Thursday, March 3, 2016

Effective Analytical Development and QC

More than one analytical laboratory within a company without clear definition of responsibilities and goals creates issues. This is particularly true if these labs are at different sites, but can also occur if they are within the same site. This can function satisfactorily if these different labs are part of one organization under the same head. However, without this each lab has its own responsibilities that are usually separate from the common goal.

Without a common head of the labs that these separate labs report to, the labs will typically go their own directions with regard to SOP's and how they plan to implement cGMP's. This includes instrument selection in order to facilitate method transfers and equivalency of testing. This is a case where one lab has all HPLC's while a newer lab starting up chooses to implement newer technology in purchasing UPLC's. One site may be more development oriented, while another is more production oriented. The lab that is more development oriented may have little interest in fully implementing cGMP's. Yet, when it comes to releasing clinical supplies, the development lab may be the only lab that has the necessary methods to test these products. Then the question comes up whether the appropriate standards have been implemented.

In the case of 2 labs conducting QC testing, there may be a difference in rounding and reporting results, particularly when it comes to impurities. These minor differences may not seem like much, but it is likely that if an auditor picks up on these differences the logical question is why are the 2 labs performing differently and which is right.

If it is possible for the different labs to come to an agreement on who will do what, how will cGMP's be implemented, which SOP's will be implemented (wording and language included depending on country), and what are the priorities?

It may, ultimately, make sense to have local laboratories with the R and D or production facilities. Responsibilities must, however, be clearly defined. It is unrealistic for different sites to constantly have to coordinate testing of products and materials. It is much better to have all of one type of testing at one site, a local manager at each site, with all analytical labs reporting into one Director.

Monday, June 9, 2014

MARK A. SCHREIBER, Ph.D.

650-739-9530

markschreiber01@gmail.com


Looking for new opportunities as:
Senior Director Analytical Chemistry
Accomplished in Analytical R&D, Quality Control, Pharmaceutical Development, Clinical Supplies, and Stability programs. Successfully led Analytical Development, CMC Development and QC teams.
Provided high quality CMC sections for IND's, NDA's, PMA's and supplements.
Strong technical abilities, leadership, flexibility, and a “get-it-done” attitude.
  • Thorough understanding of the drug development process, including all analytical support needed for initial drug substance through formulation development, including IND & NDA filing
  • Comprehensive knowledge of CMC regulatory and guidance documents
  • Goal-oriented with strong leadership capabilities.
  • Proven ability to work in unison with staff, management, and cross-functional teams – locally, nationally, and internationally.
  • Strong written and oral communication, and presentation skills
  • Highly flexible and versatile, managing change effectively
  • Directed staff of up to 35 chemists, including Ph.D.’s, managers, and junior staff
  • Successful interactions with FDA and notified body investigators and reviewers, on-site and at CDER
  • Publications and presentations (list available)


EXPERIENCE
Alexza Pharmaceuticals, Mountain View, CA
Director, Quality Control (2013-2014)
  • Lead Quality Control, Analytical Development, and Microbiology/Environmental Monitoring
  • 3 manager direct reports + 13 indirect reports, and growing
  • Transitioned the Quality Control Department from R&D to Commercial mindset
  • Improved turn-around time of testing
  • Provided backup strategies for all analytical equipment to ensure maximum “up time”
  • Close interactions with FDA during inspections
  • Increased overall compliance to be FDA inspection ready
  • Recruiting of additional staff to ensure timely turn-around times, even during peak periods
  • Work closely with a cross-functional management team, including other Directors, Sr. Directors, and VP's, trouble-shooting issues, change management, and ensuring sustainable strategic corporate growth

