guide should supplement the Guidance for Industry: Request for Quality Metrics published on 28 July 2015 which is currently still in the draft version. We have recently published a GMP News about a Quality Metrics Case Study at Aenova regarding a possible implementation. Now, the Technical Guide defines how the industry should submit Quality Metrics to the FDA. Technical standards and fields are defined. Basically, the FDA is oriented towards the data standards which are already established in other areas. FDA‘s so-called Study Data Technical Conformance Guide serves as a basis. Largely widespread in the industry, the XML format is used by the FDA and other authorities for the exchange of data and the submission of data within the marketing authorisation procedure (e.g. for eCTD).

Composed of 10 pages, the Guide primarily provides a definition of the variables necessary for the submission of Quality Metrics. The last page of the Guide refers to “Data Validation Rules”. Data Validation is defined as “a process that attempts to ensure that submitted data are both compliant and useful”. It should be ensured that the data are submitted in accordance with the required standard. The FDA recognises that the standardisation of data doesn’t ensure the quality of data, but it helps verify certain aspects of data quality thanks to automated checks. When finalising the Guidance for Industry on Quality Metrics, the FDA also wants to set validation requirements on the quality of data in the guideline and thus achieve that companies first perform a validation of their metrics before they submit them.

Source: FDA Quality Metrics Technical Conformance Guide

 

Figure 1: Types of images quality metrics.

////////

Share

Maralixibat Chloride, ماراليكسيبات كلوريد , 氯马昔巴特 , Мараликсибата хлорид

 breakthrough designation  Comments Off on Maralixibat Chloride, ماراليكسيبات كلوريد , 氯马昔巴特 , Мараликсибата хлорид
Jun 152016
 

STR1

 

2D chemical structure of 228113-66-4

Maralixibat chloride

Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

SHP625, Maralixibat chloride, Molecular Formula C40-H56-N3-O4-S.Cl, Molecular Weight, 710.4184

4-Aza-1-azoniabicyclo(2.2.2)octane, 1-((4-((4-((4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl)-, chloride (1:1)

1-[4-({4-[(4R,5R)-3,3-Dibutyl-7-(dimethylamino)-4-hydroxy-1,1-dioxido-2,3,4,5-tetrahydro-1-benzothiepin-5-yl]phenoxy}methyl)benzyl]-4-aza-1-azoniabicyclo[2.2.2]octane chloride

4-Aza-1-azoniabicyclo[2.2.2]octane, 1-[[4-[[4-[(4R,5R)-3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-, chloride

(4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt

(4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride

Cas: 228113-66-4, Free form 716313-53-0
UNII: V78M04F0XC, LUM 001, Lopixibat chloride, Treatment of Cholestatic Liver Diseases

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter, Less «
Applicant G.D. Searle & Co.

Several drawings of Maralixibat chloride

STR1

 

 

ChemSpider 2D Image | maralixibat chloride | C40H56ClN3O4S

STR1Figure imgf000053_0001

It is well established that agents which inhibit the 20 transport of bile acids across the ileum can also cause a decrease in the level of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255- 25287, discusses biochemistry, physiology, and known active agents affecting bile acids and cholesterol.

A class of ileal bile acid transport-inhibiting compounds which was recently discovered to be useful for influencing the level of blood serum cholesterol is 30 tetrahydrobenzothiepine-l,l-dioxides (THBDO compounds). (U.S. Patent Application No. 08/816,065)

Some classes of compounds show enhanced potency as pharmaceutical therapeutics after they have been enantiomerically-enriched (see, for example, Richard B. Silverman, The Organic Chemistry of Drug Design and Drug Action, Academic Press, 1992, pp. 76-82) . Therefore, THBDO compounds that have been enantiomerically-enriched are of particular interest.

A class of chemistry useful as intermediates in the preparation of racemic THBDO compounds is tetrahydrobenzothiepine-1-oxides (THBO compounds) . THBDO compounds and THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring. A method of preparing enantiomerically-enriched samples of another phenyl/seven-member fused ring system, the benzothiazepines, is described by Higashikawa (JP 59144777) , where racemic benzothiazepine derivatives are optically resolved on a chromatographic column containing chiral crown ethers as a stationary phase. Although optical resolution is achieved, the Higashikawa method is limited to producing only small quantities of the enantiomerically-enriched benzothiazepine derivatives. Giordano (CA 2068231) reports the cyclization of (2S, 3S) -aminophenylthiopropionates in the presence of a phosphonic acid to produce (2S, 3S) -benzothiazepin-4-ones . However, that preparation is constrained by the need to use enantiomerically-enriched starting materials rather than racemic starting materials. In addition, the Giordano method controls the stereochemistry of the seven-member ring of the benzothiazepin-4-one only at the 2- and 3 -positions. The 4- and 5-positions of the seven-member ring of the benzothiazepin-4-one are not asymmetric centers, and the stereochemistry at these sites therefore cannot be controlled by the Giordano method. A method by which enantiomerically-enriched 1,5- benzothiazepin-3-hydroxy-4 (5H) -one compounds have been produced is through the asymmetric reduction of 1,5- benzothiazepin-3,4 (2H, 5H) -dione compounds, reported by Yamada, et al . (J. Org. Chem. 1996, 61 (24), 8586-8590). The product is obtained by treating the racemic 1,5- benzothiazepin-3,4 (2H, 5H) -dione with the reaction product of an optically active alpha-amino acid and a reducing agent, for example sodium borohydride. Although a product with high optical purity was achieved, the method is limited by the use of a relatively expensive chemical reduction step.

The microbial reduction of racemic 1, 5-benzothiazepin- 3 , 4 (2H, 5H) -dione compounds to produce enantiomerically- enriched 1, 5-benzothiazepin-3-hydroxy-4 (5H) -one compounds is reported by Patel et al . , U.S. Patent 5,559,017. This method is limited by the inherent problems of maintaining a viable and pure bacterial culture of the appropriate species and variety. In addition, that method is limited in scale, producing only microgram quantities of the desired product. Until now, there have been no reported processes for preparing enantiomerically-enriched THBDO compounds or enantiomerically-enriched THBO compounds. Furthermore, there have been no reported processes for controlling the stereochemistry at the 4- and 5-positions of the seven- member rings of THBDO compounds or THBO compounds

FDA Grants Breakthrough Designation to Shire’s Rare GI Therapies

Tue, 06/14/2016

Shire announced that the U.S. Food and Drug Administration (FDA) has granted Breakthrough Therapy Designation for two investigational products for rare diseases: SHP621 (budesonide oral suspension, or BOS) for eosinophilic esophagitis (EoE), and SHP625 (maralixibat) for progressive familial intrahepatic cholestasis type 2 (PFIC2).

“Receiving Breakthrough Therapy Designation on two pipeline products this past week reflects the potential of our strong and innovative pipeline of more than 60 programs,” said Flemming Ornskov, M.D., MPH, and CEO, Shire. “Shire is committed to bringing innovation to the rare and specialty areas we focus on. We persevere to see compounds through the many stages of development through their challenges and successes, and always keep patients with unmet needs top of mind.”

EoE is a serious, chronic and rare disease that stems from an elevated number of eosinophils, a type of white blood cell, that infiltrate the walls of the esophagus. EoE is characterized by an inflammation of the esophagus that may lead to difficulty swallowing (dysphagia). The diagnosed prevalence of EoE ranges from approximately 15-55 cases per 100,000 persons, with high-end estimates reported by studies in Western regions.

PFIC refers to a group of autosomal-recessive liver disorders of childhood that disrupt bile formation and present with cholestasis. The symptoms of PFIC include severe itching of the skin (pruritus), and jaundice. PFIC is estimated to affect 1 in 50,000 to 1 in 100,000 births. PFIC2 is the most common type of PFIC, accounting for around half of cases.

According to the FDA, Breakthrough Therapy Designation is granted to a therapy that is intended to treat a serious or life-threatening disease or condition and preliminary clinical evidence indicates that the drug may demonstrate substantial improvement on one or more clinically significant endpoints over current standard of care. Under the designation, the FDA provides intensive guidance, organizational commitment involving senior managers, and eligibility for rolling and priority review of the application; this process helps ensure patients have access to therapies as soon as possible, pending approval. Breakthrough Therapy Designation does not guarantee that FDA will ultimately approve BOS for EoE or maralixibat for PFIC2, and the timing of any such approval is uncertain.

“On behalf of patients in the United States with EoE and PFIC2, we are so pleased that the FDA has granted Breakthrough Therapy Designation to BOS and maralixibat,” said Philip J. Vickers, Ph.D., Head of R&D, Shire. “We look forward to working with the agency to continue their development and, pending FDA approval, deliver these therapeutic options to the patients who need them most.”

