Thursday, October 29, 2009

Got animal-derived materials? Part 2

By Ray Nims

As part of a formal animal-derived materials program, biopharma companies must assess the viral and TSE risk associated with materials derived from animals or which have been in contact with animal-derived materials at some point. We have addressed the viral risk within a previous blog installment. Now let’s consider the TSE risk.

TSEs (transmissible spongiform encephalopathies) are fatal diseases believed to result from exposure of a few animal species (including various ruminants, cervids, ungulates, cats, minks, and humans) to “infectious” prion proteins. The infectious proteins (PrPSc) are capable of interacting with and altering the normal prion proteins (PrPc) within the brain and spinal cord, changing the normal proteins to the abnormal form. Since humans have contracted prion disease as a result of consuming tissues from cattle with bovine spongiform encephalopathy (mad cow disease), there is concern about the use of bovine materials and materials from other “relevant species” in the manufacture of biopharmaceuticals.

The EMEA has provided a guidance document (EMEA/410/01 Rev. 2 October 2003; http://www.emea.europa.eu/pdfs/human/bwp/TSE%20NFG%20410-rev2.pdf) describing the requirements for the use of animal-derived materials from relevant animal species (cattle, sheep, goats, and other animals which are naturally susceptible to TSEs but not including humans or non-human primates) in the manufacture of medicinal or veterinary products. The guidance applies to active substances, excipients and adjuvants, raw and starting materials and reagents, and materials which come into contact with products or equipment used to make product. The major point of the guidance is that TSE risk can be minimized, but in many cases not entirely eliminated. Where TSE risk cannot be avoided through elimination of animal-derived materials completely, the guidance provides principles for the minimization of TSE risk which include: the use of low-risk (non-relevant) animal species, the geographical sourcing of relevant species from low-risk regions, the use of low-risk tissues, the use of appropriate slaughter techniques to reduce potential for contamination of low-risk tissues with high-risk tissues, the appropriate oversight of manufacture of the animal products through Quality Assurance and self and external auditing and Quality Control testing, and the implementation of process designs to remove or inactivate the abnormal prion proteins.

Biopharma companies using animal-derived materials are instructed to perform risk assessments on those materials, which take into account the factors described above. The risk assessments must be completed as part of a formalized animal-derived materials program, documented procedurally and executed by staff that are experienced and trained to conduct such assessments. EMEA inspectors will expect to see this formal program in place. Risk assessments for individual raw materials may then be rolled up into a risk assessment for the biopharma product. The rolled up risk assessment may also consider manufacturing steps at the biopharma which may remove or inactivate the abnormal prion proteins, though these, if cited, may need to be validated. Finally, it is expected that companies will conduct a benefit/risk evaluation to assure that any benefits realized by the patient taking the product will outweigh and justify the risk associated with the use of materials derived from relevant animal species.

Friday, October 23, 2009

Got Animal Derived Materials?

By Ray Nims

Most biopharma manufacturing processes utilize a few raw materials (including cell substrates, excipients, materials which come into contact with the product, etc.) derived from animals or which have been in contact with animal-derived materials at some point. As part of a formal animal-derived materials program, the biopharma must assess the viral and TSE risk associated with such materials. Let’s consider the viral risk first. From a viral safety standpoint, it is important for each biopharma to consider such ingredients and to be aware of the inherent risk of transmitting virus into the product via the materials. Why? As Genzyme discovered in the spring of 2009 (http://www.genzyme.com/corp/media/GENZ%20PR-061609.asp), viruses can infect the upstream manufacturing processes with results devastating to both the biopharma and to the patients its products are intended to treat. Viral risk mitigation, and regulatory guidance (e.g., EP Chapter 5.1.7: Viral Safety), require that viral risk assessments be performed for animal-derived materials used to manufacture biologics. In this context, raw materials include also excipients, growth media, column packing resins, and cell substrates.


For each product, manufacturers should list the animal-derived materials utilized, and should perform a viral risk assessment for those materials. This assessment considers the animal species and tissue, the processes used to manufacture the raw material, the quality control testing performed on the raw material, and in some cases, the manufacturing process in which the raw material is to be used at the biopharma. Inspectors from the EMEA will not only expect the risk assessments to have been performed and documented, but will expect that the assessment process be formalized into a business practice or standard operating procedure. The staff performing the assessments should be qualified for this task and the assessment team should include staff knowledgeable in virology, viral inactivation and removal, and the manufacturing and purification processes employed for the specific product at the biopharma.

Viral risk assessments completed for individual raw materials may eventually be rolled up into a viral safety assessment for the product per EP Chapter 5.1.7. This product evaluation will also consider other factors, such as the patient profile and route of administration, the cell substrate, the types and pathogenicities of viral contaminants found in the cell substrate and the manufacturing process, the amount of bulk material required for a human dose, and the viral inactivation and removal capabilities of the manufacturing downstream processes.

Tuesday, October 13, 2009

Outsource it, and fuggedaboutit?

By Ray Nims

Much has been written about the rationales and advantages for outsourcing of manufacturing and/or testing services; about the selection of outsourcing partners; and about the optimization of the pharma/contractor relationship. In any pharma/contractor relationship, there are responsibilities associated with the pharma as well as contractor responsibilities. These include both business as well as compliance responsibilities. The business realities and regulatory expectations associated with the use, by a pharma company, of a contract testing organization must be considered when the decision is made to outsource. A contract testing organization desiring to provide services for a pharmaceutical must be aware of the expectations and responsibilities associated with such a partnership. The optimal and most defensible programs will be those in which the various practices to be described below are formalized within internal Quality Systems, policies, and/or standard operating procedures as well as Quality Agreements.


Responsibilities falling upon the pharmaceutical partner include: 1) the selection of the contract testing lab; 2) commissioning and providing test samples of raw materials and products for method verification (compendial methods) and method qualification (non-compendial methods); 3) instituting of a Quality Agreements, business agreement, and/or confidentiality agreement with the contractor; 4) scheduling and shipping of test samples in accordance with the requirements of the testing lab and the test system; 5) providing in-life guidance and oversight of investigations of unexpected and out of specification results; and 6) ongoing monitoring of the performance of the contract lab and its methods.

Responsibilities primarily falling upon the testing lab include: 1) attaining and maintaining GLP or GMP compliance as appropriate for the intended use of the method; 2) providing assurance that the methods offered will be available to the client over the long term; 3) responsiveness to the sponsoring pharma and adherence to the terms of the Quality and/or business agreements; 4) method validation, verification, and or qualification as appropriate for the intended use of the method; 5) control of reagent, raw material, control, and standard inventory and quality; 6) assuring secure and retrievable data archiving; and 7) retention of staff possessing the appropriate expertise for direction of operators and the methods.