Perspective Special Focus Issue: Bioanalytical laboratory management

For reprint orders, please contact [email protected]

The science of laboratory and project management in regulated bioanalysis

Pharmaceutical drug development is a complex and lengthy process, requiring excellent project and laboratory management skills. Bioanalysis anchors drug safety and efficacy with systemic and site of action exposures. Development of scientific talent and a willingness to innovate or adopt new technology is essential. Taking unnecessary risks, however, should be avoided. Scientists must strategically assess all risks and find means to minimize or negate them. Laboratory Managers must keep abreast of ever-changing technology. Investments in instrumentation and laboratory design are critical catalysts to efficiency and safety. Matrix management requires regular communication between Project Managers and Laboratory Managers. When properly executed, it aligns the best resources at the right times for a successful outcome. Attention to detail is a critical aspect that separates excellent laboratories. Each assay is unique and requires attention in its development, validation and execution. Methods, training and facilities are the foundation of a bioanalytical laboratory.

Pharmaceutical drug discovery & development Large pharmaceutical companies pursue multiple therapeutic areas and may divide efforts by aligning medical disciplines or disease areas with a specific site. This approach allows best access to fundamental science and alignment with key opinion leaders in experimental medicine. Since discovery is the foundation for innovator pharmaceutical companies, geographical concentration in selected areas near academic and medical centers has increased. Smaller pharmaceutical companies may pursue only one therapeutic area, choosing to focus on their core expertise. Even smaller, start-up companies can choose to select an individual target for which they have a unique process or intellectual property with abundant chemical libraries. Smaller companies often do not have the infrastructure needed to independently pursue all aspects of the drug discovery and development process. Therefore, they rely upon a network of consultants and contract research

10.4155/BIO.14.89  © 2014 Future Science Ltd

Steve Unger*,1, Thomas Lloyd1, Melvin Tan1, Jingguo Hou1 & Edward Wells1 Worldwide Clinical Trials, Early Phase Development, Bioanalytical Sciences, 8609 Cross Park Drive, Austin, TX 78754, USA *Author for correspondence: Tel.: +1 512 615 2280 Fax: +1 512 834 1165 [email protected]

1

organizations (CROs) to move drug candidates forward. Selection of the proper partners is critical as knowledge gaps within either organization can expose flaws, which can delay or fail the drug candidate. No drug is perfect and its flaws need to be assessed quickly, completely and carefully. Companies should have a backup compound and quickly learn from preclinical or early clinical development findings. Keeping too many poor drug candidates in the pipeline to impress investors is a poor strategy. It is best to fail fast with an early proof-of-concept study as clinical drug development is expensive. Proving the mechanism of action requires answering questions about both penetration and target engagement, and bioanalysis plays a critical role in answering these questions [1–4] . The assessment of whether a particular target protein, enzyme, receptor or gene can be affected by chemicals in a manner that impacts disease state is fundamental to drug discovery. The process of making and screening new compounds to select the best

Bioanalysis (2014) 6(10), 1357–1372

part of

ISSN 1757-6180

1357

Perspective  Unger, Lloyd, Tan, Hou & Wells

Key terms  Contract research organizations: Research organizations that serve to contract all or parts of toxicology or clinical studies. Drug development: Process of proving drug safety and efficacy in preclinical and clinical trials.

