Thursday, May 17, 2018

Cambrex Reports Expansions at its High Point, NC Facility

On May 15, 2018, Cambrex reported that it had completed a pilot plant expansion at its High Point, NC facility with the installation and commissioning of a fourth reactor suite. The new 400-sq.-ft. suite includes two 2000 L glass-lined reactors and a Hastelloy C22 filter dryer, allowing the manufacture of batch sizes ranging from 10–100kg under cGMP conditions for clinical-phase projects. The installation increased the site’s reactor capacity by approximately 30%, according to a company press release.

The company noted that it also upgraded its analytical chromatography data systems for quality control and analytical R&D to Empower 3 software (Waters).

In April 2018, the company announced it completed installation of multiple continuous flow reactor platforms at the High Point facility; the platforms will focus on the rapid development of processes to supply clinical and commercial demand for chemical syntheses.

The company also completed the construction and validation of a $3.2-million, 11,000-sq.-ft. analytical laboratory at the High Point facility to meet customer demand for analytical development and validation services in support of clinical stage cGMP products. More than 20 analytical instruments, including high-performance liquid chromatography (HPLC), mass spectrometers, and support equipment were added, and 12 analytical scientists will be hired, the company reports.

The company also reported on May 15 progress in construction for a $24-million facility at the company’s Charles City, IA plant, for the manufacture of highly potent APIs. A 4500-sq.-ft. production area, which will have a reactor capacity of 2200 gallons and will manufacture batches from 50­–300kg, will operate to an occupational exposure limit down to 0.1µg/m³.

The existing small-scale manufacturing area will be reconfigured, providing a single high-containment building to support early stage development and manufacturing. The facility is expected to be operational in the first half of 2019, the company reports.

Source: Cambrex Corporation

 

Source link

Wednesday, May 16, 2018

Irish engineering group wins major contract in Sweden

Irish engineering and construction management firm DPS has won a major contract from the global pharmaceutical company AstraZeneca.

DPS, which employs over 1,300 people, is to provide engineering, procurement, construction management and validation (EPCMV) services for small and medium sized projects on AstraZeneca’s pharmaceutical site in Södertälje, Sweden.

The contract win means that DPS will now establish a new office in Sweden to service AstraZeneca initially, as well as other clients in the Nordic region at a later stage.

The value of the contract has not been disclosed, but it is expected that over 100 DPS engineering personnel potentially will be working on AstraZeneca projects, depending on capital expenditure levels.

Frank Keogh said: “DPS is delighted to have won this contract and we are confident the combination of a substantial local office and local partners in Sweden, as well as our large pharma centre of excellence offices in Dublin and Cork will ensure a quality and cost effective delivery model for AstraZeneca. As with all our clients we will work with AstraZeneca in a true partnership, building a long term relationship”.

DPS operates in Ireland, Netherlands, Belgium, Sweden, Israel, Singapore and the United States.

Source link

Tuesday, May 15, 2018

2 Day Course: FDA Trends for Computer System Validation (CSV) Compliance and Enforcement (Boston, MA – June 25-26, 2018) – ResearchAndMarkets.com | Technology

This seminar will focus on the key areas that are most important, including security and data integrity. Implementing and following the System Development Life Cycle (SDLC) methodology is the best approach for Computer System Validation and maintaining data integrity. The life cycle approach takes all aspects of validation into account throughout the life of the system and the data that it houses. The data is a key asset for any FDA-regulated company and must be protected through its entire retention period.

This session will provide some insight into current trends in compliance and enforcement. Some are based on technology changes, and these will continue to have an impact as new innovations come into use in the industry.

Areas Covered in the Session

Computer System Validation (CSV)System Development Life Cycle (SDLC) Methodology”GxP” – Good Manufacturing, Laboratory and Clinical Practices21 CFR Part 11, Electronic Records/Electronic Signatures (ER/ES)Data Archival to ensure security, integrity and complianceValidation Strategy that will take into account the system risk assessment and system categorization (GAMP V) processesPolicies and ProceduresCritical TrainingRecent FDA findings for companies in regulated industriesRecent trends in technology that need to be addressed in the CSV approachQ&A

Laura Wood, Senior Manager

For E.S.T Office Hours Call 1-917-300-0470

For U.S./CAN Toll Free Call 1-800-526-8630

For GMT Office Hours Call +353-1-416-8900

Related Topics:Drug Discovery,Pharmaceutical Manufacturing

KEYWORD: UNITED STATES NORTH AMERICA

INDUSTRY KEYWORD: TECHNOLOGY DATA MANAGEMENT HEALTH PHARMACEUTICAL FDA

SOURCE: Research and Markets

Copyright Business Wire 2018.

PUB: 05/15/2018 07:19 AM/DISC: 05/15/2018 07:19 AM

Copyright Business Wire 2018.



Source link

DPS Group Wins Contract in Sweden from AstraZeneca

As a result of winning this contract, DPS Group is now establishing an office in Sweden, which will be used initially to service AstraZeneca exclusively but later to service other clients in Sweden.  

DPS Group CEO Frank Keogh said: “DPS are delighted to have won this contract and we are confident the combination of a substantial local office and local partners in Sweden, as well as our large pharma centre of excellence offices in Dublin and Cork will ensure a quality and cost effective delivery model for AstraZeneca. As with all our clients we will work with AstraZeneca in a true partnership, building a long term relationship.”

The value of the contract has not been disclosed but it is expected that over 100 DPS engineering personnel potentially will be working on AstraZeneca projects depending on capital expenditure levels.

About DPS Group
DPS Group is a global engineering, consulting and project management company, serving high-tech industries around the world. DPS delivers services for clients across the complete engineering and construction value chain including feasibility studies, concepts, consulting, architecture, engineering, professional outsourcing, procurement, construction management, commissioning, qualification and validation; as well as contingent staffing solutions.

DPS applies its extensive Process Engineering expertise built over 40+years, as well as significant Lean Construction experience to assist our clients in high-end process sectors such as pharmaceuticals, biotech, medical devices and semiconductors. DPS deliver their manufacturing facilities speedily, safely and cost effectively. What sets us apart are the partnerships we build with our clients through a fundamental understanding of their businesses and our own agility, flexibility, original thinking and our high-calibre people.

We have grown substantially in recent years and now employ over 1,300 people in our offices and on client sites in Ireland, Netherlands, Belgium, Sweden, Israel, Singapore, Switzerland, the United Kingdom and the United States. For more information, visit https://ift.tt/2dvSn1u.

Contact: Annette O’Hara, Group Marketing Manager  
O +353-1-466-1755
M +353-87-248-2573
E annette.ohara@dpsgroupglobal.com

SOURCE DPS Group



Source link

AstraZeneca (AZN) Partnered with DPS Group in Sweden for Construction Contract

The UK researched-based biopharmaceutical company, AstraZeneca PLC (NYSE:AZN) partnered with DPS Group, the global and European based engineering, consulting and project management company, in a construction contract.

The contract is specific for AstraZeneca’s pharmaceutical site in Swedan.

The terms and condition of contract was not disclosed by both companies even the value, however, DPs Group said that it will provide Engineering, Procurement, Construction Management and Validation (EPCMV) services for all small and medium projects of AstraZeneca’s site in Swedan.

DPS Group serving to their clients with lean construction experience in pharmaceuticals, biotech, medical devices and semiconductors from over 40 years in US and Europe. With over 1,300 employees, the construction firm presently serving their customers in Ireland, Netherlands, Belgium, Sweden, Israel, Singapore, Switzerland, the United Kingdom and the United States.

After winning the construction contract from AstraZeneca’s, DPS Group said that it is now building its office in Swedan completely for AstraZeneca which will provide services to AstraZeneca on starting and later on for other clients.

“DPS are delighted to have won this contract and we are confident the combination of a substantial local office and local partners in Sweden, as well as our large pharma center of excellence offices in Dublin and Cork will ensure a quality and cost-effective delivery model for AstraZeneca. As with all our clients we will work with AstraZeneca in a true partnership, building a long-term relationship.” CEO of DPS Group, Frank Keogh, commented on the winning contract from AsrtaZeneca (AZN).

However, in other news for AstraZeneca, the biotech firm said that it will published its financial results for the first quarter of 2018 on Friday, May 18, 2018 on its website at 07:00 UK time.

A webcast for the financial results also will take place at 12:00 UK time on Friday, May 18, 2018.

With the $90.69 market value, AstraZeneca PLC (NYSE:AZN)’s shares up 0.05% or $0.02 cents to close the Monday trading session at $36.42. The biotech company posted EPS of $1.18 for the trailing twelve months.

