Wednesday, December 30, 2009

Can Cache Valley Virus Trash Your Manufacturing?

By Dr. Ray Nims

Cache Valley virus is a single-stranded RNA virus of family bunyavirus, genus Bunyavirus. It is enveloped and nominally 80-120 nm in diameter. Cache Valley virus was first isolated in Utah in 1956 and is carried by mosquitoes. It has since been found to be a widespread virus, having been isolated in Texas, Michigan, North Carolina, Indiana, Virginia and Maryland, for example.



source: Nims et al., BioPharm. Int. 21: 89-94, 2008

Basis of Concern. Cache Valley virus is known to infect livestock, causing birth defects. There have been two reports of encephalitic disease in humans attributed to Cache Valley virus. This virus has been isolated from biologics manufacturing processes employing Chinese hamster cell substrates on a number of occasions from 2000 to 2004 (Nims et al., BioPharm. Int. 21: 89-94, 2008). The route of entry of the virus into biologics production processes has not been established with certainty, although the use of contaminated bovine serum is considered to be the most likely source. Thus far the virus has only been known to infect manufacturing processes employing bovine serum.

Regulatory Expectations. Cache Valley virus is not mentioned specifically in any regulatory guidance, as the detection of this virus in biologics production has been reported only within the past decade. It is the intent of the guidance, however, that occurrences of viral contamination in biologics manufacturing be dealt with through implementation of specific testing methods as required to assure detection of future recurrences (e.g., ICH Q5A R1). In addition, it is expected that the route of entry of the virus be established and that the process be remediated so that future recurrences are prevented where possible (e.g., 1997 Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use).

Mitigating Risk. Although many Contract Testing laboratories offer rapid nucleic acid-based detection assays for Cache Valley virus, raw material screening for this virus using such assays does not appear to be a viable means of eliminating risk. The industry experience thus far indicates that Cache Valley virus may be a low level, non-homogeneous contaminant of bovine serum. Viral screening performed on one bottle out of a large lot of serum therefore is no guarantee that this virus will not be encountered. Elimination of animal-derived materials (esp. bovine serum) from the manufacturing process may help to reduce the risk of experiencing this virus. Should this not be possible, treatment of the serum or serum-containing media should be considered. Gamma-irradiation has been demonstrated to be effective in inactivating this virus in bovine serum (Gauvin, 2009). UVC treatment of media containing bovine serum also appears to be quite effective at inactivating Cache Valley virus (Weaver, 2009).

Conclusions. Cache Valley virus infects livestock and has been found to contaminate biologics manufacturing processes employing bovine serum. It is a virus of concern for biologics manufacturers employing bovine serum which has not been gamma-irradiated. Risk of infection of biological products with Cache Valley virus through use of bovine serum may be mitigated through implementation of gamma-irradiation of the serum, or UVC- or high-temperature short-time (HTST) treatment of media containing the serum and of viral purification processes capable of removing and inactivating enveloped viruses.

Wednesday, December 23, 2009

What cell line is this anyway?

By Dr. Ray Nims

For about as long as scientists have been using cell cultures in biomedical research, there have been cases of cell line misidentification. This has been especially true for continuous cell lines, with the increased probability over time of mislabeling or cross-contamination. The primary cross-contaminant historically has been HeLa, a human cervical carcinoma cell which, given the opportunity, could outgrow most other cells in culture. More recently, the use of feeder cells for the propagation of human stem cells, and the use of xenografting for the propagation of human tumor cells, has provided additional opportunities for cell line cross-contamination and misidentification.


In the past, confirmation of cell line species of origin has been the main approach for authenticating cell lines. This was done initially by karyotyping or by immunological techniques, but more recently it has been done through the technique of isoenzyme analysis. An example of an isoenzyme analysis is shown below for Peptidase B and Aspartate Aminotransferase.  These agarose gels show a positive control, a negative control (this is the band that does not line up with the others), the test article and a standard extract.  These gels confirmed the identity of the test article as mouse derived, as expected. 




Isoenzyme analysis has the advantage that it is rapid, not very technically demanding, and may be used not only to confirm species of origin but also to detect the presence of a cross-contaminating cell if the latter is present in the culture at 10% or greater (Nims et al., Sensitivity of Isoenzyme Analysis for the Detection of Interspecies Cell Line Cross-Contamination. In Vitro Cell. Dev. Biol.-Animal 34:35-39, 1998). In fact, isoenzyme analysis is currently the primary method employed within the biopharmaceutical industry for cell line authentication in satisfaction of 1993 Points to Consider and ICH Q5D guidance.

