You are on page 1of 24

Drug Development Overview For the purpose of this summary, drug development is defined as those activities required for

advancing a biomedical concept to a medical product in commerce. This definition encompasses such endeavors as genomics, target validation, drug discovery, development candidate identification, drug safety assessment, regulatory filing, clinical studies, and registration for market. After a successful drug development effort, commercial launch, marketing, distribution, pricing, reimbursement, and product life-cycle management are the kinds of activities that occur. During drug development, collateral support work is typically ongoing. These activities include intellectual property creation and securing patents, pharmacoeconomic and pricing range assessment for the potential product, identification of physicians who would eventually prescribe the approved drug, and physician surveys on the product candidate and the medical need for it. The general steps for drug development are very well delineated. However, the specific details of drug development are extremely elastic and highly dependent upon the nature of the medical problem and various approaches to that problem, the risk tolerance of the drug development organization, the ever-changing landscape of medical care and reimbursement, the fluid but highly conservative nature and demands of regulatory bodies, and the work of other organizations on similar product candidates. Development activities are discussed below. A tiered structure is presented wherein the levels of development represent natural break points in the cost and regulatory requirements for the specific stages of drug development. Of course, there will always be some overlap between the levels as the product candidate profile evolves and as the specifics of the project demand. Most importantly, every drug development effort is unique in some way. Each will require its own roadmap to registration, the details of which are highly dependent on the specific issues that arise in the course of discovering and advancing that drug candidate. Level 1: Drug Discovery The goal of Level 1 activities is to identify a drug product candidate for subsequent evaluation in a clinical trial. Drug discovery encompasses an enormous spectrum of activities depending on the scientific maturity of a particular project. Activities that may take place at this level include a first-in-man clinical trial of an established drug in a novel, unrelated clinical indication; the identification of a novel indication for an established drug; the identification of a new dose, schedule, or route of administration or formulation for an established drug; and the identification of a new molecular (chemical) entity (NM/CE) for a medical indication. All of the above, except the last, can lead to an abbreviated, somewhat less-expensive path to regulatory review and approval of the product because the molecular entity is already in clinical use. Such products can be viewed as either pedestrian or true innovations by the regulatory and medical communities, depending on their effect on the medical condition they are intended to treat. New chemical entities present the greatest potential but the most costly challenge in drug development. Even within this category there are both true innovative medicines and entities that are derivative (me-too) agents of a first-in-class drug. Such follow-on drugs may only offer marginally, though clinically meaningful, improvements over the original novel drug. Me-too drugs can prove to be spectacular commercial successes. NME drug discovery typically begins with a biologically significant molecular target or biological process thought to be causally connected to the medical condition to be addressed. The immediate goal is to identify a molecule or collection of molecules that perturbs this key biology in the desired manner. Such a molecule is called a lead or a hit. The search for this molecule or

group of molecules is conducted by establishing a screen that measures the impact of molecules on the targeted biological activity. The biological screen can be a high-throughput one for evaluating large numbers of potential leads or a low-throughput one for detailed characterization of leads. The former tend to be simple in vitro binding/displacement/enzymatic/ target X-ray crystal/NMR-derived structure (de novo; fragment-based drug design) assays, and the latter tend to be tissue and in vivo pharmacology assays. These assays are typically layered as filters both to confirm the initially observed activity and then to assess the drug potential of leads. De novo drug design strategies involve using the molecular structure of the target derived from molecular modeling, X-ray crystallography or NMR-based structural studies as a template for design of hit molecules that will bind to the target and perturb the targets activity in the desired fashion. Some organizations parse the definitions hit and lead. Usually, a hit has an unvalidated activity in an in vitro screen, whereas a lead has some degree of validation of activity. The types of hit validation that can advance a molecule to lead status include dose-response, target selectivity, a Structure-Activity-Relationship (SAR) among analogs, activity in higher order assays such as cellular or tissue assays, and/or efficacy in relevant pharmacology models. Once the leads have been identified and activity confirmed in multiple assay systems, questions immediately arise as to the characteristics of the leads that make them less than ideal drug development candidates. When pharmacology assays were the primary source of drug leads, it was not uncommon for the lead to be the development candidate. That situation is far rarer today, as in vitro screening leads seldom have the necessary pharmacological characteristics to make them viable candidates for advancement. Hits and Leads Advancement Criteria Typically, a hit compound demonstrates activity in an in vitro biochemical screening assay in the 1 to 50 M range. At this point there are two important objectives: 1) to demonstrate activity in the next higher order assay and 2) to demonstrate a structureactivity relationship (SAR) among analogs. Ideally, the hit demonstrates positive activity in a more physiological cellular assay. The original screening may have been done in cells, so a counter screen in cells (or a biochemical counter screen) for selectivity becomes useful in validating the hit. Potency frequently erodes 10- to 100-fold between a biochemistry assay and a cellular assay because of factors such as competition with high intracellular concentrations of substrates, ligands or cofactors, or poor cellmembrane passage by the hit. An alternative to confirmation of activity in a cellular assay is the appearance of SAR among closely related analogs when they are tested in a biochemical assay. Ideally, there will be a broad range of potency among analogs, including the disappearance of activity altogether for some analogs. Optimum hits display good ligand efficiency in their biological effects. Ligand efficiency is the ratio of biological potency or efficacy per number of non-hydrogen atoms in the molecule. The lower the ratio (e.g., nM binding/heavy atom is of greater efficiency than M binding/heavy atom), the better the hit is as a starting compound for optimization. An alternative to identifying lead molecules by screening or de novo drug design is called metoo drug discovery and patent poaching or busting. Here the leads are derived from reports on competitors NME discovery efforts. The goal is to identify novel leads based upon morphing the literature leads into an analog that is patentable and may display characteristics that are improvements over the original literature compounds. Frequently these competitors compounds serve as benchmarks and comparators for identifying improved potency, selectivity, efficacy, safety, ADME/PK, etc., in a new lead series. The major advantage of me-too drug discovery is

that the uncertainty surrounding the medical viability of a first-in-class drug is resolved, so the risk of technical failure is much diminished. Once a valid lead molecule is in hand, the iterative process of drug optimization commences. The goal of this effort is to identify a patentable molecule with the optimum balance of safety and efficacy, and to do so in a relevant pharmacology model of the targeted medical condition. Alternatively the drug candidate upon dosing in an animal achieves a blood (or tissue)-level that exceeds the exposure concentration expected for efficacy in man based activity concentrations in vitro. Such an effort is accomplished by chemical modification of the original lead structure, followed by systematic testing of the new analogs in a hierarchy of biological assays. Once the biological test results have been evaluated, the structure-activity relationships derived from analysis of the data for multiple compounds are used to drive design of additional molecules for preparation and testing. The goal is to eliminate or at least diminish the biological deficiencies seen in the initial compounds. This iterative process continues until a molecule and one or more back-up molecules are identified that meet all the criteria for advancing them into clinical study. This lead optimization activity is the essence of the art and science of medicinal chemistry and by far the most laborious and expensive stage in the drug discovery process. The choice of specific molecules to be prepared is based on the experienced intuition of the chemists and on the evolving structure-activity relationships for potency, selectivity, metabolism, toxicity, absorption, clearance, and efficacy that appear when the compounds are bio-assayed. Additionally, an important consideration is the practicality of synthesizing the specific compounds evolving out of the SAR. Types of Biological Assays Assays are as varied as all of biology. In vitro assays are typically biochemical in nature or use cell-based systems. In vivo assays include animal models of disease as well as side effects/safety models and models of drug disposition (Absorption, Distribution, Metabolism and Excretion ADME). Although less frequently used today in primary screening, tissue-based assays are particularly biologically relevant. Target tissuespecific models provide excellent measurements of biological activity with fewer complications than are seen with whole-animal models. Biological features of molecules such as potency can erode as compounds are advanced from simple molecular targetbased assays to cellular assays and animal assays. The specific physicochemical properties of molecules, the route of administration, and the nature and components of rudimentary formulations can greatly affect validity and real-world meaning of data generated in a given assay system. Interpreting the specific biological data is a nuanced endeavor. The interaction of a molecule with a drug target at the protein, cellular, and in vivo levels is subject to many caveats. Factors such as competitive, un-competitive and non-competitive binding modes, binding residence times, binding kinetics, substrate concentrations, substrate/cofactor affinities, receptor/channels reserves, conformational states of ion channels, substrate processing kinetics, the targets post-translational modification state, the cellular location of the target, the intracellular location of the target, active transport into or out of cells, vascular blood flow through the target tissue, full/ partial/inverse characteristics of antagonism, various types of agonist activity, specifics of the transduction cascades, and many other factors including the often-mentioned ADME factor can disrupt the straight line from target binding to in vivo efficacy in animals and in the clinic.