Source link

AstraZeneca (AZN) Partnered with DPS Group in Sweden for Construction Contract

The UK researched-based biopharmaceutical company, AstraZeneca PLC (NYSE:AZN) partnered with DPS Group, the global and European based engineering, consulting and project management company, in a construction contract.

The contract is specific for AstraZeneca’s pharmaceutical site in Swedan.

The terms and condition of contract was not disclosed by both companies even the value, however, DPs Group said that it will provide Engineering, Procurement, Construction Management and Validation (EPCMV) services for all small and medium projects of AstraZeneca’s site in Swedan.

DPS Group serving to their clients with lean construction experience in pharmaceuticals, biotech, medical devices and semiconductors from over 40 years in US and Europe. With over 1,300 employees, the construction firm presently serving their customers in Ireland, Netherlands, Belgium, Sweden, Israel, Singapore, Switzerland, the United Kingdom and the United States.

After winning the construction contract from AstraZeneca’s, DPS Group said that it is now building its office in Swedan completely for AstraZeneca which will provide services to AstraZeneca on starting and later on for other clients.

“DPS are delighted to have won this contract and we are confident the combination of a substantial local office and local partners in Sweden, as well as our large pharma center of excellence offices in Dublin and Cork will ensure a quality and cost-effective delivery model for AstraZeneca. As with all our clients we will work with AstraZeneca in a true partnership, building a long-term relationship.” CEO of DPS Group, Frank Keogh, commented on the winning contract from AsrtaZeneca (AZN).

However, in other news for AstraZeneca, the biotech firm said that it will published its financial results for the first quarter of 2018 on Friday, May 18, 2018 on its website at 07:00 UK time.

A webcast for the financial results also will take place at 12:00 UK time on Friday, May 18, 2018.

With the $90.69 market value, AstraZeneca PLC (NYSE:AZN)’s shares up 0.05% or $0.02 cents to close the Monday trading session at $36.42. The biotech company posted EPS of $1.18 for the trailing twelve months.

Source link

CGMP Violations Found at India Facility

FDA sent a warning letter to Goran Pharma Private Limited citing inadequate quality control violations.

FDA sent a warning letter, dated April 24, 2018, to Goran Pharma Private Limited after an inspection of the company’s Sihor, Gujarat, India facility found deviations from current good manufacturing practices (CGMPs). A variety of violations found during the inspection included failure to identify ingredients from suppliers, inadequate quality control, and inappropriate equipment use. The inspection was conducted by FDA from Nov. 13–15, 2017.

Specifically, FDA stated in the warning letter that the company failed to test components from suppliers to “determine their conformance to identity, purity, strength, and other appropriate specifications. Your firm released components for use in drug product manufacturing based on certificates of analysis (COA) from your supplier without establishing the reliability of the suppliers’ analyses through appropriate validation.”

The agency requested the company provide a detailed description of how it plans on ensuring that components used in manufacturing be withheld from use until they have been tested. The company must also show how it will quality suppliers’ COAs, perform detailed risk assessments, and do a comprehensive review of laboratory practices, methods, equipment, and analyst competencies.

The warning letter also indicated that the company’s response to inspectors finding that inappropriate systems were used was lacking. The agency requested the company provide a detailed validation plan for its new system, procedures for routine monitoring of the system, a risk assessment of product manufactured on the old system, and reasons for why the company relied on past data to characterize the system’s state of control.

Inadequate control of air pressure, micro-organisms, dust, humidity, and temperature was also observed by inspectors. The agency requested the company provide an action plan for ensuring control and monitoring of temperature, humidity, and air cleanliness as well as an independent assessment of the company’s facilities and equipment.

The warning letter stated that FDA had placed the firm on Import Alert 66-40 on March 5, 2018.

FDA recommended the company hire a third-party CGMP consultant to audit the company’s operations.

Source: FDA

 

Source link

Monday, May 14, 2018

Calibration And Validation Of A Headspace Gas Chromatographic Method – Covering the Printing Inks, Coatings and Allied Industries

Pallavi Gupta, Ruchi Gupta, Pinaki Ranjan Samanta and Pradeep Shah, Uflex Ltd (Chemical Division), Noida, India05.11.18

A simple and sensitive method for the simultaneous determination of methanol, ethanol, iso propyl alcohol(IPA), methyl ethyl ketone (MEK), methyl isobutyl ketone (MIBK), n-propyl alcohol (n-PA), ethyl acetate (EA), n-butanol, n-propyl acetate (n-PAc), toluene and ortho-xylene by headspace techniques with FID detection is described. An efficient and sensitive method was successfully developed and validated for the determination of above mentioned solvents of varying polarity used as residual solvents from printing ink.

Based on Good Manufacturing Practices, measuring residual solvents is mandatory for the release testing of all active chemical ingredients. The method was validated for repeatability, linearity, limit of detection, limit of quantification and recovery according to the International Conference on Harmonization guidelines. The method validation results indicate that the method is accurate, precise, linear and sensitive for solvents assessed in the printing ink. Excellent results were obtained, within the globally accepted validation reference values, particularly taking into account the low concentration levels investigated.

Key Words: Validation, residual solvents, GC-HS, printing ink, volatiles.

* Corresponding Author: pallavi.gupta24192gmail.com

Abbreviations: Iso propyl alcohol (IPA), methyl ethyl ketone (MEK), methyl isobutyl ketone (MIBK), n-propyl alcohol (n-PA), ethyl acetate (EA), n-butanol, n-propyl acetate (n-PAc),ortho-xylene (xylene).

Residual solvents or organic volatile impurities are a potential toxic risk of pharmaceutical [1] or any other food-related products, whether it is packaging laminates of food as itself, and have been a concern of manufacturers for many years. Moreover, residual solvents can also affect the quality and stability of not only drug substances [2] but also packaging laminates of food as itself.

Thus, acceptable levels of many solvents are included in regulatory guidance documents, in particular in guideline Q3C issued by the International Conference on Harmonization of technical requirements for registration of pharmaceuticals for human use (ICH)[3].

The technique of static headspace gas chromatography has great acceptance in the chemical industry, especially for determination of solvents in printing inks and laminating films. Most chemical industries in the world have this equipment and perform this analysis on a routine basis, but in many of these laboratories, the equipment is exclusively employed to determine solvents, even when this technique can be used to determine many other substances of toxicological interest, such as volatile substances, without major changes to the equipment. We can conclude that these laboratories do not exploit all the possibilities of the technique.

Determination of residual solvents using GC with a flame ionization detector (FID) is the most common technique in the pharmaceutical industry as well as the chemical industry because of its high separation efficiency and sensitivity for volatile organic compounds. The headspace gas chromatography (HS-GC) method has been used for the determination of residual solvents in pharmaceutical compounds [4,5] and also packaging laminates of food.

The determination of volatile substances is one of the most important tests in chemical toxicology. Volatile substances can be defined as those organic compounds whose vapor pressures are greater than or equal to 0.1 mm Hg at 20°C. The determination of volatile substances in polymeric samples has been carried out through titrations, spectrophotometric methods and chromatographic methods. Titrations and spectrophotometric methods are not specific and usually lack sufficient sensitivity, besides not being able to analyze simultaneously all the volatile substances.

In contrast, gas chromatography is qualitative (by the use of retention time) and quantitative (by the use of signal strength), so it is able to analyze simultaneously several volatile substances with the adequate sensitivity and specificity necessary in chemical environments.

The authors have tried to validate a method by using gas chromatography to analyze volatile (%) and residual volatiles. This paper demonstrates a thorough study to establish a validated method for analysis involving volatiles quantitatively and qualitatively.

All gas chromatography experiments were conducted with Perkin Elmer Clarus 500, Headspace Turbo matrix 16 Auto-samplers.

The chromatographic oven temperature program was as follows: the initial temperature of 40°C was held for 8 minutes, 15°C/min to 100°C and 25°C/min up to 250°C and FID Detector temperature set at 260°C. Column used Elite-5 (Cross bond 5% diphenyl-95% dimethyl polysiloxane) length 30 meter, diameter 0.25mm ID, thickness 0.25 micrometer. Nitrogen as carrier gas with 22.8 cm/sec, flow rate of hydrogen was 40ml/min, Air was 400ml/min and sample flow rate was 1ml/min.

Headspace conditions of the oven temperature were set at 90°C and Needle temperatures were 100°C. The pressurization time was 1 min and the Transfer line temperature was 110°C.