Recent advances in molecular diagnostic techniques have made possible the authentication of human cell lines to the individual level. DNA fingerprinting technologies have matured to the point that some of them, especially single nucleotide polymorphism (SNP) typing and single tandem repeat (STR) profiling, are now considered to be viable options for standardizing human cell authentication (see ATCC SDO newsletter article, page 5. For both human and animal cells, DNA fingerprinting provides a means of determining authenticity to the individual level. However, the primary drawback is that the fingerprinting techniques as routinely performed will be less or not at all useful for detecting interspecies cocultivations or cross-contaminations. For this purpose, it may be necessary to retain isoenzyme analysis as part of the authentication armament even when the molecular technologies become the definitive authentication practices for human and animal cell lines.

Friday, December 18, 2009

Got Animal-Derived Materials? Part 3

By Dr. Ray Nims

The assessment of viral and transmissible spongiform encephalopathy (TSE) risk for animal-derived materials (ADM) used in the manufacture of biologics, which we have described in previous blogs, is just one component of an overall ADM program that should be in place at each organization producing biologics.



A formal ADM program at a biologics manufacturer ideally should be driven by an overriding SOP or policy document. This should address the procedures in place for minimizing the use of ADM, for procuring ADM with a view to minimizing viral and TSE risk, and for assessing the viral and TSE risk associated with the ADM that are used. There are specific sourcing requirements for ADM that are intended to minimize TSE risk (EMEA/410/01 Rev. 2 October 2003), and these must be followed or justification provided if deviated from. The evaluation of ADM for the presence of viruses of concern is addressed in the Code of Federal Regulations, Title 9 Part 113.53. ADM viral and TSE risk assessments should be conducted according to a formalized procedure by teams of individuals with education, training, and/or experience appropriate for these tasks. The composition of the risk assessment teams and the qualifications of their members should be described in revisable controlled documents. The risk assessments themselves should be recorded in controlled documents which may revised as new information becomes available from the ADM suppliers. The ADM information that is used as part of the risk assessment process should be archived in a manner tying it to the risk assessment itself.


The existence of a formalized ADM program, qualified risk assessment teams, as well as reports documenting the individual ADM risk assessments may be the subject of regulatory scrutiny during periodic inspections or inspections tied to a new product application. This is especially likely if the product is intended for global distribution, as these ADM issues are specifically mentioned in EP (Chapter 5.1.7) and EMEA (EMEA/410/01 Rev. 2 October 2003) guidance.

Monday, December 14, 2009

Advantages of Compendial Methods

By Dr. Lori Nixon

When you are developing a new product specification, it is usually recommended to rely on the appropriate compendial method for applicable “generic” quality characteristics such as pH, residual solvents, trace metals, bioburden, etc. By compendial method, we mean methods that are described as chapters in the United States Pharmacopeia (USP) or others that may be applicable for a specific regulatory region. The three main compendia include the USP, European Pharmacopoeia, Japanese Pharmacopeia (USP, PhEur, JP); these are the “tripartite” bodies that are involved in the International Conference on Harmonization (ICH).



photo of USP laboratories from DPR Construction Inc.

Why rely on compendial methods rather than just using your own? It is generally recommended to refer to compendial methods where applicable. The advantage to the drug sponsor is a reduced requirement for validation supporting such methods (the methods themselves are considered validated, and may only require product-specific verification in the particular testing lab). Compendial methods are “familiar” to regulatory reviewers; they are also generally expected. If you propose your own method as an alternate method, you will need to justify why your own method is equivalent or better. For the testing lab, there is some advantage in having methods that can be applied to multiple products (avoiding a multiplicity of similar methods) and where the change process is relatively well-defined and publically communicated. You may also find it simpler to transfer testing between different labs.

To reference the compendial method in your specification, you may refer simply to the test by attribute and chapter, along with the associated limit for your product. For example, your specification may include a limit of 10 EU/mL for bacterial endotoxin as measured by USP<85>. The general expectation here is that at the time of testing, the current version of USP is used. Clearly, this will require that your testing lab is aware of any potential changes to the USP and can prepare for such changes accordingly. As with any other change to an analytical method, changes to compendial methods can impact training, internal procedures, product-specific re-validation/verification, etc.

In practice, labs often rely on additional internal descriptive procedures in order to execute the compendial methods (i.e., rather than just directing analysts to follow the chapter directly). This is usually a good idea, for several reasons. It can be easier to train analysts according to a standard documentation format, and it is often necessary to describe details that may be specific to the particular lab, equipment, instrumentation, reagents, reporting requirements, etc. Again, even if there is a lab-specific procedure, it is usually best to refer directly to the compendial method (USP<85>, e.g.) in the sponsor’s product specification.