Source: Shire

Patent

WO 2003022804

It is well established that agents which inhibit the transport of bile acids across the tissue of the ileum can also cause a decrease in the levels of cholesterol in blood serum. Stedronski, in “Interaction of bile acids and cholesterol with nonsystemic agents having hypocholesterolemic properties,” Biochimica et Biophysica Acta, 1210 (1994) 255-287 discusses biochemistry, physiology, and known active agents surrounding bile acids and cholesterol. Bile acids are actively transported across the tissue of the ileum by an apical sodium co-dependent bile acid transporter (ASBT), alternatively known as an ileal bile acid transporter (IBAT).
A class of ASBT-inhibiting compounds that was recently discovered to be useful for influencing the level of blood serum cholesterol comprises tetrahydrobenzothiepine oxides (THBO compounds, PCT Patent Application No. WO 96/08484). Further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 97/33882.
Additional THBO compounds useful as ASBT inhibitors are described in U.S. Patent No. 5,994,391. Still further THBO compounds useful as ASBT inhibitors are described in PCT Patent Application No. WO 99/64409. Included in the THBO class are tetrahydrobenzo-thiepine-l -oxides and tetrahydrobenzothiepine- 1,1 -dioxides. THBO compounds possess chemical structures in which a phenyl ring is fused to a seven-member ring.

Published methods for the preparation of THBO compounds include the synthesis through an aromatic sulfone aldehyde intermediate. For example l-(2,2-dibutyl-3-oxopropylsulfonyl)-2-((4-methoxyphenyl)methyl)benzene (29) was cyclized with potassium t-butoxide to form tetrahydrobenzothiepine- 1,1 -dioxide (svn-24) as shown in Eq. 1.

Compound 29 was prepared by reacting 2-chloro-5-nitrobenzoic acid chloride with anisole in the presence of aluminum trichloride to produce a chlorobenzophenone compound; the chlorobenzophenone compound was reduced in the presence of trifluoromethanesulfonic acid and triethylsilane to produce a chlorodiphenylmethane compound; the
chlorodiphenylmethane compound was treated with lithium sulfide and 2,2-dibutyl-3-(methanesulfonato)propanal to produce l-(2,2-dibutyl-3-oxopropylthio)-2-((4-methoxyphenyl)methyl)-4-dimethylaminobenzene (40); and 40 was oxidized with m-chloroperbenzoic acid to produce 29. The first step of that method of preparing compound 29 requires the use of a corrosive and reactive carboxylic acid chloride that was prepared by the reaction of the corresponding carboxylic acid with phosphorus pentachloride.
Phosphorus pentachloride readily hydrolyzes to produce volatile and hazardous hydrogen chloride. The reaction of 2,2-dibutyl-3-(methanesulfonato)propanal with the lithium sulfide and the chlorodiphenylmethane compound required the intermediacy of a cyclic tin compound to make the of 2,2-dibutyl-3-(methanesulfonato)propanal. The tin compound is expensive and creates a toxic waste stream.
In WO 97/33882 compound syn-24 was dealkylated using boron tribromide to produce the phenol compound 28. Boron tribromide is a corrosive and hazardous material that generates hydrogen bromide gas and requires special handling. Upon hydrolysis, boron tribromide also produces borate salts that are costly and time-consuming to separate and dispose of.

An alternative method of preparing THBO compounds was described in WO
97/33882, wherein a 1,3-propanediol was reacted with thionyl chloride to form a cyclic sulfite compound. The cyclic sulfite compound was oxidized to produce a cyclic sulfate compound. The cyclic sulfate was condensed with a 2-methylthiophenol that had been deprotonated with sodium hydride. The product of the condensation was a (2-methylphenyl) (3′-hydroxypropyl)thioether compound. The thioether compound was oxidized to form an thioether aldehyde compound. The thioether aldehyde compound was further oxidized to form an aldehyde sulfone compound which in turn was cyclized in the presence of potassium t-butoxide to form a 4-hydroxytetrahydrobenzothiepine 1,1 -dioxide compound. This cyclic sulfate route to THBO compounds requires an expensive catalyst. Additionally it requires the use of SOCI2, which in turn requires special equipment to handle.
PCT Patent Application No. WO 97/33882 describes a method by which the phenol compound 28 was reacted at its phenol hydroxyl group to attach a variety of functional groups to the molecule, such as a quaternary ammonium group. For example, (4R,5R)-28 was reacted with l,4-bis(chloromethyl)benzene (?,??’-dichloro-p-xylene) to produce the chloromethyl benzyl- ether (4R,5R)-27. Compound (4R,5R)-27 was treated with diazabicyclo[2.2.2]octane (DABCO) to produce (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4-hydroxy-l , 1 -dioxido-1 -benzothiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza-l-azomabicyclo[2.2.2]octane chloride (41). This method suffers from low yields because of a propensity for two molecules of compound (4R,5R)-28 to react with one molecule of l,4-bis(chloromethyl)benzene to form a bis(benzothiepine) adduct. Once the bis-adduct forms, the reactive chloromethyl group of compound (4R,5R)-27 is not available to react with an amine to form the quaternary ammonium product.

A method of preparing enantiomerically enriched tetrahydrobenzothiepine oxides is described in PCT Patent Application No. WO 99/32478. In that method, an aryl-3- hydroxypropylsulfide compound was oxidized with an asymmetric oxidizing agent, for example (lR (->(8,9-dichloro-10-camphorsulfonyl)oxaziridine, to yield a chiral aryl-3-hydroxypropylsulfoxide. Reaction of the aryl-3-hydroxypropylsulfoxide with an oxidizing agent such as sulfur trioxide pyridine complex yielded an aryl-3-propanalsulfoxide. The aryl- 3-propanalsulfoxide was cyclized with a base such as potassium t-butoxide to
enantioselectively produce a tetrahydrobenzothiepine- 1 -oxide. The tetrahydrobenzothiepine- 1 -oxide was further oxidized to produce a tetrahydrobenzothiepine- 1 , 1 -dioxide. Although this method could produce tetrahydrobenzothiepine- 1,1 -dioxide compounds of high enantiomeric purity, it requires the use of an expensive asymmetric oxidizing agent.
Some 5-amidobenzothiepine compounds and methods to make them are described in

PCT Patent Application Number WO 92/18462.
In Svnlett. 9, 943-944(1995) 2-bromophenyl 3-benzoyloxy-l-buten-4-yl sulfone was treated with tributyl tin hydride and AIBN to produce 3-benzoyloxytetrahydrobenzothiepine-1,1 -dioxide.
In addition to forming the desired ASBT inhibitors, it is also desirable to form such

ASBT inhibitors of higher purity and having lower levels of residual solvent impurities. This is especially so with respect to ASBT inhibitors having a positively charged substituent, for example, the compounds designated as 41 (supra) and 60 (infra).
It is further desirable to provide methods for making such high purity ASBT inhibitors.

Example 11.

Preparation of (4R,5R)- 1 -((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro-4- hydroxy- 1 , 1 -diυxido- 1 -benzithiepin-5-yl)pheπoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride,
41


41

Ste l. Preparation of (4R.5R1-26.


( 4R, 5R) -26
A 1000 mL 4 neck jacketed Ace reactor flask was fitted with a mechanical stirrer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask was purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). To this was added 4.2 grams of 50% sodium hydroxide. The mixture was heated to 50°C and stiπed for 15 minutes. To the flask was added 8.3 grams of 55 dissolved in 10 mL of DMAC, all at once. The temperature was held at 50°C for 24 hrs. To the flask was added 250 mL of toluene followed by 125 mL of dilution water. The mixture was stiπed for 15 minutes and the layers were then allowed to separate at 50°C. The flask was then charged with 125 mL of saturated sodium chloride solution and stiπed 15 minutes. Layers separated cleanly in 30 seconds at 50°C. Approximately half of the solvent was distilled off under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.


( 4R, 5R) -27
Toluene was charged back to the reaction mixture of Step 1 and the mixture was cooled to 35°C. To the mixture was then added 7.0 grams of thionyl chloride over 5 minutes. The reaction was exothermic and reached 39°C. The reaction turned cloudy on first addition of thionyl chloride, partially cleared then finally remained cloudy. The mixture was stirred for 0.5 hr and was then washed with 0.25N NaOH. The mixture appeared to form a small amount of solids that diminished on stirring, and the layers cleanly separated. The solvent was distilled to a minimum stir volume under vacuum at 50°C. The residual reaction mixture contained (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 was charged with 350 mL of methyl ethyl ketone (MEK) followed by 10.5 mL water and 6.4 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 10 mL of MEK. The mixture was heated to reflux, and HPLC showed <0.5% of (4R,5R)-27. The reaction remained homogenous initially then crystallized at the completion of the reaction. An additional 5.3 mL of water was charged to the flask to redissolve product. Approximately 160 mL of solvent was then distilled off at atmospheric pressure. The mixture started to form crystals after 70 mL of solvent was distilled. Water separated out of distillate indicating a ternary azeotrope between toluene, water and methyl ethyl ketone (MEK). The mixture was then cooled to 25°C. The solids were filtered and washed with 150 mL MEK, and let dry under vacuum at 60°C. Isolated 29.8.0 g of off-white crystalline 4 Example 11a.
Alternate Preparation of (4R,5R)-l-((4-(4-(3,3-dibutyl-7-(dimemylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a 28 mm Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 100 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 4.02 grams of 50% sodium hydroxide. The mixture is stiπed for 30 minutes. To the flask is added 8.7 grams of 55 dissolved in 12.5 mL of DMAC, all at once. The charge vessel is washed with 12.5 mL DMAC and the wash is added to the reactor. The reactor is stiπed for 3 hours. To the reactor is added 0.19 mL of 49.4% aq. NaOH and the mixture is stirred for 2 hours. To the mixture is added 0.9 g DABCO dissolved in 12.5 mL DMAC. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of dilution water. The mixture is stiπed for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stiπed 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (225 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 30°C. To the mixture is then added 6.7 grams of thionyl chloride over 30 to 45 minutes. The temperature is maintained below 35°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is stiπed for 0.5 hr and is then charged with 156.6 mL of 4% NaOH wash over a 30 minute period. The addition of the wash is stopped when the pH of the mixture reaches’ 8.0 to 10.0. The bottom aqueous layer is removed at 30°C and any rag layer is retained with the organic layer. To the mixture is charged 175 mL of saturated NaCl wash with agitation. The layers are separated at 30°C and the bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-27.