A. Material GC-HS analysis was carried out by using analytical grade (AR) solvent with purity of more than 99% such as methanol (99.5%), ethanol (97.5%), IPA (99.7%), n-propyl alcohol (99.7%), MEK (99.5%), EA (99%), n-butanol (99.5%), n-propyl acetate (98%), MIBK (98%), toluene (99.5%), xylene (97%) and HPLC water used as a diluents.

B. Method of preparation of Sample

a) Preparation of Stock solution: 1 g of each solvent weighed accurately and calculated the concentration involved in the analysis shown in Table 1 used as a stock solution.

b) Dilution: – Stock solution was diluted to varying concentrations used for analysis, where varying volume (1ml, 3ml, 5ml) of stock has been taken in GC/HS (20 ml) vial and diluted with 10 ml water varying concentrations shown in Table 1. Water was used as a preferred solvent as a standard, because of its polarity and compatibility with all solvents.

c) Concentration of each solvents: Concentration of all solvents has been calculated as per the standard calculation procedure as shown in Table 1.

Transfer the above prepared diluted three base concentration into GC-HS vials (20 ml) sealed and crimped. Each of the vials contains 10-50 ppm of methanol, 10-50 ppm of ethanol, 10-50 ppm of IPA, 10-50 ppm of n-propyl alcohol and 10-50ppm of other solvents with respect to the sample. The vials have 10ml water containing solvents at different concentrations; the vials are kept at 60°C injector temperature in headspace since a sufficient flow must be maintained through the system to avoid excessive peak broadening.

In this study, a HS-GC analytical method was developed and validated for the quantitative determination of the all the solvents methanol, ethanol, IPA, MEK, MIBK, EA, n-butanol, n-propyl acetate and so on. The proposed method uses the standard addition technique with internal standard quantization for determination of eleven solvents. The method was validated as per food safety regulation defined quantitation, linearity, range, precision (system repeatability), recovery and robustness (changes in HS and GC conditions and solution stability) were determined.

Excellent results were obtained, within global validation reference values, particularly taking into account the low concentration levels investigated. The test method was validated and had good Repeatability [Table 2], relative standard deviation RSD and linearity (%) [Table 3 and 4] and linearity [Figure 01, 02 and 03 and Table 4] for the solvents used for the current study.

The recovery was good and justified the preparation of the standard in water without the product as matrix. The linear range and correlation coefficients were determined between 10-50 ppm of each solvent.

The concentration of residual solvents (ppm) determined using the formula for solvent (Equation 1).

The ELITE-5 column, in the 30 m x 0.32 mm I.D. configuration, was chosen because this column has a standard stationary phase, which is recommended by the European and American Pharmacopeias, and has provided baseline of all solvents used in the validation, including the diluent (water). The method showed good peak shape, and the narrow peak width resulted in excellent column efficiency. The blank chromatogram did not show any interference with the solvent peaks.

To carry out this study, three concentrations were prepared for each solvent. All concentrations were prepared in triplicate, by individually weighing of solvents. The experimental results were represented graphically to obtain a calibration curve and carry out the corresponding statistical study (Anova). The method is linear within a wide range for the solvents included in the validation.

The correlation coefficients were all above 0.99 and linear regression showed a positive response throughout the range [Figure 2 and 3; Table 4]. The specified range is normally derived from linearity studies and depends on the intended application of the procedure[6]. In this paper it was characterized as the interval between the lowest (8 ppm) and highest (50 ppm) concentration, which can be determined using a given method, with assumed precision, trueness and linearity. This method determine lowest concentration so it is valid for wide measurement range allows determination with adequate precision of different solvents at higher concentration too. The measurement ranges are shown in the Table 4 with the respective RSD values.

LODs were calculated at S/N ratio of ≥ 3 and LOQs were calculated as at S/N ratio ≥10 and low-residual linearity values. The sensitivity of the method was demonstrated by the low-LOD values and low LOQ obtained for all the solvents analyzed.

The mean recoveries for all the solvents were between 95.0–105.3% and were lower than tabulated [Table 4], so the recoveries and 100% values were not significantly different.

The analytical method proposed for the quality control of all selected solvents methanol, ethanol, iso propyl alcohol, MEK, MIBK, n propyl alcohol, EA, n-butanol, n propyl acetate, toluene and xylene very well complete the validation requirements and precisely fit for various polarity solvents as the method was sensitive, linear, accurate and precise. Excellent results were obtained, within globally accepted validation reference values, particularly taking into account the low concentration levels investigated. 

The authors are thankful to Uflex Chemical Division for providing infrastructure and constructive criticism which have helped to accomplish this work.

1. Y. Sitaramaraju, A. Riadi, W. Autry, K. Wolfs, J. Hoogmartens, A.V. Schepdael and E. Adams, “Static headspace gas chromatography of (semi-) volatile drugs in pharmaceuticals for topical use,” J. Pharm. Biomed. Anal., 48, 2008, 113.

2. E.M. Antolín, Y.B. Quinónez, V.G. Canavaciolo, E.R. Cruz, “Validation of an analytical method for quality control of residual solvents (n-hexane and acetone) in D-002: new active ingredient from beeswax,” J. Pharm. Biomed. Anal. 47, 2008, 646.

3. Proceedings of International Conference on Harmonization of Technical Requirements for “Impurities: Residual Solvents” Registration of Pharmaceuticals for Human Use (ICH), Tripartite harmonized guideline 3C 1997.

4. P. Mahesh, K.Swapnalee, M. Aruna, B. Anilchandra, S. Prashanti, “Analytical Method Development And Validation of Acetaminophen, Caffeine, Phenylephrine Hydrochloride and Dextromethorphan Hydrobromide in Tablet Dosage Form By RP- HPLC,” Int. J. of Pharma., 2, 2013, 2319.

5. Y. Liu, and C.O. Hua,, “Establishment of a knowledge base for identification of residual solvents in pharmaceuticals,” Anal Chem. Acta,575, 2006, 246.

6. P. Mahesh, K. Swapnalee, M. Aruna, B. Anilchandra, S. Prashanti, “Analytical Method Development And Validation Of Acetaminophen, Caffeine ,Phenylephrine v Hydrochloride And Dextromethorphan Hydrobromide In Tablet Dosage Form By RP- HPLC,” Int J. of Pharmaceutical,2, 2013, 2319 – 6718.

Pallavi Gupta, masters in instrumentation, is currently working with Uflex Limited, Noida as chemist (R&D instrumentation). She has research experience of more than two years with the expertise in the area of analysis of and cross-verification quality checks of inks, adhesives, coatings, and multi components coatings by using state-of-the-art instruments like Py- GC-MS, GC-HS, FTIR, UV-spectrophotometer, HPLC, LCMS.

Dr. Ruchi Gupta, masters in industrial chemistry and Ph.D. in chemistry, is currently with Uflex Limited, Noida as manager (R&D), and prior to this, worked as a scientist with Shriram Institute for Industrial Research.

She has research experience of more than 11 years and her areas of interest include inks, adhesives, coatings, nanomaterials, nanophotocatalysts, optical polymers, natural polymers etc. She has published 12 papers in reputed international journals and has published four chapters in national and international handbooks.

Dr. Pinaki Ranjan Samanta has more than two decades of experience in the areas associated to industrial research of coating and testing of FG and RM’s of coating-related materials. He received M. Sc and Ph. D in the field of organic chemistry, also earning his PG diploma in paint technology.

He has published more than 12 papers in national and international journals, and also two patents in his cap in the field of high performance coatings. Since the middle of 1990s, he is an accomplished analytical chemist with years of research. Dr. Pinaki is experienced in the use of several analytical techniques including mass spectrometry (GC-MS, LC-MS), atomic spectrometry (AAS) and chromatography, and is also involved in the project development and research on new technology. Dr. Pinaki’s background includes tenures at reputed organizations like Berger Paints, Shriram Institute for Industrial Research, Moser Baer and Uflex in the area of research and development.

Pradeep Shah is a science graduate and has done B. Tech. chemical technology specialization in paints from Harcourt Butler Technological Institute Kanpur – 1980 batch. He has been selected from the campus by INCOWAX Ltd., now Flint Group India Ltd. He has done post- graduate diploma in business management (PGDBM).

He joined Uflex Group Chemical Division for its ink project in 1991 to start its ink plant with initial capacity of 3,000 tons per annum in Noida. The current capacity is 25,284 ton per annum with two plants located at Noida and Jammu in India. His total experience is more than 37 years.