Development candidates will display an optimized balance of favorable and less favorable biological features. The back-up candidate should have a different liability profile from that of the nominated molecule and preferably have an entirely different molecular architecture or scaffold from the lead compound. Optimization of all the desirable features of a new drug seldom marches forward unimpeded. Compromises about the desirable features of a drug are inevitable as the medicinal chemists home in on the optimum candidate. The prize is usually a properly balanced drug, not a perfect drug. The Process of Lead Optimization With a validated hit(s), now called a lead(s), in hand, lead optimization can commence. Usually, the immediate goal for the chemists is to drive potency from double- to singledigit micromolar range into the low double-digit or lower nanomolar range in one or more biochemical or cellular assays. For agonists, low micromolar potency may be adequate for yielding drug activity in vivo. Selectivity among related targets is frequently desired, so counter screens are used to guide SAR development. Anywhere from 5- to 10,000fold selectivity for the biological target over related targets or even unrelated targets is the goal for off-target selectivity. Assay data guides the chemists in formulating an SAR to aid in the design of the next generation of analogs to be prepared for testing. Chirality has become one of those molecular features of drugs that are viewed as critically important for activity and safety. It is seldom acceptable to introduce a racemic drug into commerce. Typically all the biological activity is present in one enantiomer. However a racemic drug contains 50% of an enantiomer that typically provides no benefit to the patient but does increase the exposure to a foreign substance that may carry a significant safety liability. Depending on resources and corporate drug discovery philosophy, additional characterization studies on new leads will generate a broader range of data to help the medicinal chemists optimization efforts. These studies include compound solubility, membrane permeability, protein binding, active cellular transport, cytochrome P (CYP) 450-driven metabolism, inhibition of CYP 450, and hERG ion channel blockage. The value of these collateral profiling data is to identify features of molecules that suggest certain potential problems with the analogs that may compromise their development into safe and effective drugs. Frequently chemists will alter their analog chemistry to address these potential liabilities. However, compound optimization can be driven exclusively with only potency and selectivity data (or with only in vivo-derived efficacy and safety data). Information about molecular sites of metabolism can suggest a strategy for modification of the lead structure to block or enhance metabolism and prolong or shorten drug in vivo activity. It is imperative to begin assessing the efficacy and safety of leads in animal disease models as early as possible. Ideally, the initial hit displays pharmacological activity, as it is always more efficient to address deficiencies in an in vivo lead than in an in vitro lead that has yet to demonstrate in vivo activity and safety. Targeting potential therapeutic blood or tissue levels of leads in animals after dosing, especially if no disease model exists (as with HIV, for example), is a good alternative to in vivo efficacy. In vivo dosing should not exceed 35 mgs per kg, although this is only a guideline. The compounds are formulated in any number of modified standard animal dosing formulations. Early on, intraperitoneal (IP) dosing affords animal exposure with the minimum number of confounding variables. Dosing starts with a dose-escalation study to assess in vivo safety and perhaps efficacy. Once the maximum tolerated dose is identified, chronic dose-ranging studies are initiated. Ultimately, a standard dosing schedule is established

for efficacy determination. As improved compounds are synthesized that are effective with IP dosing, the route of drug administration is switched to that expected to be used in the clinic. All advanced molecules must then demonstrate efficacy in this standard testing format and ranked against a benchmark compound such as a literature compound or an earlier less advanced lead. The compounds that display the best in vivo efficacy and safety profile (plus the optimum in vitro profiles) become potential development candidates. Certain developmental criteria such as synthetic accessibility, bioavailability, metabolic profile, protein binding, in vivo half-life, water solubility, etc. may lead to favoring certain active compounds over others. Drug Development Candidate Criteria The designation of a development candidate is the ultimate goal of drug discovery research. The specific criteria that define a development candidate can be highly variable and depend on the target medical condition and treatment population. Typically, potency in a biochemical assay will be in the sub-1 to 100 nanomolar range. The potency in cellular assays can range from nanomolar to 10 M. The dose in a rodent disease model can range up to 150 mg per kg but is ideally less than 35 mg per kg. The minimum safety window is 10-fold, but safety windows of 3- to 5-fold are not unheard of, especially for life-threatening medical conditions. Oral bioavailability should be greater than 20%, and protein binding should be less than 80%. There is no rule of thumb for circulating drug half-life, although 8-12 hours allows for convenient dosing. Inhibition of hERG activity should be 100 M or above. CYP3A4 and CYP2D6 inhibition should also be at concentrations greater than 30 M. Water solubility greater than 50 g per mL is preferred. While these characteristics describe the ideal drug development candidate, drug discovery and development candidate selection will always be subject to trade-offs, and a candidate will seldom match the ideal. Each drug discovery program has its own unique set of issues. These issues depend on the molecular validity of the approach to the targeted disease; the relevance and robustness of the assays; the quality of data and lead compounds; the availability of funds, manpower, and time; the drug discovery philosophy of the organization; biases, experience, expertise, and insights of the scientific staff; and the onsite presence or absence of Lady Luck. Addressing the various issues and roadblocks that arise in a drug discovery process is a continuing dialog among the scientists and requires flexibility and a steely-eyed assessment of the progress and chances for a successful outcome. Scientific management is typically responsible for measuring program progress and advance/kill decisions. The final product of all this research activity is a declared development candidate that has the greatest number of desired features and no unmanageable liabilities, with the definition of unmanageable liabilities being highly dependent on the specific medical indication and the specific patient population. Level 2: Early-Stage Drug Development The goal of Level 2 activities is to advance a drug product candidate into the clinic so as to demonstrate preliminary efficacy and safety in humans in the target medical condition. While the requirements for advancing a drug through Level 2 can seem rather proscriptive, there can be some elasticity in the specific activities, including manufacturing, toxicology/safety studies, clinical trial design, regulatory requirements, and depth of efforts required to bring an NME through Phase 2 clinical studies. Again, the specifics are very much dependent on the medical indication and the specific patient population, as well as on the ever-evolving regulatory environment.

Multiple activities are initiated once the decision to advance an NME into the clinic has been made by senior managers. These activities include the manufacture of the drug, also called the Active Pharmaceutical Ingredient (API) or Drug Substance. Other activities are the development of the formulated drug, the Drug Product (DP), for human dosing; studies on the safety of the drug substance and formulated drug; preliminary toxicology studies and studies on the pharmacokinetics and disposition of the drug (Absorption, Distribution, Metabolism and Excretion [ADME]). The regulatory requirements for these activities leave the sponsor some latitude in the early stages of clinical development. This elasticity means that the degree of development, validation, and knowledge about the manufacturing process, quality controls, and safety may be fairly rudimentary at the beginning of development. The depth of knowledge about all these aspects of drug development is expected to grow substantially over time and result in ever higher quality standards and tighter control specifications for all aspects of manufacturing, distribution, storage, and clinical usage. The first step in drug development is the assignment of a project manager to coordinate and track all activities required to file an Investigational New Drug application (IND) with the regulatory authorities. Typically an IND filing is constrained by time and resources. There are always a large number of multidisciplinary activities spread over many different departments, each with other competing program demands. However, many of the studies required for an IND filing are interdependent. If completion of one task is delayed, the consequences can affect many other planned activities and thus drive up costs and lead to missed deadlines. It is the job of the project manager to make sure deadlines are not missed, commitments are kept, budgets are justified and not exceeded, hand-offs go smoothly, and the IND is successfully submitted to the regulatory authorities by the projected deadline for submission. The regulatory authorities could be the Food and Drug Administration (FDA) in the United States, European Medicines Agency (EMA) in Europe, or the Ministry of Health in Japan. The project manager seldom has line authority to make this all happen but relies on skills in cajoling, bargaining, sweet talking, horse trading, and the like to advance the NME toward the clinic. This is all tracked with a very public Gantt chart listing all aspects of the project, activity deadlines, and budgets. Regular meetings are scheduled with all responsible parties (the Project Team) where issues and progress are discussed and any changes to the plan approved. Project management is critical for the advancement of a development candidate to an IND filing and ultimately to registration for marketing. Key Early-Stage Drug Development Activities API Synthesis Process Identification and Optimization API supplies are on the critical path. Without significant quantities of API, few development activities can begin. The initial quantities of API used in development are supplied by scaling up, or worse, by performing multiple repeat production runs of the original medicinal chemistry synthesis. Rarely does the medicinal chemistry synthesis evolve into the commercial manufacturing process. The key goal of process chemists is to identify a reproducible, scalable chemical synthesis process that can supply the development effort and provide clinical grade material. Multiple routes of synthesis may be examined before the production synthesis is identified and optimized. Linear multiple step processes, solvent changes, exotic reagents and starting materials, tedious manipulations, and non-scalable steps like chromatography are to be avoided whenever possible. Also multiple steps should run together without isolating intermediates (telescoping steps) to shorten plant time utilization and reduce cost. If the API is a salt, the selection of the counter ion is a key aspect of synthesis development. Consistent production of the optimum crystal polymorph of the API for solid dosage forms and