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 325 mL of methyl ethyl ketone (MEK) and 13 mL water. Next, the reactor is charged 6.2 grams of diazabicyclo[2.2.2]octane (DABCO) dissolved in 25 mL of MEK. The mixture is heated to reflux and held for 30 minutes. Approximately 10% of solvent volume is then distilled off. The mixture starts to form crystals during distillation. The mixture is then cooled to 20°C for 1 hour. The off-white crystalline 41 (Form U) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example lib.
Alternate Preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form II of 41

A 1000 mL 4 neck jacketed Ace reactor flask is fitted with a mechanical stiπer, a nitrogen inlet, an addition funnel or condenser or distilling head with receiver, a
thermocouple, four internal baffles and a Teflon turbine agitator. The flask is purged with nitrogen gas and charged with 25.0 grams of (4R,5R)-28 and 125 mL of N,N-dimethylacetamide (DMAC). The mixture is heated to 50°C and to it is added 7.11 grams of 30% sodium hydroxide over a period of 15 to 30 minutes with agitation. The mixture is stiπed for 30 minutes. To the flask is added 9.5 grams of solid 55. The reactor is stiπed for 3 hours. To the mixture is added 1.2 g of solid DABCO. The mixture is stiπed 30 to 60 minutes at 50°C. To the flask is added 225 mL of toluene followed by 125 mL of water. The mixture is stirred for 15 minutes and the layers are then allowed to separate at 50°C. The bottom aqueous layer is removed but any rag layer is retained with the organic layer. The flask is then charged with 175 mL of 5% hydrochloric acid solution and stirred 15 minutes. Layers are separated at 50°C to remove the bottom aqueous layer, discarding any rag layer with the aqueous layer. The flask is then charged with 225 mL of water and stirred 15 minutes. The layers are allowed to separate at 50°C. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. Approximately half of the solvent is distilled off under vacuum at a maximum pot temperature of 80°C. The residual reaction mixture contains (4R,5R)-26.

Step 2. Preparation of (4R.5RV27.

Toluene (112.5 mL) is charged back to the reaction mixture of Step 1 and the mixture is cooled to 25°C. To the mixture is then added 7.3 grams of thionyl chloride over 15 to 45 minutes. The temperature of the mixture is maintained above 20°C and below 40°C. The reaction turns cloudy on first addition of thionyl chloride, then at about 30 minutes the layers go back together and form a clear mixture. The mixture is then charged with 179.5 mL of 4% NaOH wash over a 30 minute period. The mixture is maintained above 20°C and below 40°C during this time. The addition of the wash is stopped when the pH of the mixture reaches 8.0 to 10.0. The mixture is then allowed to separate at 40°C for at least one hour.

The bottom aqueous layer is removed and any rag layer is retained with the organic layer. To the mixture is charged 200 mL of dilution water. The mixture is stiπed for 15 minutes and then allowed to separate at 40°C for at least one hour. The bottom aqueous layer is removed, discarding any rag layer with the aqueous layer. The solvent is distilled to a minimum stir volume under vacuum at 80°C. The residual reaction mixture contains (4R,5R)-2 .

Step 3. Preparation of 41.
To the reaction mixture of Step 2 is charged 350 mL of methyl ethyl ketone (MEK) and 7 mL water. The mixture is stiπed for 15 minutes and the temperature of the mixture is adjusted to 25°C. Next, the reactor is charged with 6.7 grams of solid
diazabicyclo[2.2.2]octane (DABCO). The mixture is maintained at 25°C for three to four hours. It is then heated to 65°C and maintained at that temperature for 30 minutes. The mixture is then cooled to 25°C for 1 hour. The off-white crystalline 41 (Form II) is filtered and washed with 50 mL MEK, and let dry under vacuum at 100°C.

Example 12.
Alternate preparation of (4R,5R)-1 -((4-(4-(3,3-dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro- 4-hydroxy- 1 , 1 -dioxido- 1 -benzithiepin-5-yl)phenoxy)methyl)phenyl)methyl-4-aza- 1 – azoniabicyclo[2.2.2]octane chloride, Form I of 41

(4R,5R)-27 (2.82 kg dry basis, 4.7 mol) was dissolved in MTBE (9.4 L). The solution of (4R,5R)-22 was passed through a 0.2 mm filter cartridge into the feeding vessel. The flask and was rinsed with MTBE (2 x 2.5 L). The obtained solution as passed through the cartridge filter and added to the solution of (4R,5R)-2 in the feeding vessel. DABCO
(diazabicyclo[2.2.2]octane, 0.784 kg, 7.0 mol) was dissolved in MeOH (14.2 L). The DABCO solution was passed through the filter cartridge into the 100 L nitrogen-flushed reactor. The Pyrex bottle and the cartridge filter were rinsed with MeOH (7.5 L) and the solution was added to the reactor. The (4R,5R)-22 solution was added from the feeding vessel into the reactor at 37°C over a period of 10 min, while stirring. Methanol (6.5 L) was added to the Pyrex bottle and via the cartridge filter added to the feeding vessel to rinse the remaining (4R,5R)-2 into the reactor. The reaction mixture was brought to 50-60°C over 10-20 min and stiπed at that temperature for about 1 h. The mixture was cooled to 20-25°C over a period of 1 h. To the reaction mixture, methyl t-butyl ether (MTBE) (42 L) was added over a period of 1 h and stiπed for a minimum of 1 h at 20 – 25°C. The suspension was filtered through a Buchner funnel. The reactor and the filter cake were washed with MTBE (2 x 14 L). The solids were dried on a rotary evaporator in a 20 L flask at 400 – 12 mbar, 40°C, for 22 h. A white crystalline solid was obtained. The yield of 4 . (Form I) was 3.08 kg (2.97 kg dry, 93.8 %) and the purity 99.7 area % (HPLC; Kromasil C 4, 250 x 4.6 mm column; 0.05% TFA in H2O/0.05% TFA in ACN gradient, UV detection at 215 nm).

Example 12a.
Conversion of Form I of Compound 41 into Form II of Compound 41.

To 10.0 grams of Form I of 4 . in a 400 mL jacketed reactor is added 140 mL of MEK. The reactor is stirred (358 φm) for 10 minutes at 23 °C for 10 minutes and the stirring rate is then changed to 178 φm. The suspension is heated to reflux over 1 hour using a programmed temperature ramp (0.95°C/minute) using batch temperature control (cascade mode). The delta Tmaχ is set to 5°C. The mixture is held at reflux for 1 hour. The mixture is cooled to

25°C. After 3 hours at 25°C, a sample of the mixture is collected by filtration. Filtration is rapid (seconds) and the filtrate is clear and colorless. The white solid is dried in a vacuum oven (80°C, 25 in. Hg) to give a white solid. The remainder of the suspension is stirred at 25°C for 18 hours. The mixture is filtered and the cake starts to shrink as the mother liquor reaches the top of the cake. The filtration is stopped and the reactor is rinsed with 14 mL of MEK. The reactor stirrer speed is increased from 100 to 300 φm to rinse the reactor. The rinse is added to the filter and the solid is dried with a rapid air flow for 5 minutes. The solid is dried in a vacuum oven at 25 in. Hg for 84 hours to give Form II of 4

PATENT

WO 2014144650

Scheme 3:

PAPER

Journal of Medicinal Chemistry (2005), 48(18), 5853-5868

Discovery of Potent, Nonsystemic Apical Sodium-Codependent Bile Acid Transporter Inhibitors (Part 2)