Source link

Sichuan Kelun Pharmaceutical Automates Oncology Clinical Trial Set Up and Management with Clinical One Randomization and Supplies Management Cloud Service, Business News

CHENGDU, China, May 14, 2018 /PRNewswire/ — Oracle Health Sciences today announced that Sichuan Kelun Pharmaceutical Research Institute Co., Ltd (“Kelun”) has implemented Oracle Health Sciences Clinical One Randomization and Supplies Management Cloud Service to fully automate the set up and management of its multi-site, double blind colorectal cancer study. 

The China Food and Drug Administration (CFDA) has been driving reforms to improve the quality of drugs and to encourage innovation in the development of new drugs and medical devices. Kelun Pharmaceutical has been successfully conducting generics drug research since 1996 but recently launched its first, new oncology drug study. As with any new clinical trial, there is tremendous time and resources required to set up a new study including trial design and validation which can take weeks to months. ORS enables coordinators to quickly add patients to a trial, collect screening information and ensure eligibility for randomization in record time. In an effort to set up their own trial and eliminate manual, repetitive data entry, Kelun implemented Clinical One Randomization and Supplies Management Cloud Service.  

“We needed an established, standards-based cloud system to simplify the drug supply management process, and we have already determined we made a great choice by selecting Oracle’s Clinical One Randomization and Supplies Management. Within 29 days, we were fully implemented and are now in phase III of our oncology trial,” said Xiaoping Zhang, VP of Biostatistics and Data Management, Clinical Research Hub.

With Clinical One Randomization and Supplies Management, Kelun has been able to manage its trial supply and randomization more effectively and efficiently and has enabled its CRO partner and service providers, including its supply shipping specialist vendor, to automate their processes by leveraging Oracle Health Science’s cloud-based system. In addition to simplifying the drug supply management process, ORS has provided Kelun with role-based analytics to track and manage supply status in real time.

“Our eClinical platform, Clinical One and randomization and supplies management capability was designed for pharmaceutical and biopharma companies of all sizes, across all treatment areas and for all phases of research and development. We are excited to see Kelun leveraging technology to speed the setup and management of their trial and are hopeful that this oncology therapy will make it to market,” said Steve Rosenberg, general manager and senior vice president, Oracle Health Sciences.

Additional Resources

About Oracle
The Oracle Cloud offers complete SaaS application suites for ERP, HCM and CX, plus best-in-class database Platform as a Service (PaaS) and Infrastructure as a Service (IaaS) from data centers throughout the Americas, Europe and Asia. For more information about Oracle (NYSE:ORCL), please visit us at oracle.com

Trademarks
Oracle and Java are registered trademarks of Oracle and/or its affiliates. Other names may be trademarks of their respective owners.

Logo- https://ift.tt/2mPV2Zb

news POST

Purchase this article for republication.

Source link

Friday, May 11, 2018

Best Practices for Shipping Single-Use Systems

Image Courtesy of Sartorius Stedim BiotechAs single-use processing equipment becomes a more prominent part of biopharmaceutical development and manufacturing, a clear understanding of risk management and testing requirements are needed. Regulatory guidelines are not prescriptive, forcing manufacturers to develop clear strategies that will ensure that product safety and efficacy are maintained during shipment.

A thorough understanding of the distribution cycle and potential transportation risks is required. In this article, Elisabeth Vachette, senior product manager, and Jean-Marc Cappia, vice-president of marketing, both at Sartorius Stedim Biotech FMT, Aubagne, France, share with Pharmaceutical Technology some of the key issues and challenges involved, and how to meet them effectively.

Regulatory issues

PharmTech:Which regulations and standards govern long-distance shipping of liquids in single-use systems? 

Vachette:Currently, there is no dedicated regulatory guidance on the subject. FDA, the European Medicines Agency (EMA), and other regulatory bodies require that companies have qualified processes and can prove that the process will meet the quality standards of the final drug product.

[Neither FDA nor EMA] provide very specific requirements. They want end users to be in control of their processes.  What they say is that the process of drug making should be qualified, whether that involves filtration, bag or virus inactivation, transportation, or storage space during manufacturing.

It is up to end users to establish documented evidence providing assurance that processes are under control and meeting specs and defined quality attributes (i.e., that the process is stable, robust, and free of any leakage or contamination).The Parenteral Drug Association’s (PDA) technical report TR66 recommends that shipping systems be qualified for intended use through “proper design and testing in consultation with a packaging engineer.”
Table I: [CLICK TABLE TO ENLARGE]Table II: [CLICK TABLE TO ENLARGE]

Conditions for international shipment must be defined. They can be based on international standards such as the American Society for Testing and Materials’ (ASTM) D4169 or the International Safe Transit Association (ISTA) 3 series (Tables I and II). The level of severity for test conditions must be based on real-world shipping conditions.  We recommend a holistic, four-step approach (Figure 1).


Figure 1. Four-step testing approach. [Figure courtesy of Sartorius Stedim Biotech.]

Technical challenges

PharmTech: What major technical challenges do pharma and biopharma companies face when shipping liquids in single-use systems over long distances by air or sea?

Cappia: The first and main challenge is preserving product integrity within the bag, and the robustness and integrity of the system. Any leaks or bacterial ingress must be prevented.

During shipping, bags can move, resulting in water hammer and stress. The challenge for the supplier is, first, to design systems that can pass these tests. Currently, single-use systems use better films and technologies than they did in the past, so the bags can more readily pass ASTM test requirements, which are very aggressive.

Once the integrity and robustness challenge is overcome, end users must monitor the shocks and temperature variations that the pharmaceutical product can experience when it is shipped to its destination.

Vachette: For liquid shipping, there is a need first to understand the distribution cycle, what is really happening, and what kind of shipping transportation means you are using. By knowing the distribution cycle, process validation will integrate severe conditions over the normal distribution cycle in order to provide correct and meaningful qualification. This approach allows end users to validate processes in worst-case conditions. 

ASTM’s testing involves worst-case conditions, and also requires monitoring that enables the complete traceability of product, which might include tracking temperature.

The responsibility is to define the distribution cycle. End users select a supplier of transport services and a means of transporting the product (i.e., dedicated shipment, during which product is passed in a controlled way).

For exceptions, they will use a dedicated truck with cold-chain management. The approach that is used depends on the transportation supplier, although our company offers contract supply chain management and testing services for this type of process validation.
 

Outsourcing testing

PharmTech: Which aspects of testing and data collection can be outsourced for single-use shipping?

Cappia: We have qualified on ASTM standards and can supply technical data to provide a view of the regime and the constraints and stresses we have applied during testing, in terms of vibrations, shocks, and shakes. 

In addition, when manufacturers are simulating shipping conditions, they can send us the bags to check for integrity. We can either make a pressure or ink tests on the bag, or perform bacterial ingress tests on the bags. We offer these services to support customer validation, since they must simulate shipping conditions and ensure that results are within the proper framework.

The industry’s understanding of shipping requirements varies

PharmTech: Are most biopharmaceutical companies already aware of what they need to do to validate single-use shipping for liquid products, or are they leaving any vital steps out of the plannint process?

Vachette:  The level of understanding varies. Some companies, for example, the large biotech companies, are very well prepared and understand exactly what they need to include in their validation efforts.  But that is not the case for all of the smaller and mid-sized manufacturers. As a vendor, we believe that we can play a role in educating manufacturers on what is required, and providing the testing services themselves if and when needed. 

Article Details

Pharmaceutical Technology
Vol. 42, No. 5
May 2018
Pages: 55–57, 61

Citation

When referring to this article, please cite it as A. Shanley, “Best Practices for Shipping Single-Use Systems,” Pharmaceutical Technology 42 (5) 2018.

Source link

Two day Process Validation Guidance Requirements Workshop: FDA and EU Annex 15 – Qualifications and Validation (Boston, United States – July 7th-8th, 2018) – ResearchAndMarkets.com | Technology

The Process Validation Guidelines (January 2011) and the EU Annex 15: Qualification and Validation (October 2015) outline the general principles and approaches the two regulatory bodies consider appropriate elements of process validation for the manufacture of human and animal drugs and biological products, including Active Pharmaceutical Ingredients (APIs).

These guidances align Process Validation activities with a product lifecycle concept and with existing FDA and EU guidances, including the FDA/International Conference on Harmonization (ICH), Guidance for Industry, Q8 (R2) Pharmaceutical Development, Q9 Quality Risk Management, and Q10 Pharmaceutical Quality System.

The lifecycle concept, new to these Guidances, link product and process development, qualification of the commercial manufacturing process, and maintenance of the process in a state of control during routine commercial production. These guidances also support process improvement and innovation through sound science and risk management.

The new Process Validation Guideline/Practice incorporate elements of Process Validation as early as the Research and Development phase, and continues onward through Technology Transfer, into the Phase 1 IND Clinical Trial manufacturing phase, and ultimately into Phase 2 and 3, and then commercial manufacturing.

Each facility, whether producing small or large molecules requires both an overall Site Validation Plan as well as specific validation plans to manage the multiplicity of validations required to confirm the successful manufacture of each of its products.

This two day, interactive Seminar which provides a conduit to enhance your understanding of the Continued Process Verification, will be reviewed in detail: where does it begin; what is included; and, when does it end.

What FDA segments are included and excluded within the “NEW” Process Validation.Where does the Process Validation commence.What are the Three Stages and Where DThey Apply within the NEW Process Validation.How Stage 1 integrates with Phase 1.The Validation approaches that are included within this Guidance document.The Statutory and Regulatory Requirements for Process Validation.An Introduction tPhase 1 Guidance for Industry and Its Application within the “NEW” Process Validation.The Phase 1 Investigational Drug Requirements — What is and What is NOT Required.General Considerations for Process Validation – Stage 2 Process Qualification.Regulatory Strategies for Phase 2 and 3 and their Incorporation within Stages 1 and 2.General Considerations for Process Validation – Stage 3 Continued Process Verification.A Review of EU Annex 15 and its Comparison to FDA’s Process Validation Guidance.

Laura Wood, Senior Manager

For E.S.T Office Hours Call 1-917-300-0470

For U.S./CAN Toll Free Call 1-800-526-8630

For GMT Office Hours Call +353-1-416-8900

Related Topics:Pharmaceutical Manufacturing

INDUSTRY KEYWORD: HEALTH PHARMACEUTICAL

SOURCE: Research and Markets

Copyright Business Wire 2018.

PUB: 05/10/2018 12:23 PM/DISC: 05/10/2018 12:23 PM

Copyright Business Wire 2018.



Source link

Thursday, May 10, 2018

Two day Process Validation Guidance Requirements Workshop: FDA and EU Annex 15 – Qualifications and Validation (Boston, United States – July 7th-8th, 2018) – ResearchAndMarkets.com | Business

DUBLIN–(BUSINESS WIRE)–May 10, 2018–The “Process Validation Guidance Requirements (FDA and EU Annex 15: Qualifications and Validation): 2-Day Workshop ” conference has been added to ResearchAndMarkets.com’s offering.

The Process Validation Guidelines (January 2011) and the EU Annex 15: Qualification and Validation (October 2015) outline the general principles and approaches the two regulatory bodies consider appropriate elements of process validation for the manufacture of human and animal drugs and biological products, including Active Pharmaceutical Ingredients (APIs).

These guidances align Process Validation activities with a product lifecycle concept and with existing FDA and EU guidances, including the FDA/International Conference on Harmonization (ICH), Guidance for Industry, Q8 (R2) Pharmaceutical Development, Q9 Quality Risk Management, and Q10 Pharmaceutical Quality System.

The lifecycle concept, new to these Guidances, link product and process development, qualification of the commercial manufacturing process, and maintenance of the process in a state of control during routine commercial production. These guidances also support process improvement and innovation through sound science and risk management.

The new Process Validation Guideline/Practice incorporate elements of Process Validation as early as the Research and Development phase, and continues onward through Technology Transfer, into the Phase 1 IND Clinical Trial manufacturing phase, and ultimately into Phase 2 and 3, and then commercial manufacturing.

Each facility, whether producing small or large molecules requires both an overall Site Validation Plan as well as specific validation plans to manage the multiplicity of validations required to confirm the successful manufacture of each of its products.

This two day, interactive Seminar which provides a conduit to enhance your understanding of the Continued Process Verification, will be reviewed in detail: where does it begin; what is included; and, when does it end.

Learning Objectives:

What FDA segments are included and excluded within the “NEW” Process Validation.Where does the Process Validation commence.What are the Three Stages and Where DThey Apply within the NEW Process Validation.How Stage 1 integrates with Phase 1.The Validation approaches that are included within this Guidance document.The Statutory and Regulatory Requirements for Process Validation.An Introduction tPhase 1 Guidance for Industry and Its Application within the “NEW” Process Validation.The Phase 1 Investigational Drug Requirements — What is and What is NOT Required.General Considerations for Process Validation – Stage 2 Process Qualification.Regulatory Strategies for Phase 2 and 3 and their Incorporation within Stages 1 and 2.General Considerations for Process Validation – Stage 3 Continued Process Verification.A Review of EU Annex 15 and its Comparison to FDA’s Process Validation Guidance.

For more information about this conference visit https://www.researchandmarkets.com/research/22rbv4/two—day—process?w=4

View source version on businesswire.com:https://ift.tt/2rzGPAN

CONTACT: ResearchAndMarkets.com

Laura Wood, Senior Manager

press@researchandmarkets.com

For E.S.T Office Hours Call 1-917-300-0470

For U.S./CAN Toll Free Call 1-800-526-8630

For GMT Office Hours Call +353-1-416-8900

Related Topics:Pharmaceutical Manufacturing

KEYWORD:

INDUSTRY KEYWORD: HEALTH PHARMACEUTICAL

SOURCE: Research and Markets

Copyright Business Wire 2018.

PUB: 05/10/2018 12:23 PM/DISC: 05/10/2018 12:23 PM

https://ift.tt/2I7uTBF

Copyright Business Wire 2018.

Source link

MetroWest Business Digest for May 10, 2018 – News – MetroWest Daily News, Framingham, MA

Information Security Summit slated for May 24

MassBay Community College and Towerwall will hold the sixth annual Information Security Summit from 7:30 a.m. to 1:30 p.m. May 24 at the MassBay Wellesley Hills campus, 50 Oakland St. Attendees will learn from industry experts as they share their experience and knowledge regarding guiding principles of information security, user awareness, training/social engineering, cloud and security, threats and ransomware, risk management and compliance, enabling the summit participants to connect through the leaders driving innovation in the security sector. This year’s keynote address will be given by Bob Bragdon, senior vice president and publisher of CSO, the leading information resource for security, risk and privacy executives. Bragdon leads all operations for the full CSO product line, including http://CSOonline.com, the CSO portfolio of national and regional events and the Security Smart Newsletter. During his keynote, he will be discussing “Building a secure business: from culture to cloud”. Preregistration is required and a $45 registration fee does apply. To register: https://bit.ly/2FWjoqK. For information: http://massbay.edu/iss.

Great Elm Capital Corp.announce stockholder approval

Great Elm Capital Corp., of Waltham, an externally managed, business development company focused on investing in debt instruments of leveraged middle market issuers, recently announced that a majority of the stockholders of the company approved the application of the modified minimum asset coverage requirements set forth in Section 61(a)(2) of the Investment Company Act of 1940, as amended, in accordance with the Small Business Credit Availability Act (“SBCAA”) that was signed into law on March 23. As a result of such approval, and subject to satisfying certain ongoing disclosure requirements under the SBCAA, effective May 4, the asset coverage ratio test applicable to the Company has been decreased from 200 percent to 150 percent, permitting the company to incur additional leverage.

Technical Communications Corporation announces results

Technical Communications Corporation, of Concord, announced its results for the three and six month periods ended March 31. For the three months ended March 31, the company reported a net loss of $313,000, or $0.17 per share, on revenue of $930,000, compared to net income of $128,000, or $0.07 per share, on revenue of $1,385,000 for the quarter ended April 1, 2017. For the six months ended March 31, the company reported a net loss of $365,000, or $0.20 per share, on revenue of $2,046,000, compared to a net loss of $567,000, or $0.31 per share, on revenue of $2,017,000 for the six months ended April 1, 2017.

Proteon Therapeutics announces contract extension

Proteon Therapeutics, of Waltham, a company developing novel, first-in-class therapeutics to address the medical needs of patients with kidney and vascular diseases, recently announced a long-term contract extension with Lonza Pharma & Biotech for the commercial supply of investigational vonapanitase’s active pharmaceutical ingredient. Lonza has manufactured API for Proteon at its microbial manufacturing facility in Visp since 2009. Initially, a small-scale process was transferred into Lonza’s development labs for process optimization and consistency studies. The process was then scaled up to 1,000L scale cGMP manufacture to support Proteon’s early clinical studies and potential commercial requirements. As Proteon worked to complete enrollment in its ongoing phase three clinical trial, PATENCY-2, Lonza supported Proteon with three process validation batches at 1,000L commercial scale, each of which met the intended release criteria. If PATENCY-2 is successful, Proteon expects to include results from these validation runs in a potential Biologics License Application filing in the second half of 2019, which Lonza will support.