stability assessment are also very important activities early in the development program. Initially the chemists focus on the final step of the process to optimize the purity and properties of the API coming out of that step. Ideally this work establishes the quality target of the API coming from future process development efforts. API coming from these process development efforts can be used to explore potential formulations and develop quality control analytical methods, including release, stability, and in-process analytical control methods. This material may also be used to assess ADME/PK properties of the drug and for development of bioanalytical methods for measuring drug concentrations in blood and tissues. Initially, process development work and early productions are done in large glass vessels. Major impurities are identified, characterized and methodology for purifying the intermediates and final product is developed. The initial purity of the API may not be optimum but as the process evolves the API purity is expected to approach >98%. Starting materials and their suppliers are identified. In the final selection of the production synthesis, the important considerations include the costs of supplies, solvents, and reagents; the quality of the API, viability and reliability of suppliers and the manufacturer of API, if a contract manufacturer is involved; and the ongoing availability of all these suppliers and supplies. Process safety, solvent safety, and waste products and quantities are also key matters to consider in process selection, as are environmental release, including waste stream volumes and worker exposure. Avoiding unsafe intermediates (explosive, highly toxic, thermally unstable) and reaction conditions is critical to the development of a commercial process. Close attention is paid to the thermodynamics and kinetics of reactions steps. There is a strong design bias against reactions that rapidly or unpredictably generate heat. Heat is much less efficiently dissipated in large reaction vessels because of their low surface area and should heat build too rapidly, a dangerous run-away reaction can occur. Also a Freedom-To-Operate assessment of the process must be completed to avoid any patent infringement issues. Costs of goods (COGS) are evaluated for commercial viability purposes. Stability on prolonged storage under varied storage conditions and packaging configurations is assessed for multiple lots of API. The product stability results can be dependent on the specific route of synthesis. Degradation products are identified and monitored in stability studies. A rudimentary specification for acceptable API quality is developed which includes listing the expected impurities and degradation products. Failure to meet this specification causes the batch to fail, which means it cannot be used in the clinic unless it can be reworked to meet the specification. Batches fail 5-10% of the time leading to product being reworked or discarded, both at considerable cost. API Supply, Factory Manufacturing, and Scale-Up Demands for API rapidly grow, and drug supply is frequently the major bottleneck early in development. Hundreds of grams of material are usually needed and must be supplied one way or another. Once the process is selected, optimization for yield and quality begins. Scale-up is critical to supply the increasing development demands and demonstrate the commercial potential of the process. Multi-kilogram quantities of API become available to supply ongoing development activities. The transition out of glassware and into stainless steel or Hastelloy kettles is an important milestone. Demonstrating equipment compatibility with the process is crucial as manufacturing development progresses. Most importantly, the quality of API coming from production runs begins to look representative of the material that will be used in the clinic. The impurity profile should stabilize and the desired crystal form (polymorph and crystal sizes

and shapes, known to chemists as crystal habit) should begin to be consistently produced. Once the process and quality of production material are stable (or not if the time to IND is compressed), a dedicated production run commences to produce GMP (Good Manufacturing Practices) material for GLP (Good Laboratory Practices) toxicology studies and perhaps clinical use. The GMP standard is a regulatory requirement for material going into clinical use. GMP standards require detailed production record keeping; sources and lot numbers of raw materials, reagents and solvents are well documented. The proper functioning of laboratory and production equipment is verified. A batch record, essentially the detailed recipe to be followed during GMP production, is drafted and then populated with specific information including solvent weights, reaction temperature ranges, vacuum pressures, and other criteria to be adhered to during production. Deviations from and changes made in the batch record are noted, justified, and signed off by production workers and managers. The final material must meet or exceed the product specifications that were designated before production commenced. GLP standard is a regulatory requirement for preclinical laboratory work including bioanalytical support. Like GMP activities are highly documented so the quality of the results can be verified. Formulation Identification and Optimization With API in hand, pre-formulation studies commence. The specifics of these studies depend on the projected route of administration and the physicochemical properties of the API, but examples include dissolution properties and solubility; compatibility with various excipients; stability to light exposure, acid, base, and oxidants; compressibility; friability (how readily the substance crumbles); wettability; and hygroscopicity. Once the formulation properties of the API are understood, specific formulations are designed and evaluated. An injectable liquid formulation will generally contain a buffer with a pH that is close to that for blood and maybe a tiny amount of organic solvent or detergent to help solubilize the drug. For an oral liquid formulation a taste-masking agent may be added. Tablets and capsules are made of mixtures of API and solid dosage excipients. The tablet combinations work together to give a stable tablet that will maintain its integrity during handling and storage yet efficiently dissolve and deliver drug in the gastrointestinal tract. Tablet dissolution rates cannot be compromised when the tablet is formed by compression. The enormous number of drug delivery approaches and the specific demands placed on the formulation by the properties of the API make generalizations about formulations unfeasible. There is much lore among formulations scientists as well as specific experiential knowledge that is used in the design of any specific formulation. Excipients deemed acceptable by the FDA known as Generally Regarded-As-Safe are used in formulations as often as possible. Unique excipients may be required to formulate a specific drug, in which case the safety of the excipient will be evaluated to the same level as the drug itself. Excipients can have profound effects on the safety or efficacy of a drug. As a consequence, the formulated drug must be the entity studied in all key safety, bioavailability and efficacy studies. It is imperative not to make any changes in the formulation once key safety, toxicology or safety clinical trials have completed. Such changes can potentially invalidate the use of the data generated from these studies to justifying advancement of any newly formulated drug product. Drug Product Supply, Manufacturing, and Scale-Up

As development progresses the demands for formulated product substantially increase, and it is necessary to begin scaling up Drug Product production. Some processes scale easily and others are not scalable at all. Depending upon the specifics of the processes, considerable investment in the process or even in a new process may be required to supply the clinical studies with formulated drug. The scale of production can grow from tens of grams to hundreds of kilograms of formulated API over the course of development. Numbers of tablets or vials produced to support clinical studies likewise can grow from hundreds to tens of thousands. Of course, GMP standard is applied at each step of the process, and as with the API, a cookbook-like batch record outlining each step of the process is followed during production, with any deviations or changes noted and justified. This part of development is about more than supplying the clinical trials with drug. Much is being learned about the behavior of the process in an industrial production setting, taking into account steel mixing kettles, tablet presses, lyophilization driers, and so on. The process is being optimized for the specific factory equipment to be used in manufacture, and the quality of the product produced is being assessed, including the stability of the drug product during handling and storage. For injectable drugs, sterile production is mandatory. The analytical methods used to monitor the process; test DP stability; and release DP to the pharmacy are being refined and revalidated at this stage. As with API, there is a set of quality control specifications established for the Drug Product that must be met prior to release. Failure to meet these specifications leads to the batch failing. Rework of Drug Product batches to meet specifications is seldom feasible leading to substantial financial losses and potential drug shortages. Prior to registration of the drug, the FDA will do a preapproval inspection (PAI) of the factories where API and Drug Product are going to be manufactured. Part of that inspection will include a detailed description of the history of process development and justification for the final manufacturing processes for both API and Drug Product. The FDA-approved finalized batch records will be used in all commercial production from launch on. Regulatory authorities routinely audit manufacturing sites. They have the authority to cite or even shut down a manufacturer for any lapses in quality. One typical citation issued by the FDA for significant violations is called a 483 after the FDA form listing violations identified by a FDA inspector. Vendors and customers take 483 citations very seriously as such problems can compromise product productions, or worse, entire development programs. For both API and DP, the FDA will want to know the manufacturing processes are well controlled and extremely well understood by the manufacturer. Such deep understanding will ultimately help the manufacturer and the FDA readily understand source of a manufacturing problem and aid in initiating corrective actions so that no supply disruptions arise. Quality Control Methods for API and DP Quality assurance for drugs is set to a very high standard. The incorrect dose of a drug or the impurities contaminating that drug can have significant ramifications on its safety and efficacy. In order to ensure that the chemical integrity of the API and Drug Product, the desired dose of drug is administered and that any adulteration with impurities and degradation byproducts does not exceed the levels found safe in animal toxicology studies and in clinic trials, stringent specifications are placed on both the API and particularly the Drug Product. To assess the drugs quality, it is evaluated with validated analytical methods that are robust, reproducible, and accurate at measuring the features of the products delineated in the specification. The identity of both API and DP, the