candidate is a lengthy refinement, not only by chemistry, pharmacology and biology teams but also drug metabolism, bioanalysis, toxicology and pharmaceutics. The role of the discovery team is to put forward safe and effective preclinical development candidates whose mechanism of action can be quickly proven in clinical proof-of-concept studies. Once selected, the preclinical drug candidate proceeds through Investigational New Drug-enabling toxicology studies. The timeframe for these studies is short, and many larger companies keep preclinical bioanalysis or toxicokinetics in-house to ensure a fast Investigational New Drug filing. If they contract toxicology studies, bioanalysis may also be performed with that CRO. Some companies will outsource bioanalysis at the clinical development stage where good planning aligns both the clinical study and bioanalysis in the same CRO. This affords accountability and fast turnaround for the entire process, including recruitment, dosing, sample collection/shipment, bioanalysis, pharmacokinetics and data reporting. Other companies elect to support the single and multiple ascending dose studies in-house, as these studies may expose liabilities in pharmacokinetics or safety that could limit its development. Few companies continue in-house bioanalysis of later phase development candidates. Instead, they rely upon the efficiency and capacity of CROs to replicate prior work in large, Phase III studies. Getting through proof of concept and dose selection is often a shared role for bioanalytical organizations, relying upon in-house experts while transitioning to a full CRO model for bioanalysis. The management of drug development activities within pharmaceutical companies involves integrating functional organizations as part of project teams. Each discipline should have a representative who is fully knowledgeable about their organization, as well as general principles of drug development. Team leaders should have in-depth knowledge of medicinal and process chemistry, pharmacology, toxicology, drug metabolism, pharmacokinetics and pharmaceutics. The strength of the team depends upon team members, as well as its management. Governance boards should have sufficient strategic oversight without restricting teams from their tactical planning and execution.

1358

Bioanalysis (2014) 6(10)

Contract research organizations Pharmaceutical companies will engage a CRO at some stage in the lengthy and complicated process of drug development. Larger CROs reflect similar integration of teams to assist with pharmaceutical development. This can include the entire nonclinical or clinical drug development process from study design through all phases of execution. A large CRO makes use of their capacity and diversity by offering clients full service drug development, including clinical studies in both healthy and patient populations. The availability of multiple CROs affords pharmaceutical companies the opportunity to select the best and most cost-effective relationships. Within the field of bioanalysis, drug development is focused on supporting exposure measurements for pharmacokinetics, pharmacodynamics (biomarkers) or safety (i.e., toxicokinetics, immunogenicity, or therapeutic drug monitoring). While bioanalytical groups within pharmaceutical companies often ask individuals to perform numerous functions such as method development, validation, study sample analysis and data reporting, CROs generally have separate groups to perform these individual functions. When separated, the knowledge gaps across the individual functions can limit a CRO. Line and project management have a vital role in ensuring that any gaps are eliminated. Accountability and speed are critical reasons why pharmaceutical companies often employ a ‘full function’ design for bioanalysis. The counter risk is that a limited number of individuals are responsible for much of a drug product’s bioanalysis, making it essential that each individual is fully trained and compliant with procedures. Proper management and quality assurance oversight are needed to avoid having a single point of failure impact the entire product’s development. Therefore, organizations that employ a single analyst model should consider how to reduce risks. Assay problems are often exposed at method transfer. Requiring other scientists to execute some portion of the work, whether within the organization or to a CRO, is a good practice. Speed is a key requirement for a discovery CRO, which may be required to formulate, dose, analyze and report pharmacokinetic data within a few days of compound receipt. Within drug development, timelines are longer and studies more complex. As a result, quality and cost often trump speed. An obvious exception is a rapid dose-escalation study of a narrow therapeutic margin drug for which a 48-h turnaround of preliminary data may be needed. The rules of firstto-file generic bioequivalence also stress the need for speed, in addition to capacity and quality. No one

future science group

The science of laboratory & project management in regulated bioanalysis 

wants to make decisions on incorrect data; therefore, quality can never be sacrificed for speed. The separation of individual roles within a development CRO has great value, including control, compliance, training and cost. Each part of the process can be independently verified by quality assurance and managed by functional experts. This allows for optimization of individual processes, as well as the development of technical and scientific expertise within the subdiscipline. The codification of these procedures with the laboratory standard operating procedures (SOPs) is critical and serves as guidance to staff. A good laboratory will have well thought-out SOPs that are in full alignment with global regulatory standards and cover every task. Included within the SOP is best practice that reduces risks to both the laboratory and client. Leadership of individual teams by a subject matter expert helps to ensure that training and oversight meets the highest standards. Having exactly the properly skilled staff perform the function also reduces costs and therefore the price of bioanalysis. Scientific development Staff development proceeds through mastery of individual roles, then re-assignment into new ones. With bioanalysis, the minimum requirements should include a BSc degree, preferably biology, chemistry or a closely related discipline. A new scientist may learn to login or aliquot samples. Others may start by supporting the preparation of extraction solutions or mobile phases. Focus allows the individual to master each assignment. Those with sufficient talent and ambition will move into sample extraction, automation or LC-MS operation. Some laboratories will separate validation from sample analysis due to the unique requirements within a validation. Experience in both is a must for progression to more senior positions. Access to scientific literature and meetings is a critical investment and should be supported. Knowledge gaps are inexcusable as searching tools and electronic libraries have put knowledge in easy access for all. Those with several years and a breadth of experience, including the ability to troubleshoot problems, may move into the senior ranks of method development or project management. Moving from the technical into the scientific track is a significant accomplishment for a scientist and should be equivalent in grade and expectation to a PhD-level scientist. Since BSc-level scientists are often home-grown and know their organization, they can out-perform a new PhD scientist for some time. Having access to quality graduates from outstanding universities is a must for any research organization.