Be aware of compliance with compendial testing requirements when you are outsourcing testing. For example, almost any chemical testing lab with have a method for pH, but that doesn’t necessarily mean that it will comply with USP<791>. For example, in this case the USP method describes measuring the sample temperature within a certain range; often “generic” lab methods for pH do not specify control of the sample temperature. There are additional requirements such as the calibration standards chosen, etc that must also be considered. When reviewing vendor methods, check the following:

- Does the method purport to comply with any compendia? (should be clearly stated in the vendor’s procedure if so)

- Which compendia?

- Check details of the method to ensure that it does indeed comply with the current compendial procedure(s) in question

- Does the lab have a mechanism to stay current with upcoming compendial changes?

- Do they have appropriate change control system to ensure that they can prepare for method changes and associated re-validation, etc?

- Consider what verification/validation is required to ensure that the vendor method provides reliable results for your particular product/sample type.

Of course, the downside of compendial methods is that they are region-specific, and one region may not recognize the compendia of another region. There have been efforts in recent years towards harmonizing methods (ICHQ4B for bioburden testing, for example), but this process is slow and far from complete.

If you intend to market or perform clinical trials in more than one region, you may need to ensure compliance with multiple compendia. In this case, consider the following:

• Has method been harmonized through the ICH process?

• Is it possible to create an internal “harmonized” method that meets the requirements of all relevant compendia?

• Is it possible to meet the requirements of all by following the “most stringent” compendial procedure?

• Will you need to test by multiple procedures to generate results acceptable to each region?

Wednesday, December 2, 2009

Should we care about…Vesiviruses?

By Ray Nims

Vesiviruses are single-stranded RNA viruses of family calicivirus, genus Vesivirus. They are non-enveloped and 30-40 nm in diameter, and the genus includes feline calicivirus, vesicular exanthema of swine virus, rabbit vesivirus, and San Miguel sea lion virus, as well as vesivirus isolate 2117.



source: Stewart McNulty, Queens University, Belfast, UK

Basis of Concern. Vesivirus 2117 has been isolated from biologics manufacturing processes employing Chinese hamster cell substrates on a number of occasions, the first being reported in 2003 (Oehmig et al., J. Gen. Virol. 84, 2837-2845, 2003), and additional occurrences being reported in 2008 and 2009.
The susceptibility of relevant manufacturing cell lines of different animal species to infection by this virus appears to be limited to the Chinese hamster. When infected, these cells undergo a relatively rapid lytic infection. The route of entry of the virus into biologics production processes has not been established with certainty, although the use of contaminated animal-derived materials, such as bovine sera, is considered to be the most likely source.

Regulatory Expectations. Vesivirus is not mentioned specifically in any regulatory guidance, as the detection of the 2117 isolate in biologics production has been reported only within the past decade. It is the intent of the guidance, however, that occurrences of viral contamination in biologics manufacturing be dealt with through implementation of specific testing methods as required to assure detection of future recurrences (e.g., ICH Q5A R1). In addition, it is expected that the route of entry of the virus be established and that the process be remediated so that future recurrences are prevented where possible (e.g., 1997 Points to Consider in the Manufacture and Testing of Monoclonal Antibody Products for Human Use).

Mitigating Risk. At least three Contract Testing laboratories have announced rapid nucleic acid-based detection assays for vesivirus isolate 2117 within the past year. These assays are available for raw material screening and for in-process testing of biologics bulk harvest samples. Elimination of animal-derived materials (esp. bovine sera) from the manufacturing process may help to reduce the risk of experiencing this virus. Should this not be possible, treatment of the sera or sera-containing media should be considered. Studies on the inactivation of caliciviruses indicate that UV treatment may be effective (Duizer et al., Appl. Env. Microbiol. 70, 4538-4543, 2004; de Roda Husman et al., Appl. Env. Microbiol. 70, 50989-5093, 2004). Gamma-irradiation at the dosages normally used does not appear to be effective, as might be expected for a virus of this relatively small size. Studies using MMV indicate that high-temperature short-time (HTST)-treatment of medium containing bovine serum is effective in inactivating this virus (Schleh et al., Biotechnol. Prog. 25: 854-860, 2009), and would by implication be effective for vesiviruses in general.