Department of Discovery Chemistry and Department of Cardiovascular Disease, Pharmacia, 700 Chesterfield Parkway W, Chesterfield, Missouri 63017, Office of Science and Technology, Chemical Science Division, Pharmacia, 800 Lindbergh Boulevard, Creve Coeur, Missouri 63167, Department of Pharmaceutical Sciences, Pharmacia, Skokie, Illinois, and Department of Chemistry, University of Missouri, St. Louis, Missouri
J. Med. Chem., 2005, 48 (18), pp 5853–5868
DOI: 10.1021/jm0402162

http://pubs.acs.org/doi/abs/10.1021/jm0402162

Abstract

Abstract Image

In the preceding paper several compounds were reported as potent apical sodium-codependent bile acid transporter (ASBT) inhibitors. Since the primary site for active bile acid reabsorption is via ASBT, which is localized on the luminal surface of the distal ileum, we reasoned that a nonsystemic inhibitor would be desirable to minimize or eliminate potential systemic side effects of an absorbed drug. To ensure bioequivalency and product stability, it was also essential that we identify a nonhygroscopic inhibitor in its most stable crystalline form. A series of benzothiepines were prepared to refine the structure−activity relationship of the substituted phenyl ring at the 5-position of benzothiepine ring and to identify potent, crystalline, nonhygroscopic, and efficacious ASBT inhibitors with low systemic exposure.

compd R IC50 (nM)b hygroscp I wt gain (%)c hygroscp II % wt gain (%)d crystallinitye
74 OCH2C6H4(p)CH2(N+)DB 0.28 1.59 2.1 yes

(4Rcis)-1-[[4-[[4-[3,3-Dibutyl-7-(dimethylamino)-2,3,4,5-tetrahydro-4-hydroxy-1,1-dioxido-1-benzothiepin-5-yl]phenoxy]methyl]phenyl]methyl]-4-aza-1-azoniabicyclo[2.2.2]octane Chloride Salt (74). Following a similar procedure as in General Method B, the title compound 74 was prepared from the corresponding chloromethyl benzyl ether and DABCO as a white solid, mp 223−230 °C (dec); 1H NMR (CDCl3) δ 0.89 (m, 6H), 1.27−1.52 (br m, 10H), 1.63 (m, 1H), 2.20 (m, 1H), 2.81 (s, 6H), 3.06 (ABq, JAB = 15.1 Hz, J = 43.3 Hz, 2H), 3.16 (s, 6H), 3.76 (s, 6H), 4.11 (d, J = 7.7 Hz, 1H), 5.09 (s, 2H), 5.14 (s, 2H), 5.48 (s, 1H), 5.96 (s, 1H), 6.49 (d, J = 8.9 Hz, 1H), 6.99 (d, J = 8.0 Hz, 2H), 7.26 (m, 1H), 7.44 (d, J = 8.0 Hz, 2H), 7.52 (d, J = 7.4 Hz, 2H), 7.68 (d, J = 7.4 Hz, 2H), 7.87 (d, J = 8.9 Hz, 1H). HRMS calcd for C40H56N3O4S:  674.3992; found, 674.4005. Anal. Calcd for C40H56N3O4S:  ‘ C, 67.62; H, 7.95; N, 5.92; S, 4.51. Found:  C, 67.48; H, 8.32; N, 5.85; S, 4.60.

a All compounds were prepared using method B in Scheme 3.b Taurocholate is transported across the baby hamster kidney cell membrane.c % weight gain in a 25 °C, 57% humidity chamber for 2 weeks.d % weight gain in a 40 °C, 80% humidity chamber for 2 weeks.e Crystallinity as determined by X-ray powder diffraction analysis.f (N+)DB is a DABCO terminal group with the quaternary ammonium attached to the linke

ANY ERROR EMAIL amcrasto@gmail.com, +919323115463

PATENT

https://www.google.com/patents/WO1999032478A1?cl=en

Inventors James Li, Ching-Cheng Wang, David B. Reitz, Victor Snieckus, Horng-Chih Huang,Andrew J. Carpenter,
Applicant G.D. Searle & Co.

Example 10. Preparation of enantiomerically-enriched (4R.5R)-1- r.4- r _4- .3.3 -Dibutyl-7- (dimethylamino) -2.3 ,4.5- tetrahydro-4-hydroxy-1, l-dioxido-l-benzothiepin-5- yl] henoxy] ethyl] phenyl1methyl] -4-aza-l- azoniabicyclo [2.2.2] octane chloride ( (4R,5R) -XXVII) ♦

Figure imgf000053_0001

( (4R,5R) -XXVII) * = chiral center

Step 1. Preparation of 4-flUoro-2- ( (4- methoxyphenyl) methyl) -phenol To a stirred solution of 23.66 g of 95% sodium hydride (0.94 mol) in 600 mL of dry toluene was added 100.0 g of 4- fluorophenol (0.89 mol) at 0°C. The mixture was stirred at 90°C for 1 hour until gas evolution stopped. The mixture was cooled down to room temperature and a solution of 139.71 g of 3 -methoxybenzyl chloride (0.89 mol) in 400 mL of dry toluene was added. After refluxing for 24 hours, the mixture was cooled to room temperature and quenched with 500 mL of water. The organic layer was separated, dried over MgS04, and concentrated under high vacuum. The remaining starting materials were removed by distillation. The crude dark red oil was filtered through a layer of 1 L of silica gel with neat hexane to yield 53.00 g (25.6%) of the product as a pink solid: *H NMR (CDC13) d 3.79 (s, 3H) , 3.90 (s, 2H) , 4.58 (s, IH) , 6.70-6.74 (m, IH) , 6.79-6.88 (m, 4H) , 7.11-7.16 (m, 2H) .

Step 2. Preparation of 4-fluoro-2- ( (4- methoxyphenyl) methyl) -thiophenol

Step 2a. Preparation of thiocarbamate To a stirred solution of 50.00 g (215.30 mmol) of 4- fluoro-2- ( ( -methoxyphenyl) methyl) -phenol in 500 mL of dry DMF was added 11.20 g of 60% sodium hydride dispersion in mineral oil (279.90 mmol) at 2°C. The mixture was allowed to warm to room temperature and 26.61 g of dimethylthiocarbamoyl chloride (215.30 mmol) was added. The reaction mixture was stirred at room temperature overnight. The mixture was quenched with 100 mL of water in an ice bath. The solution was extracted with 500 mL of diethyl ether. The ether solution was washed with 500 mL of water and 500 mL of brine. The ether solution was dried over MgS04 and stripped to dryness. The crude product was filtered through a plug of 500 mL silica gel using 5% ethyl acetate/hexane to yield 48.00 g (69.8%) of the product as a pale white solid: XH NMR (CDC13) d 3.21 (s, 3H) , 3.46 (s, 3H) , 3.80 (s, 3H) , 3.82 (s, 2H) , 6.78-6.86 (m, 3H) , 6.90- 7.00 (m, 2H) , 7.09 (d, J = 8.7 Hz, 2H) .

Step 2b. Rearrangement and hydrolysis of thiocarbamate to 4-fluoro-2- ( (4 -methoxyphenyl) methyl) -thiophenol A stirred solution of 48.00 g (150.29 mmol) of thiocarbamate (obtained from Step 2a) in 200 mL of diphenyl ether was refluxed at 270°C overnight. The solution was cooled down to room temperature and filtered through 1 L of silica gel with 2 L of hexane to remove phenyl ether. The rearrangement product was washed with 5% ethyl acetate/hexane to give 46.00 g (95.8%) of the product as a pale yellow solid: XH NMR (CDC13) d 3.02 (s, 3H) , 3.10 (s, 3H) , 3.80 (s, 3H) , 4.07 (s, 2H) , 6.82-6.86 (m, 3H) , 6.93 (dt, J = 8.4 Hz, 2.7 Hz, IH) , 7.08 (d, J = 8.7 Hz, 2H) , 7.49 (dd, J = 6.0 Hz, 8.7 Hz, IH) . To a solution of 46.00 g (144.02 mmol) of the rearrangement product (above) in 200 mL of methanol and 200 mL of THF was added 17.28 g of NaOH (432.06 mmol) . The mixture was refluxed under nitrogen overnight . The solvents were evaporated off and 200 mL of water was added. The aqueous solution was washed with 200 mL of diethyl ether twice and placed in an ice bath. The aqueous mixture was acidified to pH 6 with concentrated HCl solution. The solution was extracted with 300 mL of diethyl ether twice. The ether layers were combined, dried over MgS04 and stripped to dryness to afford 27.00 g (75.5%) of the product as a brown oil: XH NMR (CDC13) d 3.24 (s, IH) , 3.80 (s, 3H) , 3.99 (s, 2H) , 6.81-6.87 (m, 4H) , 7.09 (d, J = 8.7 Hz, 2H) , 7.27- 7.33 (m, IH) .

Step 3. Preparation of dibutyl cyclic sulfate

Step 3a. Preparation of 2 , 2-dibutyl-l, 3-propanediol . To a stirred solution of di-butyl-diethylmalonate (Aldrich) (150g, 0.55 mol in dry THF (700ml) in an acetone/dry ice bath was added LAH (1 M THF) 662 ml (1.2 eq. , 0.66 mol) dropwise maintaining the temperature between -20 to 0°C. The reaction was stirred at RT overnight. The reaction was cooled to -20°C and 40 ml of water, and 80 mL of 10% NaOH and 80 ml of water were added dropwise. The resulting suspension was filtered. The filtrate was dried over sodium sulphate and concentrated in vacuo to give diol 598.4 g (yield 95%) as an oil. MS spectra and proton and carbon NMR spectra were consistent with the product.