future science group

Perspective

Individuals and their managers need to have regular career development discussions to ensure that staff are fully challenged and developed. Promotional review committees comprised of managers are one way to make certain that talent across an organization is both recognized and fully developed. Having a deep scientific track is another way to ensure that development is recognized and supported. Those with strong communication and organizational skills, a history of managing projects or teams, outstanding scientific leadership, and the desire to mentor or develop others may be selected to move into the management track. Within a healthy organization, great managers will also show strong scientific leadership. Risk-reward Scientists who perform method development should be encouraged to investigate new approaches. Too often, methods are duplicated copies of preceding work. While this approach may work within discovery or when developing backup chemotypes, each therapeutic class compound should be considered as a new challenge. Knowing what others know about a class of compounds is an important first step. While this should include searching for compounds within the class, structural similarity searching can uncover common chemical features across different therapeutic classes. Both approaches should be used to uncover what others already know prior to starting laboratory work. Taking unnecessary risks, such as the simultaneous determination of numerous metabolites, should be avoided. The US FDA has allowed the use of tiered assays to investigate early metabolism. This approach allows individuals to use nonvalidated assays, often with biologically sourced metabolites as reference calibrators, to estimate exposures across species [5,6] . Both the International Conference on Harmonisation and Metabolites in Safety Testing guidance require steady-state coverage in at least one primary toxicology species for circulating human metabolites [7,8] . How organizations accomplish this objective is left open until clarity is established on what must be measured to regulated standards in larger clinical trials. Unless one is trying to understand a specific toxicity that implicates unique metabolic activation in one species, there is little reason to pursue measuring numerous inactive metabolites in multiple species or tissues. Tiered assays are analogous to fit-for-purpose Key term  LC-MS: Separation of drugs or metabolites within a liquid chromatograph based upon their physical–chemical properties and measurement of concentration by response within a mass spectrometer.

www.future-science.com

1359

Perspective  Unger, Lloyd, Tan, Hou & Wells biomarker or metabolomics assessments, a screening exercise to determine what is worthy of more rigorous effort. Adding and later subtracting them from a core validated assay is a poor and inefficient strategy for drug development. There are numerous cases where a combination assay is needed and easily justified. These are mostly human plasma assays where the intent is to cover activity or a specific drug–drug interaction. Examples here include the drug–drug interaction cocktail assays that evolved from cassette dosing approaches or the use of combination therapies in oncology or virology [9–12] . Provided one has stable label internal standards, these assays are generally worth the risk of development as they accomplish numerous objectives at once. Coformulation of individual medicines into a single product is a common approach within anti-infective and oncology therapeutics and, whenever possible, should be supported by a combination assay. Innovation The FDA has recognized that failures to get new medicines to market may be related to a lack of innovation and have encouraged the use of new technology [13] . Innovation is best tried within discovery organizations and, if successful and sufficiently rugged, later transferred to development. An example of the difficulty in introducing new technology into toxicology studies includes the extensive efforts of Nicholson and others in launching the Consortium for Metabonomic Toxicology [14] . Metabolomics (or metabonomics) was first deployed in late discovery (non-GLP) toxicology studies to avoid the complexity of interpreting complex system biology results within a GLP study. It has now seen more general acceptance, with CROs having built a business model about a metabolomics platform [15] . A similar problem was seen when Caprioli and others introduced MALDI tissue imaging as a supporting technology to histopathology [16] . Acceptance within a GLP study was generally performed as a supportive technology to augment an earlier observation by traditional means. Today, there is one CRO whose entire business model is dedicated to MALDI imaging. Alternative technologies such as desorption electrospray ionization imaging have also made progress in molecular imaging [17] . Molecular imaging has the potential to answer fundamental questions regarding Key terms  Regulated bioanalysis: Measurement of drug or metabolite exposures that meets regulatory standards. Matrix management: Management of cross functional laboratory teams by Project Directors.

