What is a (35 USC §123(a)) or a (15 CFR 123(a))?
USC means United States Code; CFR means Code of Federal Regulations
These citations* are shorthand for the laws and regulations that explain in precise terms what is needed in order for the Federal government to do business. Each is a citation which refers to a particular section of the law or its implementing regulations.Each law is signed by the President after being enacted by votes of the House of Representatives and Senate. Many new laws are assigned a number in the United States Code which reflects their relationship to similar laws or laws that govern similar programs. The way laws are created follow a formal process which you can learn more about from "How Our Laws are Made." The Code of Federal Regulations is written to explain in detail how the laws are to be carried out. When a law is written, it usually does not explain in great detail what procedures are to be followed, nor does it include descriptions of the special situations which can arise. This is the job of the regulations, which govern the day-to-day business of the Federal government. Regulations are written by the government agencies responsible for the subject matter of the laws. The United States Patent and Trademark Office writes the regulations concerning patents and trademarks which are found in Title 37 of the CFR. Below are diagrams explaining how to read these notations: |
| |
Citations of United States Code
To look up a particular law: Go to USC digital libraries or FedLaw; also see Patent's 35 USC (laws) [PDF] and Trademark Rules of Practice and Federal Statutes
| ||
Citations of Code of Federal Regulations
To look up a particular regulation: Go to CFR digital libraries; also see Patent's 37 CFR (rules) [PDF] and Trademark Rules of Practice and Federal Statutes
|
All about Patents and Intellectual property by DR ANTHONY MELVIN CRASTO, Worldpeaceambassdor, worlddrugtracker, Ph.D ( ICT, Mumbai) , INDIA 30+Yrs Exp. in the feld of Organic Chemistry, Serving chemists around the world. THE VIEWS EXPRESSED ARE MY PERSONAL AND IN NO-WAY SUGGEST THE VIEWS OF THE PROFESSIONAL BODY OR THE COMPANY THAT I REPRESENT, amcrasto@gmail.com, +91 9323115463 India
HOW TO FILE INDIAN PATENT
- Home
- ABOUT ME
- GOOGLE SITE
- PATENT SEARCH ENGINE
- Patent Download Websites
- SIPO CHINA PATENT SEARCH
- WIPO
- Glossary
- IP AUSTRALIA
- FREE ONLINE PATENTS
- Intellectual Property in China
- PATENT OFFICES
- Indian patent search
- Patent expiry search
- Korea patent search in English
- HOW TO LOOK FOR PATENT TERM EXTENSION, PTA
- HOW TO FILE INDIAN PATENT
- HOW TO PREPARE PATENT LANDSCAPE
- HOW TO SEARCH PATENT FAMILY
Monday 2 February 2015
What is a (35 USC §123(a)) or a (15 CFR 123(a))?
NEW LOOK USPTO
New USPTO Website Launched on February 5. 2015
The current beta version of the USPTO.gov website will become the new website beginning February 5th. You can preview the new site and watch a one-minute video on the site’s new features.
You are viewing the BETA version of the NEW USPTO website. WATCH a 1-minute video on the site’s new features.
We encourage feedback before the site launches Feb. 5th 2015. The current USPTO web site remains available during the BETA period.
We encourage feedback before the site launches Feb. 5th 2015. The current USPTO web site remains available during the BETA period.
Sunday 1 February 2015
Firmenich's New Enantioselective Synthesis of (-)-beta-Santalol
Charles Fehr and co-workers at
Firmenich have designed the "right cat for the desired
odor". In WIPO Patent Application WO/2009/141781
(November 26, 2009), they disclose a novel, and seemingly
practical, approach to the synthesis of beta-Santalol (the
Golden Grail of Sandalwood odor).
See also - Charles Fehr,
Iris Magpantay, Jeremy Arpagaus, Xavier Marquet, Magali
Vuagnoux, Enantioselective
Synthesis of (-)-beta-Santalol by a Copper-Catalyzed Enynol
Cyclization-Fragmentation Reaction, Angewandte Chemie,
Volume 121, Issue 39, Date: September 14, 2009, Pages:
7357-7359
β-Santalol is an organic compound that is classified as a sesquiterpene. It comprises about 20% of the oil of sandalwood, the major component being α-santalol. As of 2002, about 60 tons of sandalwood oil are produced annually by steam distillation of the heartwood of Santalum album.[1]
β-Santalol is an organic compound that is classified as a sesquiterpene. It comprises about 20% of the oil of sandalwood, the major component being α-santalol. As of 2002, about 60 tons of sandalwood oil are produced annually by steam distillation of the heartwood of Santalum album.[1]
References
- Karl-Georg Fahlbusch, Franz-Josef Hammerschmidt, Johannes Panten, Wilhelm Pickenhagen, Dietmar Schatkowski, Kurt Bauer, Dorothea Garbe, Horst Surburg "Flavors and Fragrances" in Ullmann's Encyclopedia of Industrial Chemistry, Wiley-VCH, Weinheim: 2002. Published online: 15 January 2003; doi:10.1002/14356007.a11_141.
Out of the woods
Woody
fragrances such as Iso E Super, a compound known as a 'floraliser' and
used in most new fine fragrances, also rate highly, says Gautier. Some
people smell a cedarwood note in Iso E Super, while others perceive it
to be musky. The male fragrance Fahrenheit (Dior, 1988) is 25 per cent Iso E Super while Perles de lalique
(Lalique, 2007) is 80 per cent Iso E Super. The commercial Iso E Super
owes its woody smell to a 5 per cent impurity which has an odour
threshold about 100 000 times lower than that of the main product.
Alpha- (top) and beta-santalol are constituents of the prized East Indian sandalwood oil
|
In
1999, Kraft's team at Givaudan isolated this impurity by epoxidation of
a commercial Iso E Super and proposed a structure based on NMR spectra.
The team came up with a way to synthesise the compound but it was not
commercially viable and the structure turned out to be too complex for
an alternative synthetic procedure. But out of several synthesised
analogues, one had an almost equal odour threshold and smelled even
better. What is more, its industrial synthesis is straightforward and it
has become a big hit - used by Givaudan as the captive material
Georgywood.