Biotronik AG, Buelach, Switzerland
Head, Analytical and Microbiology Labs (2009-2013)
  • Construction and start-up of new analytical labs (development and QC)
  • Evaluation of analytical and formulation processes previously established at Biotronik
  • Worked closely with process development team to improve manufacturing process
  • Recruitment of entire analytical staff at the Buelach site for QC and development
  • Identification and repair of short-falls in Analytical methods and validations
  • Development and validation of analytical methods used in development projects
  • Rapid scale-up of analytical testing to successfully launch Biotronik's first Drug-Eluting Stent
  • Established Stability Review Board to put policies in place, evaluate existing stability studies and plan future registration studies
  • In-process control and release testing of commercial products
  • Microbiology testing and validation of sterilization for numerous products, including cleanroom monitoring
  • Manage budget of 1.5 million Swiss Francs

Conor Medsystems, a Johnson & Johnson Company, Menlo Park, CA
Director, Analytical Development (2006-2008)
Provided strategic and tactical direction with regard to analytical chemistry and drug formulation to ensure compliance with US and international regulatory requirements being successful in securing regulatory approvals on a timely basis, while managing 3 functional units.
  • Achieved first-in-human testing of a new drug-eluting stent (DES) in 11 months
  • Completed the CMC section of a PMA submission for CoStar within 11 months of joining Conor by personally writing, as well as managing authorship by consultants
  • Developed and validated all analytical methods for Corio and SymBio ex-US clinical trials within 2 months of joining Conor, meeting a key milestone for the company
  • Managed capital and expense budgets ensuring resources were available meeting departmental and corporate goals
  • Recruited and hired more than 15 chemists and 2 managers to support the 3 functional units
  • On-going management of staff up to 35 with organizational changes/reductions
  • Led analytical methods development, validation and transfer
  • Led in-process testing, clinical lot release, and commercial lot release
  • Providing analytical expertise to Development projects teams
  • Developed systems and procedures for sample tracking, method development/validation/transfer, stability and OOS/OOT Investigations, improving efficiency and compliance
  • Championed and led the team that developed, validated, and implemented a Laboratory Information Management System (LIMS)
  • Audited and managed contract labs for raw material testing
  • Communicated frequently with senior management and multi-disciplinary teams
Berlex Laboratories, Wayne, NJ
Section Head/Manager (2000-2006)
  • Led Analytical and CMC teams to provide laboratory activities/data/reports to support IND/NDA filings and supplements
  • Led and participated with management team to continuously improve systems including: infra-structure, budget control, compliance, training, sample flow and testing improving compliance and efficiency
  • Led method validations and transfers meeting project objectives and regulatory requirements
  • Audited and recommended selection of firms for potential acquisitions
  • Developed strategies and managed stability program for development projects and marketed products
  • Led project team to develop an ANDA product and filing the ANDA to provide a revenue stream of $5-10 million for the dermatology business unit
DuPont Pharmaceuticals, Wilmington, DE
Principal Research Scientist/Group Leader (1991-2000)
  • Led methods development/validation/transfer
  • Provided critical analytical support for drug substance and formulation/process development, including process validation, clinical release and stability testing
  • Provided analytical expertise to Project teams and CMC sub-teams
  • Wrote analytical portion of CMC sections for regulatory filings with minimal regulatory comments
  • Led the improvement of lab operations processes, including: release testing, specification setting, stability, OOS/OOT procedures, sample flow, computerized systems
  • Hands-on management of team providing departmental support for FTIR, HPTLC, and Preparatory HPLC
PREVIOUS EXPERIENCE
DuPont Pharmaceuticals, Wilmington, DE
Research Chemist/Senior Research Chemist/Group Leader
  • Developed and validated Analytical methods (HPLC and GPC/SEC)
  • Technical support for marketed products requiring extensive interactions with contract manufacturers nationally and internationally
  • Automated sample preparation using robotics (Zymark)
  • Developed and Validated Software for chromatographic data analysis
  • Management of chromatographic data system



EDUCATION
Ph.D., Analytical Chemistry, University of Virginia
B.A. , Chemistry (ACS Certified), Franklin and Marshall College
Dale Carnegie Course Graduate
    PROFESSIONAL AFFILIATIONS
American Chemical Society
American Association of Pharmaceutical Scientists (including APQ Section and Stability Focus Group)
Pharmaceutical Stability Discussion Group

Publications List
Available upon request

Monday, January 25, 2010

Identity Testing

Identity testing should be performed on all raw material ingredients in a formulation, as well as the finished product. As part of release testing, there should be 2 identity tests performed, one of which is a definitive fundamental test. This fundamental test is most frequently an infra-red spectroscopy technique (FTIR or nIR), because of the ease of collecting the spectra (with a number of different sampling techniques) and the ability to perform a quantitative comparison to a library of IR spectra. This library can either be one commercially available or one that is compiled in-house. Near IR (nIR) will require development of a learning set of a significant number of samples (typically 20 or more) in order to provide a positive confirmation of the material in question. Ultraviolet (UV) spectroscopy or HPLC retention time are frequently used for the secondary identification technique.

For an Active Pharmaceutical Ingredient (API), FTIR and HPLC retention time are very frequently used. The acceptance criteria for these tests will often be a short list of specific peaks in the FTIR spectrum that must match the reference spectrum within a few (to be specified) wave-numbers (cm-1). From a qualitative perspective, it is also advisable to inspect the entire spectrum in comparison to the reference. HPLC retention times are usually specified within 5% difference from the reference peak in the chromatogram of a reference standard, although 2% should be easily achievable. It should be noted here that the reference standard needs to be injected in the same chromatographic run as the sample of interest. This is usually done during system suitability testing of the chromatographic system.

The USP spectroscopy testing for identity <197> specifies obtaining a spectrum of the reference standard at the same time as that of the sample. This is a somewhat outdated notion with today's modern FTIR spectrophotometers, where all the data are digital and there would be no significant peak shift occurring from a spectrum taken today versus one taken months or even years earlier and stored in the digital library. Therefore, it is my opinion that it is acceptable to compare the sample spectrum with one in the spectral library to confirm identity.

There are, of course, other identity tests that can be conducted, spectroscopic, wet chemical, and chromatographic to name a few. If the compound of interest is a salt, ICH Q6A requires that an identity test be conducted for the salt. If the material is compendial, the compendial identity test(s) should be performed. ICH Q6A also says that if a drug substance is developed "as a single enantiomer, the identity test(s) should be capable of distinguishing both enantiomers and the racemic mixture."

Monday, October 19, 2009

Drug Elution

Drug release/elution is a test that is needed for any solid oral dosage form (e.g. tablets and capsules) or for any controlled release product (e.g. an injectable containing liposomes), including combination products, such as drug-eluting stents. For immediate release products, it is necessary to demonstrate the most of the drug is released into solution in a relatively short time period, typically less than 1 hour, and that the product will do this reproducibly. For controlled-release or sustained-release product designed to release the drug over an extended period of time to achieve more uniform blood levels of the drug, to reduce the number of dosage units a patient has to take in a day, often resulting in better patient compliance and better clinical efficacy. Usually, USP (United States Phamacopeia) apparatus are used (found in USP monographs <711> and <724>). Apparatus 1 through 4 are described in USP <711> and apparatus 5 through 7 are described in USP <724>.

In the special case of elution of a drug from combination products, the elution may be over an extended period of time, days (antibiotics from hip/knee implants), weeks (drug-eluting stents), or even years (contraceptive devices). In these cases, the blood levels are so low that it is not practicle to measure these levels in order to determine an in-vivo release profile in humans. Often, representative elution media are used to achieve a suitable elution profile similar to that achieved in animal studies. This elution media may be biologically relevant, in the case of serum, or more likely in the case of accelerated methods will not have any biological relevance, using surfactants and possibly organic solvents. Typically, an accelerated method is needed due to the lack of stability of the drug in the media and the necessity to have a quality control method to be able to release the product. This method must be demonstrated to be discriminatory so that any product not having the proper elution charactistics will have a different elution profile and with specification limits appropriately
set (more to come on setting appropriate specifications) will fail the specifications and not be released. As with all methods, this method must also be validated for specificity, accuracy, precision, linearity, ruggedness, and robustness. Stability of the drug in the media must also be demonstrated, as well as the solubility of the drug. The solubility is particularly important, since many drugs have very low solubility especially in aquious media. "Sink conditions" must be demonstrated, that is that the highest concentration of the drug in the media does not exceed approximately 1/3 the solubility of the drug in that media.