Step 3b. Preparation of dibutyl cyclic sulfite

A solution of 2 , 2-dibutyl-l, 3-propanediol (103 g, 0.548 0 mol, obtained from Step 3a) and triethylamine (221 g, 2.19 mol) in anhydrous methylene chloride (500 ml) was stirred at 0°C under nitrogen. To the mixture, thionyl chloride (97.8* g, 0.82 mol) was added dropwise and within 5 min the solution turned yellow and then black when the addition was 5 completed within half an hour. The reaction mixture was stirred for 3 hrs. at 0°C. GC showed that there was no starting material left. The mixture was washed with ice water twice then with brine twice . The organic phase was dried over magnesium sulfate and concentrated under vacuum 0 to give 128 g (100%) of the dibutyl cyclic sulfite as a black oil. Mass spectrum (MS) was consistent with the product .

Step 3c. Oxidation of dibutyl cyclic sulfite to 5 dibutyl cyclic sulfate

To a solution of the dibutyl cyclic sulfite (127.5 g , 0.54 mol, obtained from Step 3b) in 600 ml acetonitrile and 500 ml of water cooled in an ice bath under nitrogen was added ruthenium (III) chloride (1 g) and sodium periodate 0 (233 g, 1.08 mol) . The reaction was stirred overnight and the color of the solution turned black. GC showed that there was no starting material left. The mixture was extracted with 300 ml of ether and the ether extract was washed three times with brine. The organic phase was dried over magnesium sulfate and passed through celite. The filtrate was 5 concentrated under vacuum and to give 133 g (97.8%) of the dibutyl cyclic sulfate as an oil. Proton and carbon NMR and MS were consistent with the product.

Step 4. Preparation of aryl-3-hydroxypropylsulfide

10 To a stirred solution of 27.00 g (108.73 mmol) of 4- fluoro-2- ( (4-methoxyphenyl) methyl) thiophenol (obtained from Step 2) in 270 mL of diglyme was added 4.35 g of 60% sodium-, hydride dispersion in mineral oil (108.73 mmol) at 0°C. After gas evolution ceased, 29.94 g (119.60 mmol) of the

15 dibutyl cyclic sulfate (obtained from Step 3c) was added at 0°C and stirred for 10 minutes. The mixture was allowed to warm up to room temperature and stirred overnight. The solvent was evaporated and 200 mL of water was added. The solution was washed with 200 mL of diethyl ether and added

2025 mL of concentrated sulfuric acid to make a 2.0 M solution that was refluxed overnight. The solution was extracted with ethyl acetate and the organic solution was dried over MgS04 and concentrated in vacuo. The crude aryl-3 – hydroxypropylsulfide was purified by silica gel

25 chromatography (Waters Prep 500) using 8% ethyl acetate/hexane to yield 33.00 g (72.5%) of the product as a light brown oil: E NMR (CDC13) d 0.90 (t, J = 7.1 Hz, 6H) , 1.14-1.34 (m, 12H) , 2.82 (s, 2H) , 3.48 (s, 2H) , 3.79 (s, 3H) , 4.10 (s, 2H) , 6.77-6.92 (m, 4H) , 7.09 (d, J = 8.7 Hz,

302H) , 7.41 (dd, J = 8.7 Hz, 5.7 Hz, IH) . Step 5. Preparation of enantiomerically-enriched aryl-3 – hydroxypropylsulfoxide

To a stirred solution of 20.00 g (47.78 mmol) of aryl- 53 -hydroxypropylsulfide (obtained from Step 4) in 1 L of methylene chloride was added 31.50 g of 96% (12?) – ( -) – (8 , 8- dichloro-10-camphor-sulfonyl) oxaziridine (100.34 mmol, Aldrich) at 2°C. After all the oxaziridine dissolved the mixture was placed into a -30 °C freezer for 72 hours. The

10 solvent was evaporated and the crude solid was washed with 1 L of hexane. The white solid was filtered off and the hexane solution was concentrated in vacuo. The crude oil was purified on a silica gel column (Waters Prep 500) using 15% ethyl acetate/hexane to afford 19.00 g (95%) of the

15 enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide as a colorless oil: lH NMR (CDC13) d 0.82-0.98 (m, 6H) , 1.16-1.32 (m, 12H) , 2.29 (d, J – 13.8 Hz, IH) , 2.77 (d, J = 13.5 Hz, IH) , 3.45 (d, J = 12.3 Hz, IH) , 3.69 (d, J = 12.3 Hz, IH) , 3.79 (s, 3H) , 4.02 (q, J = 15.6 Hz, IH) , 6.83-6.93 (m, 3H) ,

207.00 (d, J = 8.1 Hz, 2H) , 7.18-7.23 (m, IH) , 7.99-8.04 (m, IH) . Enantiomeric excess was determined by chiral HPLC on a (2?,2?) -Whelk-0 column using 5% ethanol/hexane as the eluent. It showed to be 78% e.e. with the first eluting peak as the major product.

25

Step 6. Preparation of enantiomerically-enriched aryl-3- propanalsulfoxide

To a stirred solution of 13.27 g of triethylamine (131.16 mmol, Aldrich) in 200 mL dimethyl sulfoxide were

30 added 19.00 g (43.72 mmol) of enantiomerically-enriched aryl-3 -hydroxypropylsulfoxide (obtained from Step 5) and 20.96 g of sulfur trioxide-pyridine (131.16 mmol, Aldrich) at room temperature. After the mixture was stirred at room temperature for 48 hours, 500 mL of water was added to the mixture and stirred vigorously. The mixture was then 5 extracted with 500 mL of ethyl acetate twice. The ethyl acetate layer was separated, dried over MgS04, and concentrated in vacuo. The crude oil was filtered through 500 mL of silica gel using 15% ethyl acetate/hexane to give 17.30 g (91%) of the enantiomerically-enriched aryl-3-

10 propanalsulfoxide as a light orange oil: lE NMR (CDC13) d 0.85-0.95 (m, 6H) , 1.11-1.17 (m, 4H) , 1.21-1.39 (m, 4H) , 1.59-1.76 (m, 4H) , 1.89-1.99 (m, IH) , 2.57 (d, J = 14.1 Hz, IH) , 2.91 (d, J = 13.8 Hz, IH) , 3.79 (s, 3H) , 3.97 (d, J = 15.9 Hz, IH) , 4,12 (d, J = 15.9 Hz, IH) , 6.84-6.89 (m, 3H) ,

157.03 (d, J = 8.4 Hz, 2H) , 7.19 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 8.02 (dd, J = 8.7 Hz, 5.7 Hz, IH) , 9.49 (s, IH) .

Step 7. Preparation of the enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R, 5R)

20 To a stirred solution of 17.30 g (39.99 mmol) of enantiomerically-enriched aryl-3 -propanalsulfoxide (obtained from Step 6) in 300 mL of dry THF at -15°C was added 48 mL of 1.0 M potassium t-butoxide in THF (1.2 equivalents) under nitrogen. The solution was stirred at -15°C for 4 hours.

25 The solution was then quenched with 100 mL of water and neutralized with 4 mL of concentrated HCl solution at 0°C. The THF layer was separated, dried over MgS04, and concentrated in vacuo. The enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (4R,5R) was purified by

30 silica gel chromatography (Waters Prep 500) using 15% ethyl acetate/hexane to give 13.44 g (77.7%) of the product as a white solid: ‘H NMR (CDC13) d 0.87-0.97 (m, 6H) , 1.16-1.32 (m, 4H) , 1.34-1.48 (m, 4H) , 1.50-1.69 (m, 4H) , 1.86-1.96 (m, IH) , 2.88 (d, J = 13.0 Hz, IH) , 3.00 (d, J = 13.0 Hz, IH) , 3.85 (s, 3H) , 4.00 (s, IH) , 4.48 (s, IH) , 6.52 (dd, J = 9.9 5Hz, 2.4 Hz, IH) , 6.94 (d, J = 9 Hz, 2H) , 7.13 (dt, J = 8.4 Hz, 2.4 Hz, IH) , 7.38 (d, J = 8.7 Hz, 2H) , 7.82 (dd, J = 8.7 Hz, 5.7 Hz, IH) .

Step 8. Preparation of enantiomerically-enriched

10 tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R)

To a stirred solution of 13.44 g (31.07 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1-oxide (obtained from Step 7) in 150 mL of methylene chloride was added 9.46 g of 68% m-chloroperoxybenzoic acid (37.28 mmol,

15 Sigma) at 0 °C. After stirring at 0 °C for 2 hours, the mixture was allowed to warm up to room temperature and stirred for 4 hours. 50 mL of saturated Na2S03 was added into the mixture and stirred for 30 minutes. The solution was then neutralized with 50 mL of saturated NaHC03 solution.