Source link

Porvair offers validation services for pharmaceutical industry

Establishing Acceptance Limits for Uniformity of Dosage Units: Part 3

The working acceptance limits for acceptance values (AV) are determined using the critical values at, for example, 95% coverage over the corresponding AV distributions. However, validity of such limits needs to be elaborated.

Yada/shutterstock.comPart one of this article introduced the concept of sampling distribution of acceptance value (AV) in uniformity of dosage units (UDU) (1). With different sample sizes such as n= 10 and 30, their AV distributions will be different, resulting in different critical AV values (i.e., the values at the locations covering 95% of the distributions that are equal to, for example, 12.5 and 9.1 for n = 10 and 30, respectively). Such critical values will be employed as AV working limits rather than using the single compendial limit of not more than (NMT) 15 (2). 

Part two of this article described how to establish the corresponding acceptance limits for AV data for process validation batches as well as the typical characteristics of AV distributions. 

Click here to view a PDF of this article.

Peer-Reviewed

Submitted: February 22, 2018
Accepted: March 27, 2018

About the Author

Pramote Cholayudth is validation consultant to Biolab Co., Ltd. in Thailand. He is the founder and manager of PM Consult, [email protected].

Article Details

Pharmaceutical Technology
Vol. 42, No. 5
May 2018
Pages: 34–44

Citation

When referring to this article, please cite it as P. Cholayudth, “Establishing Acceptance Limits for Uniformity of Dosage Units: Part 3,” Pharmaceutical Technology 42 (5) 2018. 

Source link

Wednesday, May 9, 2018

Growing Adoption of Open Innovation Models in Pharmaceutical and Biotechnology Companies

Dublin, May 09, 2018 (GLOBE NEWSWIRE) — The “High Throughput Screening Market by Technology, Application, Product, End User – Global Forecast to 2023” report has been added to ResearchAndMarkets.com’s offering.

The global high-throughput screening (HTS) market is projected to reach USD 21.69 Billion by 2023 from USD 14.87 Billion in 2018, at a CAGR of 7.8%.

The major factors driving the growth of the HTS market include initiatives undertaken by pharmaceutical and biotechnology companies, increasing R&D spending, technological advancements in HTS, and the availability of government funding and venture capital investments.

The report analyzes the global HTS market by product & service, technology, application, end user, and region. On the basis of product & service, the reagents & assay kits segment accounted for the largest share of the global HTS market in 2017. Factors such as the large numbers of reagents and assay kits used in HTS techniques, rising prevalence of a number of diseases, increasing pharmaceutical R&D, and increased government funding for life science research are driving the growth of this segments.

Based on technology, the label-free technology segment is expected to grow at the highest CAGR during the forecast period. The major advantage offered by label-free technology is that it can help in the study of varied cell types and targets. Label-free assays also provide simple methods for studying complex biological pathways. The label-free technology is set to reduce drug failure caused by toxicity. These factors are likely to boost the growth of this technology market.

On the basis of application, the target identification & validation segment accounted for the largest share of the global HTS market in 2017. The large share of this segment is attributed to the growing number of potential drug targets for screening.

Based on end user, the pharmaceutical and biotechnology companies accounted for the largest share of the global HTS market in 2017. Factors contributing to its largest share include the increasing use of HTS techniques by pharmaceutical and biotechnology companies for drug discovery applications along with the increasing pharmaceutical R&D expenditure.

Geographically, the global HTS market is segmented into North America, Europe, Asia Pacific, Latin America, and the Middle East & Africa. In 2017, North America accounted for the largest share of the HTS market, followed by Europe and Asia Pacific. Factors such as the large spending on pharmaceutical R&D, growing adoption of HTS, availability of government funding, and the presence of major key players in the region are responsible for the large share of the North American HTS market.

The prominent players in the global HTS market are Agilent (US), Danaher (US), Thermo Fisher Scientific (US), PerkinElmer (US), Tecan (Switzerland), Axxam (Italy), Merck Group (Germany), Bio-Rad (US), Hamilton (US), Corning (US), BioTek (US), and Aurora Biomed (Canada).

Key Topics Covered:

1 Introduction

2 Research Methodology

3 Executive Summary

4 Premium Insights
4.1 High-Throughput Screening: Market Overview
4.2 HTS Market: Developed vs Developing Countries (2018 vs 2023)
4.3 Geographic Snapshot: HTS Market (2017)
4.4 Geographic Mix: HTS Market
4.5 HTS Market, By Product & Services (2018 vs 2023)

5 Market Overview
5.1 Introduction
5.2 Market Dynamics
5.2.1 Market Drivers
5.2.1.1 Growing Adoption of Open Innovation Models in Pharmaceutical and Biotechnology Companies
5.2.1.2 Government Funding and Venture Capital Investments
5.2.1.3 Increasing R&D Spending
5.2.1.4 Technological Advancements
5.2.2 Market Restraints
5.2.2.1 Capital-Intensive Nature of HTS
5.2.2.2 Complexities in the Field of Assay Development
5.2.3 Market Opportunities
5.2.3.1 Emerging Markets
5.2.3.2 Growing Research Activities in Toxicology and Stem Cells
5.2.4 Market Challenges
5.2.4.1 Dearth of Skilled Operators

6 Industry Insights
6.1 Introduction
6.2 Technological Trends
6.2.1 Label-Free Technology
6.2.2 Automation & Miniaturization
6.2.3 Microfluidics
6.3 HTS in Drug Discovery Process

7 High-Throughput Screening Market, By Product & Service
7.1 Introduction
7.2 Reagents & Assay Kits
7.3 Instruments
7.4 Consumables & Accessories
7.5 Software
7.6 Services

8 High-Throughput Screening Market, By Technology
8.1 Introduction
8.2 Cell-Based Assays
8.2.1 2D Cell Culture
8.2.2 3D Cell Culture
8.2.2.1 Scaffold-Based Technology
8.2.2.1.1 Hydrogels
8.2.2.1.1.1 Animal-Derived Hydrogels
8.2.2.1.1.1.1 Matrigel
8.2.2.1.1.1.2 Collagen
8.2.2.1.1.2 Synthetic Hydrogels
8.2.2.1.1.3 Alginate/Agarose
8.2.2.1.2 Inert Matrix/Solid Scaffolds
8.2.2.1.3 Micropatterned Surfaces
8.2.2.2 Scaffold-Free Technology
8.2.2.2.1 Microplates
8.2.2.2.2 Hanging-Drop Plates
8.2.2.2.3 Ultra-Low Binding Plates
8.2.2.2.4 Other Scaffold-Free Technologies
8.2.3 Perfusion Cell Culture
8.3 Lab-On-A-Chip
8.4 Ultra-High-Throughput Screening
8.5 Bioinformatics
8.6 Label-Free Technology

9 High-Throughput Screening Market, By Application
9.1 Introduction
9.2 Target Identification and Validation
9.3 Primary and Secondary Screening
9.4 Toxicology Assessment
9.5 Other Applications

10 High-Throughput Screening Market, By End User
10.1 Introduction
10.2 Pharmaceutical and Biotechnology Companies
10.3 Academic and Government Institutes
10.4 Contract Research Organizations (CRO)
10.5 Other End Users

11 High-Throughput Screening Market, By Region

12 Competitive Landscape
12.1 Introduction
12.2 Market Leadership Analysis
12.3 Competitive Scenario
12.3.1 Product Launches and Upgrades
12.3.2 Partnerships, Collaborations, and Agreements
12.3.3 Expansions
12.3.4 Acquisitions
12.3.5 Other Developments

13 Company Profiles

  • Agilent Technologies, Inc.
  • Aurora Biomed
  • Axxam S.P.A.
  • Bio-Rad Laboratories
  • Biotek Instruments
  • Corning Incorporated
  • Danaher Corporation
  • Hamilton Company
  • Merck Group
  • Perkinelmer, Inc.
  • Tecan Group
  • Thermo Fisher Scientific Inc.

