How To Navigate The Nonclinical Evaluation Landscape Of Biopharmaceuticals

Biotechnology-derived pharmaceuticals (biopharmaceuticals) cover a broad variety of active substances, including proteins and peptides, their derivatives, and products composed by proteins and peptides, that could be developed from cell cultures or using recombinant DNA technology.

This article presents the characteristics of biopharmaceuticals compared to conventional chemically‑synthesized pharmaceuticals (small molecules) and describes certain specifications to be considered for the design and conduct of nonclinical studies for biopharmaceuticals.

Table 1, Characteristics of Biopharmaceuticals vs Small Molecules

Biopharmaceuticals have unique characteristics that are different from small molecules, as noted in Table 1 below.

CharacteristicBiopharmaceuticalsSmall Molecules
Molecule size/structureLarge/complexSmall/uniform
ManufacturingFrom natural sources (human, animal, plant, microorganisms) by biotechnology methodsChemically synthesized
Product stabilityVery sensitive to environmental factorsGenerally, less sensitive to environmental factors
Target selectivity/specificityHighLower
Immunogenicity potentialHighLower
MetabolismCatabolized/degradedMetabolized
Route of administrationParenteralParenteral and others
High dose selection for nonclinical testingGuided by saturation of pharmacodynamic response and dose multiple to clinical doseGuided by Maximum Tolerated Dose (MTD)

Nonclinical Evaluation

The nonclinical evaluation directly depends on the type of compound. Due to the unique characteristics of biopharmaceuticals, many of the principles of conventional nonclinical evaluation require modification to achieve scientifically valid safety and efficacy outcomes for biopharmaceuticals. The properties of biopharmaceuticals that relate specifically to variation in nonclinical requirements include high selectivity and specificity as well as generally well characterized pharmacokinetic (PK) properties for certain classes. The specific applications of these factors to nonclinical requirements are described below.

Table 2, Biopharmaceuticals Nonclinical Program vs Small Molecules

Study TypeTiming Relative to Clinical StudiesBiopharmaceuticalsSmall Molecules
PharmacodynamicsBefore Phase 1YesYes
Safety pharmacologyBefore Phase 1Product specificaYes
In vitro metabolic profile; plasma protein bindingBefore Phase 1NoYes
Systemic exposureBefore Phase 1YesYes
In vivo ADMEb; in vitro drug interactionsGenerally, before Phase 3NoYes
General toxicologyBefore Phase 1, Phase 2, and Phase 3YesYes
GenotoxicityIn vitro mutagenic and clastogenic studies, before Phase 1; complete battery, before Phase 2NocYes
CarcinogenicityFor marketing approvalProduct specificdProduct specifice
Reproductive and developmental toxicityCompleted before Phase 3 or marketing approvalProduct specificfGenerally, Yes
ImmunotoxicityCompleted before Phase 3YesYes
Local toleranceBefore Phase 1YesYesg

ADME = Administration Distribution Metabolism Excretion.

  1. Evaluation as a part of toxicology/pharmacodynamic studies is often sufficient in the case of biopharmaceuticals with high target specificity. Consider a more extensive evaluation in case of a novel therapeutic class and/or no high target specificity.
  2. Comparative in vivo human and animal metabolism data.
  3. Protein or peptide administration in large quantities may generate uninterpretable results, and direct interaction with DNA or other chromosomal material is not expected with biopharmaceuticals. Studies in available and relevant systems only needed in case of cause of concern.
  4. Dependent on clinical dosing duration, patient population, product biological activity (e.g., growth factors or immunosuppressive agents), or cause of concern (weight of evidence).
  5. Chronic drugs or cause of concern.
  6. Dependent on the product, clinical indication, and intended patient population.
  7. In case of parenteral administration.

Seven Considerations for Design and Conduct of Nonclinical Studies for Biopharmaceuticals

  • Consideration 1: Animal Species Selection
    Due to the high selectivity and species specificity of biopharmaceuticals, it is important to identify and select relevant animal species for efficacy and safety evaluation. In vitro studies in mammalian cell lines that predict in vivo activity and assess species sensitivity (including human) can be used to identify and select relevant animal species. Additionally, tissue cross‑reactivity (TCR) studies can guide the selection by comparing tissue binding profiles between human tissues and animal tissues where target binding is expected.

Homologous molecules or transgenic models can be used when no relevant species can be identified (no interaction with the orthologous target in any species).

  • Consideration 2: Pharmacodynamics (PD)
    Biopharmaceuticals have limited intracellular access considering their high molecular weight and structural complexity, and therefore, their targets are extracellular.
  • Consideration 3: Pharmacokinetics/Toxicokinetics
    Typically, biopharmaceuticals have limited distribution, with limited intracellular access, and a long half-life. Classical biotransformation studies are not needed since the metabolic pathways of biopharmaceuticals are generally understood (expected to degrade to small peptides and individual amino acids).
  • Consideration 4: General Toxicity
    The toxicity of most biopharmaceuticals relates to their targeted mechanism of action and therefore, adverse effects are apparent as exaggerated pharmacology.

Usually, toxicology testing requires two different species. However, one species is acceptable for biopharmaceuticals if pharmacologically active only in one species.