potency of the drug and the impurities and their levels are generally the most important quality parameters to be examined for any API and Drug Product for the purpose of assessing drug quality for release to the next step of manufacture or for distribution to the pharmacy. Examples of other characteristics of API or Drug Product that can be measured or observed at this stage include residual solvents, heavy metals, degradation byproducts present and their levels, dissolution times for tablets, reconstitution times for solids going into solution, the presence of bacterial contamination or proof of sterility of the product for injectable drugs, polymorph, water content, inorganic contamination, LPS in injectable drugs, particulate matter, chirality, particle size, integrity of tablet coating, color, tablet hardness, infrared spectrum, and melting point. Sterility is exceedingly important for injectable drugs and the FDA wants absolute assurances of sterility for injectable drug products. Validated sterility testing is critical for this attribute. If the product tests positive for exceeding any specification, it will be rejected for use. Establishing a highly purified reference standard for quantitative comparisons is a fundamental requirement for GMP-level quality control. Samples of reference standards and production batches are supplied to regulators at the time of methods transfers into regulatory laboratories. Regulators will from time to time perform independent assessments of both API and Drug Product quality. Rejected batches or product pulled from the pharmacy shelves is exceedingly costly, so all methods are formally validated for accuracy, precision, robustness, and inter-laboratory and multiple analyst consistency, in order to avoid spurious results or release of dangerous product. Methods are refined over the entire course of development, and new methods are added as more is learned about the specific nature of the drug. Millions of dollars may be spent in developing quality control methodology. By far the most important analytical tool in assessing potency, purity, and stability is high-performance liquid chromatography (HPLC) analysis. Before registering the drug, the FDA will assess the analytical methods just as it evaluates the drug. For key methods, the FDA will actually conduct a method transfer to an FDA laboratory, thereby to evaluate not only the quality of the drug itself but also of the acceptability of the methods used to release product. Impurities and Degradation Contamination The levels of impurities and degradation byproducts are tightly controlled in both the API and the Drug Product. During process development and scale-up, as well as during storage, new impurities or higher levels of old impurities may materialize. Depending upon the clinical indication and the stage of development, these new contaminants or higher levels of contamination can cause the entire program to be shut down. The initial toxicology studies qualify these impurities and their levels in tested samples. As long as new impurities or elevated levels of previously qualified impurities do not appear in new clinical material, the drug can be dosed in people. Once something new materializes in the drug batch, the drug must be re-qualified in a new toxicology study. Bridging toxicology studies on the new batch are the easiest and cheapest way to qualify a new impurity. In the worst case, the original toxicology studies must be repeated. Alternatively, the new impurity can be isolated and subjected to an independent toxicology study. Changes in impurities levels and contamination with new impurities can be a persistent problem in development. However the FDA expects ever tighter control of this problem as the process matures and commercialization approaches. Degradation byproducts appear as the product ages. Their formation must be suppressed as long as possible. Once their levels surpass the qualification level, the

drug material must be pulled from the pharmacy or storage and discarded. Neverbefore-seen degradation byproducts present a difficult problem and can usually be traced back to a deviation in the production process or the raw material feed stocks. Occasionally even new packaging materials can lead to never-seen impurities contaminating the drug. During development, container compatibility studies and leaching studies are conducted prior to selecting containers and container closures. Regulatory authorities find one class of impurities particularly serious; these are genotoxic impurities, primarily agents that damage DNA. In recent years and outside of oncology indications, low limits for exposure to genotoxic impurities in drugs have been established by regulators. For a chronically administered drug, 5g per day is the recommended limit for patient exposure to a genotoxic impurity. For comparison, a typical human consumes 1.5 grams per day of naturally occurring genotoxins in foodstuffs such as organic fruits and vegetables. Specifications for API and DP Establishing a defined set of product specifications for both API and Drug Product is a crucial component of the development process. These specifications represent the key quality attributes that the drug must meet to be acceptable by authorities for entry into human usage. Purity and potency are the most obvious attributes of a drug substance and Drug Product, but there can be many other attributes in a specification, depending upon the specifics of the medicinal agent. The specifications established early in manufacturing optimization and scale-up can be less stringent than later stages of clinical development when it is expected that greater development and product understanding should allow for significant tightening of the specifications. Initially, some quality attributes of the product are unknown but will be tracked. The specifications for these product quality attributes will initially be set as For Information Only (FIO). As more becomes known about some attribute, FIO will be replaced with a specification that reflects the current experience with the product after multiple productions. For example, a color specification of FIO might be replaced after the third production run with white-to-tan. Later, the color could be listed as white to offwhite and the final specification might be matched against a standard color palate, for instance, white #5 to #6. Over time specific specifications may be discarded or replaced if deemed not a useful quality attribute. Some specifications refer to crucial attributes such as potency and purity. Others, like water content or color, are indications of tight control and reproducibility of the manufacturing process. The total constellation of attributes provides the regulatory and quality assurance groups with a high degree of certainty that a consistent, high quality product has been produced without any hint that something has gone awry during manufacturing, and the drug is safe to release into commerce. Again, the designated reference standard is the benchmark sample used in all these assessments. Stability of API and DP for Shelf-Life Understanding the stability characteristics for both API and particularly Drug Product are critical for developing a safe and effective pharmaceutical agent. From the very start of production of API and Drug Product, samples are set aside under highly controlled storage conditions and, using those samples, the stability of the product is evaluated. It is imperative that stability-measuring analytical methods be developed and validated to

support all stability studies. Analytical studies are conducted on the product as it is exposed to accelerated decomposition conditions in order to identify the degradation byproducts likely to appear during long-term storage and to ensure the analytical methods can detect and quantify these byproducts. Samples of each production batch are placed in a stability chamber of defined humidity and temperature. Then samples are withdrawn on a pre-specified schedule such as every week for one month, once every three months for the balance of the year, every six months for the second two years, and then yearly until the study is terminated. Two to four temperature settings are initially chosen for these studies. These storage temperatures are a cold storage temperature (20oC or 2 to 8oC, for example), room temperature (20 to 25oC), an elevated storage temperature (35 to 40oC), and an accelerated degradation temperature (>65oC). Stability studies are conducted under GMP conditions and are exceedingly expensive in terms of money and drug consumed. Early in development, the demands for stability samples can create shortages of available drug for clinical trials. As the appropriate storage conditions for stability become defined, fewer temperature ranges and time points will be necessary for populating the stability data bases, lessening the demands on API and DP. Just as surprising changes seen in product specifications after a troubled production run can indicate problems, unexpected changes in product stability can signal that the quality of the product is amiss. Once the final API and Drug Product production processes have been finalized, three consecutive production batches are produced using the final process and processing conditions. These may or may not be run at commercial scale (frequently 1/10th scale), but must be truly representative of the expected commercial process, including the specific equipment used in production. These are designated as Stability Batches, and the data arising from the stability studies on these batches will be used to establish the initial shelf-life of the commercial product. All stability data will be brought into any discussion with regulators about shelf-life, but it is data from these three representative batches of Drug Product and API (along with stability data from all subsequent production runs) that will serve as the guiding data for shelf-life approval. There is a regulatory requirement that all new stability data be reported to the FDA in the annual IND update filing. The FDA takes a very keen interest in the stability of products undergoing clinical trials. Metabolism and Metabolite Determination Metabolism is the bioprocessing of xenobiotics by the tissues of the body. The main interest in the metabolism of a drug is to understand any safety issues and biological activity arising from a drugs biotranformation. If the level of a metabolite exceeds 10% of the amount of the original drug dose, it must be characterized and its safety independently studied. Metabolite characteristics such as genotoxicity, hepatotoxicity, cardiotoxicity, fetal toxicity, carcinogenicity, and central nervous system/cardiovascular (CNS/CV) toxicity, plus metabolite accumulation, are of particular concern to regulators. Regulators are also concerned when drugs alter the metabolic efficiency of the body such that co-administered drugs display altered clearance. These drug-drug interactions can cause serious adverse events or diminished therapeutic efficacy in individuals taking multiple drugs. Different species of animals can display very different patterns of metabolites. Since toxicology studies are done in animals, it is imperative that at least one of the two species used in the toxicology studies produce the same major metabolites (that is,