20 The methylene chloride layer was separated, dried over MgS04, and concentrated in vacuo to give 13.00 g (97.5%) of the enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (4R,5R) as a light yellow solid: ‘H NMR (CDC13) d 0.89-0.95 (m, 6H) , 1.09-1.42 (m, 12H) , 2.16-2.26 (m, IH) ,

253.14 (q, J = 15.6 Hz, IH) , 3.87 (s, 3H) , 4.18 (s, IH) , 5.48 (s, IH) , 6.54 (dd, J = 10.2 Hz, 2.4 Hz, IH) , 6.96-7.07 (m, 3H) , 7.40 (d, J = 8.1 Hz, 2H) , 8.11 (dd, J = 8.6 Hz, 5.9 Hz, IH) .

30 Step 9. Preparation of enantiomerically-enriched 7-

(dimethylamino) tetrahydrobenzothiepine-1 , 1-dioxide (4R.5R) – To a solution of 13.00 g (28.98 mmol) of enantiomerically-enriched tetrahydrobenzothiepine-1, 1- dioxide (obtained from Step 8) in 73 mL of dimethylamine (2.0 M in THF, 146 mmol) in a Parr Reactor was added ca . 20 5 mL of neat dimethylamine . The mixture was sealed and stirred at 110 °C overnight, and cooled to ambient temperature. The excess dimethylamine was evaporated. The crude oil was dissolved in 200 mL of ethyl acetate and washed with 100 mL of water, dried over MgS04 and

10 concentrated in vacuo. Purification on a silica gel column (Waters Prep 500) using 20% ethyl acetate/hexane gave 12.43 g (90.5%) of the enantiomerically- enriched 7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (4R, 5R) as a colorless solid: *H NMR (CDC13) d 0.87-0.93 (m, 6H) ,

151.10-1.68 (m, 12H) , 2.17-2.25 (m, IH) , 2.81 (s, 6H) , 2.99 (d, J = 15.3 Hz, IH) , 3.15 (d, J = 15.3 Hz, IH) , 3.84 (s, 3H) , 4.11 (d, J = 7.5 Hz, IH) , 5.49 (s, IH) , 5.99 (d, J = 2.4 Hz, IH) , 6.51 (dd, J = 8.7 Hz, 2.4 Hz, IH) , 6.94 (d, J = 8.7 Hz, 2H) , 7.42 (d, J = 8.4 Hz, 2H) , 7.90 (d, J = 8.7 Hz,

20 IH) . The product was determined to have 78% e.e. by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. Recrystallization of this solid from ethyl acetate/hexane gave 1.70 g of the racemic product. The remaining solution was concentrated and recrystallized to

25 give 9.8 g of colorless solid. Enantiomeric excess of this solid was determined by chiral HPLC on a Chiralpak AD column using 5% ethanol/hexane as the eluent. It showed to have 96% e.e with the first eluting peak as the major product.

30 Step 10: Demethylation of 5- (4 ‘ -methoxyphenyl) -7-

(dimethylamino) tetrahydrobenzothiepine-1.1-dioxide (4R, 5R) To a solution of 47 g (99 mmol) of enantiomeric- enriched (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (obtained from Step 9) in 500 mL of methylene chloride at -10 °C was added dropwise a solution of boron tribromide (297 mL, 1M in methylene chloride, 297 mmol), and the resulting solution was stirred cold (-5 °C to 0 °C) for 1 hour or until the reaction was complete. The reaction was cooled in an acetone-dry ice bath at -10 °C, and slowly quenched with 300 mL of water. The mixture was warmed to 10 °C, and further diluted with 300 mL of saturated sodium bicarbonate solution to neutralize the mixture. The aqueous layer was separated and extracted with 300 mL of methylene chloride, and the combined extracts were washed with 200 mL of water, brine, dried over MgS04 and concentrated in vacuo. The residue was dissolved in 500 mL of ethyl acetate and stirred with 50 mL of glacial acetic acid for 30 minutes at ambient temperature. The mixture was washed twice with 200 mL of water, 200 mL of brine, dried over MgS04 and concentrated in vacuo to give the crude 4-hydroxyphenyl intermediate. The solid residue was recrystallized from methylene chloride to give 37.5 g (82%) of the desired (4R, 5R) -5- (4′ – hydoxyphenyl) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1- dioxide as a white solid: *H NMR (CDC13) d 0.84-0.97 (m, 6H) , 1.1-1.5 (m, 10H) , 1.57-1.72 (m, IH) , 2.14-2.28 (m, IH) , 2.83 (s, 6H) , 3.00 (d, J = 15.3 Hz, IH) , 3.16 (d, J – 15.3 Hz, IH) , 4.11 (s, 2H) , 5.48 (s, IH) , 6.02 (d, J – 2.4 Hz, IH) , 6.55 (dd, J = 9, 2.4 Hz, IH) , 6.88 (d, 8 , 7 Hz , 2H) , 7.38 (d, J – 8.7 Hz, 2H) , 7.91 (d, J = 9 Hz, 2H) .

Step 11: Preparation of enantiomerically-enriched chlorobenzyl intermediate Treat a solution of enantiomerically-enriched (4R,5R)- 5- (4′ -hydoxypheny1) -7- (dimethylamino) tetrahydrobenzothiepine-1, 1-dioxide (5.0 g, 10.9 mmol, obtained from Step 10) in acetone (100 mL) at 25 °C under N2 with powdered 5 K2C03 (2.3 g, 16.3 mmol, 1.5 eq.) and a, a’ -dichloro-p-xylene (6.7 g, 38.1 mmol, 3.5 eq.) . Stir the resulting solution at 65 °C for about 48 hours. Cool the reaction mixture to 25 °C and concentrate to 1/5 of original volume. Dissolve the residue in EtOAc (150 mL) and wash with water (2 x 150 mL) .

10 Extract the aqueous layer with EtOAc (2 x 150 mL) and wash the combined organic extracts with saturated aqueous NaCI (2 x 150 mL. Dry the combined extracts with MgS04 and concentrate in vacuo to provide the crude product . Purification by flash chromatography (5.4 x 45 cm silica,

1525-40% EtOAc/hexane) will afford the enantiomerically- enriched chlorobenzyl intermediate .

Step 12: Preparation of enantiomerically-enriched (4R.5R)- 1- r [4- [ [4- [3 , 3-Dibutyl-7- (dimethylamino) -2,3 , 4 , 5-tetrahvdro-

204 -hydroxy-1.1-dioxido-1-benzothiepin-5- yl] phenoxy] methyll phenyl! methyl] -4-aza-l- azoniabicyclo f2.2.2] octane chloride (XXVII)

Treat a solution of the enantiomerically-enriched chlorobenzyl intermediate (4.6 g, 7.7 mmol, obtained from

25 above in Step 11) in acetonitrile (100 mL) at 25 °C under N2 with diazabicyclo [2.2.2] -octane (DABCO, 0.95 g, 8.5 mmol, 1.1 eq.) and stir at 35 °C for 2 hours. Collect the precipitated solid and wash with CH3CN. Recrystallization from CH3OH/Et20 will give the desired title compound (XXVII) .

ANY ERROR,  EMAIL amcrasto@gmail.com, +919323115463

 

///////////FDA, Breakthrough Designation,  Shire, Rare GI Therapies, SHP625, maralixibat, progressive familial intrahepatic , Maralixibat chloride, 228113-66-4, UNII: V78M04F0XC, LUM 001, Lopixibat chloride, cholestasis type 2 (PFIC2), Maralixibat Chloride,  ماراليكسيبات كلوريد ,  氯马昔巴特 , Мараликсибата хлорид

CCCCC1(CS(=O)(=O)c2ccc(cc2[C@H]([C@H]1O)c3ccc(cc3)OCc4ccc(cc4)C[N+]56CCN(CC5)CC6)N(C)C)CCCC.[Cl-]

Share

FDA approves new diagnostic imaging agent FLUCICLOVINE F-18 to detect recurrent prostate cancer

 FDA 2016, Uncategorized  Comments Off on FDA approves new diagnostic imaging agent FLUCICLOVINE F-18 to detect recurrent prostate cancer
May 282016
 

FLUCICLOVINE F-18

Cyclobutanecarboxylic acid, 1-amino-3-(fluoro-18F)-, trans- [

  • Molecular FormulaC5H818FNO2
  • Average mass132.124 Da
Axumin (fluciclovine F 18)
fluciclovinum (18F)
GE-148
NMK36
trans-1-Amino-3-(18F)fluorcyclobutancarbonsäure [German] [ACD/IUPAC Name]
trans-1-Amino-3-(18F)fluorocyclobutanecarboxylic acid [ACD/IUPAC Name]
UNII-38R1Q0L1ZE
anti-1-amino-3-[18F]fluorocyclobutane-1-carboxylic acid
cas 222727-39-1
05/27/2016 11:27 AM EDT
The U.S. Food and Drug Administration today approved Axumin, a radioactive diagnostic agent for injection. Axumin is indicated for positron emission tomography (PET) imaging in men with suspected prostate cancer recurrence based on elevated prostate specific antigen (PSA) levels following prior treatment.