Conclusions. Vesivirus isolate 2117 preferentially infects Chinese hamster cells and has been found to contaminate biologics manufacturing processes employing this cell substrate. It is now a virus of concern for the biopharmaceutical industry. Risk of infection of biological products with vesiviruses through use of bovine-derived materials such as bovine sera may be mitigated through implementation of UV or HTST treatment of media containing the sera and of viral purification processes capable of removing and inactivating an even smaller non-enveloped virus such as MMV.

Tuesday, November 10, 2009

The Good Buffer

By Scott Rudge

“A Good Buffer” has a number of connotations in biochemistry and biochemical engineering. A “good buffer” would be one that has good buffering capacity at the desired pH. The best buffering capacity is at the pK of the buffer of course, although it seems buffer salts are rarely used at their pK.

Second, a good buffer would be one matched to the application. Or maybe that’s first. For example, the buffering ion in an ion exchange chromatography step should be the same charge as the resin (so as not to bind and take up resin capacity). For example, phosphate ion (negative) is a good choice for cation exchange resins (also negatively charged) like S and CM resins.

Another meaning of a “Good” buffer is a buffer described by Dr. Norman Good and colleagues in 1966 (N. E. Good, G. D. Winget, W. Winter, T. N. Connolly, S. Izawa and R. M. M. Singh (1966). "Hydrogen Ion Buffers for Biological Research". Biochemistry 5 (2): 467–477.). These twelve buffers have pK’s spanning the range 6.15 to 8.35, and are a mixture of organic acids, organic bases and zwitterions (having both an acidic and basic site). All twelve of Good’s buffers have pK’s that are fairly strongly temperature dependent, meaning that, in addition to the temperature correction required for the activity of hydrogen ion, there is an actual shift in pH that is temperature dependent. So, while a buffer can be matched to the desired pH approximately every 0.2 pH units across pH 7 ± 1, the buffers are expensive and not entirely suited to manufacturing applications.

In our view, a good buffer is one that is well understood and is designed for its intended purpose. To be designed for its intended purpose, it should be well matched to provide adequate buffering capacity at the desired pH and desired temperature. As shown in the figure, the buffering capacity of a buffer with a pK of 8 is nearly exhausted below pH 7 and above pH 9.




It’s easy to overshoot the desired pH at these “extremes”, but just such a mismatch between buffering ion and desired pH is often specified. Furthermore, buffers are frequently made by titrating to the desired pH from the pK of the base or the acid. This leads to batch to batch variation in the amount of titrant used, because of overshooting and retracing. In addition, the temperature dependence of the pK is not taken into account when specifying the temperature of the buffer. Tris has a pK of 8.06 at 20°C, so a Tris buffer used at pH 7.0 is already not a good idea at 20°C. The pK of Tris changes by -0.03 pH units for every 1°C in positive temperature change. So if the temperature specified for the pH 7.0 buffer is 5°C, the pK will have shifted to 8.51. Tris has 3% of its buffering capacity available at pH 7.0, 5°C, it’s not well matched at all.

A good buffer will have a known mass transfer rate in water, so that its mixing time can be predicted. Precise amounts of buffering acid or base and cosalt are added to give the exact pH required at the exact temperature specified. This actually reduces our reliance on measurements like pH and conductivity that can be inexact. Good buffers can be made with much more precision than ± 0.2 pH units and ± 10% of nominal conductivity, and when you start to make buffers this way, you will rely more on your balance and understanding of appropriate storage conditions for your raw materials, than making adjustments in the field with titrants, time consuming mixing and guessing whether variation in conductivity is going to upset the process.

Friday, November 6, 2009

Enzyme induction…done pharmacodynamically

By Ray Nims

Pharmacodynamics is the study of a specific effect of a drug as related to drug concentration at the putative active-site for that effect. Pharmacodynamics is sometimes used to model quantitatively the effect of a drug over time as drug concentration at the active-site rises and falls. Another type of pharmacodynamic study entails exposing the animal or in vitro system to graded doses of a drug and monitoring the effect associated with each active-site concentration. From the latter type of study, one is able to estimate both potency for the effect (given in terms of the active-site drug concentration at the half-maximal effect for that drug, or EC50) and its efficacy (given in terms of percentage of maximal response compared to other drugs causing the same effect through the same mechanism). In receptor theory, EC50 is considered to reflect the affinity of the drug for a receptor, while efficacy is a measure of the bound drug’s ability to cause the specific response.


The induction of drug-metabolizing enzymes, such as the cytochromes P450, may be considered to represent an effect of a drug or xenobiotic. It is common for investigators to measure such induction at one or a few dose levels and to compare the resulting enzyme induction with that of a prototype inducer. These comparisons are sometimes described in terms of the test xenobiotic causing “strong” (“potent”) or “weak” induction in comparison with the prototype inducer. As already pointed out quite elegantly by D. A. Smith and coworkers (Letter to the Editor: The Time to Move Cytochrome P450 Induction into Mainstream Pharmacology is Long Overdue. Drug Metab. Dispos. 35:697-698, 2007; http://dmd.aspetjournals.org/cgi/content/full/35/4/697), such statements are both misleading and inaccurate. As with any drug effect, enzyme induction must be described in terms of both potency and efficacy. It is possible for an inducer to be very potent but to display little efficacy. In fact, a xenobiotic having high potency and little or no induction efficacy might represent a competitive inhibitor for this effect. In contrast, there may be inducers which are very effective, but not very potent.

It is possible for efficacy and potency for enzyme induction to be estimated on the basis of studies using intact animals, provided that certain assumptions are made (e.g., that total plasma drug concentration is a suitable proxy for drug concentration at the induction active site, which cannot be sampled directly). An example of such a study is that of R.W. Nims and coworkers (Comparative Pharmacodynamics of Hepatic Cytochrome P450 2B Induction by 5,5-Diphenyl- and 5,5-Diethyl-substituted Barbiturates and Hydantoins in the Male F344/NCr Rat. J. Pharmacol. Exp. Therap. 270: 348-355, 1994; http://jpet.aspetjournals.org/cgi/content/abstract/270/1/348). A more straightforward approach is offered through in vitro enzyme induction studies, in which enzyme induction can be related to drug concentration in the culture medium (e.g., Kocarek and coworkers: Differentiated Induction of Cytochrome P450b/e and P450p mRNAs by Dose of Phenobarbital in Primary Cultures of Adult Rat Hepatocytes. Mol. Pharmacol. 38:440-444, 1990; http://molpharm.aspetjournals.org/cgi/content/abstract/38/4/440).

Measurement of the induction of the cytochromes P450 and other drug-metabolizing enzymes following drug treatment in animals and humans is an important aspect of drug characterization. The studies should be performed and reported in a manner consistent with other drug effects, that is, in a manner consistent with the principles of pharmacology.

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.

Tuesday, August 4, 2009

Do You Use Risk Assessments in Auditing?

Audits are a critical component of quality systems, but are they guided by formal assessments of risk to your products? In this world of ICH Q9, can you offer even a semi-quantitative justification for your audit priorities? We have spoken to many people in the industry, and almost all mention a risk assessment being undertaken prior to an audit. But we have not found many people that formalize that risk assessment, or keep it updated from audit to audit. Even fewer communicate their scoring of risk to either their internal clients or the vendor that has been audited.

A new trend in auditing is to use a form of risk assessment both before and after the audit. A popular form is the Failure Modes and Effects Assessment, or FMEA (see, for example, http://www.sre.org/pubs/Mil-Std-1629A.pdf). In a traditional FMEA, risks of failure are identified in a detailed fashion, and scored in three categories related to the failure’s probability, detectability and severity. Scoring is done on a semi-quantitative or relative basis using an arbitrary scale such as 1-10. For an audit, you might use the same categories as they relate to a particular vendor's (or department's) ability to deliver a product or service, failure free. You could organize your FMEA according to the critical quality attributes of the product or service being delivered or according to a list of requirements from a guideline or the CFR's. Your FMEA should receive input from affected departments, and should be used for prioritization of audit items. You should have the FMEA in mind as you conduct your audit, and remember why various items received high prioritization. You may change ratings for probability or detectability based on what you observe. If instead, you confirm your evaluation, you should probe remediations that decrease your firm's primary concern. A remediation that addresses detectability, when the issue was probability, likely won’t mitigate the risk of failure.
When you return from your audit, rescore the FMEA with assessments based on your observations and data that you collected. Make sure that you share your analysis with the stakeholders. And monitor the performance of the vendor until the next audit; the data will help inform your next FMEA.

Do you already use FMEA's in audit preparation and reporting? Let us know your practices.

Tuesday, June 23, 2009

Why is Quality by Design so heavy with statistics?

Why is the literature on Quality by Design so laden with statistics and experimental design space jargon? After all, the definition of the term “design” doesn't seem to include the analysis of messy data leading to rough correlations with results that are valid only over a limited range. So what gives?

The idea behind QbD was to use mathematical, predictive models to predict process outcomes. This concept can be applied directly to simple unit operations, such as drying, distilling, heating and cooling. However, unlike in the petrochemical business, the thermodynamic properties of most active pharmaceutical ingredients are not known and are difficult to measure. The unit operations used to manufacture common biotechnology products, such as cell culture, chromatography and fermentation have been modeled, but the models are very sensitive to unknown or unmeasureable adjustable parameters. The batch nature of these operations also makes their control difficult, as classical control theory relies on the measurement of an output to make an adjustment to an input to correct the output back towards the design specification.

Since there does not appear to be a clear path to using models, an approach has been chosen that emphasizes getting as much phenomenological information from as few experiments as possible. This is the Design of Experiments approach, where input conditions or operating parameters are systematically varied over a range and the process outputs measured, with statistics used to deconvolute the results. The combined ranges tested become the “design space”, and the process performance outputs with the variations closest to the process failure limit become the critical performance parameters. The results are useful, but only within the design space, and only with the certainty that the statistics report. Also, since the results are phenomenological, the effect of scale is often unknown.

The statistical approach is acceptable, and for the immediate future it's probably the best that we can expect. But the focus on this approach seems to drown out the more pressing need for good process models and physical properties data. These are the elements that allowed the petrochemical and commodity chemicals industries to scale up processes with assurance that quality specifications would be met. There are countless models available for bioprocessing's more complicated unit operations, but they have parameters that we don't know and can't calculate from first principles. There is no question that we need to find ways to collect this data, and a commitment to publish or share it. There are also simpler unit operations that we can model, scale up and scale down with complete assurance. These include operations such as mixing and storing solutions, filtration, diafiltration, centrifugation and some reactions. We shouldn't let the more complicated operations that still require statistical DOE approaches prevent us from applying the true principles of QbD to our simpler unit operations.

Wednesday, June 3, 2009

Ten Steps to Choosing your Contract Manufacturer

For many young companies, choosing a contract manufacturer, or CMO, for their lead pharmaceutical candidate is critical. Choose the wrong contractor, and you could be faced with delays and cost overruns with which your investors and patients won’t be very sympathetic. While there is no guarantee that you will always make the right decision, here are some tips that can help you make your choice in an organized, thoughtful, meaningful and objective way:

1) Make a list of all the possible suppliers. Such lists may be purchased, but they are also easily assembled from internet searches. In fact, you can do a little pre-screening with your own internet search.

2) Screen potential suppliers with a phone call. You will probably speak with a business development or sales person representing the contractor, but usually these people are quite knowledgeable about their company’s capabilities, and common problems encountered in the industry. We recommend you not reveal too many details about your project, and be prepared mostly to listen. However, you should have three or four key capabilities or proficiencies that you are looking for in all of the potential vendors. If possible, try to rule out vendors who do not meet your “must-have” requirements at this stage. Stay tuned for a blog on how to establish your showstopper list, it’s a critical exercise, and may extend beyond key capabilities and proficiencies.

3) Keep a matrix, and record the date you first contacted the vendor, when they responded to you, the status of any confidentiality agreements, and all the contact information that you can gather (email addresses, cell phone numbers, main switchboards and extension numbers). Also note the responses that each contact had relative to your three or four show-stopper criteria.

4) Meet with your team, and select three to five potential vendors to request a proposal from. We don’t recommend more than five: getting good, comparable proposals is a lot of work, like 2n times the work, where n = the number of proposers. Not to mention the work that contract manufacturers go through to read your RFP and prepare a proposal. Your three or four showstopper criteria should help you limit the number of proposers; if necessary you can begin to narrow down based on “nice-to-have” criteria as well. You may also deliberately choose to look at a range of vendors that represent different strengths/weaknesses (for example, do you prefer a “one-stop shop” that is convenient, but maybe not the best at everything, or a “specialty” vendor that provides higher levels of expertise, but will require you to select and manage multiple vendors?).

5) Solicit proposals. Most contract manufacturing seekers have a Request for Proposals (RFP) process that includes a document. These RFP documents vary from one page requirements descriptions, to lengthy, legalistic documents that require a team, and a month, to respond to. You should do what is comfortable for your organization. There needs to be enough information so that the vendor is able to respond with a meaningful proposal. There is some legal danger, particularly with intellectual property, so it’s not a bad idea to get your RFP reviewed by your legal counsel. And you should only send an RFP to a vendor after they have signed a mutually agreed confidentiality agreement.

6) Score your proposals. Find some basis to make apples to apples comparisons. RMC uses a modified Kepner Tregoe analysis, but many forms of analysis will work. You should have determined how you will assess and weight qualitative data before you begin. And in doing so, you should not under-estimate intangible factors: the ability to communicate, time zone differences, good references (you’ve checked, right?) are some examples. At this stage you should be ranking and scoring on “nice-to-have” criteria as well as comparing cost/timeline, capabilities, capacity, and viability of the business. You might form your opinion of the viability of the business by reading annual reports and press releases, and by assessing how busy the manufacturing area and support labs look during your visit.