metabolites that occur at levels of 10% or more of the original drug dose) expected to be observed in humans. These human metabolites are initially identified from in vitro human liver tissue and ex vivo plasma exposure studies. The major metabolites are identified and characterized prior to initiating clinical trials, using validated bioanalytical methods developed specifically to track the parent drug and its major metabolites in test animal and human blood. Sometimes this work is carried out using very expensive specially synthesized radiolabelled drug to track radioactive metabolites. These days, HPLC-MS (HPLC-mass spectrometry) techniques are preferred for measuring metabolites, but radiolabel studies remain the gold standard. Metabolism can occur in any tissue, but the liver is the primary organ for human metabolism. There are two basic types of metabolism chemistries: 1) those metabolic reactions that alter the parent drug, or Phase 1 metabolism; and 2) those metabolic reactions that add (conjugate) some fragment to the drug, or Phase 2 metabolism. Frequently, these two processes occur sequentially during the metabolism of drug, first to place a conjugation handle on the drug, then to conjugate some fragment that aids in clearing the drug. Lipophilic drugs tend to be extensively metabolized, whereas hydrophilic drugs are cleared directly through the kidneys with little metabolism. Listed below are the metabolic reaction types and the enzymes involved. Redox reactions (adds oxygen, removes hydrogen, adds hydrogen) Phase 1 metabolism o Cytochrome P450 - CYP o Flavin Monooxygenases o Other Oxidoreductases; i.e., dehydrogenases, aldo-keto reductases, amine oxidase (MAO), peroxidases, quinone reductases Hydration reactions (adds water hydrolases) Phase 1 metabolism o Esterase (esters and lactones) and thiolesterases o Amidases (proteases); nitrilase o Epoxide hydrolases o Inorganic estrases; i.e., phosphatases, sulfatases, nitrases o Glycosylases Conjugation reactions Phase 2 metabolism o Methylation (O, N, S) o Sulfonation and phosphorylation o Glucuronidation (O, S, C and N glucuronosyl transferase) o Acylation o Glutathione conjugation o Amination of carbonyls Metabolism can be compromised in patients with diseased livers, and regulators can require narrow clinical studies in patients with marginal liver function to gauge any potential safety issues in patients with limited liver function. Also, various ethnic groups, men/women and older people can show different metabolism profiles. If a specific ethnic population lacks a key drug-metabolizing enzyme, individuals in that population can suffer from adverse events or lack of drug efficacy. Key metabolic enzymes may be elevated in certain individuals, or key metabolic enzyme isoforms can display either very low or high levels of activity in certain populations, adversely impacting drug safety or efficacy. Sometimes very low doses of drugs that inhibit specific metabolic enzymes

maybe added to a particular populations dosing regimen along with the therapeutic agent, in order to block that agents rapid clearance. This practice is called boosting, and it is used, for example, to elevate the circulating blood levels of certain antiviral agents used to treat AIDS. The most important in vitro metabolism studies are conducted in order to explore drugdrug interactions. These are primarily liver cytochrome p450 isozyme inhibition, substrate and inductions studies with a special focus on the two major drug-metabolizing CYP isozymes, CYP3A4 and CYP2D6. These studies are now routinely conducted prior to lead selection, although some companies may perform such studies just prior to entering the clinic. If a drug appears to inhibit any other key drug metabolism enzymes, including conjugating enzymes, special drug-drug interaction studies can be required with these specific enzymes. There are two particular classes of drugs that actually utilize the metabolism machinery to liberate the biologically active agent or to facilitate the removal of a circulating active agent with a long half-life. These are called prodrugs and soft drugs, respectively. Pharmacokinetics (PK), Including Absorption, Distribution, and Excretion Pharmacokinetics deals with the changes in in vivo drug concentration over time. The simplest situation involves bolus intravenous drug dosing. The drug enters circulation, establishing the maximal blood concentration, called the Cmax, within seconds. The drug is rapidly distributed throughout the circulatory system, and subsequently begins to disappear from circulating blood until it is no longer detectable. Multiple processes are occurring as the drug concentration in blood drops. These processes include metabolism, absorption into tissues and passage from the tissues back into circulation, excretion through the kidneys and liver, and, for volatile agents, exhaled from the lungs. Ideally, the dynamic kinetics of drug exposure will correlate with the appearance and decay of efficacy (pharmacodynamics) and toxicity (toxicokinetics). This information can guide drug dose schedules. For example, in chronic drug exposure it is useful to identify a dose schedule that balances the appearance and disappearance of drug such that the concentration stays within an efficacy manifold and below a side-effect threshold. This is known as steady-state dosing. A vast field of special drug delivery formulations and devices has evolved over the decades with the goal of prolonging dose exposure and evening out fluctuations in circulating drug concentrations. Enterohepatic circulation is a particular type of pharmacokinetics in which the drug is excreted through the liver then reabsorbed, creating a biphasic decay/concentration curve. Enterohepatic circulation delays the clearance of the drug which may either be useful or a problem. Drug recycling can also occur via reabsorption from the bladder after excretion by the kidneys, but this is a much rarer phenomenon. Drug distribution is complicated by the fact that drugs can partition at different rates into and out of various tissues. The classic example is the blood-brain barrier, which blocks the passage of many drugs into the CNS compartment. Different tissues can also metabolize and excrete the same metabolites at different rates. Finally, different tissues have different blood-flow rates, leading to variable drug exposure levels among different tissues and giving rise to the concept of multiple-compartment kinetics for distribution. Certain drugs can be sequestered in some tissues and then slowly released from these

depots, prolonging drug exposure, whereas other tissues rapidly take up and rerelease drugs, leading to rapid clearance. Absorption is the process by which a drug passes through a tissue barrier and into circulation or into a target tissue. Typically, drugs are dosed in an oral formulation, and the phenomenon of absorption refers to the movement of drug out of the GI tract and into circulation. For topical drugs, absorption refers to passage of the drug from the surface of the skin into the underlying tissues or even into circulation. Drugs administered via the lungs, mouth, eyes, nasal passages, rectum, vagina and depots directly under the skin or intramuscularly all display special absorption pharmacokinetics. Many factors affect oral drug absorption. Dissolution properties of a drug molecule are the primary feature chemists can affect during drug design. If the drug does not dissolve, it will not be absorbed. Chemists can engineer features into a molecule that greatly enhance solubility in gastric and intestinal fluids, particularly bile. The standard practice is to introduce a functional group that forms a salt that dissolves in water. However, if the charge is a permanent fixture of the molecule, absorption may be compromised, because the drug will then not traverse lipophilic membranes of the cells lining the GI tract. In fact, those features which enhance water solubility in general are the same features that inhibit membrane solubility and permeability. The ideal drug molecule balances these opposing properties to maximize water and lipid solubility. Formulation chemists use certain tricks to enhance solubility, including atomizing the drug to maximize water-exposed surface area, adding detergents to formulations to aid in dispersing lipophilic drugs, or even dissolving the drug in an acceptable fatty solvent for administration as a preformed liquid. Other factors that affect oral drug absorption include transporters in the lining of the GI tract, which drives absorption across cell membranes. On the other hand there are transporters, such as the P-glycoprotein pump (PGP/MDR transporter), that inhibit absorption by facilitating efflux out of GI cellular lining and back into the intestines. Regulatory authorities are interested in these efflux systems, because they too can lead to drug-drug interactions with loss of efficacy or increased toxicity of drugs competing for the transport machinery. The PGP/MDR pump is of particular note for its protective role in preventing toxic xenobiotics from entering general circulation and ejecting chemotherapeutic drugs from resistant tumors. If one drug inhibits the MDR pump, toxic levels of a second drug normally cleared by the MDR system may accumulate to create a safety problem. The PGP/MDR system is also a key protective feature in the bloodbrain barrier. This system lessens exposure of the brain to potentially neurotoxic xenobiotics but can also inhibit CNS drug access to the brain. When designing a drug that targets brain biology, considerable attention must be paid to PGP/MDR interactions with that drug. Once a drug enters the lining of the GI tract it may be metabolized, pumped back out into the GI tract, or enter vascular circulation. Drugs absorbed from the GI promptly pass through the portal vein in the liver where they can be metabolized away (first-pass metabolism) or absorbed by the liver tissues for direct excretion back into the GI tract in bile. These processes can severely limit the amount of drug entering circulation and compromise a drugs efficacy in the clinic. After entering circulation, the drug is distributed throughout the body, where it is absorbed into tissues and, for some drugs, subsequently into cells and cellular organelles. At each step, the physicochemical properties of the molecule affecting