However, on strictly cash terms, the
most valuable fragrances are the natural products. For example, East
Indian sandalwood oil is one of the most precious and expensive
perfumery raw materials.
Beta-santalol is the main
olfactory constituent of sandalwood oil. It wasn't until 1990 that
beta-santalol could be prepared in the lab - this took an 11-step
reaction that was unsuitable for industrial scale-up. The best
sandalwood oil substitutes are derivatives of alpha-campholenic
aldehyde, prepared from inexpensive alpha-pinene, most of which is a
byproduct of the paper industry.
'The components in
sandalwood oil are not easy to make,' acknowledges Gautier. 'But what
Firmenich has done is screened all around that kind of structure to try
to find woody notes. We have a lot of substitutes so that perfumers can
create something close to sandalwood without needing sandalwood oil.'
Kraft agrees that there are 'excellent synthetic substitutes' out there.
For example, the campholenic aldehyde derivatives typically have a good
sandalwood smell. 'The other olfactory parts of natural sandalwood oil -
such as the smokey, cedar-like aspects - can easily be added using, for
example, cedarwood oil,' says Kraft.
Sandalwood was initially quite cheap (Guerlain's Samsara
contained 25 per cent sandalwood) but overharvesting has led to Indian
sandalwood trees being listed on the World Conservation Union's
threatened species red list.
'There's still a good
market for sandalwood oil and the price has gone, in the past six or
seven years, from $1000 [£680] per kg to over $2000,' says Leffingwell.
One Australian firm, TFS Corporation, has taken the initiative and
planted over 1700 hectares of the endangered Indian sandalwood (Santalum album ) in plantations in Western Australia.
http://www.google.com/patents/WO2009141781A1?cl=en
A solution of butyllithium in hexanes (1.35 M, 11.7 ml, 15.8 mmol) was added over a 15 minutes period to a stirred suspension of ethyltriphenylphosphonium iodide (6.61g, 15.8 mmol) in THF (125 ml) at 00C. The resultant red solution was cooled to -78°C and 3-(2-methyl-3-methylene-bicyclo[2.2.1]hept-2-exo-yl)-propanal (2.55 g, 14.33 mmol) in solution in THF (16 ml) was added over a 15 minutes period. After further 5 minutes at -78°C, a solution of butyllithium in hexanes (1.35 M, 12.7 mL, 17.2 mmol) was added over a 5 minutes period and the mixture was further stirred for 20 minutes at -78°C before allowing to reach 00C in 2 hours. Dry paraformaldehyde (2.60 g, 86.0 mmol) was added in one portion to the deep red homogeneous solution and the mixture was stirred for 30 minutes at 00C and was allowed to reach room temperature. After 1 hour at room temperature the mixture was poured into 5.2 ml of saturated aqueous solution of NH4Cl and extracted twice with CH2Cl2. The organic layer was washed with water and brine, and dried with Na2SO4. The mixture was filtered through a short pad of silica gel with dichloromethane as eluent and solvents were removed under pressure to give a crude. Purification of crude compound was performed by flash chromatography on silica gel with cyclohexane/AcOEt 90/10) as eluent to give pure β-santalol as pale yellow oil. Further bulb to bulb distillation under reduced pressure afforded β-santalol in 50% yield (Z:E=95:5). 1H NMR (CDCl3, 400 MHz): 5.29 (t, J=7.5, IH), 4.73 (s, IH), 4.45 (s, IH), 4.14 (s, 2H), 2.66 (d, J=3.8, IH), 2.12-1.94 (m, 3H), 1.78 (d, J=I.2, 3H), 1.71-1.60 (m, 3H),
1.44-1.36 (m, 2H), 1.32 (br s, IH), 1.27-1.17 (m,3H), 1.04 (s, 3H). 13C NMR (CDCl3, 125 MHz): 166.2, 133.9, 129.0, 99.7, 61.6, 46.8, 44.7, 44.6, 41.5, 37.1, 29.7, 23.7, 23.2, 22.6, 21.2.
B) Preparation of β-santalol (via [1,4] hydrogenation) i) Preparation of a compound (V): 2-methyl-5-(2-methyl-3-methylene-bicyclo[2.2.1] hept-exo-2-yl)-pent-2-enal
3-(2-Methyl-3-methylene-bicyclo[2.2.1]hept-2-exo-yl)-propan-l-al (274.0 mg,
1.54 mmol) was dissolved in toluene (15.0 ml) at room temperature under nitrogen. The mixture was heated to reflux and propionaldehyde (0.4 ml, 1.96 mmol) and aqueous catalytic solution of hexamethyleneimine and benzoic acid (0.12 ml, 0.616 mmol) was separately added in one portion. Once that addition was finished, the mixture was further heated at reflux for 6 hours. The mixture was cooled down to room temperature and extracted twice with brine, the organic layer was dried over MgSθ2, filtered off and concentrated to give a crude which was further purified by flash chromatography with cyclohexane/ AcOEt (95/5) to afford the title compound in 80% yield.
1H NMR: 9.38 (s, IH), 6.48 (dt, J1=?^, J2=1.2, IH), 4.78 (s, IH), 4.49 (s, IH), 2.69 (d, J=3.9, IH), 2.40-2.29 (m, 2H), 2.12 (d, J=3.1, IH), 1.75 (s, 3H), 1.72-1.64 (m, 3H), 1.59-1.51 (m, IH), 1.47-1.36 (m, 2H), 130-1.21 (m, 2H), 1.09 (s, 3H). 13C NMR: 195.2, 165.5, 155.2, 139.1, 100.3, 46.8, 44.8, 44.7, 39.4, 37.1 , 29.6, 24.9, 23.7, 22.6, 9.1.