1360

Bioanalysis (2014) 6(10)

metabolism-mediated toxicity and target engagement [18] . While traditional means of harvesting and analyzing tissues will continue to be used, the speed and spatial information that one can obtain using imaging will allow comparative studies to provide great value. In bioanalysis, innovation can be as simple as the introduction of new column chemistry, such as monolithic, core shell and hydrophilic interaction chromatography, or the introduction of new instrumentation such as ultra-high performance liquid chromatography (UPLC) or multiplexed HPLC [19–22] . New technologies require a champion for successful transition into regulated bioanalysis. Most individuals within regulated bioanalysis have intolerance to risk, and there are always skeptics of anything different that creates resistance to change. In addition to direct costs of new technology, the amount of time to qualify instrumentation, validate software, revise procedures and train staff presents high hurdles. To foster innovation, managers must reward individuals who take the risk of introducing new technology, particularly within the field of regulated bioanalysis. One recent example is that provided by GlaxoSmithKline in championing the broader use of dried blood spot technology in routine toxicokinetic and clinical bioanalysis [23] . As a result, there has been considerable investigation into microsampling, including highlighting the complexity and uniformity of dried blood, as well as approaches to overcome sampling problems. Matrix management of projects & laboratory resources Project management is a critical part of any matrix management organization. By allowing selected individuals to manage projects, they can focus entirely upon study requirements, timelines and deliverables. Within a bioanalytical CRO, the Project Director or Project Manager serves a role that is similar to the Study Director. They are the central point of contact and responsibility for the study. When assigned to specific clients, a strong relationship between the CRO Project Director and the client Study Director or Study Monitor generally develops. This relationship of trust is never given; it is earned by daily execution of high quality work in a timely manner. The CRO Project Director must fully disclose and quickly overcome any laboratory limitations. They serve as both a champion and consultant for the client, reducing risks with proper planning, explaining CRO procedures, ensuring laboratory compliance with client-specific requirements and resolving any problems. Regular and accurate communication with laboratory staff or management and the client is a primary requirement.

future science group

The science of laboratory & project management in regulated bioanalysis 

Matrix management requires regular and effective communication between the project managers and resource providers. When properly executed, matrix management aligns the best resources at the best times for a successful outcome. Project Directors have critical roles in representing their client’s needs, while Laboratory Managers need to fully represent their resources on an ongoing basis to support rapidly changing timelines. This process requires weekly planning and daily adjustments to fully optimize effectiveness. The Project Director is fully dependent upon laboratory execution, which is the critical foundation for a successful study. Safety Laboratories are both fascinating and dangerous work environments. Laboratory Managers need to ensure that their staff work in a safe environment. The analytical laboratory will have exposure to chemicals, which requires compliance with federal, state and local agencies. A well-written chemical hygiene plan and an effective safety committee are tools that help to ensure safe operation. Having the right culture for safety includes the support of co-workers to call out noncompliance and to take immediate corrective action. Staff must know how to access and comply with safety data sheets, which detail the required personal protective equipment, as well as the disposal and shipping requirements for the chemical. When working with chemicals, adherence to engineering and work practice controls, as well as proper protective equipment should always be enforced. Besides gloves, laboratory coats, safety glasses and chemical fume hoods, ventilated cabinets around balances and devices to reduce static should be used to avoid the accidental inhalation of particulate. Regular monitoring of air exposure to volatile organic chemicals should be performed to ensure compliance with permissible exposure levels set by Occupational Health and Safety. The chemical fume hood plays a major role in protecting laboratory staff and should be regularly monitored for proper operation. The largest source of solvent vapors occurs when liquids are heated within evaporators or MS ion sources. Generally, these systems have good containment with powerful blower fans to exhaust vapors. Building design should include sufficient exhaust with wide diameter tubing to avoid restricting flow. Liquid handlers that manipulate large volumes of solvents at room temperature should also be enclosed within ventilated cabinets, which are regularly tested for proper operation. Analysis of volatile organic chemicals badges worn by staff provides an 8-h sampling of their work environment and will illustrate any procedural problems.