Wednesday, August 12, 2009

Particle Size

Particle size is one test that the FDA requires for API (Active Pharmaceutical Ingredients), especially if this is in a solid oral dosage form. The particle size will affect the release properties of the drug from the product, since the surface area to volume of the particle will change in relation to the particle size. This may also be considered critical for true solutions, such as injectables, as the process may be affected by how fast the API particles dissolve in the solvent. There are a variety of techniques for measuring particle size, some of which are microscopic analysis, laser-light scattering (often using a Malvern particle sizer) of either dry powder or a liquid suspension, and light obscuration. The results are a statistical measure and are often reported as the particles size of 10%, 50%, and 90% of the particles to indicate the size distribution.

Particle sizing is dependent on not only the specific technique that is used to measure the particles, but also on the specific method used with that technique. Therefore, a specification must be developed using data from the technique and method that will be used to control particle size. If the particle size can be related to the rate of dissolution of the API, product, or API in the process, this will be strong supportive data for the specification.
Short courses in particle size analysis and dissolution testing are available from the American Chemical Society (www.chemistry.org) and the American Association of Pharmaceutical Scientists (http://www.aapspharmaceutica.com/).



Mark A. Schreiber, Ph.D.
http://www.linkedin.com/in/maschreiber

Wednesday, March 4, 2009

Spreadsheet for USP <905> Content Uniformity Calculation

At first glance, the acceptance criteria for the content uniformity calculation in USP chapter <905> seem somewhat daunting. These criteria are different from the old criteria which were based on performing a simple arithmetic mean and standard deviation calculation. Once one examines how the criteria are determined, it actually makes sense. If the mean deviates more than 1.5% from the target label claim of the product, this is seen as a significant change and the criteria for the permitted deviation of the individual units (using the standard deviation) become tighter. Ten (10) units are still analyzed in the initial testing and used for comparison with acceptance criteria. If outside of the acceptance criteria, an additional 20 units are tested and the calculations repeated.

I have gone through these calculations and acceptance criteria and created an spreadsheet for the general case. A view of this spreadsheet is shown below. If you email me, I will gladly sent this to you in Excel format. If you have online access to the USP-NF, sign on at
hhttp://www.uspnf.com/ and go to general chapter <905> Uniformity of Dosage Units. There is a good article in Pharm Tech on the probability of passing CU. http://pharmtech.findpharma.com/pharmtech/data/articlestandard//pharmtech/382004/122726/article.pdf
The equations are also included for plugging into Excel. I've done the work for you.






































http://www.linkedin.com/in/maschreiber
maschreib@att.net

Friday, February 27, 2009

Analytical Methods for Drug Product

Very often the HPLC methods (assay and impurities) developed for the API are directly transferable to use with the drug product, depending upon the complexity of the product. The methods to consider for the drug product are:
  • Identity
  • Appearance
  • Assay
  • Degradation products
  • Dissolution/Disintegration (for solid oral dosage forms)
  • Uniformity of content (content uniformity using USP <905)>
  • Residual solvents (if they are used in processing the dosage form)
The degradation products of drug products do not include the impurities from the API unless the degradation products of the product are the same as the impurities. In this case, the compounds need to be tracked.

Reporting of impurities/degradation products is a recurring topic of discussion between industry and the FDA. The FDA seems to always be pushing for lower and lower reporting limits. However, ICH Q3B clearly defines the reporting limits for deg products in drug product. This is dependent on the daily dosage and the reader is refered to the guidance for industry, http://www.fda.gov/cder/guidance/1317fnl.pdf




___
http://markaschreiber.blogspot.com/
http://www.linkedin.com/in/maschreiber
http://www.markaschreiber.com