7) Visit the top two or three vendors in person. Vendors may not allow a formal quality audit prior to signing a contract, but be sure to bring your quality representatives even for an informal “technical visit”. If the project is large, you may take the resistance to a pre-use quality audit as a red flag. Again, spend as much time as possible listening, rather than talking. Get a tour, and copies of all presentations. Ensure that you have a meeting between the decision makers for both sides as well as aligning discussions between key technical and quality personnel. If there are disputes or further work to do, your decision makers must have a good working relationship.
Your visit is also your best opportunity to break past the business development group and take a true measure of the business. Chat with the people in the lab or production area if you can. Look over the state of the equipment, the cleanliness of the facility and the stock in the warehouse. Check their flexibility-- what can they make happen for you, vs. what will have to be run past someone in another building, or another city? Ultimately, you should think about hiring a contract manufacturer similarly to how you hire an employee, by hiring for expertise as well as fit with your team.

8) Consider entering contract negotiations with at least two vendors. Things can go wrong in negotiations, and your position is stronger if you can legitimately walk away. We typically don’t let any of the final three candidates off the hook until the ink is dry on the final contract. If your budget can justify it, having a second contractor performing development work and verifying the primary contractor’s results is an excellent idea. It may ultimately spread your risk in supply chain, and give you leverage in negotiating commercial supply agreements later on.

9) Revisit your analysis tool. You may learn new things in your contract negotiations that cause you to adjust your evaluation. Don’t be afraid to be frank if you feel like terms have been changed since the selection was made. This is a good reason to keep a back up vendor.

10) You must now manage the project according to the criteria by which you selected your supplier. Hold your supplier and yourself accountable to these criteria. For example, after selecting a vendor because they can meet a very aggressive timeline, do not put the project timeline at risk by failing to order back up critical path supplies, in case the primary order doesn’t arrive, or fails to meet specifications on arrival. We will have more to say about managing a contract manufacturer in a future blog.

Choosing the right manufacturing partner is critical for your success as a drug developer. Spend the time required to make a good decision. This time should be spent gleaning as much tangible and non-tangible information on all your options as possible, and then objectively comparing it. You should have an idea about how you’re going to evaluate and weight non-tangible factors into your decision. And once you have made the choice, manage according to your criteria. Although everyone has their own methods for vendor selection, these are some suggestions that have worked well for us and our clients. If you have questions or comments, please visit http://www.rmcpharma.com/ or email us at info@rmcpharma.com.

Thursday, May 21, 2009

How do you establish a reference standard for release of your first GMP lot?

There are some different approaches depending on situation, but here is one general approach that many people use:

* Preclinical material that was used to support IND-enabling tox studies is characterized and used as an interim reference standard. This doesn't have to be manufactured by GMP; could even be research grade material. Characterization tests should be performed and include methods to determine content, primary structure, bioactivity, tentative release tests, etc. Important to have a good handle on content from this standard (i.e., for a protein drug you might look at total protein content by elemental analysis.) Potency units could be defined here as well.
* Use this interim reference standard as a comparator to release your initial GMP material.
* Use material from the first GMP campaign to create a primary reference standard. Again, this material will need to be subjected to a panel of characterization tests in addition to the release tests (typically at least or more in-depth than the characterization done on initial reference standard).
* The primary reference standard supercedes the interim reference standard after it is created and qualified. This typically happens during or immediately after the first GMP campaign.

From an assay validation standpoint, you will always have to start with some initial standard (typically, the interim reference standard, which is all you may have available before GMP manufacture), and you can get into some circularity with accuracy (i.e., assaying the standard against itself). So for your initial standard and assay validation you rely somewhat on orthogonal methods of analysis. Also, as per ICH Q2, accuracy can be inferred from precision, linearity and specificity.

By Lori Nixon, RMC Pharmaceutical Solutions, Inc.

Find out more at www.rmcpharma.com

Friday, May 15, 2009

RMC Pharma is 5!

RMC Pharmaceutical Solutions celebrated its 5 year anniversary today. We have grown from a couple people looking for their first consulting opportunity each in our spare bedrooms or kitchen tables to an integrated team capable of taking almost any health care product from conception to commercialization. Today, we are 10 regular employees, with 6000 square feet of office and lab space in Longmont, CO. We've helped 40 clients on three continents with projects ranging from producing pre-clinical proof of concept materials to overseeing pre-approval inspections and quality audits from the biggest pharmaceutical companies in the world. Our capabilities have expanded beyond those of a typical consulting group, to include specialized software such as Slimstat, Minitab, Omnisign, SuperPro and DPL decision modeling software, to laboratory capabilities, including zetasizing, HPLC and Akta based process development. Still, our most valuable resource is our people and the relationships that we've built in our five years. The experience has been very rewarding for me personally, and I hope that my partners, associates and clients have benefited from it as I have.