future science group

Perspective

Poorly designed and densely populated LC-MS laboratories have noise problems. Long-term hearing losses can be reduced using ear protection and by isolating or soundproofing vacuum pumps. Some LC-MS laboratories use carpet tiles to reduce noise, but spill contamination can be a problem. When servicing LC-MS systems, exposed high voltage and gas pressures also present hazards, which lock-out practices should address. Staff should be well trained to respond to spills of limited size. Organizations often develop Hazmat Teams with specialized training and equipment that are capable of handling larger spills. Emergency Responders are generally needed for large spills or for more dangerous chemicals. Secondary containment is critical to reducing accidents, particularly the breakage of large solvent bottles. Significant spills have significant impact. Laboratories should also have a pollution prevention plan that will reduce or recycle its waste stream. Chemicals are expensive to purchase and expensive to dispose. Substitution of less toxic compounds, reduction in use, and access to a chemical inventory to reduce the purchase of redundant chemicals are ways to reduce costs and waste. All solvent waste must be properly stored until being disposed by a hazardous waste disposal service that is properly licensed to accept such wastes. Laboratories performing bioanalysis have additional safety requirements. Universal precaution assumes that all human blood products to which one is exposed are infected with blood-borne pathogens. Managers must use the ‘universal precautions’ approach to protect employees by implementing the use of important techniques: special engineering controls such as biosafety cabinets, work practice controls and personal protective equipment to help the employee avoid being infected by blood-borne pathogens. The CDC is an effective source of information to assist any laboratory in the development of their blood-borne pathogen guidelines. Vaccinations and immediate medical treatment of incidents are also important tools to reduce the impact of biological infection. Whether risks are chemical, electrical or biological in nature, vigilance is required to maintain a safe laboratory. Requiring staff to rotate through safety inspections is a good means to ensure that each individual assimilates group values for safety. Many organizations may have health and safety experts, but safety is every scientist’s responsibility. Laboratory design Unless they are starting a new laboratory or moving one, few managers have the opportunity to design a

www.future-science.com

1361

Perspective  Unger, Lloyd, Tan, Hou & Wells laboratory. More typically, a Laboratory Manager may modify or expand a present laboratory. In addition to guidance from engineers and planners, soliciting advice from other scientists is a wise first course of action. Staff needs to be actively engaged in establishing the user specifications for the project as they will be required to work in this environment. The proper storage of precious samples should be top consideration (walk-in vs standalone freezers; or centralized vs individual laboratory freezers). The conduct of the study is more expensive than sample analysis and samples need to be protected at any cost. No laboratory freezer or refrigerator should be without temperature monitoring, sample tracking and generator backup power. Generators should be regularly tested. All freezers must be on temperature monitoring with automated calling in the event of a power failure. When performed at a CRO, centralized storage with restricted access is the norm. High density storage systems are routinely used in newer walk-in freezers to improve capacity, while reducing footprint and energy costs. Uninterrupted power supplies generally last for less than 20 min and are used only as a bridge to generator power. If deployed throughout the LC-MS laboratory, uninterrupted power supplies are expensive and often unnecessary. Provided there is sufficient extract stability, re-injection is always possible. If not, re-analysis of a fresh aliquot is needed. Many CROs have continuous 24-7 operation of the LC-MS laboratory and will respond quickly in the event of power failure. Line power should be conditioned and be able to withstand a power surge that could damage systems. Monitoring of noise or voltage fluctuations is required by most manufacturers prior to the installation of new instrumentation. Spectral (frequency) analysis should be performed if noise is present to determine its source. MS systems using analog electron multipliers are more sensitive to low frequency (1/f) electronic noise than those using pulse counting electron multipliers. Wet chemistry laboratories are very expensive and should be well thought out. Hoods are also costly; therefore, the location and access to chemical fume hoods and biological safety cabinets need to be carefully planned for both function and safety. Working too closely can result in accidents and contamination; allow at least three linear feet of hood space and an overall laboratory space of 100 sq ft per person. In largely populated LC-MS laboratories, heating and noise can be greatly reduced by locating roughing pumps in contained areas. This can include commercially available vacuum pump enclosures or the design of a separate corridor for the pumps. Sufficient air conditioning needs to be provided to handle the high heat load of instrumentation.