For more information about this report visit https://ift.tt/2K52CIw

CONTACT: ResearchAndMarkets.com
Laura Wood, Senior Manager
press@researchandmarkets.com
For E.S.T Office Hours Call 1-917-300-0470
For U.S./CAN Toll Free Call 1-800-526-8630
For GMT Office Hours Call +353-1-416-8900
Related Topics: Drug Discovery

Source link

Deploying the Cloud in GxP Environments

Meeting the stringent cloud compliance and regulatory requirements in pharma

 

The traditional IT infrastructure for most life sciences organizations was not designed to meet the business challenges that companies are faced with today. It can take significant, sustained, and hugely disruptive investment in new technologies and infrastructure to bring internal systems to the required security, performance, and compliance level. At the same time, a life sciences company must do much more than maintain “business as usual.” It must reduce costs and increase productivity and innovation against a backdrop of continually changing market pressures and regulatory requirements. This is the reason that we’re seeing greater cloud adoption in other parts of the life sciences business. However, good practice quality guidelines (GxP) environments have their own unique requirements. There are very strict guidelines around application and system usage in key business functions, such as research and development, clinical trials, quality, and manufacturing, set by the FDA and other global regulators. This article looks at the cloud deployment models available for GxP environments and how to select the right one for a pharmaceutical company’s cost constraints and regulatory profile.

Three types of cloud service

The strengths and weaknesses of internal IT deployments are similar across industries. They are, however, exacerbated in the regulatory environment. A large life sciences company can have thousands of different IT architecture combinations and a large proportion of its overall IT budget is taken up with simply operating, maintaining, and supporting these existing systems. More importantly, the result can often be a lack of agility, if it takes IT too long to respond to changing business requirements. With the additional compliance constraints, it can take many months to deploy a new module or just add extra computing or storage capacity. In addition, users are often faced with slow and inefficient legacy systems and, worse, much of their data remains under-utilized, due to its storage in inaccessible silos throughout the organization.

Cloud services can help overcome many of the drawbacks of existing internal systems. There are infinite combinations of cloud deployments, however; generally, the following delivery types can enable a company to decide which elements of its IT infrastructure to continue to operate internally and which to have executed by a cloud service provider.

  • Infrastructure as a service (IaaS). IaaS provides a service to establish and run virtualized computer resources over the internet. Virtualization is the creation of virtual—rather than actual—versions of IT infrastructure, such as operating systems, servers, or storage devices. The services provider is responsible for managing and delivering hardware, storage, servers, and data center space that form the foundation of a cloud environment. 
  • Platform as a service (PaaS). PaaS is a cloud computing service that provides all the platform—hardware, middleware, and operating system—components needed for a company to develop, run, and manage applications. The cloud technology provider takes care of all the infrastructure while the pharma company manages its own application portfolio.
  • Application as a service (AaaS). Also known as software as a service, AaaS provides a completely hosted—and managed if required—IT package. The provider makes applications available to the company over the internet via a thin client PC.

 

Four types of cloud deployment 

Before looking at the four cloud deployment models, it’s worth considering the characteristics that all cloud services have in common. Using the internet allows many companies to connect securely to the same service, enabling collaboration and information sharing. Companies using the cloud service have access to shared resources that are continually improving so that they should always have access to the latest and best performing systems. With some cloud service providers, the service is delivered on-demand. Life sciences companies access the service as required and usage can be metered or architected in such a way that they only pay for what they use.

A major benefit of the cloud is its virtually limitless scalability and geographic agnosticism—that can be applied extremely quickly to meet demand. One life sciences company found that it would require 250 internal servers to meet peak processing times during certain phases of global clinical trials. This meant waiting for internal resource to be freed up, and as the project was estimated to cost $150 per second, that was a very costly delay.1 Switching to a cloud service meant that the company not only could meet its computing requirements quickly, but it could scale up for peak processing and scale down afterwards—only paying for the resource they used.

Further qualifying the virtualization tools themselves can greatly reduce qualification time, especially in the scenario where the underlying specifications of the servers are identical, allowing the rapid deployment of pre-qualified server packages.

The cloud deployment models available allow a company to access the benefits of cloud computing while ensuring that its working within the performance, security, and risk levels of the organization’s requirements.

Hosted public internet

A public cloud is a publicly accessible cloud environment owned by a third-party cloud service provider (CSP). Services are provisioned in a multi-tenant environment where many customers are using the same service. The infrastructure may be hosted on the premises of the service provider, a third-party data center, or, possibly, multiple third-party facilities and, further, may reside on equipment owned or leased by the CSP. It is vital before engaging with such a provider that a pharma company fully understands its provider’s architecture, the layers of service-level agreements (SLAs), and the relationships between all of the delivery partners. Ultimately, though, the environment will be operated by whoever is making use of it, be it life sciences companies, government organizations, or academic institutions. 

The service is delivered across the public internet and accessed via thin clients at the customer site. The main features of hosted public cloud include:

  • Fast and easy deployment of standardized solutions.
  • Easy to connect and collaborate with external customers, partners, and suppliers.
  • Complete management and support of IT infrastructure.
  • System performance and continuity guaranteed under SLA.
  • Reasonable levels of security.
  • Lack of auditability—while most public cloud providers will offer standard third-party audited accreditations, such as ISO27001 or SOC 2, they will not generally permit traditional GxP audits.

While companies have access to the latest web security standards, the hosted public cloud will not deliver the highest levels of security possible and is likely not to be up to the companies’ requirements if this is a foremost concern. 

In addition, the cloud provider is responsible for the creation and ongoing maintenance of the public cloud and its IT resources. It is more difficult to control patching and upgrade frequency and it is likely that the user will have little-to-no transparency over what happens below the operating system. 

Where application and infrastructure qualification and validation assurance is essential, a pharma company will need to find ways of working with the cloud provider to gain all the information it needs to meet the organization’s compliance requirements. Appendix 11 of the ISPE GAMP Good Practice Guide for IT Infrastructure Control and Compliance2 provides strategies for qualifying the suppliers for each of the different engagement types.

 

Hosted private network

A private cloud, as the name suggests, is solely owned by the cloud service provider. Deployed internally or externally, a hosted private network offers high levels of security using the provider’s private cloud and delivers data management and business continuity services. It is the ideal choice for organizations that need to manage their host applications and other applications used by their customers. The main features of a hosted private network are:

  • Ability to retain existing IT system customizations.
  • Flexibility to modify systems as required.
  • Flexibility on the control of upgrade and patch frequency.
  • Maximum levels of reliability and scalability.
  • Maximum levels of security.
  • Greater control over cloud infrastructure.
  • Typically running on dedicated hardware (though private clouds can be virtualized).

There isn’t a great deal of difference in the design structure between hosted public cloud and hosted private network. The biggest difference for the latter is that the provider is, effectively, delivering a single tenant service over a multi-tenant architecture. It is essential that the provider can prove complete customer and data isolation—that a company’s applications and data are completely isolated from that of any other customer using the provider’s services. As such, the security, performance, and compliance benefits of the private model will come at an increased cost.

Hybrid cloud

A hybrid cloud contains the best parts of the hosted public cloud and hosted private network models. In a hybrid cloud deployment, the cloud environment is comprised of two or more different cloud deployment models. For example, one may choose to deploy cloud services processing sensitive data to a private cloud and other, less-sensitive cloud services to a public cloud. A hybrid cloud delivers superior data management, security, scalability, and performance, but adds complexity in terms of management and reliability due to the diverse configurations that this model can create. The hybrid model potentially provides the best opportunity of balance for a GxP-regulated entity; higher-risk GxP applications and services can reside in a qualified cloistered environment, while non-GxP applications can exist outside of the more constrictive GxP control set. The main features of hybrid cloud are:

  • Ability to deploy primary solution on premise.
  • Ability to retain existing IT system customizations.
  • Flexibility to modify systems as required.
  • Flexibility on the control of upgrade and patch frequency.
  • Flexibility to deploy business continuity and disaster recovery capabilities externally.
  • High levels of reliability and scalability.
  • High levels of security.
  • Greater control over cloud infrastructure.

Hybrid cloud deployments can be complex and challenging to create and maintain due to the potential disparity in cloud environments. Life sciences companies need to work closely with the cloud service provider to know exactly who is responsible for managing every element of the IT infrastructure. Where qualification and validation is important, the cloud service provider must be able to demonstrate and record that all its activities meet a company’s GxP compliance requirements. 

On-premise cloud

Where security and control are paramount concerns, on-premise cloud deployments are preferred. In this model, all IT infrastructure remains within the organization. With on-premise cloud, a company uses cloud computing technology as a means of centralizing access to IT resources by different parts, locations, or departments of the organization. 

Even though the cloud infrastructure physically resides on the company’s premises, the IT resources it hosts are still considered “cloud-based,” as they are made remotely accessible via the cloud to both internal and external users. The service provider delivers the level of management and maintenance skills the pharma customer requires to operate the system. The main features of on-premise cloud are:

  • Ability to qualify the data center infrastructure, cloud stack, and virtualized architectures.
  • Ability to remain using existing hardware.
  • Ability to maintain system on-premise.
  • Ability to retain existing IT system customizations.
  • Flexibility to modify systems as required.
  • Flexibility on the control of upgrade and patch frequency.
  • Ability to use provider to flexibly resource IT infrastructure.
  • Maximum levels of security.
  • Maximum control over cloud infrastructure.

From the standpoints of data integrity, security, and software validation, on-premise cloud represents an attractive option. However, it does have drawbacks. Unsurprisingly, this cloud type suffers from some of the key weaknesses of internal IT systems. Key among these is the potential lack of scalability. A company is still bounded by the capabilities of its existing servers and can’t take advantage of the unlimited potential to quickly and securely scale computing capacity as business requires.

Further, with a hardware refresh rate of three to five years, and the internal costs of managing the solution and any associated regulated expectations, this deployment type can soon exceed the perceived value of an on-premise architecture.

 

The regulatory paradox

To meet the criteria for computing in a GxP environment, software applications have to be carefully validated and other IT infrastructure components—data center facilities, network components, and infrastructure software and tools—needed to be properly qualified. The life sciences industry had become very comfortable with using the GAMP 5 for the validation of applications. Until recently, similar guidance for cloud deployments was in short supply, but the International Society for Pharmaceutical Engineering (IPSE), the creator of GAMP 5, has addressed this with the publication of the GAMP Good Practice Guide: IT Infrastructure Control and Compliance rev 2.2 The guide directly addresses the vastly increased risk profile for cloud computing and provides a roadmap for transitioning from an internal self-managed relationship to a model for working with a qualified supplier, such as a CSP. 

The IPSE guidance for achieving compliance now places new emphasis on:

  • Supplier assessment and management.
  • Installation and operational qualification of infrastructure components (including facilities).
  • Configuration management and change control of infrastructure components and settings in a highly dynamic environment.
  • Management of risks to IT Infrastructure.
  • Involvement of service providers in critical IT Infrastructure processes.
  • SLAs with XaaS (i.e., IaaS, PaaS, SaaS) providers and third-party data center providers.
  • Security management in relation to access controls, availability of services, and data integrity.
  • Data storage, and in relation to this, security, confidentiality, and privacy.
  • Backup, restore, and disaster recovery.
  • Archiving.

This new guidance comes at a critical time, as regulatory pressure elsewhere in the business are likely to encourage life sciences companies to investigate cloud services. A slew of recent and forthcoming regulations across the European Union (EU) place an emphasis on information sharing and improved data management. The EU General Data Protection Regulation (GDPR), which deals with the management of personal information; the ISO Identification of Medicinal Products (IDMP), which involves improving information sharing and reporting of medicinal products; and the EU Clinical Trials Regulation (CTR) will affect every company that sells, markets, or works in Europe.

In all three cases, the regulations require enterprise-level of control and visibility of data within an organization—and, in some cases, its suppliers, partners, and customers. It involves bringing together different data in different formats from different parts of the business. In many cases, existing legacy systems will labor to meet performance, security, and transparency requirements to comply with these regulations. The scalability, reliability, and proven security capabilities of the cloud make it an increasingly attractive option. 

What to expect from a cloud provider

Delivering cloud services into a regulated environment places extra responsibility on service providers. Often, as the GAMP Cloud Special Interest Group has pointed out, this will involve them being willing to adapt their business model, as “it involves even greater movement of control toward the supplier, but still leaves the responsibility for the data and process within the regulated company. …The compliance concerns are just as valid, on infrastructure, platform, and application level, with little or nothing that we as life sciences companies can influence with regard to the provider’s management processes.”3

While true, many service providers have made significant efforts to tailor their service to meet GxP requirements. In addition to meeting all the latest cloud standards, such as SSAE and ISO 27001, some deliver against qualification standards and include validation packages that let a company take a risk-based approach to application development, delivery, and amendment. They will all provide the most stringent security, access, and change controls to meet the needs of regulated environments.

Where some providers differ is in their willingness or ability to deliver the level of audit rights and documented processes that life sciences companies require to meet their GxP compliance responsibilities. It is essential that companies are sure that the change control and documentation processes of the provider meet their requirements, especially within their qualification documentation practices.

Ready to go

The cloud is not an immature technology. Properly architected, built, and managed, it is a highly resilient, scalable, and secure platform that has been proven to successfully host mission-critical applications. More and more industries—even the US government—are quickly moving to adopt a “cloud-first” strategy. The GxP environment, like other regulated environments, has very stringent requirements and that has certainly slowed adoption. 

The lack of clear implementation guidance has been an issue. However, with the new IPSE guidance and a risk-based approach to cloud deployment backed by a cloud service provider whose services are designed for regulated environments, companies can now begin to benefit more fully from the cloud. Today, the cloud is better suited to deliver GxP-compliant services that will help life sciences organizations meet their key business challenges. As the GAMP Special Interest Group says: “We all know it’s the way to go.”3

 

Jaleel Shujath is Director, Life Sciences Strategy, at OpenTextStephen Ferrell is a Partner at Promedim Ltd.

 

References

1. https://ift.tt/2jKZIgA

2. ISPE, GAMP Good Practice Guide: IT Infrastructure Control and Compliance (Second Edition), 2017

3. https://ift.tt/2I8nmyr

Source link

Validation by Numbers | Pharmaceutical Technology


Validation by Design, Lynn Torbeck, PDA Books, Bethesda, MD, 2010, 200 pp., ISBN: 193372238X

How many samples should I take? Is the assay method validated? Is this result really out of specification? Should I adjust the tablet weight? Is this process under control? Should I reject the batch? The pharmaceutical industry has always faced questions such as these, and they may be answered best from a statistical perspective.

Unfortunately, many quality-assurance workers and production managers charged with answering these types of questions do not have an adequate working knowledge of statistics. They find it difficult to answer these questions and to understand answers that statisticians provide.

The book Validation By Design: The Statistical Handbook for Pharmaceutical Process Validation, by Lynn Torbeck, a member of Pharmaceutical Technology‘s Editorial Advisory Board, contains information useful to people who are new to statistics and to employees responsible for implementing statistical techniques that monitor and control pharmaceutical production processes and quality-assurance activities. The book was written specifically to address the statistical issues contained in the US Food and Drug Administration’s November 2008 draft guidance for industry titled Process Validation: General Principles and Practices, which is reproduced in its entirety in an appendix. Torbeck’s book can form the basis for interdepartmental discussions and for an understanding of the statistical techniques that are consistent with the intent of the guidance.

An important feature of the book is its interpretation of the guidance’s statistical implications. The author rewrote the guidance’s statistical content as a series of self-audit questions that cite specific lines in the guidance document. For example, lines 27–29 of the guidance state, “The lifecycle concept links the product and process development, qualification of the commercial manufacturing process, and the maintenance of the process in a state of control during routine commercial production.” The associated self-audit question asks, “Is the process in a state of control during routine commercial production?” The question is followed

by a brief explanation of the term “state of control” and a reference to the chapter of the book that describes the concept. This self-audit format leads the reader through the various statistical techniques that enable compliance with various sections of the guidance.

A second important feature of the book is the chapters that elucidate statistical methods and concepts. Each of these chapters is written in a standard format that contains subtopics such as “Other Names,” “Acronyms,” “Definition,” “Related Topics,” “Calculation,” “Illustration,” “Cautions,” “Advice,” and “References.” This consistent categorization enables the reader to understand the statistical concepts and decide whether their use is appropriate in a particular situation. The chapters explain simple statistical concepts such as average and relative standard deviation, as well as complicated ones such as control charts, root-cause analysis, process mapping, interquartile range, and Plackett–Burman designs.

The book should prove useful to employees charged with developing a self-audit program to measure the company’s level of compliance with the process-validation guidance. It also would be a solid basis for writing validation protocols.

The book cannot be considered an introductory text about statistics because the topics are not presented in depth. Still, its references provide necessary information for readers who wish to delve further into any of the subjects. According to the author’s preface, the book is for “those engaged in meeting the requirements of the FDA process-validation guidance.” The questions that interpret the guidance, the author’s explanations, and the chapters about statistical methods and techniques should do much to help personnel meet those requirements.


Russell Madsen is president of The Williamsburg Group, 18907 Lindenhouse Rd., Gaithersburg, MD 20879, tel. 301.938.4266, fax 301.869.5016, [email protected]
. He also is a member of Pharmaceutical Technology‘s Editorial Advisory board.

Source link

Pharmaceutical Validation Documentation Requirements

Pharmaceutical validation is a critical process that ensures that pharmaceutical products meet the desired quality standards and are safe fo...