Thursday, May 14, 2009

RMC Expands Board of Directors

FOR IMMEDIATE RELEASE May 04, 2009
Contact: Kathy Rudge
RMC Pharmaceutical Solutions, Incorporated
info@rmcpharma.com
303 776 5200

Longmont, CO. Shareholders of RMC Pharmaceuticals Solutions, Inc. recently approved the expansion of its Board of Directors and appointed Dr. Mark Young and W. Todd Myers as additional members to the Board. The move brings outside directors to the company’s leadership, and adds invaluable expertise and experience as RMC Pharmaceuticals, Inc. continues to build its brand and business. In a related action, the shareholders unanimously elected Dr. Scott Rudge to the position of Chairman of the Board of Directors.
“I’m pleased to welcome Dr. Young and Mr. Myers to our leadership team at RMC Pharmaceutical Solutions,” said Dr. Rudge. “These gentlemen are very highly regarded in the biotechnology and entrepreneurial pharmaceutical business space. Their advice and guidance will help us to leverage and promote our “Bolt-on CMC” services as we continue to grow and expand our capabilities.” Timothy Joy, RMC Pharmaceutical Solutions’ President and CEO said, “Our collaborations with Mark and Todd have been very positive over the years. It made sense to formalize these relationships and take full advantage of their talents. Our clients will derive extended benefits from their addition to our team.”
Dr. Young is an experienced executive specializing in process development and manufacturing of biopharmaceutical products. He started his career as a process development scientist with large international pharmaceutical companies (Hoffmann LaRoche and Upjohn), and then joined the biotech industry in 1985. He has served as a Senior Vice President of three biotech companies (Synergen, Protein Design Labs, ZymoGenetics) and as Chief Operating Officer of Biomira. Dr. Young received degrees at the University of Nebraska, Columbia University, and The University of Michigan, all in Chemical Engineering. He is currently a consultant, specializing in bioprocess development and manufacturing issues.
W. Todd Myers, C.P.A., is a veteran financial executive with significant management experience in both public and private drug discovery/development companies. Mr. Myers is currently a member of the Board of Advisors of BioLaurus, Inc., a contract research organization specializing in cutting edge molecular imaging and high content analysis services for the biotechnology and pharmaceutical industry. He also provides coaching and guidance to start-up life science companies as an Entrepreneur-in-Residence at CONNECT, a non-profit organization dedicated to creating and sustaining the growth of innovative technology and life science businesses in San Diego. Most recently, Mr. Myers was the Chief Financial Officer of SGX Pharmaceuticals, Inc., a publicly-traded biotechnology company focused on the discovery, development and commercialization of innovative cancer therapeutics, and was instrumental in the sale of SGX to Eli Lilly & Company at a premium of 119% to its then-current trading price. Prior to SGX, as the Director of Finance at CombiChem, Inc, a publicly-traded computational drug discovery company, Mr. Myers managed all financial operations and was the senior financial representative on the team responsible for the sale of the company to DuPont Pharmaceuticals. He was also the Chief Financial Officer of FeRx Incorporated, a privately-held, clinical stage company dedicated to the development of oncology products, and has held positions with Premier Inc. and with Ernst & Young LLP. Mr. Myers received his B.S. in Accounting from the University of Illinois.

About RMC Pharmaceutical Solutions, Inc.
RMC Pharmaceutical Solutions, Inc. is a privately-held provider of expert services to the pharmaceutical, biotechnology, medical device and food industries. The company was founded by Timothy Joy and Scott Rudge in 2004 to provide comprehensive services to companies developing products for the health care industry. Since its inception, RMC Pharmaceutical Solutions has served more than 50 different clients in North America, Europe and Asia. More recently, RMC has rolled out its “Bolt-on CMC” service offering. Bolt-on CMC support provides an experienced integrated team to support Chemistry, Manufacturing and Control areas such as process, analytical and formulation development; quality control/quality assurance; and oversight of GMP manufacturing. As part of this service, clients can access both the considerable expertise of the RMC team, as well as physical assets, such as an analytical and process development laboratory; document management and storage; and advanced software tools. For more information, please visit www.rmcpharma.com.