1362

Bioanalysis (2014) 6(10)

Ways to control the storage and release of reference compounds in a manner similar to what is done for radiolabeled and controlled substances should be considered. Contamination can be reduced by storing dry powder and making primary stock preparations remote to the laboratory where extraction or LC-MS analysis is performed. Having a separate, isolated reference standard laboratory will greatly reduce contamination when performing trace analysis. Whenever possible, once-only use of reference compounds from ampules can also reduce contamination. Laboratories should have a disaster recovery plan, which defines exactly how to re-establish operations in as little time as possible in the event of an emergency. With many software applications storing data directly to servers, a laboratory interruption may have limited impact. However, a server failure can precipitate a major incident. New clients should audit both laboratory and IT facilities, looking at data backup and archival practices, as well as instrument qualification and computer software validation practices. When planning, laboratory and server facilities should be separated. Each laboratory is unique and will have its own requirements. Taking the time to plan and build facilities will determine how effective the laboratory will operate. Once completed, the Laboratory Manager must decide how to equip the laboratory. Technology Since a large portion of the cost of a bioanalytical laboratory is its capital investments; managers need to be aware of the potential for changing technology to impact their laboratory investments. Limitations here include intellectual property rights, which can block their competitors from introducing better or more costeffective models to users. Laboratory Managers need to keep a close eye on emerging technology and patents to avoid having a laboratory with antiquated instrumentation. The enforcement of patents has caused buyer’s remorse when shipments were impounded or engineers performed field modifications that restricted instrument performance. Becoming accidentally entangled in intellectual property fights is something that every Laboratory Manager should avoid. Leasing rather than capital purchasing has been increasingly used to reduce the risks of new or changing technology, with numerous third-party leasing and re-sale companies entering the market. Terms for leasing are tied to the residual market value of the asset. With the large amount of equipment made available from pharmaceutical downsizing, the value of instrumentation has declined. Therefore, leasing terms are not as attractive as they were 15 years ago. This glut

future science group

The science of laboratory & project management in regulated bioanalysis 

of used equipment has also decreased the value of new instrumentation, particularly when manufacturers are unable to improve the performance of new systems from past models. In an effort to keep the cost of new instrument as high as possible, some vendors have blocked used instrumentation from re-entering the market by refusing to provide software licenses to buyers and intimidating owners over license agreements. Software cannot be transferred so instrument companies ultimately influence the fate of used equipment. In addition to software licenses, buyers who purchase used instrumentation generally also need to secure service contracts from the vendor. Whether it is new or used instrumentation, buyers need to heed investor’s advice: ‘Caveat emptor’. Within the field of LC-MS, the cost of instrumentation dominates capital acquisitions. Generally, the highest performance triple quadrupole LC-MS systems have maintained a cost of at least US$300,000, while the less sensitive or prior model instrumentation has been priced at approximately US$200,000. Laboratory Managers need to have a long-term plan regarding their capital budgeting, which considers the lifetime and depreciation of their investments. In the past, it was common to have in-field upgrades of mass spectrometers. Thermo Finnigan once offered single-to-triple quadrupole upgrades in your laboratory. Due to the cost and time to ship, high-resolution mass spectrometers were routinely also upgraded in the field. Today, systems are returned to be refurbished and re-sold. Applied Biosystems Sciex has introduced a total of seven new models since 1995, and only the API 300/365 was offered as an in-field upgrade. Most scientists expect at least 10 years of service from a mass spectrometer. Maintaining capacity and a state-ofthe-art fleet is a challenge with model changes every 2–3 years. It is therefore important that Laboratory Managers fully consider technology improvements and decide whether vendor claims actually translate to performance. In the field of immunoassay or ligand binding analysis (LBA), equipment costs can be relatively low unless buying patented technology, such as microfluidics, bead-based assays or laser fluorescence and electrochemiluminescence detection. The cost of LBA is generally tied to reagents (e.g., antibodies and receptors), with some kits selling for as much as several thousand dollars per 96-well plate. Exact replication by others is not possible, keeping reagent costs high. Vendors often provide technology that requires a laboratory to use their products. Luminex-, Mesoscale Discovery- or Gyrolab-based assays are examples. Luminex offers their bead-based xMAP® technology as a means to