High dose selection for toxicology studies can be determined using PK/PD approaches that identify a dose which provides:

  1. The maximum intended pharmacological effect in the nonclinical species; and
  2. An approximately 10-fold exposure over the maximum exposure to be achieved in the clinic.

The higher of these two would be the high dose, unless the use of a lower dose is justified (e.g., maximum feasible dose [MFD]). In case in vivo/ex vivo PD endpoints are not available, PK data and available in vitro binding and/or pharmacology data can be used to select the high dose.

The duration of repeat-dose studies for biopharmaceuticals is based, as for small molecules, on the intended clinical treatment duration and disease indication. Duration usually ranges between 1 to 3 months for biopharmaceuticals, and the recommended duration for short-term and chronic use/indications is included in Table 3, as follows:

Intended Clinical UseDuration
Short-term use (< 7 days) and acute life-threatening diseasesUp to 2 weeks
Chronic indicationsGenerally, 6 months
Chronic useScientifically justified

Source: ICH S6 Preclinical Safety Evaluation of Biotechnology derived Pharmaceuticals.

Because of their high selectivity and specificity, biopharmaceuticals have less potential for off‑target toxicity. Since TCR studies can supplement knowledge of target distribution and potential unexpected binding, it is recommended to perform a TCR study with a panel of human tissues to support the initial clinical dosing; knowing that tissue binding per se is not indicative of in vivo biological activity, and that binding to areas not typically accessible in vivo to biopharmaceuticals (e.g., cytoplasm) is normally not relevant.

  • Consideration 5: Carcinogenicity
    A weight of evidence approach can be followed (review of relevant data, additional endpoints in toxicology studies, understanding target biology related to potential carcinogenic concern) and if there is no evidence of cause of concern, no carcinogenic studies are needed.
  • Consideration 6: Reproductive and Developmental Toxicity
    The species-specific profile of embryo-fetal exposure to biopharmaceuticals should be considered since high molecular weight proteins do not cross the placenta by simple diffusion (e.g., the neonatal Fc receptor [FcRn], a transport mechanism, varies across species).
  • Consideration 7: Immunogenicity
    There are potential risks of allergic reactions and immunotoxicity considering the origin and nature of biopharmaceuticals; therefore, antidrug antibody (ADA) responses and the impact on PD activity, exposure, or safety due to immune-mediated reactions should be characterized (typically as part of toxicology studies).

Conclusions

The aforementioned considerations and specifications are important for the optimal drug development of biopharmaceuticals to best support the clinical evaluation and marketing authorization. Overall, nonclinical development of biopharmaceuticals requires a scientific and rational approach focusing on the unique characteristics of the product and an understanding of the current regulatory environment.

Please click here to start a conversation with our nonclinical experts today .

References

  • American College of Toxicology. eLearning Seminar Basic Topics in Toxicology American College of Toxicology. November 2022.
  • Food and Drug Administration. Webinar Nonclinical Safety Assessment for Small Molecules and Biologic Drug Development. May 2020.
  • ICH Harmonised Tripartite Guideline M3(R2) Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals. June 2009.
  • ICH Harmonised Tripartite Guideline Q5C Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products. November 1995.
  • ICH Harmonised Tripartite Guideline S1A Guideline on the Need for Carcinogenicity Studies of Pharmaceuticals. November 1995.
  • ICH Harmonised Tripartite Guideline S6(R1) Preclinical Safety Evaluation of Biotechnology‑derived Pharmaceuticals.
  • ICH Harmonised Tripartite Guideline S7A Safety Pharmacology Studies for Human Pharmaceuticals. November 2000.

This article was authored by :

  • Pinar Merino-Pascual, Senior Medical Writer I, Regulatory and Medical Writing
  • Margaret Studzinska, Senior Director, Regulatory and Medical Writing
  • Wai-Fung Chau, Senior Medical Writer, Regulatory and Medical Writing

Suggested For You

perspectives

October 15th, 2024

Putting the Action in Diversity Action Plans and the Real-Time Data Visualization Technology Needed to Ensure It Happens

perspectives

October 8th, 2024

Diversity Action Plan Guidance Part I: Implications for Sponsors

perspectives

September 30th, 2024

Meet the Leaders Driving MMS’s European Growth

perspectives

September 30th, 2024

The Future of Data Management and Biostatistics: Trends and Technologies Shaping the Industry

perspectives

September 24th, 2024

Embracing Quality Management Maturity (QMM) at the Cornerstone of the Pharmaceutical Industry

perspectives

September 11th, 2024

From Historical Precedent to Modern Approvals: Lessons Learned on OTC Drug Products for FDA OMORs

perspectives

September 4th, 2024

Email Security – Navigating Through the Process of Validation and Compliance with Healthcare Business Stakeholders

perspectives

August 27th, 2024

Optimizing Oncology Drug Development: FDA Expedited Pathways, Real-Time Review, and Global Programs

perspectives

August 20th, 2024

Clinical Study Protocols: A Comprehensive Guide to Best Practices From A Senior Medical Writer

perspectives

August 5th, 2024

Ensuring Robust Data Privacy and Protection: An Overview of the MMS Framework

perspectives

July 30th, 2024

The Critical Role of Quality Control (QC) – Medical Writing and Beyond

perspectives

July 23rd, 2024

PSI 2024 Ignited Conversations on External Data Sources, Requirements for Estimands, and Bayesian Methodology for Statisticians in Pharma