Tag Archives: Raw Materials Testing

A Survey of Material Science and Physical Characterization Techniques and Equipment for Small Molecules – Part I 

Material science and physical characterization are crucial to ensure drug products’ or drug substances’ identities with measurements and analysis. There are many types of techniques and equipment to do this, each with its own features and qualities. In this blog, the first part of a series, we will review the advantages of a range of techniques and equipment. 

We’ll begin with X-Ray Powder Diffraction (XRPD) or XRD analysis. It is a powerful, non-destructive, and rapid technique for analyzing a wide range of materials (1 µm to 100 mm), including metals, polymers, catalysts, plastics, pharmaceuticals, and other materials. It is used for identification, crystal form characterization, and crystalline content in amorphous products.  

Inductively Coupled Plasma-Mass Spectrometry (ICP-MS) is useful for the detection of elemental content and elemental impurities. ICP-MS is also 

efficient as it is a multi-element technique. Ions are directly detected by an MS detector rather than by emission of light, as in the case of ICP-OES (Inductively Coupled Plasma-Optical Emission Spectroscopy). The ions are separated by a Quadrupole based on the mass-to-charge ratio. The best detection limits are available for most of the elements in ICP-MS as the number of ions produced is high, and although some spectral interference is seen, these are defined and limited.   

Dynamic Vapor Sorption (DVS) is a gravimetric technique used to measure the change in

 mass of a material in response to changes to surrounding conditions such as temperature or humidity. DVS is primarily used with water vapor but can be applied to other organic solvents as well for the physicochemical characterization of solids.  Some of the most common uses of DVS include:  

  • To determine the sorption isotherm;   
  • To evaluate the hygroscopicity of an API powder;   
  • To compare the hygroscopicity of different solid-state forms: solvates, polymorphs, salts, amorphicity, and cocrystals;  
  • To determine the deliquescence point of a material;  
  • To quantify and qualify the amorphous content in drug substance or excipient, and 
  • To evaluate the efficacy of packaging materials. 

We will wrap up with Differential Scanning Calorimetry (DSC). It is one of the most used thermoanalytical techniques for determining freezing and melting points and phase transitions. Specifically, it measures the heat flow produced by a sample when it is either heated, cooled, or held isothermally at an unchanging temperature. Crystallization behavior and chemical reactions are some other characteristics that can be measured by this technique. 

Our next entry will conclude with a look at Thermogravimetric Analysis (TGA), Fourier-Transform Infrared Spectroscopy (FTIR), Nuclear Magnetic Resonance (NMR), and, of course, the definitive analytical technique, Mass Spectroscopy (both GCMS and LCMS). 

Resources 

Setting the Standard with Reference Standard Qualifications

The term Reference Standard Qualifications (RSQ) “is defined as a list of tests, references to analytical procedures, and appropriate acceptance criteria,”1 and incorporate these quantitative reference standards to do so: purity, potency, identification, impurities content, and generally full characterization 

Identity Testing

There are a range of tests to ensure the identity of the drug product or drug substance. These can be done by a variety of equipment, including LC-MS, 1H NMR, 13C NMR, FTIR (Fourier Transform Infrared Spectroscopy), TGA (Thermal Gravimetric Analysis)/DSC (Differential Scanning Calorimetry), and UV-Vis. Appearance testing can be achieved by chiral or ion chromatography, elemental analysis (identifying C, H, and N), TGA/DSC, and XRPD.  

Quantitative Testing
For quantitative reference standards, there is a standard formula that should utilized (HPLC Purity) x (100% − % Residual Solvents − % Inorganic Impurities) = The Determination and Confirmation of the Assay Value % (i.e., weight %).  These component values can be quantified by using HPLC for the purity analysis, residual solvents by LOD or KF Water Content and HSGC, and inorganic impurities by ROI (Residue on Ignition), ICP-MS/OES ((Inductively Coupled Plasma Mass Spectrometry or Inductively Coupled Plasma Optical Emission Spectroscopy). (For a limited number of sample quantities of both residual solvent or inorganic impurities, they can alternatively be tested by TGA.) Finally, to access the weight percentage confirmation, titration and qNMR are excellent techniques. 

 Concurrent with identity and quantitative testing, storage conditions and stability should be established.  This includes storage (container/closure), expiration dating, re-testing procedures, and usage and tracking.  

We Can Assist with your Reference Standard Qualifications Needs  

SK pharmteco has years of experience and numerous experts in Reference Standard Qualifications. Compendia methods (USP/EP/BP/JP) are utilized in tandem with analytical method development, validation, and transfer to ensure optimal success with your drug substances and drug products. Please contact us with any specific questions or to receive a quote for your RSQ needs.   