future science group

Perspective

Key term  Ligand binding analysis: Assay that measures concentrations using a binding reagent such as an antibody or receptor and a means of detecting the amount of bound drug or ligand.

multiplex fit-for-purpose biomarkers assessments using proprietary assays. The Mesoscale Discovery SECTOR® Imager incorporates multiarray electrochemiluminescence detection for wider dynamic range assays, but requires the purchase of proprietary reagents. The Gyrolab™ platform offers advantages when sample volume is limited or reagents are expensive and the chance to build your own assays. However, the microfluidic disks add significant cost to an assay. Others may charge little for their instrumentation but make money from its use. Included in this are 96-well kit manufacturers whose assays can be adapted for regulated bioanalysis [24] . This is a good model for a CRO since they can control their upfront, capital investments while maintaining a fixed cost when assaying samples. Often, this cost is charged back to the client. However, the risk of buying large numbers of expensive LBA plates only to have a cancelled study is something that needs to be managed. LC-MS instrumentation It is difficult to understand the present or see the future without knowing the past. Mass spectrometry dates to the Cavendish Laboratory at Cambridge University of JJ Thompson and FW Aston. There have been enormous improvements over the past 40 years, making it the workhorse of a modern bioanalytical laboratory. MS/MS evolved first as a means to study ion chemistry and second as an analytical alternative to GC-MS [25] . A mixture of ions representing the individual components was separated using a mass analyzer, dissociated in a high pressure collision, and the fragments resolved in a second analyzer prior to detection. While MS/MS was first performed using magnetic sectors, the quadrupole mass spectrometer was developed as a less expensive, fast scanning MS [26,27] . Unlike magnets that suffer from hysteresis, voltages are easily ramped allowing quadrupoles to be scanned at high speed. By adding a radiofrequency-only collision quadrupole and another mass-analyzing quadrupole to a single quadrupole, the triple quadrupole was born [28] . Prior to the advent of LC-MS, the ion source was located within the high vacuum chamber. Due to the high gas load of liquid flow stream, a simple interface could not be achieved on most commercial instrumentation. Numerous interfaces were designed and tested to overcome this limitation. Henion was the first to promote the use of the Sciex TAGA™ atmospheric pressure source as the best LC-MS instrument [29] . It

www.future-science.com

1363

Perspective  Unger, Lloyd, Tan, Hou & Wells was the only quadrupole designed to operate at atmospheric pressure, using a large cryopumping system with molecular skimmers to handle the high gas load. Early atmospheric pressure ionization (API) sources put demands on vacuum requirements and losses were great when transferring ions made at atmospheric pressure to the high vacuum mass analyzer [30] . Much effort over the past 20 years has been spent trying to improve the transmission of ions through this region. Two fundamentally different sources are used for most bioanalysis, electrospray (ESI) and atmospheric pressure chemical ionization [31,32] . Since ESI is the most commonly used interface, we will consider it as an example of changing technology. Analytes do not need to be thermally stable, so there is no need to vaporize the sample to transfer it into the gas phase. A sustained ESI is possible at flow rates

The science of laboratory and project management in regulated bioanalysis.

Pharmaceutical drug development is a complex and lengthy process, requiring excellent project and laboratory management skills. Bioanalysis anchors dr...
894KB Sizes 0 Downloads 6 Views