May 27, 2016

Release

The U.S. Food and Drug Administration today approved Axumin, a radioactive diagnostic agent for injection. Axumin is indicated for positron emission tomography (PET) imaging in men with suspected prostate cancer recurrence based on elevated prostate specific antigen (PSA) levels following prior treatment.

Prostate cancer is the second leading cause of death from cancer in U.S. men. In patients with suspected cancer recurrence after primary treatment, accurate staging is an important objective in improving management and outcomes.

“Imaging tests are not able to determine the location of the recurrent prostate cancer when the PSA is at very low levels,” said Libero Marzella, M.D., Ph.D., director of the Division of Medical Imaging Products in the FDA’s Center for Drug Evaluation and Research. “Axumin is shown to provide another accurate imaging approach for these patients.”

Two studies evaluated the safety and efficacy of Axumin for imaging prostate cancer in patients with recurrent disease. The first compared 105 Axumin scans in men with suspected recurrence of prostate cancer to the histopathology (the study of tissue changes caused by disease) obtained by prostate biopsy and by biopsies of suspicious imaged lesions. Radiologists onsite read the scans initially; subsequently, three independent radiologists read the same scans in a blinded study.

The second study evaluated the agreement between 96 Axumin and C11 choline (an approved PET scan imaging test) scans in patients with median PSA values of 1.44 ng/mL. Radiologists on-site read the scans, and the same three independent radiologists who read the scans in the first study read the Axumin scans in this second blinded study. The results of the independent scan readings were generally consistent with one another, and confirmed the results of the onsite scan readings. Both studies supported the safety and efficacy of Axumin for imaging prostate cancer in men with elevated PSA levels following prior treatment.

Axumin is a radioactive drug and should be handled with appropriate safety measures to minimize radiation exposure to patients and healthcare providers during administration. Image interpretation errors can occur with Axumin PET imaging. A negative image does not rule out the presence of recurrent prostate cancer and a positive image does not confirm the presence of recurrent prostate cancer. Clinical correlation, which may include histopathological evaluation of the suspected recurrence site, is recommended.

The most commonly reported adverse reactions in patients are injection site pain, redness, and a metallic taste in the mouth.

Axumin is marketed by Blue Earth Diagnostics, Ltd., Oxford, United Kingdom

Patent

http://www.google.com/patents/WO2014023775A1?cl=en

The non-natural amino acid [ F]-l-amino-3-fluorocyclobutane-l-carboxylic acid

([18F]-FACBC, also known as [18F]-Fluciclovine) is taken up specifically by amino acid transporters and has shown promise for tumour imaging with positron emission tomography (PET).

A known synthesis of [18F]-FACBC begins with the provision of the protected precursor compound 1 -(N-(t-butoxycarbonyl)amino)-3 –

[((trifluoromethyl)sulfonyl)oxy]-cyclobutane-l-carboxylic acid ethyl ester. This precursor compound is first labelled with [18F]-fluoride:

II before removal of the two protecting groups:

IT III

EP2017258 (Al) teaches removal of the ethyl protecting group by trapping the [18F]- labelled precursor compound (II) onto a solid phase extraction (SPE) cartridge and incubating with 0.8 mL of a 4 mol/L solution of sodium hydroxide (NaOH). After 3 minutes incubation the NaOH solution was collected in a vial and a further 0.8 mL 4 mol/L NaOH added to the SPE cartridge to repeat the procedure. Thereafter the SPE cartridge was washed with 3 mL water and the wash solution combined with the collected NaOH solution. Then 2.2 mL of 6 mol/L HCl was then added with heating to 60°C for 5 minutes to remove the Boc protecting group. The resulting solution was purified by passing through (i) an ion retardation column to remove Na+ from excess NaOH and Cl~ from extra HCl needed to neutralise excess of NaOH to get a highly acidic solution before the acidic hydrolysis step, (ii) an alumina column, and (iii) a reverse-phase column. There is scope for the deprotection step(s) and/or the

purification step in the production of [18F]-FACBC to be simplified.

Example 1: Synthesis of f FIFACBC

No-carrier- added [18F]fluoride was produced via the 180(p,n)18F nuclear reaction on a GE PETtrace 6 cyclotron (Norwegian Cyclotron Centre, Oslo). Irradiations were performed using a dual-beam, 30μΑ current on two equal Ag targets with HAVAR foils using 16.5 MeV protons. Each target contained 1.6 ml of > 96% [180]water (Marshall Isotopes). Subsequent to irradiation and delivery to a hotcell, each target was washed with 1.6 ml of [160]water (Merck, water for GR analysis), giving approximately 2-5 Gbq in 3.2 ml of [160]water. All radiochemistry was performed on a commercially available GE FASTlab™ with single-use cassettes. Each cassette is built around a one-piece-moulded manifold with 25 three-way stopcocks, all made of polypropylene. Briefly, the cassette includes a 5 ml reactor (cyclic olefin copolymer), one 1 ml syringe and two 5 ml syringes, spikes for connection with five prefilled vials, one water bag (100 ml) as well as various SPE cartridges and filters. Fluid paths are controlled with nitrogen purging, vacuum and the three syringes. The fully automated system is designed for single-step fluorinations with cyclotron-produced [18F]fluoride. The FASTlab was programmed by the software package in a step-by-step time-dependent sequence of events such as moving the syringes, nitrogen purging, vacuum, and temperature regulation. Synthesis of

[18F]FACBC followed the three general steps: (a) [18F]fluorination, (b) hydrolysis of protection groups and (c) SPE purification.

Vial A contained K222 (58.8 mg, 156 μπιοΐ), K2C03 (8.1 mg, 60.8 μπιοΐ) in 79.5% (v/v)

MeCN(aq) (1105 μΐ). Vial B contained 4M HC1 (2.0 ml). Vial C contained MeCN

(4.1ml). Vial D contained the precursor (48.4 mg, 123.5 μιηοΐ) in its dry form (stored at -20 °C until cassette assembly). Vial E contained 2 M NaOH (4.1 ml). The 30 ml product collection glass vial was filled with 200 mM trisodium citrate (10 ml). Aqueous

[18F]fluoride (1-1.5 ml, 100-200 Mbq) was passed through the QMA and into the 180-

H20 recovery vial. The QMA was then flushed with MeCN and sent to waste. The trapped [18F]fluoride was eluted into the reactor using eluent from vial A (730 μΐ) and then concentrated to dryness by azeotropic distillation with acetonitrile (80 μΐ, vial C). Approximately 1.7 ml of MeCN was mixed with precursor in vial D from which 1.0 ml of the dissolved precursor (corresponds to 28.5 mg, 72.7 mmol precursor) was added to the reactor and heated for 3 min at 85°C. The reaction mixture was diluted with water and sent through the tC18 cartridge. Reactor was washed with water and sent through the tC18 cartridge. The labelled intermediate, fixed on the tC18 cartridge was washed with water, and then incubated with 2M NaOH (2.0 ml) for 5 min after which the 2M NaOH was sent to waste. The labelled intermediate (without the ester group) was then eluted off the tC18 cartridge into the reactor using water. The BOC group was hydrolysed by adding 4M HC1 (1.4 ml) and heating the reactor for 5 min at 60 °C. The reactor content with the crude [18F]FACBC was sent through the HLB and Alumina cartridges and into the 30 ml product vial. The HLB and Alumina cartridges were washed with water (9.1 ml total) and collected in the product vial. Finally, 2M NaOH (0.9 ml) and water (2.1 ml) was added to the product vial, giving a purified formulation of [18F]FACBC with a total volume of 26 ml. Radiochemical purity was measured by radio-TLC using a mixture of MeCN:MeOH:H20:CH3COOH (20:5:5: 1) as the mobile phase. The radiochemical yield (RCY) was expressed as the amount of radioactivity in the [18F]FACBC fraction divided by the total used [18F]fluoride activity (decay corrected). Total synthesis time was 43 min.

The RCY of [18F]FACBC was 62.5% ± 1.93 (SD), n=4.

/////FDA,  diagnostic imaging agent,  recurrent prostate cancer, fda 2016, Axumin, marketed, Blue Earth Diagnostics, Ltd., Oxford, United Kingdom, fluciclovine F 18

C1[C@@](C[C@H]1[18F])(N)C(=O)O

Share

FDA´s new policy regarding grouping of supplements for CMC changes

 regulatory  Comments Off on FDA´s new policy regarding grouping of supplements for CMC changes
May 192016
 

The US Food and Drug Administration’s (FDA) Office of Pharmaceutical Quality (OPQ) released a new document outlining how supplements can be grouped together and submitted concurrently for the same chemistry, manufacturing and controls (CMC) changes. Find out more about Policy and Procedures regarding the Review of Grouped Product Quality Supplements.

http://www.gmp-compliance.org/enews_05320_FDA%B4s-new-policy-regarding-grouping-of-supplements-for-CMC-changes_15173,Z-RAM_n.html

On April 19, 2016 the US Food and Drug Administration’s (FDA) Office of Pharmaceutical Quality (OPQ) released a new document outlining how supplements can be grouped together and submitted concurrently for the same chemistry, manufacturing and controls (CMC) changes to multiple approved new drug applications (NDAs), abbreviated new drug applications (ANDAs) and biological license applications (BLAs) submitted by the same applicant.