References  

  1. https://www.ema.europa.eu/en/documents/scientific-guideline/ich-q-6-test-procedures-acceptance-criteria-new-drug-substances-new-drug-products-chemical_en.pdf 

Some Background and Concerns About PFAS

PFAS

The Background and Concerns of PFAS

PFSA structure(Per- and) PolyFluoroAlkyl Substances (PFAS) are a class of ubiquitous chemicals that have been found in water, air, fish, and soil across the nation and worldwide. Known as “Forever Chemicals,” there are thousands of different PFAS, and they are present in consumer, commercial, and industrial products.1 Having one of the strongest bonds in organic chemistry, their structures proved to be resistant to heat, water, oil, and degradation.2 They are found in “food packaging and non-stick cookware, cosmetics, waterproof and stain-proof textiles and carpet, aqueous film forming foam (AFFF) to fight Class B fires, and as part of metal plating processes.”3 Teflon and Scotchgard were two of the pioneering products to utilize these fluoropolymers. The two most common PFAS are perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS).

Health Concerns of PFAS

Some of the most frequently cited health concerns associated with PFAS include adverse cardiovascular, immunity, developmental, and hepatic effects.3,4The most commonly heard refrain to minimize these concerns is that if they are so prevalent, why are there
not more health issues associated with them? In fact, “The Lancet Commission on Pollution and Health reported that pollution was responsible for 9 million premature deaths in 2015, making it the world’s largest environmental risk factor for disease and
premature death.” This was updated in 2019, and those numbers held steady, accounting for one in six deaths worldwide.5/i> While this number includes all types of pollution, the impacts are clear.

The Exposure Concerns of PFAS Are Regulatory and Legal

Due to their combination of persistence, pervasiveness, mobility, and the ability of some to bioaccumulate (or build up in animals and humans), they have been in the news recently too.6 Predictably, they are also now moving through the courts.7-9 Some of the most common areas of litigation are directed at PFAS found in drinking water and firefighting foam. The regulatory initiatives are also increasing. These address a range from water and soil to numerous manmade products including food packaging.10,11 The European Chemicals Agency (ECHA) and the NIH have a wealth of guidance and regulations that apply to PFAS.12,13

PFAS Detection

The EPA has useful direction for analytical methods development and sampling research that outlines the “laboratory validation process following a particular rulemaking or guidance effort and are available to support regulatory or guidance activities.”14 For
technique and equipment, PFAS are typically analyzed by mass spectrometry, coupled with gas chromatography or liquid chromatography (GCMS and LCMS), which enables detection in the low parts per billion.

AMPAC Analytical has years of experience and numerous experts in trace analysis by mass spectrometry who can assist with method development for high-volume analyses for both common and atypical sample matrices that will allow you to stay ahead of evolving regulatory concerns. Please contact us with specific questions or to receive a quote for PFAS quantitation.

References

  1. PFAS Explained | US EPA
  2. Understanding Organofluorine Chemistry. An Introduction to the C–F bond – Chemical Society Reviews (RSC Publishing)
  3. PFAS Health Effects Database: Protocol for a Systematic Evidence Map – ScienceDirect
  4. Toxicological Profile for Perfluoroalkyls (cdc.gov)
  5. Pollution and Health: a Progress Update – The Lancet Planetary Health
  6. ‘Forever Chemicals’ Are Everywhere. What Are They Doing to Us? – The New York Times (nytimes.com)
  7. PFAS Settlements: Future of PFAS Litigation Landscape to be Determined by Upcoming Decision | Reuters
  8. PFAS: The New Frontier of Product Liability – ProQuest
  9. DuPont, Corteva, and Chemours Announce Resolution of Legacy PFAS Claims | DuPont
  10. Trends in the Regulation of Per- and Polyfluoroalkyl Substances (PFAS): A Scoping Review
  11. PFAS in Food Packaging: State-by-State Regulations – September 2023 | Bryan Cave Leighton Paisner – JDSupra
  12. Per- and Polyfluoroalkyl Substances (PFAS) – ECHA (europa.eu)
  13. Guidance on PFAS Exposure, Testing, and Clinical Follow-Up – NCBI Bookshelf (nih.gov)
  14. PFAS Analytical Methods Development and Sampling Research | US EPA