membrane permeability, transporter binding, and metabolism can affect exposure of the target to the drug and the drugs efficacy. While it is in circulation the drug may bind to cells and proteins in plasma. High protein binding (usually 98% or higher) removes free drug from circulation, making it less available for binding/distribution to its target and thus compromising the drugs efficacy. Drugs that compete for the same protein binding sites can create yet another source of adverse drug-drug interactions affecting patient safety. The classic example of this kind of safety problem involves the anticoagulant, Coumadin. Coumadin is highly protein bound, and it has a narrow safety window for blood concentrations of circulating free drug. If a second drug displaces Coumadin from its protein-bound state, effective concentrations of free Coumadin in circulation may rise above a safe level, and the patient could bleed to death. However, protein binding is not always bad. The most significant drug-binding protein in circulation is albumin, which possesses many binding nooks for lipophilic drugs. A circulating protein such as albumin can serve as a depot for a drug, releasing it over time and extending the drugs efficacy. This may diminish the number of daily drug doses required and improve patient compliance or moderate exposure to high, potentially unsafe levels of drug at the time of dosing. The percentage of an orally dosed drug that actually makes it past the many hurdles described above and into circulation is referred to as the drugs bioavailability (F). If a drug is delivered by intravenous injection, 100% of the drug enters circulation. Typically, the maximal concentration of circulating drug is reached at the conclusion of intravenous dosing, and then the concentration of drug in blood decays away as the drug is taken up and rereleased by tissues, metabolized, and excreted. On the other hand, an orally delivered drug is affected by multiple processes prior to entering circulation. Blood levels rise over time to a maximum blood concentration, even as circulating concentrations of drug are decaying, because of the same processes that act on the drug in circulation after injection. The area under the absorption-decay curve (AUC) for blood concentration is proportional to the amount of drug absorbed into circulation from an oral or intravenous dose. Since the intravenous dose is by definition 100% bioavailable, the ratio of the oral AUC to that for intravenous dose, times 100%, yields the percent of drug entering circulation after oral administration. Ideally, the bioavailability for oral dosing is 100%, although frequently that is not the case; oral bioavailability greater than 20% is highly desirable. The oral bioavailability of a drug will affect the oral dose size, dosing schedule, efficacy, drug safety, tolerability, patient compliance, and cost of goods. The most serious problem with very low oral bioavailability is the large variability of drug blood levels encountered in the general patient population taking the drug and its effect on patient safety. For example, if a drug is 50% available in the normal patient population and 100% available in an outlier patient, the patient experiencing the higher absorption is exposed to twice the amount of drug as compared with the amount of drug to which the normal population is exposed. If the therapeutic safety window is 5-fold, then the higher absorber is still well within the safety manifold for the drug. However, if the drug is only 1% orally bioavailable in the normal population it can easily jump to 5% or 10% in outlier patients and create serious safety concerns. Assuming the drug is efficacious at 1% and should the bioavailability drop to 0.1%, the circulating concentration easily falls below the efficacy threshold in some patients. Bio-Analytical Methods Development

Analytical methods are needed to track the amount of circulating drug and its metabolites. Understanding human PK is a primary concern for doctors and regulators. In order to reliably measure blood levels, validated methods must be developed for both the toxicology species and for humans. The process usually entails mixing the drug or its metabolites in whole blood from the target species and then processing the blood to remove the extraneous blood components and liberating the free drug and its metabolites. The process generally stabilizes the drug and its metabolites in some matrix, preventing degradation. The blood extract is analyzed against a reference standard to yield a quantitative assessment of blood concentrations of the drug and its key metabolites. Multiple samples are taken over time so the kinetics in blood can be determined. This work is highly documented as part of GLP. The data are used to ascertain the toxicokinetics and pharmacodynamics for the drug whenever possible, along with any significant drug accumulation after chronic dosing, as drug accumulation over time can lead to serious safety issues. A distinct type of PK study involves sampling different patients at different time courses for blood draws. The individual data are pooled and analyzed leading to an understanding the PK of the drug in the treatment population. This is called population PK, which yields data that are thought to better represent the PK attributes of the drug than would a PK study of only a few individuals, even if that study were more thorough. Safety Pharmacology Drugs display many signs and symptoms associated with their side-effect profile. During both discovery and development, the safety profiles of the lead compounds are evaluated to identify potential safety liabilities and gauge the safety window (therapeutic window) for dosing. There is a large battery of potential safety assessment assays available for evaluating side effects of new agents, ranging from a simple lethal dose (LD50) determination to tests designed to pick up subtle signs of neurotoxicity or adverse immunological responses. The Erwin test is a general CNS symptom survey screen. The hERG K-channel assay and related electrophysiological assays are used to evaluate leads liabilities for cardiac arrhythmias. There are numerous assays available to assess the hormonal effects, carcinogenicity, genotoxicity, embryo-fetal toxicity, teratogenicity, and eye and skin contact irritability. There exist safety assessment assays for nearly every possible adverse physiological symptom. Toxicology and Clinical Starting Dose Determination The primary concern of regulatory agencies is for the safety of patients taking drug. Specific toxicology studies for all new drugs are mandated by the regulatory authorities before the drug is allowed to enter clinical study. The goals of these toxicology studies are to assess risk, determine likely early markers for toxicity so as to guide clinicians conducting trials, and identify a safe starting dose for the Phase 1 trial in humans. Toxicology studies are designed to provide data that support dose scheduling and prolonged exposure to the drug. Regulations require GLP toxicology studies supporting clinical trials to be conducted in two species, usually a rodent and a larger non-rodent. Rats and beagle dogs or monkeys are the species of choice for most IND-enabling toxicology studies. An acute dose-escalation study determines a safe dose range and provides data upon which to base a subsequent study, a repeat-dose study covering a range of doses. The animals are monitored for any signs and symptoms of toxicity, including weight loss and lethargy, and clinical blood work looks for adverse findings in blood chemistry. At the end

of the trial, the study animals are sacrificed, a necropsy performed, and samples of body tissues are analyzed for pathological changes. Tissue slices are microscopically examined for pathological effects caused by the drug. A control group and a washout group of animals are incorporated into the study. The results are highly documented per GLP reporting requirements and are used to guide clinicians on the selection of starting clinical dose. Traditionally, fourteen-day, twenty-eight-day, and ninety-day repeat dosing schedules in animals are performed to justify repeat dosing in humans for an extended time. The starting human dose will be one-tenth of the maximum tolerated dose in the more sensitive of the two species in the repeat dosing toxicology study. Sometimes the toxicology study will be designed to mimic the expected clinical trial, for example by mimicking some cyclic pattern of dosing in the clinic. If toxicology studies reveal that the drug is unsafe at doses below the expected efficacy dose, demonstrate that the drug has significant toxicity after drug exposure has ended (delayed toxicity), show that drug toxicity is irreversible, or indicate unpredictability (idiosyncratic toxicity), chances are that development of the drug will be terminated. Although the toxicology roadmap described above seems formulistic, there is some latitude in these studies, especially in the interpretation of toxicology results and how potentially adverse results can be managed going forward in the clinic. It is always advisable to use a highly experienced toxicologist in planning, executing, interpreting data and applying those results to a clinical plan. Such a person can provide favorable guidance on a problematic toxicological result or better yet avoid an adverse toxicological outcome in the first place. Clinical Plan, Clinical Protocol, and Clinical Brochure The most critically important aspect of any drug development program is the design of the clinical trials. Clinicians develop a clinical plan based on the expected therapeutic indication, the treatment population, the dosing format, the safety profile, and the expected treatment outcomes, and they must include a supporting statistical plan. From the clinical plan, very specific treatment protocols (essentially a detailed clinical roadmap) will be drafted for approval by regulatory and medical authorities. Ultimately, these protocols will be incorporated in a clinical support document called a Clinical Brochure. This document is provided to all clinical investigators conducting the clinical trial. The Clinical Brochure includes summaries on medical justification for the trial, preclinical pharmacology and PK (and metabolism), toxicology data and potential safety concerns, description of the drug and the drug container label, stability data on the drug and any reconstituted drug formulation, pharmacy instructions for storage, handling (including reconstitution), distributing and disposal of the drug, instructions on administering the drug, details on monitoring effects of the drug in patients, informed consent forms for patients, restrictions on the treatment population, the endpoints for the trial, the statistical analysis plan, and justification for the size of the trial and stopping criteria. The IND An Investigational New Drug (IND) application is the documentation presented to the FDA requesting permission to study a new drug, drug formulation, new dosage form, drug combination, or new drug indication in a human clinical trial. An IND includes summaries, original data, and formal reports covering chemistry, formulation, stability data, container enclosure and storage information, manufacturing and control methods

for the drug and drug formulation, specifications, preclinical pharmacology, safety and toxicology on the drug, the clinical plan, statistical plan and clinical brochure, and the required forms for filing an IND. This documentation is filed with the FDA who assigns an IND number to the study. The FDA has 30 days during which to comment on the application. It can remain silent, ask specific questions, ask for more data, or block initiation of the trial. After 30 days, the sponsor may commence with the clinical trial unless the FDA directs otherwise. Some sponsors have pre-IND discussions with the FDA to address any concerns the sponsor has with the FDAs guidance or perspective on the proposed trial, including manufacturing, trial design, and safety. Once opened, an IND must be updated annually 90 days after the anniversary date of the IND submittal. Significant changes to the IND, such as changes to the manufacturing process, formulation, storage conditions, stability, clinical trial, and new safety and toxicology information, must be submitted to the FDA as IND amendments whenever such changes occur. Once opened, the IND remains active at the FDA until product development ceases and the drug is abandoned by the sponsor. Institutional Review Board (IRB) Approval Institutions that perform clinical trials are required to establish a committee to review the ethics, scope, and merits of any clinical trial conducted within their institution. The committee is composed of physicians not involved in the trial under review, medical ethicists, and lay people. It is their charge to review all the documentation supporting the trial, especially the patient consent form and the medical and scientific rationale for the study. They take a keen interest in trial procedures that can cause distress to trial participants and make a medical and ethical assessment of the risks and benefits of the trial. Once they approve the trial and budgets are agreed upon between the institution and the sponsor, the trial can commence and recruitment of patients begins. The IRB will continue to monitor the trial for any issues affecting patient welfare. Phase 1 Safety Trials The Phase 1 trial design is pretty straightforward. The study population may be defined as normal volunteers or patients diagnosed with the disease that is expected to be treated with the drug under evaluation. The trial population should be reflective of the treatment population in terms of age, sex, or any other characteristic defining the targeted patient population. The initial dosing schedule, the starting dose, and a definition of safety factors to be monitored during the trial are key aspects of Phase 1 trial design. The goal of a first-in-man trial is to delineate the safe dose and schedule to be used in future trials and to identify any safety issues to be monitored in future trials. Combinations of new drugs with old drugs are assessed for safety in a Phase 1 setting as well. These trials tend to be small in size, with only 20 to 30 patients enrolled. Patient blood and occasionally tissue samples are collected to analyze for drug and generate a human PK profile. Sometimes special patient populations are used in radiolabelled drug studies and in studies evaluating drug clearance rates in patients with compromised kidney and/or liver function. If the trial is conducted in patients with the indicated medical condition for the drug, occasionally some patients will show a medically meaningful drug efficacy response while on the trial. Phase 2 Efficacy, Dose, and Schedule Trials Phase 2 trial designs are quite varied, but all have the primary goal of demonstrating some level of drug efficacy in the target patient population. Each trial design is very

dependent on the specific disease or medical condition, stage of the disease, and the type of response to the drug expected from the preclinical data. These trials can be blinded, placebo-controlled, or open label designs. Trial size can range from 20 to 300 patients and include a patient control arm if desired. A key feature of Phase 2 trials is to establish the optimum dose and schedule for achieving the desired therapeutic outcome. The most important aspect of Phase 2 trial design is the incorporation of crisp, clear endpoints of drug efficacy. Well defined patient entry criteria can be critical for the success or failure of these trials. A statistical plan will be required to size the trial and interpret the trial results. Once sufficient positive Phase 2 data are available to define the scope of a drugs activity in patients with the target condition, a drug registration trial can be designed. The registration trial design with its justification and all supporting data is submitted to the FDA for discussion at an end-of-Phase 2 meeting. The sponsor will try to solicit agreement from the FDA regarding acceptable endpoints for the Phase 3 trials that will lead to registration approval to market the drug. However, the FDA is never bound by such agreements and can change registration endpoints should it choose or if warranted by circumstances such as changes in the treatment landscape for the originally targeted medical condition. Level 3: Late-Stage Drug Development The goals of Level 3 activities are to confirm efficacy and safety in humans in at least two well controlled Phase 3 clinical trials. Additionally, Level 3 activities include completion of support work for supplying the market with drug that meets an acceptable quality standard and providing a dossier for review by regulatory authorities, called a New Drug Application (NDA), which documents all these activities. During late-stage drug development, the Chemistry, Manufacturing Controls (CMC) work efforts are completed, including finalizing the production processes and scale-up of API and Drug Product to commercial scale. Additionally, in order to finalize the specifications, batch records and quality control methods, the manufacturing process is run at scale three consecutive times, adhering to specifications in the batch records (called validation production runs). Other key activities include building launch supplies and establishing distribution channels. Late-stage toxicology studies are completed, including six or nine-month repeat-dosing studies in rodents and nine-month repeat-dosing studies in beagle dogs or monkeys, two-year carcinogenicity studies, and any product-specific safety studies, including studies that provide guidance for worker exposure. The manufacturing plants are readied for a preapproval inspection (PAI) by the FDA. The suppliers of feedstock chemicals and advanced intermediates are qualified, and specifications for supplies and intermediates are finalized. Suppliers of advanced intermediates prepared under GMP conditions submit Drug Master Files (DMF) to the FDA for review. A DMF describes methods of production, specifications, and quality control methods and supports the NDA review. The Phase 3 trials are called registration trials because the data arising from these trials will be used by regulatory authorities to decide for or against approval of the drug for commercialization. Per prior agreement with the FDA, specific statistically significant endpoints for registration trials must be achieved before an NDA can be submitted. The entire clinical efficacy and human safety profile for the drug must be documented and reviewed and the data submitted to the FDA for review. If the data are equivocal but significant secondary endpoints are met, the FDA can have a committee of medical advisers review the data and make recommendations regarding approval. For certain life-threating conditions such as cancer, significant efficacy data from a single Phase 3 trial or even a Phase 2 trial may be sufficient for regulatory approval of the drug.

Level 4: Regulatory Review and Approval The goal of Level 4 activities is authorization from regulatory authorities to market the drug within the specified label claims. The drug industry is the most highly regulated enterprise in the world. The reasons are obvious, given the intimate exposure of humans to these items of commerce and the potential effect they can have on human health. There are a number of significant laws and administrative regulations that have evolved over the past 100 years that affect drug research and development either directly or indirectly. These major laws are listed below: 1906 - Pure Federal Food and Drug Act adulterated food and drugs 1938 Federal Food, Drug and Cosmetic Act drug safety 1944 Public Health Service Act regulation of biological agents such as blood products, hormones and vaccines 1962 Kefauver-Harris Drug Amendments drug efficacy 1984 Drug Price Competition and Patent Term Restoration Act Hatch-Waxman generic products 1990 International Conference on Harmonization (ICH) 1992 Prescription Drug User Fee Act (PDUFA) 1996 Health Insurance Portability and Accountability Act (HIPPA) patient privacy 1997 Food and Drug Administration Modernization Act accelerated fasttrack approval 2003 Pediatric Research Equity Act mandated drug testing in children

The specific goals of each these laws are varied, but the overarching goal is to ensure that drugs are properly labeled, pure, safe, effective, and available to the appropriate patient populations. Regulatory authorities become engaged with the sponsor once an IND is implemented. However, most of the pre-IND activities necessary to support an IND on a development candidate are governed by regulation, as well. Once it has been decided that a development candidate will be advanced into the pre-IND process, the sponsors regulatory group becomes involved in a development program. The regulatory group reviews and advises on all documents and procedures involved with manufacturing both API and Drug Product, quality control methods, production runs, raw materials sourcing, compliance with GMP, GLP compliance in safety and toxicology studies, GMP storage and distribution, all aspects of clinical study, clinical site compliance, and reporting of adverse events (AEs) and severe adverse events (SAEs) in the trial within the mandated reporting time window. Further, this group is responsible for controlling all the documentation supporting the IND and for filing timely clinical safety updates, IND amendments, and annual reports with the FDA. The regulatory group drafts the IND and ultimately the NDA and schedules meetings and manages all contact and communication with the FDA. They provide advice and recommendations to members of the development team regarding regulatory and statutory guidance from the FDA for any particular development issue, and they facilitate the resolution of regulatory issues arising between the sponsor and the FDA. They also draft and file documents supporting clinical trials and drug registration in jurisdictions outside the US and provide guidance on ex-US regulatory compliance. Additionally, the regulatory group must also help rectify any manufacturing and marketing issues that arise with the FDA. The label claim is of particular interest to the marketing group, as no claims whatsoever can be made in marketing materials that are not specifically in the FDAallowed label claims. The specifics of a label claim can make or break the commercial success

of the drug. It is the responsibility of the regulatory group to ensure that the NDA is in compliance with FDA guidelines. The regulatory group in consultation with the legal, marketing, clinical and safety groups negotiates the label claims at the time of registration. The regulatory group addresses any issues with the FDA once the product enters commerce, including the compliance of all marketing materials and marketing activities within FDA guidelines. The NDA consists of multiple sections. The currently acceptable NDA format, called the Common Technical Document (CTD), came into existence to harmonize new drug application documentation around the world. The CTD sections consist of: Quality (CMC reports and summaries), nonclinical (ADME, safety, toxicology, biology MOA and pharmacology reports, overview, and summaries), and clinical (reports, overview, and summaries). An NDA can have millions of pages. The NDA is now submitted electronically to the FDA. The NDA may be submitted in total or in sections. Each section of the NDA will be reviewed by the experts at the FDA. Those sections completed early can be reviewed prior to the submission of late-arriving sections. Such a scenario is referred to as a rolling NDA. The sponsor will need to satisfy the FDA that the drug is safe and effective for the registered indication. More data may be requested, up to and including additional clinical trials and/or additional CMC work, to address any FDA concerns. Once the drug is approved by the FDA, distribution to pharmacies for sale can commence, but research and development in support of the drug will continue in the form of Phase 4 clinical trials to support expanded label claims or identify new indications, troubleshooting production issues, qualifying new sources of production materials and secondary suppliers for API and Drug Product to ensure no shortages of drug develop, improving the production process to drive down cost of goods (COGS), and expanding the biological understanding of the drug. Intellectual Property Without strong intellectual property protection laws and vigorous enforcement of these laws, new drug invention and development would not be commercially viable and few new drugs would be brought into existence. Patents are granted by government patent offices around the world and typically expire 20 years from the initial filing date. Provisional patent applications lock down a filing date for one year, by which time the provisional application must be converted to a standard patent application. Patent officials usually take several years to evaluate the patent, and during that time they may ask for restrictions on the scope of the patent if the patent includes multiple inventions, or they may issue Office Actions, which call on the sponsor to justify all or part of the patent claims. The patent office will grant exclusivity only to the specific claims listed within the patent. Inventors may end up with multiple claims or only a single claim granted. The narrower the scope of the claim, the more likely it is that the patent office will approve it. However, narrow claims make it easier for competitors to find work-around solutions. Even if broad allencompassing claims are not granted, the act of revealing large numbers of structures in a claim creates prior art. All competitors will need to take this prior art into account when they file their own patent applications on similar molecules. Patent applications for inventions that are already covered by another inventors patent or for inventions that are obviously based on prior art or known to be in the art will not be granted. Patents can also be invalidated if the legal inventors are not accurately identified or prior art is not revealed. For pharmaceuticals, by far the most important patents are composition-of-matter patents. These patents cover the specific chemical/molecular composition of the drug. Because of the exacting nature of chemical structural information such patents are impossible to directly

circumvent. The second most significant type of patent claims specific uses for a molecule, such as for the treatment of cancer. Beyond these key patents, a large number of inventive aspects applied to drugs can be covered by patents, if the aspects are not obvious or in the prior art. Those aspects include the method and process of production; formulations, routes, and sequence of administration; physiochemical properties of the molecule such as crystal habit, polymorphs, solvation state, and chirality; treatment of different medical conditions; sequence of use; composites/extracts of molecules; plant sources of drugs; drug-device hybrids; certain metabolites; methods of purifying natural-product drugs; combinations of drugs; packaging configurations; drug delivery methods, genomic profile of drug treatment population and many other aspects. Much of what passes for new drug invention is really research performed to circumvent patents. This practice has been called patent busting, patent poaching, and me-too drug research. This is standard approach to drug discovery. Once a specific molecular composition has been successfully patented, the owner of that patent can prevent anyone else from using that specific molecule for any purpose other than for research in pursuit of a new drug or development of a generic version of the patented compound. Patents must be invalidated by the courts before anyone can sell the patented molecule prior to the patents lapsing or expiring. Many drugs have enormous sales volumes in the billions of dollars with very high profit margins. The only way that these high profit margins can be maintained is through the monopolistic pricing patents afford inventors. Once drug patents expire, drugs become nothing more than expensive regulated specialty chemicals. Profit margins then collapse, an event known as a patent cliff. A patent cliff can be catastrophic for any company that relies on a key patent to protect a major drug product and can even lead to the companys demise. Aside from patents, governments can grant exclusive use privileges for some finite time. If the drug is registered but no intellectual property is available to protect it, the sponsor is granted five years of data exclusivity, which prevents competitors from using the original data to register the drug. The competitors can recreate the data for a new NDA. However, this is very costly and usually takes five years or more to complete. Designation as an orphan drug (meaning, the drug is intended to treat a rare disease of fewer than 200,000 patients in the US) grants seven years of data exclusivity for use only in the orphan indication. Bibliography Pharmaceutical Manufacturing Handbook: Regulations and Quality (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2008 Clinical Trials Handbook (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2008 Pharmaceutical Manufacturing Handbook: Production and Processes (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2008! Preclinical Development Handbook: Toxicology (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2008 Preclinical Development Handbook: ADME and Biopharmaceutical Properties (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2008 Drug Safety Evaluation (Pharmaceutical Development Series); ed. Shayne C. Gad; Wiley-Interscience; 2009 Pharmaceutical Manufacturing Handbook: Regulations and Quality; ed. Shayne Cox Gad; Wiley-Interscience; 2008! Development of Therapeutic Agents Handbook; ed. Shayne Cox Gad; WileyInterscience; 2011!

Drug Discovery Handbook (Pharmaceutical Development Series); ed. Shayne Cox Gad; Wiley-Interscience; 2005! Drug Discovery and Evaluation: Methods in Clinical Pharmacology; ed. H. Gerhard Vogel, Jochen Maas and Alexander Gebauer; Springer; 2011! Drug Discovery and Evaluation: Safety and Pharmacokinetic Assays; ed. H. Gerhard Vogel, Franz Jakob Hock, Jochen Maas and Dieter Mayer; Springer; 2006 Drug Discovery and Evaluation: Pharmacological Assays, 3rd edition; ed. Hans Vogel; Springer; 2007 Real World Drug Discovery: A Chemist's Guide to Biotech and Pharmaceutical Research; Robert M. Rydzewski; Elsevier Science; 2008 Early Drug Development: Strategies and Routes to First-in-Human Trials; ed. Mitchell N. Cayen; Wiley; 2010 Integration of Pharmaceutical Discovery and Development : Case Histories Pharmaceutical Biotechnology Volume 11; ed. Borchardt, R. T., Freidinger, R. M., Sawyer, T. K., Smith, P. L.; Plenum Press; 1998 Burger's Medicinal Chemistry, Drug Discovery and Development, Volumes 1-8 ; ed. Donald J. Abraham and David P. Rotella; Wiley-Interscience; 2010 Comprehensive Medicinal Chemistry II, Volumes 1-8; ed. David J Triggle and John B Taylor; Elsevier Science; 2006 The Practice of Medicinal Chemistry, 3rd Edition; Camille Georges Wermuth; Elsevier Science; 2008! Process Understanding: for Scale-Up and Manufacture of Active Ingredients; ed. Ian Houson; Wiley-VCH Verlag; 2011 Active Pharmaceutical Ingredients Development, Manufacturing, and Regulation (Drugs and the Pharmaceutical Sciences, Volume 151); ed. Stanley Nusim; Marcel Dekker; 2005 Pharmaceutical Dosage Forms, Volumes 1-3; ed. Herbert Lieberman, Martin Rieger, Gilbert S. Banker; Informa Healthcare;1996 Practical Process Research & Development; Neal G. Anderson; Academic Press; 2000 Drug Discovery and Development, Drug Discovery Volumes 1 and 2; ed. Mukund S. Chorghade, Wiley; 2006/2007 Guide to Drug Development: A Comprehensive Review & Assessment; Bert Spilker; Lippincott; 2008! ADMET for Medicinal Chemists: A Practical Guide; ed. Katya Tsaioun and Steven A. Kates; Wiley; 2011 Drug Development: From Laboratory to Clinic; Walter Sneader; Wiley-Blackwell; 1986 Drug Discovery. A History; Walter Sneader; Wiley; 2005 !

You might also like