H) Preparation of a compound (IV): Acetic acid 2-methyl-5-(2-methyl-3-methylene- bicyclo[2.2.1]hept-2-exo-yl)-penta-l,3-dienyl ester To a stirred solution of 2-methyl-5-(2-methyl-3-methylene-bicyclo[2.2.1]hept-2-yl)- pent-2-enal (268.0 mg, 1.23 mmol) in toluene (3.0 ml) were added Ac2O (0.35 ml, 3.70 mmol), Et3N (0.70 ml, 5.02 mmol), and a catalytic amount of DMAP (15.0 mg, 0.1 mmol). The resulting mixture was heated to reflux for 22 hours. The mixture was cooled down to room temperature and quenched with brine, extracted twice with Et2O, dried over MgSO4, filtered off and concentrated to give a crude which was further purified by flash chromatography with cyclohexane/AcOEt (98/2) to afford the title compound in 82% yield (E:Z=79:21). 1H NMR: 7.18 (s, IH), 5.99 (d, J=12.4, IH), 5.72 (dt, il=\2A, J2=6.1, IH), 4.76 (s, IH), 4.47 (s, IH), 2.68 (d, 3.4, IH), 2.17 (s, 3H), 2.12-2.01 (m, 2H), 1.81 (d, J=I.0, 3H), 1.73-1.63 (m, 3H), 1.43-1.39 (m, 2H), 1.27-1.18 (m, 2H), 1.02 (s, 3H).
13C NMR: 167.9, 165.5, 134.4, 130.6, 126.9, 120.7, 100.0, 46.9, 45.3, 45.0, 44.5, 37.0, 29.7, 23.6, 23.0, 20.8, 10.4.
Hi) Preparation of(2Z)-Acetic acid 2-methyl-5-(2-methyl-3-methylene-bicyclo[2.2.1] hept-2-exo-yl)-pent-2-enyl ester (VII)
Acetic acid 2-methyl-5-(2-methyl-3-methylene-bicyclo[2.2.1]hept-2-exo-yl)-penta-l,3- dienyl ester (6.80 g, 93% pure; 24.3 mmol0.18 mmol) was treated with
[(Cp*)Ru(COD)]BF4 (52 mg, 0.122 mmol) and maleic acid (230 mg, 1.95 mmol) in dry and degassed acetone (20 ml) at 600C under 4 bars of H2 for 24 hours. The product was extracted with pentane/5% NaOH, washed twice with saturated aqueous NaCl, dried
(Na2SO4) and bulb-to-bulb distilled: 6.80 g (81% Z and 5% E by GC; 92%). 1H NMR: 5.38 (t, J=7.1, IH), 4.73 (s, IH), 4.59 (s, IH), 4.45 (s, IH), 2.66 (br s, IH),
2.12-2.04 (m, 4H), 2.07 (s, 3H), 1.73 (d, J= 1.0, 3H), 1.69-1.61 (m, 3H), 1.45-
1.37 (m, 2H), 1.27-1.17 (m, 3H), 1.04 (m, 3H).
13C NMR: 171.1, 166.1, 131.4, 129.4, 99.7, 63.2, 46.8, 44.7, 44.6, 41.2, 37.1, 29.7, 23.7,
23.4, 22.6, 21.5, 21.0.
β-Santalol | |
---|---|
(2Z)-2-Methyl-5-[2-methyl-3-methylene-bicyclo[2.2.1]hept-2-yl]pent-2-en-1-ol
|
|
Identifiers | |
CAS number | 77-42-9 |
EC number | 201-027-2 |
Jmol-3D images | Image 1 |
Properties | |
Molecular formula | C15H24O |
Molar mass | 220.35 g mol−1 |
Appearance | Liquid |
Density | 0.9717 g/cm3 |
Boiling point | 177 °C (351 °F; 450 K) |
Solubility in water | Practically insoluble |
Solubility in ethanol | Soluble |
Solubility in diethyl ether | Soluble |
Chiral rotation ([α]D)
|
−87.1° |
Refractive index (nD)
|
1.5100 |
Related compounds | |
Related terpenes | α-Santalol |
Except where noted otherwise, data are given for materials in their standard state (at 25 °C (77 °F), 100 kPa) |
see
http://pubs.rsc.org/en/content/articlelanding/2014/ob/c3ob42174k#!divAbstract
Biocatalyst mediated regio- and stereo-selective hydroxylation and epoxidation on (Z)-α-santalol were achieved for the first time, using a fungal strain Mucor piriformis. Four novel metabolites were characterized as 10,11-cis-β-epoxy-α-santalol, 5α-hydroxy-(Z)-α-santalol, 10,11-dihydroxy-α-santalol and 5α-hydroxy-10,11-cis-β-epoxy-α-santalol. Using Amano PS lipase from Burkholderia cepacia, α- and β-isomers of 10,11-cis-epoxy-α-santalol were resolved efficiently.
Friday 30 January 2015
Data exclusivity for medicinal products in Europe...8+2+1 approach
Data exclusivity for medicinal products in Europe
The pharmaceutical sector is heavily regulated, with significant costs associated with both developing a new medicinal product and generating the data required to get a product to market. Protecting that data is therefore important. Data exclusivity is a form of product exclusivity right for medicinal products in Europe, and market exclusivity is a related form of additional protection.
These two rights are in addition to any granted patent exclusivity right covering a medicinal product.
Why is data exclusivity granted?
The rationale for granting data and market exclusivity is to compensate the innovator company for the investment it has put in to developing the new medicinal product and to generating the data required to obtain a marketing authorisation.
Regulatory approval for medicinal products requires applicants to provide information about the efficacy and safety of their product to regulatory authorities. The first applicant for approval of a new medicinal product must provide a substantial body of data relating to the product (including the results of pre-clinical tests and clinical trials).
The regulatory regime permits generic companies, who subsequently wish to gain their own approval for the same drug substance, to rely on information filed by the innovator company that made the first application. In order to be able to benefit from the data provided by the innovator in their regulatory filings for that medicinal product - the "reference medicinal product" - a generic company must show that their product has the same qualitative and quantitative composition as that product and that it is bioequivalent.
So what is the impact of a product enjoying "data exclusivity"?
An innovator company enjoys a period of "data exclusivity" during which their pre-clinical and clinical trials data may not be referenced in the regulatory filings of another company(typically a generic company) for the same drug substance.
For marketing authorisation applications made from November 2005 onwards, the period of data exclusivity in Europe has been harmonised as 8 years from the date of first authorisation in Europe. For marketing authorisation applications made before November 2005, the period of data exclusivity varies from EU member state to EU member state, and is either 6 years or 10 years.
For marketing authorisation applications made from November 2005 onwards, there is an additional period of 2 years of "market exclusivity". This is the period of time during which a generic company may not market an equivalent generic version of the originator's pharmaceutical product (although their application for authorisation may be processed during this period, such that they are in a position to market their product on the expiry of this additional 2 year period).
An extra year of protection for new indications
As noted above, the rules determining exclusivity changed in 2005. Under the "old rules" data exclusivity lasted either 6 or 10 years. The "new rules" follow an "8 + 2 + 1" year approach:
"Under the 'old rules' data exclusivity lasted up to 10 years. The 'new rules' follow an '8 + 2 + 1' year approach."
- During the first 8 years from the grant of the innovator company's marketing authorisation, data exclusivity applies.
- After the 8 years have expired a generic company can make use of the pre-clinical and clinical trial data of the originator in their regulatory applications, but still cannot market their product.
- After a period of 10 years from the grant of the innovator company's marketing authorisation, the generic company can also market their product, unless the innovator product qualifies for a further one year of exclusivity.
- This additional 1 year may be obtained in a number of circumstances, such as where the innovator company is granted a marketing authorisation for a significant new indication for the relevant medicinal product. In such a situation the generic company can only market their product after 11 years from the grant of the innovator company's marketing authorisation.
How to Calculate Data Exclusivity Periods in Europe
An application for approval of a New Chemical Entity must contain data to allow assessment of the
safety and efficacy profile. These data include pharmacological and toxicological tests and the results of
clinical trials.
To avoid repeating such tests and trials, applications for generic pharmaceutical substances are not
required to include these data; instead they may rely on the data provided in relation to the NCE
application. However, EC directive 2001/83/EC prevents regulatory authorities from accepting
applications for approval of generics that rely on this data until a data exclusivity period has expired.
This period starts on the day of the first marketing authorisation in the European Community (including
Liechtenstein/Switzerland), and expires either six or ten years thereafter, depending on the country in
which the application is to be filed and the procedure used to file it.
Data exclusivity relates to the active ingredient per se, new periods of data exclusivity are not applied to
later approval of new dosage forms, routes of administration or indications.
1) To calculate the expiry of the data exclusivity period for centralised applications add 10 years to
the corresponding European First Marketing Authorisation Date shown in the Pipeline Selector Report.
2) To calculate the expiry of the data exclusivity period for national or mutual recognition
procedure applications, add the term from the following table to the corresponding European First
Marketing Authorisation
Exclusivity .......Period ...........Country
10 Years
Belgium (BE)
France (FR)
Germany (DE)
Italy (IT)
Luxembourg (LU)
Netherlands (NL)
Sweden (SE)
United Kingdom (UK)
6 years
Austria (AT)
Denmark (DK)
Finland (FI)
Greece (GR)
Iceland (IS)
Ireland (IRL)
Norway (NO)
Portugal (PT)
Spain (ES)
Other, acceding countries*
* Poland has a data exclusivity period of 3 years for approvals prior to 1/5/2004 and 6 years thereafter. Hungary,
Slovenia, Slovakia and Malta have requested transitional data exclusivity periods.
New Data Exclusivity Provisions for Europe
A New Data protection directive (2004/27/EC) has been introduced in Europe, which provides eight
years of protection from the first European Community marketing authorisation date, after which a
European regulatory body may accept an application for approval of a generic. However, the product
may not be marketed within ten years of the first European Community marketing authorisation date.
An additional one year period of data exclusivity is allowed in respect of new indications with ‘significant
clinical benefit’ over the existing indications. Approvals for other product line extensions, including
different pharmaceutical forms, are regarded as a bundle of approvals for which only one data
exclusivity period is available.
This new directive applies only to data relating to NCE products submitted for approval after 31 October
2005, hence do not affect any of the current Pipeline Patent Intelligence products.
Generic applications in the EU, patents and exclusivity
Types of protection on the originator molecule that have to be taken into account by generic drug manufacturers when filing applications for generic drugs in the EU include the following:
- patents at the time of marketing
- supplementary protection certificates at the time of marketing
- data/regulatory exclusivity at the time of regulatory submission
- market exclusivity at the time of marketing.
Patents
Patents in the EU last for 20 years. Pharmaceutical products are normally covered by a number of patents, sometimes by as many as 30 to 40 patents or more. For pharmaceuticals these patents can be extended with a maximum of five years via a supplementary protection certificate.
Evergreening
A patent on a new use or ‘indication’, formulation, salt or ester can block the registration or marketing of a generic medicine for treatments where the original patent has already expired. This strategy is known as ‘evergreening’ aims to prevent or delay competition from generic medicines by extending market protection through patents on minor changes to the original product.
Data exclusivity
Data exclusivity is a separate and additional provision to patent protection for the originator medicine. With data exclusivity, there can be neither disclosure of regulatory data to a competitor nor regulatory reliance upon the data. Data exclusivity is the period during which no generic drug application can take place.
Therefore, generic medicines can only be evaluated and approved by the medicines regulatory authorities after the data exclusivity period has expired.
In the EU there is now an 8+2(+1) formula for data and marketing exclusivity, which means that originator's data is protected for 8 years and they have marketing exclusivity for a maximum of 11 years from first marketing approval in the EU.
Exclusivity period: 8+2(+1)
The 8+2(+1) exclusivity period came into effect in the EU in late 2005.
8 years data exclusivity dating from the European Commission authorisation decision: before that, no generic applications may be filed.
+2 years marketing protection: no generic applications may be approved.
+1 year: new indication(s) if it constitutes a significant clinical benefit.
It applies for all products, regardless of whether they followed a centralised or national approval procedure.
It is not retroactive; it does not affect exclusivity periods for products for which applications were submitted before the effective date—late 2005.
Market exclusivity
With marketing exclusivity, the agency cannot allow a competing product to enter the market during the time period in which an innovator has a right of exclusivity.
Submission of generic applications is only possible after the expiry of the data exclusivity at year 8. However, there is no linkage of patent protection with registration. Approval of the application and launch of the generic can then take place at year 10 if there is no patent protection, and if there is no additional 1 year of marketing exclusivity due to a significant new indication registered for the reference product during the first 8 years of license, see figure below.
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE
Join me on google plus Googleplus
amcrasto@gmail.com
Thursday 29 January 2015
Marketing Authorisation in Europe
Authorisation Procedures for medicinal products
Procedures for evaluating medicinal products and granting marketing authorisation
The European system for the authorisation of medicinal products for human and animal use was introduced in January 1995 with the objective of ensuring that safe, effective and high quality medicines could quickly be made available to citizens across the European Union.
The European system offers several routes for the authorisation of medicinal products:
- The centralised procedure, which is compulsory for products derived from biotechnology, for orphan medicinal products and for medicinal products for human use which contain an active substance authorised in the Community after 20 May 2004 (date of entry into force of Regulation (EC) No 726/2004) and which are intended for the treatment of AIDS, cancer, neurodegenerative disorders or diabetes. The centralised procedure is also mandatory for veterinary medicinal products intended primarily for use as performance enhancers in order to promote growth or to increase yields from treated animals.
Applications for the centralised procedure are made directly to the European Medicines Agency (EMA) and lead to the granting of a European marketing authorisation by the Commission which is binding in all Member States. - The mutual recognition procedure, which is applicable to the majority of conventional medicinal products, is based on the principle of recognition of an already existing national marketing authorisation by one or more Member States.
- The decentralised procedure, which was introduced with the legislative review of 2004, is also applicable to the majority of conventional medicinal products. Through this procedure an application for the marketing authorisation of a medicinal product is submitted simultaneously in several Member States, one of them being chosen as the "Reference Member State". At the end of the procedure national marketing authorisations are granted in the reference and in the concerned Member States.
Purely national authorisations are still available for medicinal products to be marketed in one Member State only.
Special rules exist for the authorisation of medicinal products for paediatric use, orphan drugs, traditional herbal medicinal products, vaccines and clinical trials.
The EMA and the authorisation procedure
In 1993 the European Medicines Agency (EMA) was founded with the primary task of providing scientific advice of the highest possible quality to the Community Institutions on all matters relating to medicinal products for human and veterinary use. EMA's main task is to co-ordinate the scientific evaluation of the safety, efficacy and quality of medicinal products which undergo either procedure. All scientific questions arising in these procedures are dealt with by the EMA.
The agency has today established itself as a world-leading agency for the evaluation of medicinal products. It constitutes a major asset in making Europe an attractive location for new pharmaceuticals and allows for speedy and robust authorisation of new innovative medicines.
EMA's key tasks are to:
- provide Member States and Community institutions with the best possible scientific advice on questions about the quality, safety and efficacy of medicinal products for human and veterinary use;
- establish a pool of multinational scientific expertise (by mobilising existing national resources) in order to achieve a single evaluation via the centralised or mutual recognition marketing authorisation procedures;
- organise speedy, transparent and efficient procedures for the authorisation, surveillance and where appropriate, withdrawal of medicinal products in the EU;
- advise companies on the conduct of pharmaceutical research;
- reinforce the supervision of existing medicinal products (by co-ordinating national pharmacovigilance and inspection activities);
- create databases and electronic communication facilities as necessary to promote the rational use of medicines.
What is a Marketing Authorisation needed for?
In accordance with EU Directive 2001/83 and Regulation (EC) No. 726/2004 governing medicinal products, in order to legally place a medicinal product on the market in the European Economic Area (EEA), a Marketing Authorisation (MA) or ‘licence’ must first be approved. To obtain an MA, a Marketing Authorisation Application (MAA) is submitted to the appropriate Competent Authority(s) (CAs), for assessment and MA approval.
What does an MAA consist of?
An MAA is a comprehensive dossier of information and data describing all aspects (Administrative/Quality/Safety/Efficacy) of a medicinal product demonstrating that it is appropriate for use in patients. There is an internationally agreed standard for the overall content and format for this dossier which is referred to as the Common Technical Document (CTD).
The CTD format is applicable for all MAA regulatory submission routes and all product types, although some modifications may be required for certain application/product types. CTD format is also applicable to other submission types including variations and renewals.
At the top level, a CTD dossier is split into five modules:
Module 1: Administrative information and prescribing information (any additional region-specific information not specified in the CTD is also included in this module)
Module 2: CTD Summaries
Module 3: Quality
Module 4: Non-clinical Study Reports
Module 5: Clinical Study Reports
The overall organisation of the CTD is fixed and should not be changed. Documents and data should be assigned to the most appropriate sections in Modules 1 to 5. The only exceptions to this are the non-clinical and clinical summaries, within which individual formats/tables can be modified as required to best present the data for assessment.
What information and data is required in each CTD Module?
Guidance on the top level structure and content of the CTD dossier can be found in Eudralex Volume 2B of EC Notice to Applicants “Presentation and format of the dossier – Common Technical Dossier”.
CTD does not specify the detailed content in terms of what studies and data are required. There are European Community guidelines regarding the quality, safety and efficacy of medicinal products which must be considered and accounted for in preparation of the MAA dossier. In addition, for medicinal product quality, the general chapters and monographs of the European Pharmacopoeia or other national pharmacopoeias should also be accounted for as appropriate.
Not all data requirements are mandatory or required for each and every application or product type. If an element is considered to be not relevant or not applicable, the absence of such should be fully justified.
There may also be regional differences that will need to be taken into account in dossier preparation.
The following is a summarised description of the information to be included in each Module.
Module 1
This Module includes all of the administrative information relating to the application and the concerned medicinal product. Key items for inclusion are:
- Cover letter
- Application form and annexes (e.g. manufacturing licences, proof of payment of fees, letters of access)
- Product Information including the Summary of Product Characteristics (SmPC), labelling, patient leaflet, braille, results of readability testing
- Information about the experts
- Specific requirements for different types of applications
- Environmental Risk Assessment
- Pharmacovigilance system description
- Risk Management Plan
Module 2
This Module presents summary information for the quality, non-clinical and clinical Modules with so-called expert review of the information and data in the context of the MAA and regulatory framework.
Module 3
Module 3 is the Quality Module which presents all of the information regarding drug substance and drug product development, characterisation, manufacturing and testing to demonstrate that the medicinal product is of suitable quality.
Module 4
This is the Non-clinical Module which includes all of the non-clinical study reports and literature addressing the complete battery of non-clinical testing required for the medicinal product and application type.
Module 5
This is the Clinical Module which includes all of the clinical study reports and literature addressing the clinical requirements for the medicinal product and application type
Are there specified requirements for formatting such as filenames, margins, font type and size, hyperlinking, etc?
There is some general but limited guidance on formatting available, however the aim is obviously to facilitate dossier navigation and review. Margins need to be large enough to ensure that bindings do not obscure any data. The selected font and font size should be easily legible. Times New Roman and 12-point font are recommended for narrative text, although variations to this are accepted, for example for tabulated data where it might be necessary to reduce the font size slightly to ensure a table fits on the page.
In terms of pagination and segregation, documentation should be prepared in line withCHMP/ICH/2887/99 Revision 1 Organisation CTD recommendation.
National-only submissions must consider any Member State-specific requirements. For example in the UK, the MHRA recommends file naming conventions through Special Mail 5. European submissions would be in accordance with Non-eCTD electronic Submissions (NeeS) or fully electronic (eCTD) format.
Is there a requirement to re-format old dossiers?
There is no obligation to reformat approved dossiers. However it is possible to re-format Quality documentation into Module 3 format in order to facilitate ongoing life-cycle management of the licence, as variations, renewals, etc., will need to be submitted in CTD format. Re-formatting is not recommended for Modules 4 and 5 for Non-clinical and Clinical data.
When re-formatting into Module 3, all approved variations must be fully integrated into the re-formatted Module. In terms of the regulatory procedure to be followed to submit the re-formatted Module 3, this does not constitute a variation in itself. Therefore it is recommended that it is submitted as part of another regulatory procedure (e.g. variation or licence renewal), subject to agreement from the concerned CA.
If however, an old format licence is to be used as the basis for MAAs to be submitted via the Mutual Recognition Procedure (MRP), the current approved documentation will need to be reformatted intoCTD format. It is recommended that Applicants discuss any MRP plans with the proposed Reference Member State (RMS) in advance to determine the best means of doing this, as different Member States may prefer different approaches.
Are there different National administrative requirements (i.e. delivery addresses, number of copies, paper/electronic, etc)?
There are likely to be a variety of Member State-specific administrative requirements, some of which are documented in the available EU Guidances. However, there is still the possibility of additional requirements or nuances not readily available in the standard sources of information. In the absence of direct, recent experience of an MAA submission to the concerned Member States (MSs), it is highly recommended that contact be made with the relevant CA to establish any specific national requirements in advance of submission.
Requirements for final submission to the MSs again may vary and therefore needs to be checked in advance. In terms of final publication and dispatch of the MAA, this aspect of MAA preparation is often dismissed as minor, yet it requires the same level of care and attention as preparation of the dossier itself. Timelines frequently do not allow much time for this part of the project. Consequently any delays ahead of publication can mean publication and dispatch activities need to be completed at speed, potentially compromising the quality of the final submission.
What are the different types of MAA procedures?
Medicinal products can only be placed on the European Economic Area (EEA) market when a Marketing Authorisation has been issued by the competent authority of a Member State for its own territory (National Procedure) or when an authorisation has been granted in accordance with a community authorisation (Centralised Procedure (CP), Mutual Recognition Procedure (MRP) and Decentralised Procedure (DCP)).
National Procedure
The competent authorities of the Member States (MS) are responsible for granting MAs for products which are placed on their markets, except for medicinal products which are authorised under DCP,MRP and CP. In order to obtain a national MA, a single application is submitted to the competent authority of the MS.
Mutual Recognition Procedure
The Mutual Recognition Procedure aims at facilitating access to a single market by relying upon the principle of mutual recognition. A marketing authorisation or the assessment in one MS (the Reference Member State (RMS)) will in principle be recognised by the competent authorities of the other MSs (the Concerned Member States (CMS)), unless there are grounds for supposing that the authorisation of the medicinal product concerned may present a potential serious risk to public health.
After the first marketing authorisation is granted in one of the EU Member States, the MA Holder (MAH) may request one or more Member State(s) to recognise an authorisation granted by the RMSby submitting a MR application.
The Mutual Recognition Procedure is divided into the following steps:
Decentralised Procedure
The Decentralised Procedure is to be used in order to obtain marketing authorisations in several Member States where the medicinal product in question has not yet received a MA in any Member State at the time of application.
The Decentralised Procedure is divided into the following steps:
Centralised Procedure
Marketing Authorisation Applications for products that fall under the ‘mandatory’ or ‘optional’ scope of the Centralised Procedure, facilitate simultaneous approval in all EU Member States (including Iceland, Liechtenstein and Norway). The European Medicines Agency (EMA) will act as the Competent Authority for CP submissions.
Mandatory Scope – the Centralised Procedure is ‘mandatory’ for human medicines that are:
- Derived from biotechnology processes, such as genetic engineering
- Intended for the treatment of HIV/AIDs, cancer, diabetes, neurodegenerative disorders or autoimmune diseases and other immune dysfunctions
- Orphan medicinal products
Optional Scope – medicinal products can be accepted for consideration under the ‘optional’ route by the EMA, on the basis that the product contains either a new active substance or constitutes a significant therapeutic, scientific or technical innovation, or the authorisation is in the interests of patients within the community.