Resources

Gas Chromatography – An Introduction

A Brief History of Gas Chromatography
Gas chromatography (GC) is one of the most important and prevalent analytical tools available to chemists. It was invented in 1952 by A.T. James and A.J.P. Martin as an outgrowth of research dating back to the previous decade.1-6 Their early techniques on adsorption and partition enabled some of the later developments that A.J.P. Martin spoke about at the 1957 Lansing Symposium, entitled, “Past, Present and Future of Gas Chromatography.” He concluded his address with the following prediction: “If we tie the gas chromatograph to other pieces of laboratory equipment, we have the possibility of almost the automatic chemist…”2 While the idea of an “automatic chemist” hasn’t quite come to fruition, Martin’s belief “that the uniting instrument of the gas chromatograph in the center” of his lab of the future certainly has.2 With the meteoric rise of GC7, its adoption and adaptation continued unabated in the ensuing decades. Then, “The introduction of robust, efficient, and reproducible fused-silica capillary columns and the provision of relatively inexpensive but reliable equipment for GC-MS provided a crucial new impetus in the 1980s.”1 Interestingly, the GC column saw some major advancements within just a few miles of AMPAC Analytical Laboratories. At that location, Walter Jennings and the company he co-founded, J&W Scientific, were instrumental in the development and manufacturing of capillary GC columns. The company was eventually purchased by Agilent in 2001.8,9 

GC Capabilities
Some of the analyses that GC can do include the separation of compounds in mixtures based on the polarity of the compounds, testing for purity – or for impurities, e.g., and detection of residual solvents. Conversely, with a technique known as preparative chromatography, GC can be used to prepare pure compounds from a mixture.  In pharmaceutical analysis, there are additional applications: 

  • Analysis of various functional groups. 
  • Determining purity of pharmaceutical compounds. 
  • Analysis of drugs that are commonly abused. 
  • Determination in pharmaceutical R & D the identity of natural products that contain  complex mixtures of similar compounds. 
  • Use in metabolomics studies.10 

GC for Testing Residual Solvents
The testing of residual solvents is necessary to ensure potency and, with some solvents, to determine their potential negative effects. GC is an excellent choice to do this. Residual solvents are separated into three classifications by the ICH (International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use): 

Class 1 solvents: Solvents to be avoided – Known human carcinogens, strongly suspected human carcinogens and environmental hazards. The Permissible Daily Exposure (PDE) of these solvents used in pharmaceutical products can range from 2 to 1500 parts per million (PPM).  

Class 2 solvents: Solvents to be limited – Non-genotoxic animal carcinogens or possible causative agents of other irreversible toxicity such as neurotoxicity or teratogenicity and solvents suspected of other significant but reversible toxicities. The PDE for these solvents ranges from 50 to 3880 ppm. 

Class 3 solvents: Solvents with low toxic potential – low toxic potential to man requiring no health-based exposure limit. Class 3 solvents have PDEs of 50 mg or more per day.11 

Within these three classifications, some of the most commonly used solvents are listed: 

  • Benzene (class 1)  
  • Acetonitrile, Cyclohexane, Hexane, and Methanol (class 2) 
  • Acetic Acid, Acetone, and Heptane (class 3).11,12 

Headspace GC (HSGC) and Direct Injection GC
In addition to the numerous GC developments such as column type, phase and coating techniques, and multidimensional GC, two types of sampling methods arose: direct injection (“DI”) and headspace (“HSGC”).  In the former, the sample is injected “directly” into a typical sample injector of the GC column. In HSGC, when heated, “the more volatile compounds will tend to move into the gas phase (or headspace) sample. The more volatile the compound, the more concentrated it will be in the headspace. Conversely, the less volatile (and more GC-unfriendly) components that represent the bulk of the sample will tend to remain in the liquid phase.“13 

Therefore, by extracting the “headspace vapor and injecting it into a gas chromatograph, there will be far less of the less-volatile material entering the GC column, making the chromatography much cleaner, easier, and faster.”13 Generally, HSGS is much cleaner and results in less wear on the column. For more on comparative techniques with DI and HSGC, please see the first entry in the “Resources” section below. 

We Can Assist with your GC Needs  

AMPAC Analytical has years of experience and numerous experts in Gas Chromatography who can assist with method development for analyses for both common and atypical sample matrices that will allow you to stay ahead of evolving regulatory concerns. Please contact us with any specific questions or to receive a quote for your GC needs.   

References  

  1. History of Gas Chromatography – ScienceDirect 
  2. The Development of Gas Chromatography – ScienceDirect 
  3. Gas-Liquid Chromatography: the Separation and Identification of the Methyl Esters of Saturated and Unsaturated Acids from Formic Acid to n-Octadecanoic Acid (PDF) 
  4. James A T & Martin A J P. Gas-liquid partition chromatography: the separation and microestimation of volatile fatty acids from formic acid to dodecanoic acid. Biochem. J. 50:679-90, 1952. (upenn.edu) 
  5. A New Form of Chromatogram Employing Two Liquid Phases – PMC (nih.gov) 
  6. Gas Chromatography | SpringerLink 
  7. Three Early Symposia Showing the Direction for the  Evolution of Gas Chromatography.pdf   
  8. Co-Founder of J&W Scientific, Gas Chromatography Pioneer Walter Jennings Dies | Agilent 
  9. Professor Walter Goodrich Jennings: A Remembrance (chromatographyonline.com) 
  10. Pharmaceutical Applications of Gas Chromatography 
  11. https://database.ich.org/sites/default/files/ICH_Q3C-R8_Guideline_Step4_2021_0422_1.pdf 
  12. ICH_Q3C-R8_Guideline_Step4_2021_0422_1.pdf 
  13. An Introduction to Headspace Sampling in Gas Chromatography Fundamentals and Theory (perkinelmer.com)  

Resources  

 

 

Nitrosamines – An Update

NDMA

The linkage between nitrosamines and cancer was first postulated by William Lijinsky in 1970. Then, in 2018, N-nitroso-dimethylamine (NDMA)) was detected in an active pharmaceutical ingredient, Valsartan (an Angiotensin-II-receptor antagonist).  Finally, the FDA issued a guidance for the industry, “Control of Nitrosamine Impurities in Human Drugs”, in the fall of 2020. However, the guidelines continue to evolve. There has been an update in March of 2021, with ongoing risk assessments.  Other regulatory agencies have instituted their own, along with updates. For example, since our blog series on nitrosamines, there have been some regulatory updates from the European Medicines Agency (EMA). Their new guidelines are outlined in a document entitled, “Questions and Answers for Marketing Authorization Holders/Applicants on the CHMP Opinion for the Article 5(3) of Regulation (EC) No 726/2004 Referral on Nitrosamine Impurities in Human Medicinal Products.”1 The updated section answers the crucial question, “Which limits apply for nitrosamines in medicinal products?” 

 The answers are provided with a definition of nitrosamines and acceptable exposures: 

 “The ICH M7(R1) guideline defines N-nitrosamines as substances of the “cohort of concern” for which limits in medicinal products refer to the so-called substance-specific acceptable intake (AI) (the Threshold of Toxicological Concern, TTC, value of 1.5 ug/day cannot be routinely applied) which is associated with a negligible risk (theoretical excess cancer risk of <1 in 100,000 over a lifetime of exposure). The calculation of AI assumes a lifelong daily administration of the maximum daily dose of the medicinal product and is based on the approach outlined in the ICH M7(R1) guideline as well as the principles described in relation to the toxicological evaluation in the assessment report of the CHMP’s Article 5(3) opinion on nitrosamine impurities in human medicinal products.”1,2 (A previous blog examined TTC, here.)  

A Useful List of Nitrosamine Limits  

In Appendix 1 found on the EMA site, there is a list of more than eighty nitrosamines listed, along with CAS numbers, known medicinal sources, CPCA (Carcinogenic Potency Categorization Approach) categories, and their guidance publication dates.3,4  

Some Caveats 

There are some exceptions that should be considered. For example, the EMA states, “The ‘less than lifetime’ (LTL) approach should not be applied in calculating the limits as described above but can only be considered after consultation with competent authorities as a temporary measure until further measures can be implemented to reduce the contaminant at or below the limits defined above.”1  

Additionally, those medications intended for advanced cancers also have some exceptions. For example, “If the active substance itself is mutagenic or clastogenic at therapeutic concentrations, N-nitrosamine impurities should be controlled at limits for non-mutagenic impurities according to ICH M7(R1).”  

There is also guidance when one or more than one nitrosamines may be present. For the latter, the guidance advises one of two approaches: 

  1. The total daily intake of all identified N-nitrosamines is not to exceed the AI of the most potent N-nitrosamine identified. 
  1. The total risk level calculated for all identified N-nitrosamines is not to exceed 1 in 100,000. The approach chosen needs to be duly justified by the MAH (Marketing Authorization Holder)/Applicant.1 

Final Thoughts on Nitrosamines 

Nitrosamine guidance worldwide is ever-evolving, yet the impetus to quantify and regulate them is clear. There will doubtlessly be further updates to regulations. AMPAC Analytical Laboratories – an SK pharmteco company (AAL) is an industry leader in the detection of nitrosamines and other genotoxic impurities (GTI), We have the specialized expertise, equipment, and methodologies to detect these impurities by gas chromatography or high-performance liquid chromatography coupled with mass spectrometry to support your API project.  Also, importantly, AAL can assist in navigating those projects within today’s regulatory landscape. Please contact us with any specific questions or to receive a quote for nitrosamines or other GTIs.  

References  

  1. https://www.ema.europa.eu/en/documents/referral/nitrosamines-emea-h-a53-1490-questions-answers-marketing-authorisation-holders/applicants-chmp-opinion-article-53-regulation-ec-no-726/2004-referral-nitrosamine-impurities-human-medicinal-products_en.pdf 
  2. ICH M7 Principles – Impurity Identification and Control (europa.eu) 
  3. https://www.ema.europa.eu/en/human-regulatory/post-authorisation/referral-procedures/nitrosamine-impurities 
  4. Appendix 1 Nitrosamine AIs (europa.eu) 

Resources 

AMPAC  Analytical

General Information on Nitrosamines 

Nitrosamine and Pharmaceuticals 

Regulatory Experiences with Root Causes and Risk Factors for Nitrosamine Impurities in Pharmaceuticals
https://doi.org/10.1016/j.xphs.2022.12.022 

Dynamic Vapor Sorption

Dynamic Vapor Sorption (DVS) is a gravimetric technique used to measure the change in mass of a material in response to changes to surrounding conditions such as temperature or humidity. DVS is primarily used with water vapor but can be applied to other organic solvents as well for the physicochemical characterization of solids. 

DVS was developed by Daryl Williams, the founder of Surface Measurement Systems Ltd., in 1991. The company then delivered the first working DVS instrument to Pfizer in 1992.1 Since then, many other equipment manufacturers have entered the field. 

Figure 1: A DVS isotherm plot indicating sorption and desorption rates and hysteresis. The isotherm shows a typical hysteresis curve where the adsorption phase is almost identical to the desorption phase (i.e. reversible). Note that at 80 %RH, there is net sorption of 0.9% between adsorption and desorption traces. The material appears to be slightly hygroscopic according to the definition in Ph. Eur.2 

Sorption, Desorption, Absorption, and Adsorption
There are five main physical processes that occur during the DVS experiment. The first, sorption, is when a material takes on moisture due to increased humidity. Conversely, desorption is the process that occurs when the material loses moisture due to decreasing humidity. Sorption can be classified as one of two types. Adsorption is moisture that is observed on a surface of a material, while absorption is moisture that has penetrated the surface of a material. The fifth term and the term that relates sorption and desorption is called hysteresis. The overall chart that includes and tracks the sorption and desorption rates and hysteresis is called the isotherm. These curves are crucial for understanding the physicochemical characteristics of a solid, such as porosity, polymorphic change, or liquefying of a sample.2 

Applications 

There are numerous reasons to utilize DVS, and some of the most common within the active pharmaceutical ingredient (API) industry are: 

  • To determine the sorption isotherm;  
  • To evaluate the hygroscopicity of an API powder;  
  • To compare the hygroscopicity of different solid-state forms: solvates, polymorphs, salts, amorphicity, and cocrystals; 
  • To determine the deliquescence point of a material
  • To quantify and qualify the amorphous content in drug substance or excipient,3 and 
  • To evaluate packaging materials. 

Of course, there are a variety of other applications in other industries, including for building materials, food science, cosmetics, coatings, and sealants. 

DVS Analysis of APIs
For pharmaceutical development, DVS is used for a variety of applications, including screening early drug and excipient candidates, establishing processing parameters, and identifying packaging and storage requirements (Figure 2).4,5  

 

Figure 2: A DVS isotherm of an API showing that the material started to gain significant mass after exposure to relative humidity values of more than 60 %RH. The change here was irreversible, as demonstrated by the desorption curve. DVS could be a useful tool to suggest storage conditions in terms of humidity contents in the surrounding environment. 

However, due “to the typically slow establishment of an equilibrium, DVS experiments are rather time-consuming.”4 Nevertheless, “water content of solid active pharmaceutical ingredients and excipients, individually and when formulated in pharmaceutical dosage forms, is a parameter that should be monitored throughout the drug lifecycle.”5  

As an analytical technique for APIs, DVS has become a necessary step within drug development and production, reducing issues that can arise during manufacturing, packaging, transportation, solubility, dissolution rate, stability, or storage. AMPAC Analytical’s sister company, SK biotek Ireland Analytical Services, has the experience, technology (including a Surface Measurement System’s DVS Resolution Dual Vapor Gravimetric Sorption Analyser), and support, to assist with this vital testing. Both companies are part of SK pharmteco and can easily transfer your project from either business unit to ensure the most optimal solution and logistical support are provided to meet your product timeline. We invite you to contact our team members and discuss how we can assist with your sorption testing requirements.  

Figure 3: A SMS DVS instrument like the one located at SK biotek Ireland. 

References  

  1. https://surfacemeasurementsystems.com/our-story/ 
  1. Ph. Eur., 2023, 11.2 Edition, Chapter 2.9.39  
  1. https://www.sciencedirect.com/science/article/abs/pii/S0022354915303348 
  1. https://www.sciencedirect.com/science/article/abs/pii/S0022354918302193 
  1. https://www.sciencedirect.com/science/article/abs/pii/S0022354916325230 
  •  

Keys To Effective Method Development

Effective method development is crucial for the quality control of Active Pharmaceutical Ingredients (API) and Drug Products (DP). Thorough method development enables successful downstream method validation. 

The regulatory guidance  specifies that: 

  • Method development and validation vary by application (quantitative, qualitative, etc.). 
  • It is phase appropriate. 
  • The client may provide additional guidance/validation criteria. 
  • The validation guidance directs how AMD (analytical method development) is conducted. 

Early Adoption of Forced Degradation Analysis
It is recommended that forced degradation be performed early in the method development lifecycle and that method parameters are suitable for mass spectrometry. This will prevent many issues that could occur in later stages and ensures the primary purity method is stability-indicating (specificity). When performing forced degradation, these considerations should be weighed: 

  • Always utilize a control sample without exposure to stressors.  
  • The stressors generally consist of acid, base, peroxide, heat, and photolytic conditions. Other stressors may be used based on known material incompatibility 
  • After exposing the compound to these stressors, target 5-20% degradation of the main peak.  
  • If degradation is not observed under reasonable conditions, then the material can be considered stable under those conditions. 

Impurity Genesis and Identification Assessment
Acceptance criteria should be scaled to impurity levels. How can the method unequivocally assess the analyte of interest in the presence of likely impurities, degradants, and the sample matrix? Additional considerations include: 

  • Is the method capable of identifying and/or quantifying a specific compound? 
  • Are there solvents present that can interfere with potential impurities? 
  • Known impurities? Are the known impurities stable under the method conditions? 
  • Is the method specific for degradation byproducts for stability-indicating methods? 

Cross-Platform Method Robustness
Robustness refers to a method’s ability to meet its analytical requirements (system suitability requirements) despite small variations of the method’s parameters, such as discrete changes to a column or sample tray temperature, percent organic modifier, flow rate, and detector wavelength. This capability is typically built into the method during method development.  

Precision or Accuracy – You Need Both in Strong AMD
AMPAC Analytical development strategy involves the early adoption of forced degradation studies with the goal of every primary purity method being stability-indicating (specificity) and mass spectrometry compatible. The validation strategy is phase-appropriate and application-specific and guides the development strategy. The validation acceptance criteria guidelines are specific to the test methodology, intended use, and level. Finally, method lifecycle performance is assessed, and reevaluation or revalidation can occur.  

 

 

 

 

Contact us today to learn how we can create accurate, precise method development for your API and DP pipeline.  

Phase-Appropriate Method Development 

The Cost of Drug Discovery and Development and How to Mitigate It 

The path to successful drug discovery and development is extremely long, expensive, and risky and can take between 10 to 15 years at an average cost of more than $1–2 billion for every new drug that is approved for clinical use.1,2 In fact, preclinical drug discovery alone “typically takes five and a half years and accounts for about one-third of the cost of drug development.”3,4 Therefore, even during the earliest stages of a drug product or active pharmaceutical ingredient project, phase-appropriate method development should be instituted to manage costs. This bolsters the chances for success and ensures reliable results, quality management, and reproducibility while avoiding “unreliable results (that) might not only be contested in court but could also lead to unjustified legal consequences for the defendant or to wrong treatment of the patient.”5 At its most basic, phase-appropriate method development maps the “what is needed” to “when it is needed.”6 Effective phase-appropriate method development can provide long-term product support by introducing mass spectrometry compatibility and forced degradation development to ensure your methods are stability-indicating and amenable to unknown impurity identification. By instituting a phase-appropriate method development process, combined with a quality-by-design approach around each logical sequence of events – and rigorously following it – it is more likely to create a cost-effective, successful outcome as you take the drug product through the regulatory process. 

It Can Pay to Outsource 

As the incentives for strong phase-appropriate method development increase, so too has the recognition of its value. Unfortunately, “it is not uncommon…to find pharmaceutical companies and contract research organizations (CROs) that are not taking advantage of the phase-appropriate approach and simply reference the typical ICH guidance for analytical items, such as method validation.”7 However, while FDA guidance encourages the use of a phase-appropriate approach, it is lacking in details and requirements. This leaves many companies to seek out ICH guidance as an alternative, conservative approach. Also, within their CGMP quality system, they may find it difficult to accommodate differing levels of CGMP compliance throughout the various clinical phases of development. This is when it might be an opportune moment to consider an outside expert that specializes in phase-appropriate method development processes for drug discovery and validation.  A successful yet robust phase-appropriate method development program can balance competing interests and requirements and still provide a regimen that meets the overall development goals without sacrificing any of the requirements of the program.  

AMPAC Analytical Laboratories (AAL), an SK pharmteco company, has decades of experience in providing a wide array of release testing services for raw materials, intermediates, APIs, and drug products. Our labs are equipped to handle hazardous, cytotoxic/high potency compounds as well as controlled substances for schedule II through V. Additionally, we have not only the expertise to conduct forced degradation experiments but also appropriate instrumentation like mass spectrometers to support later phases of development for your products. Please contact us to discuss how we can ensure the success of your drug discovery and development project and simultaneously reduce risks. 

References  

  1. https://www.sciencedirect.com/science/article/pii/S2211383522000521 
  2. https://www.frontiersin.org/articles/10.3389/fphar.2020.00770/full 
  3. https://www.frontiersin.org/articles/10.3389/fphar.2020.00770/full 
  4. https://www.nature.com/articles/nrd3078 
  5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3658022/ 
  6. https://www.pharm-int.com/2020/08/27/phase-appropriate-drug-development-validation-process/ 
  7. https://www.pharmtech.com/view/designing-phase-appropriate-cmc-analytical-programs 

Resources 

 

Forced Degradation Studies Can Reduce Stress(ors)

Forced Degradation is an important addendum to our previous post on Stability and Storage. Stressors are applied to new APIs and drug products to determine their degradation pathways and products under a variety of environmental conditions, including acid, base, light, heat, and oxidation. Forced degradation studies are also known as stress testing, stress studies, stress decomposition studies, and forced decomposition studies. These conditions “…are more severe than accelerated (stability) conditions and thus generate degradation products that can be studied to determine the stability of the molecule.”1  

Regulatory requirements for forced degradation were enacted by the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) in 1993.2   However, these guidelines are very general in (the) conduct of forced degradation and do not provide details about the practical approach towards stress testing. Although forced degradation studies are a regulatory requirement and scientific necessity during drug development, it is not considered as a requirement for (a) formal stability program.”1 However, stability studies have become a requisite for new drug moieties. In the absence of specific guidelines, the amount of stress needs to be representational: “Overstressing a molecule can lead to degradation profiles that are not representative of real storage conditions and perhaps not relevant to method development. Therefore, stress-testing conditions should be realistic and not excessive.”3 

AMPAC Analytical (AAL), an SK pharmteco company, can assist with forced degradation studies for products at all phases of development, in tandem with stability, storage, and method development, to ensure the viability of the drug products as they were designed. We introduce forced degradation studies early in method development to ensure your product quality throughout the development lifecycle. Contact AAL today to learn more.  

References 

  1. https://www.sciencedirect.com/science/article/pii/S2095177913001007 
  2. http://www.columbiapharma.com/reg_updates/international/ich/q1a.pdf 
  3. https://www.researchgate.net/profile/Dan-Reynolds-3/publication/279607256_Available_Guidance_and_Best_Practices_for_Conducting_Forced_Degradation_Studies/links/5afd6a2ca6fdcc3a5a44c50f/Available-Guidance-and-Best-Practices-for-Conducting-Forced-Degradation-Studies.pdf 

Learn more: https://ampacanalytical.com/laboratory-services/stability-program/ 

The Background, Advantages of, and Considerations for Radiolabeled Peptides

Integrin αvβ3 and RGD Binding

Integrin αvβ3 and RGD Binding

The Background, Advantages of, and Considerations for Radiolabeled Peptides
The use of radiolabeled peptides is a well-established tool in researching and treating many diseases and conditions. Selective receptor-targeting peptides are utilized as agents due to their rapid circulatory and tissue clearance and the high affinity and specificity to their targets. Peptides also have a relatively small size and low molecular weight compared to proteins and antibodies. There have been innovations and improvements in the design of peptides that incorporate chemical modifications with “impressive diagnostic accuracy and sensitivity.”1 Coupling these peptides with radiolabeling for peptide receptor radionuclide imaging (PRRI) and therapy (PRRT) has yielded remarkable results. In fact, a historical summary of radiolabeled peptides asserts, “The emergence of radiolabeled peptides for use with PET/CT such as 68Ga, 18F, and 64Cu, and the use of new receptor binding ligands…, have revolutionized PRRI and improved its diagnostic power beyond expectation.”2  

“Criteria for a successful peptide tracer,” to be utilized for PRRI, “are a high target specificity, a high binding affinity, long metabolic stability, and a high target-to-background ratio.”3 

Oncology and Radiolabeled Peptides
Oncology has benefited from the “tumor-philic” properties of Arg-Gly-Asp (RGD) peptides “because of their high affinity and selectivity for integrin αvβ3 – one of the most extensively examined targets of angiogenesis. Since the level of integrin αvβ3 expression has been established as a surrogate marker of angiogenic activity, imaging αvβ3 expression can potentially be used as an early indicator of the effectiveness of antiangiogenic therapy at the molecular level.”4 In addition to integrin αvβ3 expression, “tumor angiogenesis…has been well recognized as an essential hallmark for tumor growth, invasion, and metastasis.”4 All this has made RGD-containing peptides “promising molecular agents for imaging angiogenesis.”5  

Beyond Oncology
Since integrins are “involved in adhesion between cells and the extracellular matrix” and, as such, are associated with normal and pathological states, this family of receptors is useful for targeting a range of diseases.6 Current applications include “cardiovascular imaging, atherosclerosis imaging, remodeling after myocardial infarct or stroke, imaging of myocardial ischemia, identification of abdominal aortic aneurysm, imaging of chronic inflammation, pulmonary inflammation, assistance with bone marrow evaluations, and tissue engineering.”6  

A Brief History of RGD Peptides and Radiolabeling
Although radiopharmaceutical therapies, which can concurrently detect and treat tumors (i.e., theranostics, a portmanteau of therapeutics and diagnostics), have been around for eighty years, it was not until the combination of these payloads was combined with peptides that the potential for better targeting became a reality. The use of the RGD peptide sequence to attach to cells was first reported by M.D. Pierschbacher and E. Ruoslahti in Nature, nearly forty years ago in 1984, as a feature of fibronectin.5,7 Next, targeting tumors with radioactive peptides began, initiated by OctreoScan’s breakthrough in the early 1990s, wherein somatostatin receptor subtype 2 (SST-2) positive tumors were identified.8,9 After that, the first monomeric integrin-specific PET tracer used in patients was F-Galacto-RGD, a glycosylated RGD-peptide.5,10 Since then, the tripeptide R-G-D sequence has generally been utilized as a tracer, carrying the isotope to integrins that are expressed on both tumor cells and activated endothelial cells of tumor neo-vasculature. In the ensuing years, advancements have continued apace, and going forward, the intersection of PRRI/PRRT from radiolabeled peptides combined with AI, precision, and personalized medicine assures transformative innovations. 

Obstacles and Numerous Options for Radiolabeled Peptide Production
Because of their established safety, development, and design history, and “the fact that there are many RGD-based tracers with known pharmacokinetics, it can be useful to use them in the imaging of diseases that currently have no accurate method of diagnosis available.”6 However, there are barriers to synthesizing radiolabeled peptides: it can be a time-consuming, complex, multi-step process. It is also highly variable based on the peptide. Other drawbacks include the intricacies of radiolabeling and the lack of automation for some of these protocols.  

Fortuitously, RGDs radiopeptides are not the only ones exhibiting exciting potential for diagnostic imaging and targeted radionuclide therapies. An extensive review article from Paweł Kręcis et al. that appeared in Bioconjugate Chemistry is recommended, as it presents some developments and perspectives in both aforementioned areas regarding the research on somatostatin, bombesin, vasoactive intestinal peptide, gastrin, neurotensin, and exendin peptide analogs, among others.11 It includes the application of radiolabeled peptides and antibodies and an analysis of clinical trials reported in 2008-2018. 

Finally, just a few of the current companies doing interesting things in radiopharmaceuticals include Aktis Oncology, Mariana Oncology, Point Biopharma, and Rayze Bio. AMPAC Fine Chemical and AMPAC Analytical, both SK pharmteco companies, have been at the vanguard of small molecule and analytical developments utilizing innovative technologies and techniques directed by teams with decades of experience. If you have a radiolabeled peptide targeted for development, our teams can design, validate, optimize, and analyze your project, ranging from research to commercial quantities.  Contact us today for more information.     

References 

  1. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5235994/ 
  2. https://link.springer.com/article/10.1007/s00259-012-2064-5 
  3. https://link.springer.com/article/10.1007/s00259-011-2038-z 
  4. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4679356/ 
  5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8924613/ 
  6. https://link.springer.com/article/10.1007/s00259-020-04975-9 
  7. https://pubmed.ncbi.nlm.nih.gov/6325925/ 
  8. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6789871/ 
  9. https://www.cancer.gov/news-events/cancer-currents-blog/2018/lutathera-fda-gastrointestinal-nets 
  10. https://pubs.acs.org/doi/10.1021/ja9603721 
  11. https://pubs.acs.org/doi/full/10.1021/acs.bioconjchem.0c00617 

Related Articles and Resources