The agency says the goal of its new policy is to make the process more efficient and consistent when reviewing grouped supplements.The term “grouped supplements” is used to describe two or more supplements reviewed and processed using the procedures set forth in the new document, though FDA makes clear that supplements cannot be grouped if submitted by a different applicant or if the supplements provide for different CMC changes. “The supporting data necessary for the review of the CMC changes should be the same for each of the grouped supplements,” FDA says. “Any supplement that provides for the same CMC changes but necessitates the review of data that is unique to that supplement (e.g., product-specific data) should not be grouped.”

Supplements can be grouped when the following criteria are met:

  • The cover letter for the supplements clearly states the purpose of the proposed CMC changes and indicates that the supplement is one of multiple submissions for the same change.
  • Each supplement includes a list of the application numbers (NDA, BLA, and ANDA, as appropriate) and identifies the drug products that will be covered by the CMC changes.
  • The supplements have the same submission date on Form FDA 356h.

“On a case-by-case basis, the Center may also group supplements that do not meet some or any of the criteria described above, if grouping the supplements is advantageous to the review process,” FDA says.

Circumstances where this may occur include cases when an applicant submits a group of supplements for the same CMC change and then, at a later date, submits additional supplements for the same change and requests FDA officials to include the second set of supplements in the group.

The Regulatory Business Project Manager (RBPM) and Branch Chief (BC) of the relevant review division will decide on a case-by-case basis whether such changes will be allowed, though FDA notes that “consideration will be given to whether the goal date for the original group of supplements could still be met if the second set of supplements is added to the review.”

Additionally, seven new procedures were outlined by FDA in the MAPP (Manual of Policies and Procedures).

Source: Regulatory Affairs Proffessional Society – See more at:  OFFICE OF PHARMACEUTICAL QUALITY Review of Grouped Product Quality Supplements

 

//////// supplements, FDA, MAPP, supplements for CMC changes

Share

FDA releases draft guidance on the use of comparability protocols for post approval changes

 regulatory  Comments Off on FDA releases draft guidance on the use of comparability protocols for post approval changes
Apr 292016
 

 

 

The US FDA released a draft guidance for industry “Comparability Protocols for Human Drugs and Biologics: Chemistry, Manufacturing, and Controls Information”. The guidance replaces the draft guidance published in February 2003. It provides recommendations on implementing postapproval changes through the use of comparability protocols (CPs). Read more about FDA´s draft guidance for industry “Comparability Protocols for Human Drugs and Biologics”.

On April 19, 2016, the US Food & Drug Administration (FDA) released a draft guidance for industry “Comparability Protocols for Human Drugs and Biologics: Chemistry, Manufacturing, and Controls Information”. Comments and suggestions regarding the draft guideline should be submitted within 60 days of publication.

The guidance replaces the draft guidance published in February 2003. It provides recommendations on implementing postapproval changes through the use of comparability protocols (CPs). A CP is a comprehensive, prospectively written plan for assessing the effect of proposed CMC postapproval changes on the identity, strength, quality, purity, and potency of a drug product or a biological product. Using a CP in an original application or prior approval supplement (PAS) will, in many cases, facilitate the subsequent implementation and reporting of CMC changes. This could result in moving a product into distribution or facilitating a proactive approach to reinforcing the drug supply chain sooner than without a submitted protocol.

The guidance emphasizes that it is intended to establish a framework to promote continuous improvement in the manufacturing of quality products by encouriging applicants to employ tools of  ICH Q8 to Q11:

  • Effective use of knowledge and understanding of the product and manufacturing process;
  • A robust control strategy;
  • Risk management activities over a product´s life cycle;
  • An effective pharmaceutical quality system.

An FDA approved submission containing a CP provides an applicant with an agreed-upon plan to implement the proposed change(s), and in many cases, justification to report the implementation of the proposed change(s) in a reduced reporting category.

FDAs recommendations for the CP content: The CP submission should provide a comprehensive, detailed plan for the implementation of proposed changes and should include the information described below:

  • Summary;
  • Description of and Rationale for the Proposed Changes;
  • Supporting Information and Analysis (based on knowledge and risk assessments, information from development);
  • Comparability Protocol for the Proposed Change(s) – the CP should describe the specific tests and studies to be performed, including analytical procedures to be used and criteria to be achieved for the expected results. The level of detail that should be provided will depend on the complexity of the change and the specific risks associated with the change to product quality;
  • Proposed Reduced reporting category (i.e., an annual report, CBE, or CBE-30);
  • Other Information.

Additionally, the draft guidance provides a “Questions and Answers” section on CPs in the Appendix, which covers general questions and questions regarding formulation, manufacturing site and process, specification (including analytical methods), packaging, and process analytical technology (PAT) changes.

CPs together with “established conditions” may be effective tools for the overall product life cycle management. They can also facilitate the management of post-approval CMC changes in a more predictable and efficient manner, as it is the intention of the planned ICH Q12 Guideline “Lifecycle Management”. Steps 1 and 2 a/b of ICH Q12 are expected for June 2017.

For more information please visit the ICH website and see the FDA draft guidance for industry “Comparability Protocols for Human Drugs and Biologics: Chemistry, Manufacturing, and Controls Information“.

///////draft guidance for industry, Comparability Protocols for Human Drugs and Biologics, Chemistry, Manufacturing, Controls Information, fda

Share

FDA approves new drug Venclexta (venetoclax) for chronic lymphocytic leukemia in patients with a specific chromosomal abnormality

 Uncategorized  Comments Off on FDA approves new drug Venclexta (venetoclax) for chronic lymphocytic leukemia in patients with a specific chromosomal abnormality
Apr 122016
 
Venetoclax.svg
Venclexta (venetoclax)
04/11/2016 12:12 PM EDT
The U.S. Food and Drug Administration today approved Venclexta (venetoclax) for the treatment of patients with chronic lymphocytic leukemia (CLL) who have a chromosomal abnormality called 17p deletion and who have been treated with a least one prior therapy. Venclexta is the first FDA-approved treatment that targets the B-cell lymphoma 2 (BCL-2) protein, which supports cancer cell growth and is overexpressed in many patients with CLL.

April 11, 2016

Release

The U.S. Food and Drug Administration today approved Venclexta (venetoclax) for the treatment of patients with chronic lymphocytic leukemia (CLL) who have a chromosomal abnormality called 17p deletion and who have been treated with at least one prior therapy. Venclexta is the first FDA-approved treatment that targets the B-cell lymphoma 2 (BCL-2) protein, which supports cancer cell growth and is overexpressed in many patients with CLL.

According to the National Cancer Institute, CLL is one of the most common types of leukemia in adults, with approximately 15,000 new cases diagnosed each year. CLL is characterized by the progressive accumulation of abnormal lymphocytes, a type of white blood cell. Patients with CLL who have a 17p deletion lack a portion of the chromosome that acts to suppress cancer growth. This chromosomal abnormality occurs in approximately 10 percent of patients with untreated CLL and in approximately 20 percent of patients with relapsed CLL.

“These patients now have a new, targeted therapy that inhibits a protein involved in keeping tumor cells alive,” said Richard Pazdur, director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research. “For certain patients with CLL who have not had favorable outcomes with other therapies, Venclexta may provide a new option for their specific condition.”

The efficacy of Venclexta was tested in a single-arm clinical trial of 106 patients with CLL who have a 17p deletion and who had received at least one prior therapy. Trial participants took Venclexta orally every day, beginning with 20 mg and increasing over a five-week period to 400 mg. Results showed that 80 percent of trial participants experienced a complete or partial remission of their cancer.

Venclexta is indicated for daily use after detection of 17p deletion is confirmed through the use of the FDA-approved companion diagnostic Vysis CLL FISH probe kit.

The most common side effects of Venclexta include low white blood cell count (neutropenia), diarrhea, nausea, anemia, upper respiratory tract infection, low platelet count (thrombocytopenia) and fatigue. Serious complications can include pneumonia, neutropenia with fever, fever, autoimmune hemolytic anemia, anemia and metabolic abnormalities known as tumor lysis syndrome. Live attenuated vaccines should not be given to patients taking Venclexta.

The FDA granted the Venclexta application breakthrough therapy designation, priority review status, and accelerated approval for this indication. These are distinct programs intended to facilitate and expedite the development and review of certain new drugs in light of their potential to benefit patients with serious or life-threatening conditions. Venclexta also received orphan drug designation, which provides incentives such as tax credits, user fee waivers and eligibility for exclusivity to assist and encourage the development of drugs for rare diseases.

Venclexta is manufactured by AbbVie Inc. of North Chicago, Illinois, and marketed by AbbVie and Genentech USA Inc. of South San Francisco, California. The Vysis CLL FISH probe kit is manufactured by Abbott Molecular of Des Plaines, Illinois.

///FDA,  approves,  new drug, chronic lymphocytic leukemia,  specific chromosomal abnormality, Venclexta, venetoclax,  fda 2016
Share
Follow

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

Join other followers: