AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

Selection and justification of starting materials: new Questions and Answers to ICH Q11 published

 regulatory  Comments Off on Selection and justification of starting materials: new Questions and Answers to ICH Q11 published
Dec 082016
 

 

The ICH Q11 Guideline describing approaches to developing and understanding the manufacturing process of drug substances was finalised in May 2012. Since then the pharmaceutical industry and the drug substance manufacturers had time to get familiar with the principles outlined in this guideline. However, experience has shown that there is some need for clarification. Thus the Q11 Implementation Working Group recently issued a Questions and Answers Document.

http://www.gmp-compliance.org/enews_05688_Selection-and-justification-of-starting-materials-new-Questions-and-Answers-to-ICH-Q11-published_15619,15868,S-WKS_n.html

str0

The ICH Q11 Guideline describes approaches to developing and understanding the manufacturing process of drug substances. It was finalised in May 2012 and since then the pharmaceutical industry and the drug substance manufacturers had time to get familiar with the principles outlined in this guideline. However, experiences during implementation of these principles within this 4 years period have shown that there is need for clarification in particular with regard to the selection and justification of starting materials.

On 30 November 2016 the ICH published a Questions and Answers document “Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities)” which was developed by the Q11 Implementation Working Group. This document aims at addressing the most important ambiguities with respect to starting materials and at promoting a harmonised approach for their selection and justification as well as the information that should be provided in marketing authorisation applications and/or Drug Master Files.

In the following some examples of questions and answers from this document:

Question:
ICH Q11 states that “A starting material is incorporated as a significant structural fragment into the structure of the drug substance.” Why then are intermediates used late in the synthesis, which clearly contain significant structural fragments, often not acceptable as starting materials?

Answer:
The selection principle about “significant structural fragment” has frequently been misinterpreted as meaning that the proposed starting material should be structurally similar to the drug substance. However, as stated in ICH Q11, the principle is intended to help distinguish between reagents, catalysts, solvents, or other raw materials (which do not contribute a “significant structural fragment” to the molecular structure of the drug substance) from materials that do. … The presence of a “significant structural fragment” should not be the sole basis for of starting material selection. Starting materials justified solely on the basis that they are a “significant structural fragment” probably will not be accepted as starting materials by regulatory authorities, as the other principles for the appropriate selection of a proposed starting material also require consideration.

Question:
Do the ICH Q11 general principles for selection of starting materials apply to processes where multiple chemical transformations are run without isolation of intermediates?

Answer:
Yes. The ICH Q11 general principles apply to processes where multiple chemical transformations are run without isolation of intermediates. In the absence of such isolations (e.g., crystallization, precipitations), other unit operations (e.g., extraction, distillation, the use of scavenging agents) should be in place to adequately control impurities and be described in the application. The drug substance synthetic process should include appropriate unit operations that purge impurities.
The ICH Q11 general principles also apply for sequential chemical transformations run continuously. Non isolated intermediates are generally not considered appropriate starting materials.

Question:
Is a “starting material” as described in ICH Q11 the same as an “API starting material” as described in ICH Q7?

Answer:
Yes. ICH Q11 states that the Good Manufacturing Practice (GMP) provisions described in ICH Q7 apply to each branch of the drug substance manufacturing process beginning with the first use of a “starting material”. ICH Q7 states that appropriate GMP (as defined in that guidance) should be applied to the manufacturing steps immediately after “API starting materials” are entered into the process … . Because ICH Q11 sets the applicability of ICH Q7 as beginning with the “starting material”, and ICH Q7 sets the applicability of ICH Q7 as beginning with the “API starting material”, these two terms are intended to refer to the same material.
ICH Q7 states that an “API Starting Material” is a raw material, intermediate, or an API that is used in the production of an API. ICH Q7 provides guidance regarding good manufacturing practices for the drug substance; however, it does not provide specific guidance on the selection and justification of starting materials. When a chemical, including one that is also a drug substance, is proposed to be a starting material, all ICH Q11 general principles still need to be considered.

With the recent publication of this draft Q&A Document with the complete title “Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities) Questions and Answers (regarding the selection and justification of starting materials)” on the ICH website it reached Step 2b of the ICH Process and now enters the consultation period.  Comments may be provided by e-mailing to the ICH Secretariat at admin@ich.org.

Image result for ICH Q11

Image result for ICH Q11

extra info…………
A PRESENTATION

 

 

 

Ever since the FDA issued its landmark guidance Pharmaceutical GMPs-A Risk Based Approach in 2004, the industry has been struggling with how to demonstrate process understanding as a basis for quality. Bolstered by guidance from ICH, specifically Q6-Q10, the pieces have long been in place to build a solution that is philosophically consistent with these best practice principles. Even so, the evolution to process understanding as a basis for quality has been slow. Pressure to accelerate this transformation spiked in 2011 when the FDA issued its new guidance on process validation that basically mandated the core components of ICH Q6-10 as part of Stages 1 and 2. To be fair, enforcement has been uneven and that fact has further impeded adoption, with the compliance inspectors themselves struggling to acquire the necessary skills to fully evaluate statistical arguments of process control and predictability.

One area debated since 2008 is the application of GMPs and demonstration of control for drug substances. Drug substance suppliers and drug product manufacturers have used the tenets of ICH Q7A as the foundation for deciding where GMPs can be reasonably implemented, to establish the final intermediate (FI) and the regulatory starting material (RSM). However, the ability to support the quality of the drug substance has a profound impact on the ability to defend the drug product quality. In the last few years it has become apparent that it was not reasonable to apply the same requirements for drug products to drug substances because the processes can be markedly different. In response to this need, the ICH issued a new guidance; Q11: Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities). The key ICH documents that impact Q11 are shown in Figure 1.


Figure 1. Guidances Impacting ICH Q11.

The FDA formally adopted ICHQ11 in November 2012 and its purpose is two-fold. First, it offers guidance on the information to provide in Module 3 of the Common Technical Document (CTD) Sections 3.2.S.2.2 – 3.2.S.2.6 (ICH M4Q). Second, and perhaps most importantly, it attempts to clarify the concepts defined in the ICH guidelines on Pharmaceutical Development (Q8), Quality Risk Management (Q9), and Pharmaceutical Quality System (Q10) as they pertain to the development and manufacture of drug substances.

What makes ICH Q11 so important is its emphasis on control strategy. This concept was introduced in ICH Q10 as “a planned set of controls, derived from current product and process understanding that assures process performance and product quality.”

Within the drug product world, the control strategy concept has been elusive as industry grapples with moving from a sample-and-test concept of quality to one of process understanding and behavior. This concept is even more removed for drug substance manufacturers and, in some cases, is more difficult to implement. But Q11 is much more than a mere framework for control strategy. The guidance is structured very similarly to the concepts discussed in the new 2011 Process Validation guidance. Looking closely, Q11 addresses:
• Product Design/Risk Assessment/CQA Determination
• Defining the Design Space and establishing a control strategy
• Process validation and analysis
• Information required for Sections 3.2.S.2.2 – 3.2.S.2.6 of the eCTD
• Lifecycle management

Product design/Risk assessment/CQA determination

Within the context of process development, the guidance defines similar considerations to those defined in the Stage 1 activity of Process Validation. Understanding the quality linkage between the drug substance’s physical, chemical, and microbiological characteristics, and the final drug products’ Quality Target Product Profile (QTPP), is the primary objective of the product and process design phase. The product’s QTPP is comprised of the final product Critical to Quality Attributes (CQAs). Identifying the raw material characteristics of the drug substance that can impact the drug product is a critical first step in developing a defensible control strategy. Employing risk analysis tools at the outset can help focus the process development activities upon the unit operations that have the potential to impact the final product’s CQAs. In the case of biological drug substances, any knowledge regarding mechanism of action and biological characterization, such as studies that evaluate structure-function relationships, can contribute to the assessment of risk for some product attributes.

Drug substance CQAs typically include those properties or characteristics that affect identity, purity, biological activity, and stability of the final drug product. In the case of biotechnological/biological products, most of the CQAs of the drug product are associated with the drug substance and thus are a direct result of the design of the drug substance or its manufacturing process. When considering CQAs for the drug substance, it is important to not overlook the impact of impurities because of their potential impact on drug product safety. For chemical entities, these include organic impurities (including potentially mutagenic impurities), inorganic impurities such as metal residues, and residual solvents.

For biotechnological/biological products, impurities may be process-related or product-related (see ICH Q6B). Process-related impurities include: cell substrate-derived impurities (e.g., Host Cell Proteins [HCP] and DNA); cell culture-derived impurities (e.g., media components); and downstream-derived impurities (e.g., column leachable). Determining CQAs for biotechnology/biological products should also include consideration of contaminants, as defined in Q6B, including all adventitiously introduced materials not intended to be part of the manufacturing process (e.g., viral, bacterial, or mycoplasma contamination).

Defining the design space and establishing a control strategy

ICH Q8 describes a tiered approach to establishing final processing conditions that consists of moving from the knowledge space to the process design space and finally the control space. ICH Q8 and Q11 define the Design Space as “the multidimensional combination and interaction of input variables (e.g., material attributes) and process parameters that have been demonstrated to provide assurance of quality.” In the drug product world the terminology typically applied to the design space is the Proven Acceptable Range (PAR) that used to equate to the validated range.

Here is why this is important: the ability to accurately assess the significance and effect of the variability of material attributes and process parameters on drug substance CQAs, and hence the limits of a design space, depends on the extent of process and product understanding. The challenge with drug substance processes is where to apply the characterization. ICH Q7A recognizes that upstream of the RSM does not require GMP control. The design space can be developed based on a combination of prior knowledge, first principles, and/or empirical understanding of the process. A design space might be determined per unit operation (e.g., reaction, crystallization, distillation, purification), or a combination of selected unit operations should generally be selected based on their impact on CQAs.

In developing a control strategy, both upstream and downstream factors should be considered. Starting material characteristics, in-process testing, and critical process parameters variation control are the key elements in a defensible control strategy. For in-process and release testing criteria the resolution of the measurement tool should be considered before making any conclusions.

Process validation

ICH Q11’s description of process validation mimics the same description in ICH Q7A but offers up an alternative for continuous verification that mirrors the concepts in ICH Q8 and the new process validation guidance. As mentioned, the enforcement of the new guidance by the FDA has been uneven, but positioning the process validation to satisfy the new guidance requires the drug substance manufacturer to formally implement characterization and validation standards, just as a drug product manufacturer would be required to do.

Life-cycle management

The quality system elements and management responsibilities described in ICH Q10 are intended to encourage the use of science-based and risk-based approaches at each lifecycle stage, thereby promoting continual improvement across the entire product lifecycle. There should be a systematic approach to managing knowledge related to both drug substance and its manufacturing process throughout the lifecycle. This knowledge management should include but not be limited to process development activities, technology transfer activities to internal sites and contract manufacturers, process validation studies over the lifecycle of the drug substance, and change management activities.

Conclusion

The new ICH Q11 guidance represents the most recent example of the FDA’s commitment to the principles of QbD to define an integrated framework for implementing the principles of ICH Q6-Q10. Although the guidance does not mandate adopting ICH Q8, the considerations required to create a defensible control strategy require a much higher level of process understanding than the conventional approach of sample and test, once the foundation of product development. Defining the requirements is another example of where the FDA is going in terms of expectations for drug substance and drug product understanding. If effectively enforced, this can be a significant step forward, pushing the industry toward a QbD philosophy for process and product development.

/////////Selection, justification, starting materials,  ICH Q11 , ich, qbd

Share

Statistical DoE Approach to the Removal of Palladium from Active Pharmaceutical Ingredients (APIs) by Functionalized Silica Adsorbents

 QbD, regulatory  Comments Off on Statistical DoE Approach to the Removal of Palladium from Active Pharmaceutical Ingredients (APIs) by Functionalized Silica Adsorbents
Nov 032016
 

Abstract Image

The influence of four parameters (temperature, scavenging time, amount of scavenger, and concentration of palladium in the solution) on the efficiency of Pd removal from a cross-coupling reaction, using a commercially available Pd scavenger, SPM32, was studied. The DoE-based method employed yielded more information than is readily attainable from standard adsorption isotherms and kinetics experiments. The optimal regime of scavenging was identified; intuitive and nonintuitive effects of temperature, scavenging time, and scavenger amounts were highlighted; and a mathematical model quantifying predicted Pd removal from the synthetic intermediate was built.

link http://pubs.acs.org/doi/abs/10.1021/op5000336

Statistical DoE Approach to the Removal of Palladium from Active Pharmaceutical Ingredients (APIs) by Functionalized Silica Adsorbents

PhosphonicS Ltd., 44c Western Avenue, Milton Park, Abingdon, OX14 4RU, United Kingdom
Org. Process Res. Dev., 2014, 18 (5), pp 626–635
DOI: 10.1021/op5000336
Publication Date (Web): April 14, 2014
Copyright © 2014 American Chemical Society
str0

Preparation of tert-butyl 2-[(4-cyanophenyl)amino]propanoate (3).

4-Bromobenzonitrile (18.20 g, 100 mmol), L-alanine tert-butyl ester hydrochloride (21.73 g, 120 mmol), ±BINAP (1.25 g, 2 mmol) and cesium carbonate (48.87 g, 150 mmol) were added to a 3-neck round bottom flask containing a magnetic stirrer. Toluene (167 mL) was added and a reflux condenser, thermometer and a rubber septum were attached. Argon gas was bubbled through as the heterogeneous mixture was warmed to reflux temperature with slow agitation from the magnetic stirrer. Palladium acetate (0.45 g, 2 mmol) was added quickly through one of the side-arm joints and de-gassing was continued for 5 min. The reaction mixture was kept under argon at reflux and the disappearance of 4-bromobenzonitrile was monitored by GC-MS. After 16-24 h, the reaction mixture was filtered through a sinter funnel, washed with toluene and then filtered through a nylon membrane (Sigma Aldrich catalogue no. Z290793, 0.45 µm pore size) and washed with toluene. This crude reaction mixture was used in the DoE matrix without further purification.

Experimental results

NMR δC (62.9 MHz, CDCl3): 172.7, 150.0, 133.7, 120.4, 112.7, 99.3, 82.3, 51.7, 28.0, 18.5 ppm.

NMR δH (250 MHz, CDCl3): 7.39 (2H, d, J = 8.8 Hz, Ph), 6.52 (2H, d, J = 8.8 Hz, Ph), 4.85 (1H, d, J = 7.4 Hz, NH), 4.01 (1H, quintet, J = 7.4 Hz, CH(Me), 1.43 (9H, s, tBu), 1.42 (3H, d, J = 7.4 Hz, Me).

GC/MS: GC method used: hold at 50 °C for 4 min; increase temperature from 50 to 280 °C at 30 °C / min; hold at 280 °C for 5 min. Peaks were recorded and identified as follows: 4-Bromobenzonitrile: 10.05 min. Molecular peak observed at m/z = 181 for the 79Br isotope, m/z = 183 for the 81Br isotope L-alanine tert-butyl ester hydrochloride: not observed. Retention time less than 5 min, peak lost within the solvent front. Product: 13.64 min. Molecular peak observed at m/z = 246, main fragment at m/z = 146 (M – CO2 t Bu) Side product (not identified or quantified, minor peak): 14.56 min.

str0 str2

Jan Recho

Jan Recho

Jan Recho

Systems developer at Clearsy

Corresponding Author *E-mail: jan.recho@phosphonics.com.

Experience

Systems Developer

Clearsy

– Present (1 year 6 months)

Scientist

PhosphonicS

(3 years 6 months)

• Design and synthesis of silica supported transition metals scavengers and catalysts (Pd, Rh, Ru) for the pharmaceutical industry.
• Management of fine chemistry customer projects
• Process optimisation (Quality by Design – QbD, Design of Experiments – DoE).
• Business development activities for the French market

Junior researcher

Institut des Matériaux de Nantes

(4 years 8 months)Nantes Area, France

Synthesis and characterisation of a cellulose derived, organosilane-based, bio-material for cartilage growth.
Work in imidazolium and pyridinium ionic liquids.

Share

QbD Sitagliptin

 QbD  Comments Off on QbD Sitagliptin
Oct 302016
 

 

str0

Image result for sitagliptin qbd

 

Application of On-Line NIR for Process Control during the Manufacture of Sitagliptin

Global Science, Technology and Commercialization, Merck Sharp & Dohme Corporation P.O. Box 2000, Rahway, New Jersey 07065, United States
Org. Process Res. Dev., 2016, 20 (3), pp 653–660
DOI: 10.1021/acs.oprd.5b00409
Publication Date (Web): February 12, 2016
Copyright © 2016 American Chemical Society

Abstract

Abstract Image

The transamination-chemistry-based process for sitagliptin is a through-process, which challenges the crystallization of the active pharmaceutical ingredient (API) in a batch stream composed of multiple components. Risk-assessment-based design of experiment (DoE) studies of particle size distribution (PSD) and crystallization showed that the final API PSD strongly depends on the seeding-point temperature, which in turn relies on the solution composition. To determine the solution composition, near-infrared (NIR) methods had been developed with partial least squares (PLS) regression on spectra of simulated process samples whose compositions were made by spiking each pure component, either sitagliptin free base (FB), water, isopropyl alcohol (IPA), dimethyl sulfoxide (DMSO), or isopropyl acetate (IPAc), into the process stream according to a DoE. An additional update to the PLS models was made by incorporating the matrix difference between simulated samples in lab and factory batches. Overall, at temperatures of 20–35 °C, the NIR models provided a standard error of prediction (SEP) of less than 0.23 wt % for FB in 10.56–32.91 wt %, 0.22 wt % for DMSO in 3.77–19.18 wt %, 0.32 wt % for IPAc in 0.00–5.70 wt %, and 0.23 wt % for water in 11.20–28.58 wt %. After passing the performance qualification, these on-line NIR methods were successfully established and applied for the on-line analysis of production batches for compositions prior to the seeding point of sitagliptin crystallization.

http://pubs.acs.org/doi/abs/10.1021/acs.oprd.5b00409?journalCode=oprdfk

 

Next…………..

A biocatalytic manufaturing route for januvia – Society of Chemical …

www.soci.org/~/media/Files/…/2011/…/Jake_Janey_Presentation.ashx

Nov 2, 2011 – 9 Steps, 52% overall yield, >100Kg of sitagliptin prepared ….. FDA filings requires “Quality by Design”: A way to allow process changes within.

A PRESENTATION

 

 

A PRESENTATION

Example of QbD Application in Japan

https://www.pmda.go.jp/files/000213677.pdf

Aug 11, 2016 – QbD assessment experience in Japan … Number of Approved Products with QbD … Active Ingredient : Sitagliptin Phosphate Hydrate.

WILL BE UPDATED WITH MORE, WATCH OUt

/////////

Name Explanation
Active Pharmaceutical Ingredient (API) An active pharmaceutical ingredient (API) is a substance used in a finished pharmaceutical product, intended to furnish pharmacological activity or to otherwise have direct effect in the diagnosis, cure, mitigation, treatment or prevention of disease, or to have direct effect in restoring, correcting or modifying physiological functions in human beings.

 

Annual Product Reviews (APR) The Annual Product Reviews (APR) include all data necessary for evaluation of the quality standards of each drug product to determine the need for changes in drug product specifications or manufacturing or control procedures. The APR is required by the U.S. Code of Federal Regulations.
ANVISA The Brazilian Health Surveillance Agency (in Portuguese, Agência Nacional de Vigilância Sanitária) is a governmental regulatory body in Brazil. Similar to the FDA in the United States, it oversees the approval of drugs and other health products and regulates cosmetics, food products, and other health-related industries.
Biologic License Application (BLA) The Biologics License Application (BLA) is a request for permission to introduce, or deliver for introduction, a biologic product into commerce in the U.S.
CFDA The China Food and Drug Administration is similar to the FDA in the United States and is responsible for regulating food and drug safety.
cGMP Current Good Manufacturing Practices govern the design, monitoring, and control of manufacturing facilities and processes and are enforced by the US FDA. Compliance with these regulations helps safeguard a drug’s identity, strength, quality, and purity.
COFEPRIS The Federal Commission for Protection against Sanitary Risks (in Spanish, Comisión Federal para la Protección contra Riesgos Sanitarios) is a government agency in Mexico. It regulates food safety, drugs, medical devices, organ transplants, and environmental protection.
Common Technical Document (CTD) The Common Technical Document (CTD) is the mandatory common format for new drug applications in the EU and Japan, and the U.S. The CTD assembles all the Quality, Safety and Efficacy information necessary for a drug application.
European Medicines Agency (EMA) The European Medicines Agency (EMA) is a decentralised agency of the European Union (EU), located in London. It began operating in 1995. The Agency is responsible for the scientific evaluation, supervision and safety monitoring of medicines developed by pharmaceutical companies for use in the EU.
Food and Drug Administration (FDA) The Food and Drug Administration (FDA) is an agency within the U.S. Department of Health and Human Services. The FDA is responsible for the approval of new pharmaceutical products for sale in the U.S. and performs audits at the companies participating in the manufacture of pharmaceuticals to ensure that they comply with regulations.
Human growth hormone A growth hormone (GH or HGH) is a peptide hormone produced by the pituitary gland that stimulates growth in children and adolescents. It is involved in several body processes, including cell reproduction and regeneration, regulation of body fluids, and metabolism. It can be produced by the body (ie, somatotropin) or genetically engineered (ie, somatropin).
In-Process Control (IPC) In-Process Controls (IPC) are checks performed during production in order to monitor and if necessary to adjust the process to ensure that the product conforms its specification.
Interferons (INFs) Interferons are proteins produced by the body as part of the immune response. They are classified as cytokines, proteins that signal other cells to trigger action. For example, a cell infected by a virus will release interferons to stimulate the defenses of nearby cells.
Interleukins Interleukins are proteins produced by cells as an inflammatory response. Most interleukins help leukocytes communicate with and direct the division and differentiation of other cells.
Investigational Medicinal Product Dossier (IMPD) The Investigational Medicinal Product Dossier (IMPD) is the basis for approval of clinical trials by the competent authorities in the EU. The IMPD includes summaries of information related to the quality, manufacture and control of the Investigational Medicinal Product, data from non-clinical studies and from its clinical use.
Investigational New Drug (IND) An Investigational New Drug application is provided to the FDA to obtain permission to test a new drug in humans in Phase I – III clinical studies. The IND is reviewed by the FDA to ensure that study participants will not be placed at unreasonable risk.
Marketing Authorization Application (MAA) The Marketing Authorization Application (MAA) is a common document used as the basis for a marketing application across all European markets, plus Australia, New Zealand, South Africa, and Israel. This application is based on a full review of all quality, safety, and efficacy data, including clinical study reports.
Master batch records These general manufacturing instructions, which are required by cGMP, are the bases for a precise, detailed description of a pharmaceutical manufacturing process. They ensure that all proper ingredients are included, each process step is completed, and the process is controlled.
Medicines and Healthcare Products Regulatory Agency (MHRA) The Medicines and Healthcare products Regulatory Agency (MHRA) regulates medicines, medical devices and blood components for transfusion in the UK. MHRA is an executive agency, sponsored by the Department of Health.
MFDS The Ministry of Food and Drug Safety (formerly the Korean Food & Drug Administration) is a government agency that oversees the safety and efficacy of drugs and medical devices in South Korea.
Monoclonal antibodies Monoclonal antibodies are antibodies made in a laboratory from identical immune cells that are clones of a single cell. They are distinct from polyclonal antibodies, which are made from different immune cells.
NDA A New Drug Application (NDA) is the vehicle submitted to the FDA by drug companies in order to gain approval to market a new product. Safety and efficacy data, proposed package labeling, and the drug’s manufacturing methods are typically included in an NDA.
New Drug Application (NDA) The New Drug Application (NDA) is the vehicle through which drug sponsors formally propose that the FDA approve a new chemical pharmaceutical for sale and marketing in the U.S.

 

Oligonucleotides These short nucleic acid chains (made up of DNA or RNA molecules) are used in genetic testing, research, and forensics.
Parenteral Parenteral medicine is taken or administered in a manner other than through the digestive tract. Intravenous and intramuscular injections are two examples.
Peptide hormones Peptide hormones are proteins secreted by organs such as the pituitary gland, thyroid, and adrenal glands. Examples include follicle-stimulating hormone (FSH) and luteinizing hormone. Similar to other proteins, peptide hormones are synthesized in cells from amino acids.
PMDA The Pharmaceuticals Medical Devices Agency is an independent administrative agency that works with the Ministry of Health, Labour and Welfare to oversee the safety and quality of drugs and medical devices in Japan.
Process Analytical Technology (PAT) These analytical tools help monitor and control the manufacturing process, including accommodating for variability in material and equipment, in order to ensure consistent quality.
Product Quality Reviews (PQR) The Product Quality Reviews (PQR) of all authorized medicinal products, is conducted with the objective of verifying the consistency of the existing process, the appropriateness of current specifications for both starting materials and finished product, to highlight any trends and to identify product and process improvements. The PQR is required by the EU GMP Guideline.
Quality by Design (QbD) This concept involves a holistic, proactive, science- and risk-based approach to the development and manufacturing of drugs. At the heart of QbD is the idea that quality is achieved through in-depth understanding of the product and the process by which it is developed and manufactured.
Restricted Access Barrier System (RABS) This advanced aseptic processing system provides an enclosed environment that reduces the risk of contamination to the product, containers, closures, and product contact surfaces. As a result, it can be used in many applications in a fill-finish area.
Scale-up Scale-up involves taking a small-scale manufacturing system developed in the laboratory to a commercially viable, robust production process.
Six Sigma Six Sigma is a set of quality management methods, techniques, and tools used to improve manufacturing, transactional, and other business processes. The goal is to enhance quality (as well as employee morale and profits) by identifying and eliminating the cause of errors and process variations.
Target Product Profile (TPP) This key strategic document summarizes the features of an intended drug product. Characteristics may include the dosage form, route of administration, dosage strength, pharmacokinetics, and drug product quality criteria.
TFDA The Taiwan Food & Drug Administration is a governmental body devoted to enhancing food safety and drug quality in that country.
Share

QbD in Pharma Development World Congress 2017, SelectBio, 20-21 April, 2017, Radisson Hyderabad HITEC City, India

 CONFERENCE, QbD  Comments Off on QbD in Pharma Development World Congress 2017, SelectBio, 20-21 April, 2017, Radisson Hyderabad HITEC City, India
Oct 212016
 

str0

 

http://selectbiosciences.com/images/DEL_QBD_20Oct.html

QbD in Pharma Development World Congress 2017 Registration

3 for 2 Offer

SELECTBIO are offering 3 for the price of 2 on all delegate passes. To take advantage of this offer contact us by email, phone or click the Contact Us button below. Looking for more than 3 passes? Contact us for more information on our special rates for large groups.

Radisson Hyderabad HITEC City

Radisson Hyderabad HITEC City

Flights

To find low cost flights, try the following websites:

Flightchecker
Travelsupermarket
Kayak

The refined Radisson Hyderabad Hitec City features the prompt services, such as a concierge, and comfortable, air-conditioned rooms you need for a satisfying visit. You can stay fit with laps in the swimming pool and fitness centre, or relax in your suite with 24-hour room service and free Wi-Fi access. The aminities include Satellite TV, Work desk, Wi-Fi access, Tea and coffeemaker, Bottles of mineral water, Large bathrooms with separate rain showers, Large wardrobe, Mini barWrap up a day of meetings with authentic Indian cuisine at Cascade or exotic Asian specialties at The Oriental Blossom. Treat your colleagues to drinks at Zyng lounge bar, or if the weather is nice, gather outside at Poolside Grill for a barbecued meal.
A business centre is also available to help you complete work while staying at this Hitec City hotel in the heart of Gachibowli, a new-age IT suburb of Hyderabad, near the Hyderabad International Convention Centre.
SELECTBIO has negotiated special rates (see below) to include buffet breakfast, and Wi-Fi. Standard Room Single/Double – INR 4500/5000+Tax
To make your reservation at these discounted rates please contact Sakshi Modgil at s.modgil@selectbio.com. We recommend early booking to avoid disappointment.

Sakshi Modgil's Profile PhotoSAKSHI MODGIL

Visa Requirements
International visitors travelling from outside India will require a Business visa.
PLEASE NOTE, THIS EVENT IS NOT ASSOCIATED WITH THE INDIAN GOVERNMENT, THEREFORE CONFERENCE VISA IS NOT APPLICABLE
International visitors will require an invitation letter to obtain their Business visa. We will only provide invitation letters to customers that are fully registered for the event. In the event of an unsuccessful visa application we will refund the full delegate fee paid.
http://selectbiosciences.com/media/VISA_Invitation_Letter_Requirements.pdf

Visa Invitation Requirement
Please ensure that the above form is duly complete, as it will expedite the preparation of an invitation letter. Also mention clearly, to whom the invitation letter should be addressed as per the requirement of the country of origin.
For more information on Indian visa’s applications for International visitors, please contact your local Indian embassy.
Please plan sufficiently in advance because processing of Indian Visa Application may take 4-6 weeks.

Copyright © 2016 SELECTBIO, All rights reserved.

This email was sent from SELECTBIO Ltd to amcrasto@gmail.com.

SELECTBIO Ltd, Woodview, Bull Lane, Sudbury, CO10 0FD, United Kingdom.

//////////QbD,  Pharma Development,  World Congress,  2017, SelectBio, Radisson Hyderabad HITEC City, India

Share

QbD: Controlling CQA of an API

 QbD  Comments Off on QbD: Controlling CQA of an API
Sep 132016
 

The importance of Quality by Design (QbD) is being realized gradually, as it is gaining popularity among the generic companies. However, the major hurdle faced by these industries is the lack of common guidelines or format for performing a risk-based assessment of the manufacturing process. This article tries to highlight a possible sequential pathway for performing QbD with the help of a case study. The main focus of this article is on the usage of failure mode and effect analysis (FMEA) as a tool for risk assessment, which helps in the identification of critical process parameters (CPPs) and critical material attributes (CMAs) and later on becomes the unbiased input for the design of experiments (DoE). In this case study, the DoE was helpful in establishing a risk-based relationship between critical quality attributes (CQAs) and CMAs/CPPs. Finally, a control strategy was established for all of the CPPs and CMAs, which in turn gave rise to a robust process during commercialization. It is noteworthy that FMEA was used twice during theQbD: initially to identify the CPPs and CMAs and subsequently after DoE completion to ascertain whether the risk due to CPPs and CMAs had decreased.

 

 

 

Image result for Quality by Design in Action 1: Controlling Critical Quality Attributes of an Active Pharmaceutical Ingredient

Image result for Quality by Design in Action 1: Controlling Critical Quality Attributes of an Active Pharmaceutical Ingredient

Quality by Design in Action 1: Controlling Critical Quality Attributes of an Active Pharmaceutical Ingredient

CTO-III, Dr. Reddy’s Laboratories Ltd, Plot 116, 126C and Survey number 157, S.V. Co-operative Industrial Estate, IDA Bollaram, Jinnaram Mandal, Medak District, Telangana 502325, India
Department of Chemistry, Osmania University, Hyderabad, Telangana 500007, India
Org. Process Res. Dev., 2015, 19 (11), pp 1634–1644
*Telephone: +919701346355. Fax: + 91 08458 279619. E-mail: amrendrakr@drreddys.com (A.K.R.)., *E-mail:sripabba85@yahoo.co.in (P.S.).

str1

 

str2

 

str3 str4

 

str5 str6

str1 str2
str3
str4
str5
str6
str1
str2
str3
str4
str6
////// QbD, DoE, FMEA, ANOVA, Design space.
Share

New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach

 Formulation, regulatory  Comments Off on New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach
Sep 022016
 

Image for unlabelled figure

The current work outlines the application of an up-to-date and regulatory-based pharmaceutical quality management method, applied as a new development concept in the process of formulating dry powder inhalation systems (DPIs). According to the Quality by Design (QbD) methodology and Risk Assessment (RA) thinking, a mannitol based co-spray dried formula was produced as a model dosage form with meloxicam as the model active agent.

The concept and the elements of the QbD approach (regarding its systemic, scientific, risk-based, holistic, and proactive nature with defined steps for pharmaceutical development), as well as the experimental drug formulation (including the technological parameters assessed and the methods and processes applied) are described in the current paper.

Findings of the QbD based theoretical prediction and the results of the experimental development are compared and presented. Characteristics of the developed end-product were in correlation with the predictions, and all data were confirmed by the relevant results of the in vitro investigations. These results support the importance of using the QbD approach in new drug formulation, and prove its good usability in the early development process of DPIs. This innovative formulation technology and product appear to have a great potential in pulmonary drug delivery.

Fig. 1

Fig. 1.

Steps and elements of the QbD methodology completed by the authors and applied in the early stage of pharmaceutical development.

“By identifying the critical process parameters, the practical development was more effective, with reduced development time and efforts.”

Edina Pallagi, our QbD evangelist from Hungary shares her team’s experience applying QbD to Dry Powder Inhalation Formulation.

The paper covers:

  • QbD methodology the researchers applied
  • Formulation of dry powder inhalation – API and excipients
  • QTPP, CQA and CPPs  identified for pulmonary use along with target, justification and explanation
  • Characterization test methods
  • Knowledge Space development
  • QbD software used

New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach

  • a Institute of Drug Regulatory Affairs, University of Szeged, Faculty of Pharmacy, Szeged, Hungary
  • b Department of Pharmaceutical Technology, University of Szeged, Faculty of Pharmacy, Szeged, Hungary

///////

Share
Jul 092015
 

Areas of discussion included how expectations for raw material control are evolving within changing regulatory and business paradigms including quality by design (QbD), counterfeiting, complex supply chains, and sourcing changes. discussed risk assessment and mitigation strategies along with supplier risk management plans.

Regulatory Considerations

the lack of a consistent definition of raw materials in regulations pertaining to the pharmaceutical industry. In its Q7 guideline, the International Conference on Harmonisation of Technical Requirements for the Registration of Pharmaceuticals for Human Use (ICH) defines raw materials as “starting materials, reagents, and solvents intended for use in the production of intermediates or APIs.” However, the term as defined by different speakers could cover a wide range of materials including the following:

• starting or source materials (cell lines, viral or bacterial stocks, media components, chemicals, tissues, serum, water)

• in-process materials (resins, buffers, filters, column housings, tubing, reagents)

• excipients

• packaging components, both primary and secondary (syringes, vials, stoppers, plungers, crimps, boxes, trays, and labels)

• device/delivery components (pen/ injector components, IV bags, filters). Some regulations directly consider the control of raw materials, but they are not comprehensive and are scattered among the US Code of Federal Regulations (CFR), ICH, and other regulations/guidances. Although the regulations are not extensive, the need to control raw materials was clear from all presenters and is implicit in the sources cited below:

• 21 CFR 610.15 regarding constituents

• 21 CFR 211.80 regarding components and containers/closures

• 21 CFR 211.110 regarding control of in-process materials • ICH Q5A/D for cell substrates and viral safety

• ICH Q7 discussing the need to control materials with appropriate specifications

• ICH Q10 stating that a biomanufacturer is responsible for the quality of purchased materials

• the US bill “Country-of-Origin Labeling for Pharmaceutical Ingredients,” proposed in September 2008

• QbD principles requiring an understanding of the criticality of quality attributes for raw materials and their effect on processes and products.

Developing Control

Strategies Control of raw materials is essential to maintaining safety. Thorough knowledge of raw materials can mitigate the potential for contamination derived from such sources as microbes, chemicals, prions, and pyrogens. Raw material control for safety also includes identification — being able to verify that you have received the correct material — because the presence of an incorrectly identified material in a manufacturing process could compromise safety.

Control of raw materials is essential to ensure lot-to-lot consistency because variation in them can directly affect the variation of both product and process. So manufacturers must understand the critical material attributes (CMAs) of their raw materials and which of those affect variability — as well as how to control that variability.

You must show that you are using appropriate analytical methods to characterize raw materials. Raw materials such as polyethylene glycol (PEG) isomers, trace materials in media and water, container and closure materials, and chromatography resins all have the potential to affect lot-to-lot consistency. An effective raw material control program will also ensure consistent supplies.

A single source for a vital raw material can be a significant financial and quality-assurance risk. If a supplier goes out of business or experiences quality problems, can that raw material be obtained elsewhere? Has a second source been qualified in case the primary source is no longer available? Does the second source have the capacity to meet your needs? A QbD approach to raw material control requires that you understand the impact on your product’s critical quality attributes.

You will need to show that you understand the effect of raw material variability on your product as well as on your manufacturing process. Use of multiple lots during development can provide data on raw material lot-to-lot variability and its related effects on process and product. When that is not feasible, a manufacturer may consider including different lots of raw materials during bench-scale studies. In addition to the raw materials themselves, you should gain an understanding of whether and how raw material degradants might affect your process or product.

A QbD approach can use relevant knowledge to help you define how to go about setting specifications, in-process controls (IPCs), and handling conditions. Testing of Raw Materials The forum discussed what levels of testing are important for specific raw materials. A supplier’s certificate of analysis (CoA) is never sufficient for raw materials because good manufacturing practices (GMPs) require appropriate testing, and at a minimum, testing for identity. The material ordered may include additives, preservatives, degradation products, or contaminants. You must verify that the CoA is appropriate for control of the raw material, but you can’t assume that at the outset.

Similarly, CoA verification may be necessary only once a year once your experience with a given supplier has shown that quality is consistent. Vendor qualification is an important factor in defining your testing needs. To ensure the quality of raw materials against adulteration, identity testing is essential. Currently, tests with fingerprint techniques — e.g., nuclear magnetic resonance (NMR) imaging and Raman, nearinfrared (NIR), and Fourier-transform infrared (FTIR) spectroscopy — are used to assure the identity and quality of raw materials.

Whatever techniques you use, it is important to retain samples for future investigations. Photographic libraries of materials and their packaging have also proven useful for identifying and preventing use of counterfeit products. How often and in how much depth you need to verify a CoA through independent testing is an important consideration, especially for environments in which counterfeiting or contamination can occur.

Once you understand the CMAs of your raw materials, you need to identify which tests are relevant for testing specific quality attributes (QAs) of those raw materials. Sampling plans need careful consideration and should be risk based, dependent on the nature and use of the RM, and any regulatory requirements. Such plans should always be justified in a report available for inspection and/or filing.

It is important to consider RM stability and whether any special tests for degradants are needed for release of the material over time. A stability profile will dictate the purchasing program (storage of large quantities or buying as needed) as well as affect the associated testing strategy.

Supply Quality Management:

Ensuring Quality and Availability It is becoming increasingly evident in the current supply chain environment that management of suppliers and the “cold chain” is essential to assuring the quality of raw materials. How often and how thoroughly you perform vendor audits depends on your experience with a given vendor.

A manufacturer’s general experience with a vendor (prior knowledge) is an important criterion used to evaluate that vendor’s suitability to supply raw materials. Items to consider when selecting a vendor include its quality systems and its solvency, as well as its length of time in business, its geographic area, and whether it supplies multiple industries or just one or two drug manufacturers. Those form part of a risk assessment relating to suppliers to be described in more detail below.

Ensuring both the availability and qualification of secondary suppliers is important as well. Practices such as split purchasing may help ensure that you have good working relationships with multiple vendors. Strict change control sections should be included in supplier agreements and should include details requiring a vendor to notify you of changes in its product or suppliers. Such agreements should also provide for impact assessments from both supplier and manufacturer in the event that a supplier makes any changes. Supply chain traceability is not as straightforward as it might seem.

Although most manufacturers use country-of-origin (COO) questionnaires, those often prove less than ideal in revealing what you need to know. It is critical to craft questions that get the in-depth answers you need. For example, rather than asking “Do you purchase supplies from any high-risk countries?” you might ask “From what countries do you purchase supplies?” If the specified countries include any you consider to be high risk, you can follow up or choose another supplier.

It is critical to use risk-assessment techniques for determining traceability to avoid a false sense of security that can lead to costly or even deadly errors. It is sometimes unclear exactly what roles are played by whom in a supply chain.

Which companies are manufacturers, which are distributors, and which are intermediaries is not necessarily clear. A company that simply repackages a raw material from 55-gallon drums into smaller containers may consider itself a manufacturer. Due diligence will help ensure that you really know where your raw materials originated. As part of assessing supply chain complexity, forum participants were informed of a proposed program whereby industry creates a system of cooperative audits in which vendors would be audited by a selected team representing all industry rather than multiple auditors from each company continuously auditing suppliers.

The resulting audits would lead to certification that would assure all purchasers that each vendor meets certain defined criteria. Such a “360° Rx” program would enable increased depth of supplier audits and save manufacturers time and money (see box, right). The Role of Compendial Standards: Compendia provide some assurance of minimum quality standards for specified materials. However, compendial standards may differ among the pharmacopoeias.

Few of the complex raw materials (e.g., culture media, soy, yeastolates, most growth factors) used in biotechnology manufacturing are compendial, and those that are (e.g., insulin) may not have the appropriate compendial limits on specific quality attributes — or even test for quality attributes necessary to control pharmaceutical manufacturing. Even for standard chemical raw materials (e.g., trace metals), compendial standards may not focus on quality attributes relevant for biotechnology process and product quality assurance.

Those may be product- and/or process-specific. Furthermore, compendial standards do not necessarily help control for contamination, counterfeiting, or supply chain issues because a supplier can simply state it meets compendia — a statement that currently requires no certification

Risk Management

Risk assessments are an important tool for ensuring the safety, efficacy, consistency, and supply of pharmaceutical products. Many companies in both the United States and the European Union are using ICH Q9 as a basis for risk assessment, control, communication, and future review.

Risk assessments should begin by identifying all raw materials and assessing their criticality to product safety, efficacy, and supply. RM risk assessments require cross-functional input from all departments including supply, product development, manufacturing, quality control, quality assurance, clinical, and any other contributors. It was clear from this forum’s discussions that risk assessments are only as good as the people who carry them out. Having the right expertise over a spectrum of areas is vital if any risk assessment is to be meaningful. Multiple risk assessment tools exist, but in general, a good risk assessment must address concepts such as impact/ severity and likelihood/detectability.

One tool discussed at the forum (Figure 1) used nine blocks to score a supplier’s performance against material risk levels for audits, supplier qualification, supplier monitoring, change control, material specifications and testing, quality agreements, supplier certification, and sourcing, or other appropriate combinations of factors. Risk assessment should also be performed in relation to country of origin. It is critical to be able to trace your raw materials to their source. Just as a biopharmaceutical manufacturer audits its suppliers, those suppliers must also know, audit, and qualify their own distributors.

It is now well known that there are high-risk geographic areas where additional caution should be exercised to assure purity and identity of sourced materials. A potentially overlooked risk assessment issue is that manufacturers need to evaluate their raw materials and products in relation to opportunities for someone to make a profit through adulteration (e.g., by diluting a product to increase volume, and thus sales income). Any materials identified in such an evaluation should be managed with particular caution.

Risk assessments ensure that appropriate control strategies and raw materials (e.g., grade, origin) have been selected, which is relevant to a QbD approach. For regulatory filings, acceptable specifications, raw materials, and control strategies are tested with the necessary acceptance criteriia to ensure the performance of a process and the quality of its ultimate products. A periodic risk review should include more than a mere cursory review of individual risk assessments. It should reevaluate the risk program itself based on experience and lessons learned. Your risk assessment should be phase-appropriate, and as such it will change as data become available throughout development.

Early on, your raw materials risk assessment can be based on platform and previous knowledge, on the quality assurance of your suppliers, and adventitious agent introduction. As a manufacturing process develops, you will need to reevaluate that risk assessment including commercial considerations of scale and production frequency, highrisk raw materials control strategy, and handling and storage requirements.

During commercialization, design of experiments (DoEs) and collated knowledge will further define the CQAs of both product and RMs as well as potential and actual interactions among RMs, process, and product. At that point, you will be able to define and justify the raw materials for your commercial process and refine their specifications.

By the time your product is ready for market launch, you will have updated the failure modes and effects analysis (FMEA), completed your raw materials specifications, set your sourcing strategy, put in place your supplier qualification program, defined your raw material control strategy, and made your risk assessment ready for filing. The morning’s session resulted in a list of elements to be included in a raw materials risk assessment

 

Elements of Raw Material

Risk Assessments Is the raw material biological, chemical, or physical (such as tubing or stoppers, materials that are not actual components of the end product)? How likely is the raw material to introduce biological or chemical contamination?

Is the raw material or are its degradants able to directly affect the safety and/or efficacy of a drug substance (e.g., toxicity, chemical modifications)?

How complex is the raw material itself or its impurity profile? How much prior knowledge (e.g., historical or published knowledge, current experience) do you have regarding the raw material? What is the Intended use of the raw material (e.g., as a buffer, reagent, or excipient)?

Where in the manufacturing process will this raw material will be used (upstream/ downstream)?

What is the extent of supply chain traceability (considering country of origin, supply chain complexity, and supply chain security)?

What is the extent of supplier quality assurance (from audits, monitoring, historical experience)?

How extensive is the characterization of the raw material (how well can the raw material be characterized; standard existing methods or novel techniques; the RM’s impact on test methods)?

How stable is the raw material? Is the raw material new to the process or a result of a change to an existing raw material (if a change, what studies have been executed to assure comparability)?

What is the depth of knowledge of the RM’s own manufacturing process to assess the risk associated with its use (e.g., process contaminants)?

Does the use of the raw material in a manufacturing environment present safety and/or handling risks? Does your process have the ability to clear the raw material?

Are there associated business risks (e.g., a solesource or multiple-source material, unique or not to the pharmaceutical industry, custom-made or not, and the supplier’s ability to consistently meet specific requirements)?

What is your level of understanding of the raw material CMAs?

Benefits of Implementing QbD

Benefits for the FDAEnhances scientific foundation for review
Provides for better coordination across review, compliance, and inspection
Improves information in regulatory submissions Provides for better consistency
Improves quality of review (establishing a quality management system for CMC)
Provides for more flexibility in decision making
Ensures decisions made on scientific and not on empirical information
Involves various disciplines in decision making
Uses resources to address higher risks
Benefits for Industry
Ensures better design of products with fewer problems in manufacturing
Reduces number of manufacturing supplements required for postmarket changes; relies on process and risk understanding and risk mitigation
Allows for implementation of new technology to improve manufacturing without regulatory scrutiny
Allows for possible reduction in overall costs of manufacturing; creates less waste
Ensures less hassle during review, reduces deficiencies, speeds approvals Improves interaction with the FDA; operates on a scientific rather than on a process level
Allows for continuous improvements in products and manufacturing processes
Allows for better understanding of how APIs and excipients affect manufacturing
Relates manufacturing to clinical during design
Provides a better overall business model

Frequently Used QbD Terms 

 

Quality Attribute: A physical, chemical, or microbiological property or characteristic of a material that directly or indirectly alters quality Critical Quality Attribute (CQA): A quality attribute that must be controlled within predefined limits to ensure that a product meets its intended safety, efficacy, stability, and performance
Real-Time Release (RTR): Ability to evaluate and ensure acceptable quality of an in-process and/or final product based on process data, including valid combination of assessment of material attributes by direct and/or indirect process measurements and assessment of critical process parameters and their effects on in-process material attributes Process Parameter: An input variable or condition of a manufacturing process that can be directly controlled in the process. Typically, such parameters are physical or chemical (e.g., temperature, process time, column flow rate, column volume, reagent concentration, or buffer pH).
Critical Process Parameter (CPP): A process parameter whose variability has an influence on a CQA and therefore should be monitored or controlled to ensure a process produces a desired quality. Process Performance Attribute: An output variable or outcome that cannot be directly controlled but is an indicator that a process performed as expected
Key Process Parameter (KPP): An input process parameter that should be carefully controlled within a narrow range and is essential for process performance; a key process parameter does not affect product quality attributes. If the acceptable range is exceeded, it may affect the process (e.g., yield, duration) but not product quality. Non-Key Process Parameter: An input parameter that has been demonstrated to be easily controlled or has a wide acceptable limit. Such parameters may influence quality or process performance if acceptable limits are exceeded.
Design Space: The multidimensional combination and interaction of input variables (e.g., material attributes) and process parameters that have been demonstrated to provide assurance of quality; working within a design space is not considered to be a change requiring regulatory approval. Movement out of a design space is considered to be a change and would normally initiate a regulatory postapproval change process. Design space is proposed by an applicant and is subject to regulatory assessment and approval (ICH Q8). Control Strategy: A planned set of controls, derived from current product and process understanding, that ensures process performance and product quality; such controls can include parameters and attributes related to drug substance and drug product materials and components, facility and equipment operating conditions, in-process controls, finished-product specifications, and associated methods, and frequency of monitoring and control (ICH Q10).
Quality Target Product Profile (QTPP): A prospective summary of the quality characteristics of a drug product that ideally will be achieved to ensure desired quality, taking into account safety and efficacy of a drug product
Share

Key steps in implementation of QbD for a biotech product…….Quality by design for biopharmaceuticals

 regulatory  Comments Off on Key steps in implementation of QbD for a biotech product…….Quality by design for biopharmaceuticals
Aug 052014
 

Unfortunately we are unable to provide accessible alternative text for this. If you require assistance to access this image, or to obtain a text description, please contact npg@nature.com

 

 

Identifying target product profile (TPP). TPP has been defined as a “prospective and dynamic summary of the quality characteristics of a drug product that ideally will be achieved to ensure that the desired quality, and thus the safety and efficacy, of a drug product is realized”. This includes dosage form and route of administration, dosage form strength(s), therapeutic moiety release or delivery and pharmacokinetic characteristics (e.g., dissolution and aerodynamic performance) appropriate to the drug product dosage form being developed and drug product-quality criteria (e.g., sterility and purity) appropriate for the intended marketed product. The concept of TPP in this form and its application is novel in the QbD paradigm.

Identifying CQAs. Once TPP has been identified, the next step is to identify the relevant CQAs. A CQA has been defined as “a physical, chemical, biological, or microbiological property or characteristic that should be within an appropriate limit, range, or distribution to ensure the desired product quality”10. Identification of CQAs is done through risk assessment as per the ICH guidance Q9 . Prior product knowledge, such as the accumulated laboratory, nonclinical and clinical experience with a specific product-quality attribute, is key in making these risk assessments. Such knowledge may also include relevant data from similar molecules and data from literature references. Taken together, this information provides a rationale for relating the CQA to product safety and efficacy. The outcome of the risk assessment would be a list of CQAs ranked in order of importance. Use of robust risk assessment methods for identification of CQAs is novel to the QbD paradigm.

Defining product design space. After CQAs for a product have been identified, the next step is to define the product design space (that is, specifications for in-process, drug substance and drug product attributes). These specifications are established based on several sources of information that link the attributes to the safety and efficacy of the product, including, but not limited to, the following:

  • Clinical design space
  • Nonclinical studies with the product, such as binding assays, in vivo assays and in vitro cell-based assays
  • Clinical and nonclinical studies with similar platform products
  • Published literature on other similar products
  • Process capability with respect to the variability observed in the manufactured lots

The difference between the actual experience in the clinic and the specifications set for the product would depend on our level of understanding of the impact that the CQA under consideration can have on the safety and efficacy of the product. For example, taking host cell proteins as a CQA, it is common to propose a specification that is considerably broader than the clinical experience. This is possible because of a greater ability to use data from other platform molecules to justify the broader specifications. On the other hand, in the case of an impurity that is unique to the product, the specifications would rely solely on clinical and nonclinical studies.

In QbD, an improved understanding of the linkages between the CQA and safety and efficacy of the product is required. QbD has brought a realization of the importance of the analytical, nonclinical and animal studies in establishing these linkages and has led to the creation of novel approaches.

Defining process design space. The overall approach toward process characterization involves three key steps. First, risk analysis is performed to identify parameters for process characterization. Second, studies are designed using design of experiments (DOE), such that the data are amenable for use in understanding and defining the design space. And third, the studies are executed and the results analyzed to determine the importance of the parameters as well as their role in establishing design space.

Failure mode and effects analysis (FMEA) is commonly used to assess the potential degree of risk for every operating parameter in a systematic manner and to prioritize the activities, such as experiments, necessary to understand the impact of these parameters on overall process performance. A team consisting of representatives from process development, manufacturing and other relevant disciplines performs an assessment to determine severity, occurrence and detection. The severity score measures the seriousness of a particular failure and is based on an estimate of the severity of the potential failure effect at a local or process level and the potential failure effect at end product use or patient level. Occurrence and detection scores are based on an excursion (manufacturing deviation) outside the operating range that results in the identified failure. Although the occurrence score measures how frequently the failure might occur, the detection score indicates the probability of timely detection and correction of the excursion or the probability of detection before end product use. All three scores are multiplied to provide a risk priority number (RPN) and the RPN scores are then ranked to identify the parameters with a high enough risk to merit process characterization.  FMEA outcome for a process chromatography step in a biotech process. RPN scores are calculated and operating parameters with an RPN score >50 are characterized using a qualified scaled-down model. For the case study presented here, these include gradient slope, temperature, flow rate, product loading, end of pool collection, buffer A pH, start of pool collection, volume of wash 1, buffer B pH, buffer C pH and bed height. Process characterization focused on parameters such as temperature, that have a high impact on the process (severity = 6), occur frequently in the manufacturing plant (occurrence = 6) and are difficult to quickly correct if detected (detection = 7). In contrast, parameters such as equilibration volume, with a low impact on the process (severity = 3), low occurrence (occurrence = 2) and a limited ability to detect and correct (detection = 5), were not examined in process characterization.

I liked this pic

this is the check" class="grayscale

SEE AT

http://www.tevapharm.com/Media/EventsUpdates/Pages/Quality-by-Design.aspx

Share

Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design

 regulatory, Uncategorized  Comments Off on Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design
Feb 032014
 

Figure

 

A strategy for the risk assessment of potentially genotoxic impurities is described that utilizes Quality by Design in an effort to furnish greater process and analytical understanding, ultimately leading to a determination of impurity criticality. By identifying the risks and parameters that most influence those risks, an enhancement of both product and process control is attained that mitigates the potential impact of these impurities. This approach calls for the use of toxicological testing where necessary, chemical fate arguments when possible, multivariate analyses to develop design space, and use of spiking data to support specifications. Strong analytical support, especially with the development of low-level detection methods, is critical. We believe that this strategy not only aids in the development of a robust API process but also delivers on the identification and subsequent mitigation of risks to a class of impurities that are of high interest in the field.

Risk Assessment of Potentially Genotoxic Impurities within the Framework of Quality by Design

Adam R. Looker, Michael P. Ryan, Bobbianna J. Neubert-Langille and Redouan Naji
Org. Process Res. Dev., 2010, 14 (4), pp 1032–1036
pp 1032–1036
Publication Date (Web): April 7, 2010 (Communication)
DOI: 10.1021/op900338g
Figure
Share
Follow

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

Join other followers: