de-risk your drug development - covance

9
DE-RISK YOUR DRUG DEVELOPMENT Centralized Biopharmaceutical Chemistry, Manufacturing & Control Analysis

Upload: others

Post on 13-May-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: DE-RISK YOUR DRUG DEVELOPMENT - Covance

D E - R I S K Y O U R D R U G D E V E L O P M E N T

Centralized Biopharmaceutical Chemistry, Manufacturing & Control Analysis

Page 2: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

Lead to Clinical Candidate SelectionTaking the Right Molecule Forward

Taking this lifecycle approach also prepares the program for potential acceleration under regulatory schemes such as ophan drug designation or break through therapy status. In this instance, the central analytical laboratory would have a central repository of experience and expertise on the molecule and its associated methods to apply to the preparations for commercial release.

Introduction

According to recently conducted market research, 85% of respondents outsourced some or all of their manufacturing and analytical testing activities.1 Depending on your outsourcing strategy, a typical drug development program may involve 5-6 significant process changes at which evaluation can occur.

This introduces the potential for problems at BLA submission where a recent analysis of FDA complete response letters showed that 47% of the products that received them had CMC analytical issues. In many cases the drug had shown sufficient evidence of clinical efficacy but was delayed entering the market due to issues with the CMC package.

An alternative to moving analytical methods as the process moves to new facilities is to use a central analytical laboratory to provide all of the characterisation, method development, method validation and quality control activities required throughout the process. Since analytical methods under pin all of the activities in CMC, this will save time, money and ameliorate future regulatory issues.

Page 3: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

Developability involves stressing the protein to determine intrinsic molecular stability. As a result, the biopharmaceutical is exposed to a range of stress conditions, which normally include:

Stable Biologic

• Low risk

• Liquid formulation (High conc)

• Generic manufacturing process

As a component of the QbD approach, developability risk assessment studies are designed to de-risk process development at a very early stage. Ideally this based on a preliminary TPP and the anticipated MoA. These studies aim to assess the stability profile of a candidate biopharmaceutical, or rank the stability profiles of a series of similar biopharmaceuticals, from a manufacturing point of view, prior to formulation or process development.

Expansion of the risk assessed approach to an analytical control strategy ranks the CQAs related to the MoA, pharmacokinetics, anti-drug antibody liabilities, safety and stability profile.

Developability is a holistic risk assessment approach that takes a broad range of parameters into account. The stability of these parameters are then assessed in response to a range of stress conditions (see stability testing).

With this approach the risk of failure during manufacturing can be assessed and the biopharmaceutical can go directly into further development; be structurally modified to improve stability; formulated to improve stability or deselected as a potential drug candidate.

Developability Risk Assessment

• Thermal denaturation

• Humidity

• pH acid/base

• Ionic strength

• Freeze/thaw

• Oxidation

• Light

• Mechanical shear

• Glycation

Less Stable Biologic

• Medium risk

• Liquid formulation (Lower conc)

• Modified manufacturing process

Unstable Biologic

• High risk

• Lyophilisation

• Lyo manufacturing process

• Re-engineer

Risk assessment

Physicochemical Characterization

Primary structure/ product-related impurities:

• Mass spectrometry – Multi-attribute monitoring

• Intact mass/peptide mapping/ N- & C-termini sequencing/ disulphide bonds/glycan analysis

• Fragmentation, oxidation and deamidation degradation products

• HCP monitoring

• CE-SDS - molecular size and truncated forms

• Isoelectric focusing - charged isoforms/ deamidation/ C-terminal lysine

• Reversed phase HPLC - hydrophobicity/ hydrophilicity profile

• Ion exchange chromatography – charged variant profile

Primary structure/ product-related impurities:

• Circular dichroism - secondary and tertiary structural analysis

• Intrinsic and extrinsic fluorescence - tertiary structural analysis

• Differential scanning calorimetry - protein thermodynamics/tertiary structure

• Isothermal titration calorimetry - thermodynamics of protein binding

• Surface plasmon resonance - protein binding

• SEC-MALLS - quaternary structure/aggregation

• Dynamic light scattering - quaternary structure/ aggregation

• Analytical ultracentrifuge - quaternary structure/ aggregation

Far from being a complete list, other analytical methods may be required. In addition, advances in analytical technology mean that new techniques are continually being made available. Ultimately, the selection of techniques in a protein characterization package will depend on the nature of the biopharmaceutical and the potential impurities that arise from the various routes of degradation that it undergoes.

Listed below are some of the analytical techniques that can be used to provide protein characterization data. As no one analytical technique can provide a comprehensive understanding of a specific protein parameter, an approach using orthogonal techniques is usually required.

The ICH Q6B guidelines describe the need for a comprehensive understanding of the molecular structure of any therapeutic protein, over and above the testing required for batch release. In essence, protein characterization involves analytical techniques that can determine the physicochemical properties of the biopharmaceutical as well as evaluating the primary structure and higher order structure (secondary/ tertiary and quaternary structure) of the protein. Protein characterization also involves determining product-related impurities arising from spontaneous chemical reactions, enzymatic degradation or aggregation.

Page 4: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

Biological Characterization

With biologics drug discovery, the active biopharmaceutical is generally selected based on the knowledge of normal and diseased biochemical systems and the known physiological function of the protein molecule.

Once the biopharmaceutical(s) has been selected, it is essential to understand with some detail the MoA of the protein in treating the disease state: there is increased focus on understanding the pathology of a specific disease, identifying and validating appropriate biomarkers and providing more effective proof of therapeutic concept, to increase the overall success rate.

It is also important to correlate the MoA with any known structure-function relationship.

Such studies can involve in vitro and/or in vivo analysis. Examples of such studies include:

Antibody Biological Characterization

Currently, the majority of the biopharmaceuticals in development are monoclonal antibodies or antibody-related molecules. In addition to their ligand binding characteristics through the complementary-determining region (CDR), such monoclonal antibodies also can manifest biological activity through the Fc region of the protein. This biological activity occurs by the Fc region binding to specific Fcγ receptors on effector cells to induce immune responses such as ADCC, CDC and/or ADCP.

Monoclonal antibodies can be selected or designed to possess or be deficient in Fcγ receptor functionality. Irrespective of this, regulatory authorities have an expectation that effector function will be evaluated to determine whether this will have an impact on the product safety and efficacy. Specifically, they require ADCC, CDC, ADCP and cytotoxic

properties along with Fc receptor and Complement C1q binding activities to be evaluated.

Characterization studies usually start early in the drug discovery/preclinical phases of development. Initially a limited set of characterization methods can be applied to the molecule, but as the biopharmaceutical progresses down the development pipeline, increasing a full comprehensive package of characterization techniques is required.

Labcorp has a standard screen for all Fc receptors (e.g., FcγIII for ADCC, FcRn) by SPR. Additional functional cell-based assays will be performed based on antibody class and need to support further characterization (e.g., ADCC, CDC, ADCP).

Class I MOA:Cell-bound antigen with depletion

Class II MOA:Cell-bound antigen with blocking

Class III MOA:Soluble antigen with blocking

CDCApoptosis

NK Cell ADCC

• Cell proliferation

• Apoptosis/necrosis

• Effector function

• Receptor/ligand binding

• Cell signalling

• Migration, transmigration

• Microbial challenge

These studies provide critical insight into the appropriate potency assay strategy of your drug which provides confidence a consistently manufactured product is administered during all phases of a clinical investigation. This is then linked to clinical efficacy being demonstrated by substantial evidence from adequate and well-controlled clinical investigations with a consistently manufactured product.

Characterising your drug throughout development is therefore a critical success factor to reach market and to ensure your potency assay strategy is fit for purpose.

Page 5: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

BiopharmaceuticalAnalysisThe Road to Quality Control Testing

Formulation Development

Pre-Formulation Risk Assessment

Formulation development is the process by which the active biopharmaceutical is combined with inactive excipients to produce a final drug product that is efficacious, stable and can readily be administered to the patient. The excipients each have a specific function to ensure product performance and conformance.

Formulation development can range from the simple mixing of excipients with the drug substance to additional processing steps such as lyophilisation to produce a dry formulation.

Formulation development involves evaluating a range of excipients and determining the effect upon the protein structure and stability profile using a series of physico-chemical analytical techniques.

Stress testing with a range of factors is generally applied to evaluate the effectiveness of the excipients and for optimisation purposes. Moreover, in the search for the formulation that provides the optimal stability and delivery characteristics, modern approaches to formulation development use DoE approaches in order to evaluate a wide range of excipients and physical conditions yet minimise the size of the studies to a manageable level.

The elements of a pre-formulation risk assessment are sequence analysis, conformational stability and colloidal stability.

With this knowledge, an early assessment can be made with regard to formulation development. The data directs what formulation format would be most appropriate for the molecule, e.g. liquid or lyophilised powder. It also indicates which formulation conditions would be favorable for the molecule

and identify any potential need for stabilzing excipients.

The use of such upfront risk assessment studies has cost and resource implications early in development; however, the outputs of such studies leads to reduced issues during product development, process development and cGMP manufacturing resulting in significant cost, resource and timeline savings at a later stage.

Sequence analysis

In silico assessment of potential degradative events:

• Deamidation

• Oxidation

• Proteolytic hydrolysis sites

Conformational stability

• Isoelectric point (pI)

• Thermal stability (Tm)

• Hydrophobicity/ hydrophilicity

Colloidal stability

• Polyethyleneglycol (PEG) solubility

• Diffusion interactive parameter (KD)

• Viscosity at high concentration

• Aggregation on-set temperature

During pre-formulation risk assessment, lead candidates are screened for conformational and colloidal stability as well as the potential for post-translational modifications (PTMs) that could perturb the structural integrity of the biopharmaceutical. Pre-formulation assessment is performed in the R&D drug discovery phase, prior to preclinical development activities.

Page 6: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

To be able to manufacture, release and demonstrate stability, a biopharmaceutical requires a range of appropriate analytical techniques. Irrespective of whether the method is a physicochemical assay or bioassay, such techniques need to be robust, reproducible and capable of operating in a GMP environment.

Analytical development starts with information gathering and selection of a technique capable of measuring the relevant biopharmaceutical parameter. This selection should be based on sound scientific principles and ‘fit for purpose’.

Over the last decade the paradigm for analytical development has been shifting from the classical ‘one factor at a time’ (OFAT) approach to the multivariate ‘quality by design’ (QbD) approach.

With OFAT, the concept is to vary only one variable at a time while keeping the other variables fixed. In contrast, the QbD approach uses the statistical design of experimental (DoE) method to vary two or more key variables to produce a multivariate design space (with analytical QbD known as method operable design region or MODR), which is the operating range for the key variables. This gives an early

assessment of robustness parameters that will contribute to our understanding of phase-appropriate validations.

With analytical QbD, risk assessment tools are used to determine the key input variables to experimentally test. While both approaches are acceptable, regulators are increasingly expecting the QbD approach to be adopted. Analytical development is performed in the R&D space and the outcome should be an assay, the characteristics of which are well understood, and is suitable for qualification or validation for cGMP operations.

Method Development and Optimization

Method Establishment or Method Transfer (Early Phase Method Establishment)

Where an assay has already been developed and the method needs to be operated in an alternative laboratory, method establishment or method transfer comes into play. Method establishment occurs at an earlier stage of development prior to a method undergoing qualification/validation.

Method transfer is a defined regulatory process that transfers a qualified or validated analytical method, along with appropriate expertise and documentation from one site to an operating site.

An important part of moving an assay from the originator laboratory to the recipient laboratory is to demonstrate that the transferred method is operating to an appropriate standard. Since these methods underpin the entire CMC package it is important to plan these transfers well in advance and to follow the appropriate type of method transfer to avoid issues in the future.

Comparative testing:

Both originating and receiving labs participate in method transfer exercise. Results are compared to pre-defined acceptance criteria.

Co-validation:

The receiving laboratory participates in the initial method validation activities.

Method validation or revalidation:

Complete or partial validation is performed by the receiving laboratory.

Transfer waiver:

Typically not applicable to non-compendial analytical test methods for biologics.

Method Validation

Before an analytical method can be used in a cGMP setting it is necessary to show that it is fit-for-purpose. This is demonstrated by a phase appropriate validation assay, which is performed in line with the regulatory guidelines described in ICH Q2 (R1) Validation of analytical procedures.

Qualification is a phase-appropriate validation to qualify the critical parameters of a method and is typically performed without pre-specified criteria. The ICH-defined process of validation establishes documented evidence to provide assurance that an analytical method will consistently produce a result meeting its predetermined specifications and quality attributes.

As outlined in ICH Q2(R1), the assay parameters that need to be considered in a qualification/validation exercise are:

• Specificity

• Linearity

• Range

• Accuracy

• Precision

• Repeatability

• Intermediate precision

• Detection limit

• Quantitation limit

• Robustness

Validation exercises are performed by QC analysts and regulatory compliance is overseen by QA.

The decision of when to qualify or validate a particular assay is at the discretion of the developer, provided it occurs before licensure. Nevertheless, generally with cGMP manufacture of Phase I clinical batches qualified assays are acceptable, whereas with later development the general expectation is that assays should be validated.

It should also be noted that changes to the drug substance/drug product manufacturing processes or formulations would generally require re-qualification or re-validation of the relevant analytical methods. The same applies if the method itself is changed or modified.

Testing for Impurities

Characteristic Identification Quantitative Qualitative Potency

Accuracy

Repeatability

Intermediate Precision

Specificity

Detection limit

Quantitation limit

Linearity

Range

Prec

isio

n

Page 7: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

QC Release Testing

A crucial component of any cGMP manufacture is the QC release testing of the batch/lot. With each GMP batch or lot, specific critical quality attributes of the drug substance or drug product need to be demonstrated and are required to fall with in an acceptance range outlined in a prescribed release specification.

Guidance regarding the release of drug substance and drug product is provided in ICH Q6A Test Procedures and Acceptance Criteria for New Drug Substances and New Drug Product: Chemical substances or ICH Q6B: Test Procedures and Acceptance Criteria for Biotechnological/ Biological Products.

With release testing analytical methods are required to address the following:

• Appearance and description

• Identity

• Purity and impurities

• Potency

• Quantity

• General/compendial tests

While the guidelines advise on the general tests that need to be applied for QC release, there is no stipulation of the specific methods to be employed.

These are selected by the biopharmaceutical developer based on sound scientific principles; moreover, as described above, the methods need to be qualified or validated, depending on the phase of development.

Generally, the methods used for QC release testing use well-established physicochemical techniques, such as:

• Compendial assays

• Liquid chromatography

• Electrophoretic chromatography

• UV/visible spectroscopy

• Biophysical analysis

At Labcorp, with a view to the future, we have developed GMP-compliant LC-MS MAM workflows for use in release testing.

For the determination of biological activity and more specifically potency a biological assay needs to be included in the release specification. While animal-based assays can be used for QC release, cell-based potency assays are more preferable for assay variability, cost and ethical reasons.

Stability Testing

Forced Degradation

An essential component of any development program is stability, requiring a range of stability studies to demonstrate that the biopharmaceutical remains intact and potent during storage. The importance of such studies is highlighted in a set of regulatory guidance documents ICH Q1A(R2) Stability testing of new drug substances and products, ICH Q1B Photostability testing of new drug substances and products and ICH Q5C Stability of biotechnology/biological products.

During the drug discovery or preclinical phases, real time stability data is generally unavailable, therefore in order to gain some understanding of drug substance/drug product stability and stability-indicating capability of the assays and to understand any product stability liabilities, it is necessary to perform forced degradation (i.e., stress testing).

The outcomes of any biopharmaceutical forced degradation study are:• To determine the intrinsic stability of

the biopharmaceutical

• To determine the degradation pathways

• To identify and elucidate the structures of the degradative products

• To establish which analytical techniques are truly stability- indicating

There are essentially four types of stability study used in evaluating a biopharmaceutical:• Real time stability study

• Accelerated stability study

• Forced degradation stability study

• In-use stability studies

Real-time and accelerated stability studies are generally performed in association with the manufacture of GMP grade material, to determine shelf life of the product and provide some surety in case of temperature

Through LC-MS MAM, it is possible to monitor amino acids from various sections of molecule and makes it possible to monitor multiple degradation pathways at specific sites of modification.

These studies involve exposing the drug substance/drug product to severe conditions in order to accelerate any degradation reactions which normally include:• Thermal denaturation

• Humidity

• pH acid/base

• Ionic strength

• Freeze/thaw

• Oxidation

• Light

• Mechanical shear

excursions from the standard storage conditions.

Real time stability studies are also performed in association with clinical studies, where their role is to demonstrate that any biopharmaceutical that is being evaluated in the clinic has not undergone degradation and remains potent. Globally, Labcorp has ICH-stability chambers to support hundreds of stability time points annually.

Possible alternatives are immunological, ligand binding or enzymatic assays, depending on the nature of the biopharmaceutical. QC release testing is performed by trained QC analysts under the auspices of the Quality Assurance team to ensure GMP compliance.

The in-use study is a short term study that is intended to demonstrate the stability of the drug product for its use in the clinic setting during the administration to patients.

Although several of the stability studies can be carried out for R&D purposes, the formal real-time and accelerated studies are performed under GMP with qualified or validated analytical methods.

Page 8: DE-RISK YOUR DRUG DEVELOPMENT - Covance

De-Risk with Centralized Analytical Services

Centralized CMC Analytical Services from LabcorpLabcorp Overview

Labcorp’s global CMC analytical activities spans the development cycle from initial R&D discovery studies through to QC batch release testing for clinical trials, and on to support commercial release.

With a focus on your goals, a pragmatic risk assessed approach enables milestone achievements. The target product profile (TPP), mechanism-of-action (MoA) and critical quality attributes (CQAs) assist in developing an analytical control strategy.

Tap into experience and insight from work across biotherapeutics including: recombinant proteins, vaccines and cell and gene therapies. Plus, global expertise in correlating structure-function relationships, including in-vitro analyses, structural characterization, formulation development and forced degradation stress testing, with supporting method development, technology transfer, and phase-appropriate validation.

We have successful completed global regulatory audits (FDA, MHRA, EMA) and offer a range of both non-cGMP and cGMP CMC biopharmaceutical development studies.

Scientific Review

Labcorp’s Lead Analytical Scientist works directly with you to either design the suite of studies you need or, where appropriate, review the existing data on your molecule to help specify and design any additional (or repeat) studies.

Program management

The overall program will be managed by a dedicated program manager, who will oversee the timelines and delivery of the program.

Experimental conduct

Experienced Analytical Scientists will be allocated to each individual study in the program. These scientists will oversee the conduct of the experimental phases in the laboratories and will collate and report the data.

Program communication

Labcorp’s customers have direct access for detailed discussions. The program manager also updates customers frequently with study status and results.

Program report

Each Lead Analytical Scientist will issue a QC-checked, peer-reviewed and fully interpreted draft report for your studies within 8 weeks of the experimental phase completing.The final report will be fully QC- checked and QA-audited (where applicable) prior to issue.

Program review

At the end of the program a full review of all the data and reports will be conducted by the Lead Analytical Scientist. This review will indicate if possible additional studies may be required and is conducted with reference to the latest regulatory guidance(s) from the EMA, FDA and JMHLW. The program review can include a visit by our Senior Scientist (and/or other senior employees) to your company to present an overall summary of our findings (if required).

Over

750CMC projects including...

...45that have gone into NDAs/BLAs

Over

2,000Biologics development studies conducted over the last 5 years

Regulatory Compliance

Labcorp’s global CMC services division capabilities can operate both to non-GXP and cGMP. We satisfy all international regulatory requirements, and work to relevant cGMP, ICH, FDA and EU guidelines.

For CMC these include:

• ICH Q1A (R2) stability testing of new drug substances and products

• ICH Q2 (R1) validation of analytical procedures

• ICH Q6B specifications: Test procedures and acceptance criteria for biotechnological/ biological products

• ICH Q6B Appendices for physio-chemical characterization and impurtities

• ICH Q5C stability testing of biotechnological/ biological products

Resources

Facilities in Greenfield, IN, Harrogate, Huntingdon and York, UK house approximately 400 scientists working to provide full CMC services in both an R&D and cGMP manner.

Access highly-experienced and specialist teams - holding PhDs in relevant fields - for physicochemical analysis, bioassay and genomics, who operate in facilities including a cell culture suite, PCR suite and an ICH-compliant stability suite. By keeping abreast of scientific

advancements and regulatory guidelines, we help your team ensure that the science behind your discoveries is fully reflective of modern day pharmaceutical requirements.

Leverage 30+ years of experience working with NCEs and NBEs with particular expertise in monoclonal antibodies, ADCs, gene therapies, cell therapies and vaccines.

Pursue your goals using knowledge gained from conducting over 750

CMC projects including 45 that have gone into NDAs/BLAs in the last 5 years alone.

The CMC services division at Labcorp is a cGMP-compliant facility using fully validated industry standard software and instruments and we partner with the world’s largest and smallest product development companies.

For clients who wish to follow the quality by design (QbD) approach to product and process development, we work to the guidelines outlined in:• ICH Q8 (R2) pharmaceutical

development

• ICH Q9 quality risk management

• ICH Q10 pharmaceutical quality system

All cGMP studies are conducted under the auspices of Labcorp’s GMP quality assurance groups to ensure regulatory compliance. This QMS has been audited by all of the principle regulatory authorities and has never received a critical comment in a regulatory audit.

Page 9: DE-RISK YOUR DRUG DEVELOPMENT - Covance

Originally authored in 2020. ©2022 Laboratory Corporation of America® Holdings All rights reserved. BROCMC001-0222

Learn more at drugdevelopment.labcorp.com/CMC