What are the differences between the MA procedures?
Centralised Procedure | Decentralised and Mutual Recognition Procedures | National Procedure |
---|---|---|
Less flexible procedure as only one tradename and one MA holder can be used | More flexible procedure as different tradenames and MA holders can be used | One tradename and oneMAH |
The EMA chooses the Rapporteur / Co-Rapporteur for the assessment | The applicant chooses theRMS with the most relevant expertise | The applicant chooses the country |
All or nothing procedure as the MAA is either accepted or rejected in all EU Member States | Can withdraw MAA from Member States that are causing difficulties, whilst continuing with applications in other states | Can withdraw the MAA at anytime |
One application and one assessment | Multiple assessments and complex administration procedures | One application and one assessment |
Well defined timescale throughout the procedure | Although the main assessment has a defined timescale, the national phase can be long and undefined | Undefined assessment timeline |
What types of application (legal basis) can be submitted?
The legal provisions covering all procedures for human medicinal products are contained in Directive 2001/83/EC (as last amended by Directive 2004/24/EC and by Directive 2004/27/EC). This Directive provides a number of different legal bases by which to apply for an MA.
- according to Article 8(3) of Directive 2001/83/EC, relating to full applications containing all appropriate quality, nonclinical and clinical data;
- according to Article 10 of Directive 2001/83/EC, relating to generic medicinal products and similar biological medicinal products;
- according to Article 10a of Directive 2001/83/EC, relating to applications relying on well-established medicinal use supported by bibliographic literature;
- according to Article 10b of Directive 2001/83/EC, relating to applications for new fixed combination products;
- according to Article 10c of Directive 2001/83/EC, relating to informed consent from a marketing authorisation holder for an authorised medicinal product.
Orphan Medicinal Product Designation in the EU
What is an Orphan Medicinal Product?
What is an Orphan Medicinal Product Designation?
An Orphan Medicinal Product Designation (OMPD) is granted by the European Commission, subsequent to an initial opinion being made by the European Medicines Agency’s (EMA’s) Committee for Orphan Medicinal Products (COMP) that the product qualifies as an orphan medicinal product.
What are the criteria for orphan designation?
To qualify for orphan designation (as set out in Article 3 of (EC) No 141/2000), a product must meet one of the following criteria:
- It is intended for the diagnosis, prevention or treatment of a life-threatening or chronically debilitating condition affecting no more than 5 in 10,000 people in the EU at the time of submission of the application.
- It is intended for the diagnosis, prevention or treatment of a life-threatening, seriously debilitating or serious and chronic condition and, without incentives, it is unlikely that the revenue after marketing of the medicinal product would cover the investment in its development.
Additionally, irrespective of the criteria met above, either of the following must apply:
- No satisfactory method of diagnosis, prevention or treatment of the condition concerned is authorised, or;
- If a satisfactory method exists, the medicine in question must be of significant benefit to individuals affected by the condition.
When can an orphan designation be applied for?
An application for orphan medicinal product designation can be submitted to the EMA at any stage of development; however it must be before an application for marketing authorisation is made.
A sponsor can request orphan designation for a product even if a separate sponsor already holds a designation covering the indication in question. In this instance, the first to gain marketing authorisation would benefit from market exclusivity.
Orphan designation can be requested for new chemical entities (NCEs) and new biological entities (NBEs), but also for existing approved products when the designation concerns a new, potentially orphan, indication for that product.
Any application for orphan designation must be supported by a robust scientific justification of the medical plausibility of the intended use.
How much does an application for OMPD cost?
The EMA do not charge a fee for the designation procedure.
What are the incentives of gaining Orphan Medicinal Product Designation?
Gaining orphan designation for a medicinal product entitles sponsors to the following benefits and/or incentives:
10 years’ market exclusivity: Orphan medicinal products benefit from market exclusivity in the EU for 10 years after receiving a marketing authorisation. An extension of 2 years’ market exclusivity may be granted if paediatric studies are included in the marketing authorisation.
During the 10 (+2) year period, directly competitive similar products for the same indication cannot normally be placed on the market.
Protocol assistance: Protocol assistance (PA) is similar to the EMA Scientific Advice procedure. However, additional benefits are included for PA, and sponsors can take advantage of the following:
- EMA Scientific Advice procedure, designed to help sponsors optimise development of their orphan medicinal product within the scope of the orphan indication
- Protocol assistance, also enables sponsors to ask questions related to:
- significant benefit over other satisfactory methods of treatment, diagnosis, or prevention of the indication in question
- development of medicinal products for rare conditions (e.g. whether two products are deemed ‘similar medicinal products’)
- study design related to clinical superiority over similar products in the same indication
This procedure can help applicants maximise the chances of their marketing authorisation application being successful.
Fee reductions: A special fund from the European Commission, agreed annually by the European Parliament, is used by the EMA to grant fee reductions for orphan medicinal products. Reduction of fees will be considered for all types of centralised activities, including applications for marketing authorisation, pre-authorisation inspections, variations and protocol assistance. For organisations holding both orphan designation and ‘SME status’ enhanced fee reductions are applicable.
Automatic access to centralised procedure: Medicinal products holding orphan designation are now required to apply through the centralised procedure in accordance with Article 3.1 of (EC) Regulation 726/2004.
EU-funded research: Sponsors developing orphan medicinal products may be eligible for grants from the EU and Member States’ programmes and initiatives supporting research and development, including the European Commission framework programme.
Points to consider in gaining orphan designation
There are several misconceptions about orphan designation for medicinal products, two of which are detailed below:
- In theory, regulators require the same level of proof of efficacy and safety for orphan drugs as for other drugs. There are examples of pivotal studies being accepted with reduced patient numbers; however, this is condition or drug-specific. Any development plans should be discussed with the EMA through Protocol Assistance procedures.
- Orphan designation will not automatically allow accelerated review for marketing authorisation, conditional approval or approval under exceptional circumstances. Applicants would still need to apply for such procedures.
How to obtain Orphan Medicinal Product Designation?
In order to benefit from the incentives provided for orphan products a sponsor must obtain orphan designation for the product by compiling an OMPD application and submitting this to the EMA. Applications are reviewed by the Committee for Orphan Medicinal Products (COMP) and an opinion on whether the medicinal product meets the criteria for orphan designation will be issued. The opinion is forwarded to the European Commission who makes the final decision on whether orphan designation will be granted.
What is the procedure for gaining orphan designation?
Notification of intent to submit: A sponsor should notify the EMA of their intent to submit an OMPDapplication in the form of a Letter of Intent at least two months in advance of the planned submission date; the Letter of Intent (and ultimately the final application package) should be submitted in accordance with EMA published timelines.
Pre-submission meeting: The OMPD application procedure also offers sponsors the opportunity of a pre-submission meeting with the EMA. It is highly recommended to use this free service as feedback from the EMA on draft OMPD applications can significantly improve the success of OMPDapplications. If a pre-submission meeting is required, this should be stated in the Letter of Intent.
Submission of OMPD application: Following the pre-submission meeting the application is refined and a final package submitted to the EMA. The EMA will validate the submission and, if necessary, the sponsor is given three months to address any validation issues. The date of successful validation is taken as Day 1 of the procedure.
OMPD evaluation: The COMP will evaluate applications within 90 days of the start of procedure (i.e. post-validation) and will adopt either a positive or negative opinion at monthly held COMPmeetings.
At Day 60 the COMP will either:
- Adopt a positive opinion, or
- If a negative opinion is being considered, COMP will forward a list of questions to the sponsor. The sponsor will be advised as to whether written responses or an oral explanation will be required in time for the next COMP meeting
At Day 90 the COMP will either:
- Adopt a positive opinion to be forwarded to the European Commission, or
- In the absence of sufficient justification in the written response or oral explanation, adopt a negative opinion
On receipt of a negative opinion a sponsor can appeal. However, the intent to do so should be notified without delay. Negative opinions will be published on the EMA website, but the EMA offer the possibility of withdrawing applications likely to receive a negative opinion before adoption takes place at the COMP meeting.
Commission Decision: A positive opinion is forwarded to the European Commission and a Decision granted, typically within 30 days; in practice receipt of a Commission Decision can take 2-3 months following adoption of the COMP opinion.
Following receipt of the European Commission Decision a Public Summary of Opinion will be published on the EMA website and the orphan product will be included in the Community Register of Orphan Medicinal Products.
Timeline: The minimum timeframe from Letter of Intent to European Commission Decision is five months, although additional time may be required for validation responses, an oral/written explanation and European Commission Decision.
What information is required for OMPD applications?
In order to submit an application for orphan designation a sponsor (who can be an individual or an organisation) must have a fixed address in the European Union and is required to include the following documentation:
Key documentation
- Cover Letter – identifying sponsor, medicinal product, proposed orphan indication
- Application form – available on EMA website
The common FDA/EMA application form can be used. However, it is important to note that there are key differences in the required supportive documentation for application in the two regions. Additionally, applications for designation will be reviewed separately by the respective Authorities. - Core dossier – Parts A-F in accordance with guidance document ENTR/6283/00
Part A: Description of the Condition
Part B: Prevalence of the Condition
Part C: Potential for return on Investment
Part D: Existence of other methods of diagnosis, prevention or treatment
Part E: Description of stage of development
Part F: Bibliography - Proof of Establishment in the EU
- Letter of Authorisation (if applicable)
- Translations of product name and proposed orphan indication into all official EU languages, plus Icelandic and Norwegian
Key components of application
The key components of the orphan designation application include the following:
Indication: the orphan indication must be a clearly defined, medically plausible, condition or disease. Sub-setting (or salami slicing) can be considered but only if it can be shown that the sub-set is medically plausible, and it can be demonstrated that the medicinal product would have no beneficial effects in the wider population. The orphan indication can be broader than the therapeutic indication proposed for a Marketing Authorisation Application.
Part A – Medical plausibility: this section must include a summary of the medicinal product and the proposed mechanism of action. The condition must be well defined and supported by published literature. Preliminary clinical and/or nonclinical data is required to support the notion that the product may be beneficial in the proposed orphan indication.
Part A – Justification of the life-threatening or debilitating nature of the disease or condition: the sponsor is expected to prove that the proposed orphan indication is life-threatening or debilitating in nature through the use of published literature.
Part B – Prevalence of the condition: in order to meet the prevalence designation criteria, the sponsor is required to demonstrate that less than 5 in 10,000 persons in the EU are affected by the disease or condition at the time of designation application. For products intended for diagnosis or prevention sponsors are required to show that less than 5 in 10,000 persons in the EU will receive or use the diagnostic. Prevalence data should be considered for all EU countries, as well as Iceland, Liechtenstein, and Norway.
Part D – Justification of significant benefit: in the application the sponsor must identify if there are any existing methods of diagnosis, treatment or prevention for the orphan indication. These could include Competent Authority approved methods or those commonly accepted by clinicians within the EU. If such methods exist, sponsors are required to demonstrate how the medicinal product under consideration could offer significant benefit in terms of improved safety, efficacy and/or improved pharmacokinetic properties.
What is required to maintain an OMPD?
In order to maintain orphan designation, sponsors are required to submit an Annual Report to theEMA within two months following the anniversary of the grant of the designation. The report should include details of:
- the progress of development since the initial application
- updated details of the current regulatory status
- any incentives received for development of the product
\When submitting a marketing authorisation application for a product with OMPD, applicants are expected to demonstrate that the OMPD criteria are still met, particularly with regards to significant benefit (if applicable). If these criteria are no longer met, the designation will be removed.
DRUG APPROVALS BY DR ANTHONY MELVIN CRASTO .....FOR BLOG HOME CLICK HERE
Join me on google plus Googleplus
amcrasto@gmail.com
Subscribe to:
Posts (Atom)