article

Exploring the potential of ADCs beyond oncology

Posted: 6 August 2024 | | No comments yet

Antibody Drug Conjugates (ADCs) are already deployed as part of the roster of oncology treatments, but here, Mike Ward, Head of Thought Leadership for Life Sciences & Healthcare, Clarivate, looks into their potential to help treat a whole range of other diseases and conditions. He discusses the barriers and enablers that will impact ADCs’ ability to make the shift from oncology to a broader use spectrum.

Antibody drug conjugates

What are the main therapeutic areas beyond oncology where ADCs show potential?

A number of areas represent significant opportunities for Antibody Drug Conjugates (ADCs) beyond oncology, leveraging their ability to deliver therapeutic agents specifically to diseased cells or tissues while minimising off-target effects. The development of ADCs in these areas is ongoing, with several preclinical and clinical studies exploring their potential.

The main areas where ADCs are demonstrating potential include:

Infectious diseases:

  • Bacterial Infections: ADCs can target specific bacterial pathogens, delivering antibiotics directly to the bacteria, which may help reduce antibiotic resistance and protect beneficial microbiota.
  • Viral infections: ADCs can be designed to target viral proteins, offering a new approach to treating viral infections like HIV and hepatitis.

Autoimmune diseases:

  • Rheumatoid Arthritis: ADCs targeting specific immune cells or inflammatory mediators can provide more precise treatment options with potentially fewer side effects.
  • Systemic Lupus Erythematosus (SLE): Targeting specific immune pathways with ADCs can help reduce disease activity and improve patient outcomes.

Neurological disorders:

  • Alzheimer’s Disease: ADCs targeting amyloid-beta plaques or tau protein aggregates could offer new treatment options for neurodegenerative diseases.
  • Multiple Sclerosis: ADCs could be used to selectively target and modulate pathogenic immune cells or inflammatory processes in the central nervous system.

Cardiovascular diseases:

  • Atherosclerosis: ADCs could be engineered to target and deliver drugs to atherosclerotic plaques, potentially stabilising them and preventing cardiovascular events.
  • Heart Failure: Targeting specific pathways involved in cardiac remodelling and fibrosis with ADCs might offer new therapeutic strategies for heart failure.

Metabolic disorders:

  • Diabetes: ADCs could target and modulate insulin resistance or inflammation associated with diabetes.
  • Obesity: By targeting adipose tissue or specific metabolic pathways, ADCs could offer new treatments for obesity and related metabolic disorders.

Hematologic disorders:

  • Haemophilia: ADCs could be used to deliver clotting factors or other therapeutic agents directly to the site of bleeding, improving efficacy and reducing side effects.
  • Anaemia: Targeted delivery of erythropoiesis-stimulating agents to the bone marrow using ADCs could enhance the treatment of various forms of anaemia.

Fibrotic diseases:

  • Pulmonary Fibrosis: ADCs can target and modulate fibrotic pathways in the lungs, offering potential treatment for conditions like idiopathic pulmonary fibrosis.
  • Liver Fibrosis: Targeted therapies using ADCs could help reduce liver fibrosis and prevent the progression to cirrhosis.

How do companies prioritise their R&D investment between oncology and other therapeutic areas for ADCs?

This prioritisation is based on a combination of market demand, scientific feasibility, regulatory environment, competitive landscape, potential for breakthrough therapies, strategic fit, partnerships, and financial considerations.

Oncology receives substantial investment due to the high incidence of cancer and significant unmet needs in this area. Oncology also offers high returns and the opportunity to address severe, life-threatening conditions. However, other therapeutic areas, such as autoimmune diseases, infectious diseases and neurological disorders, also attract considerable investment if they present significant unmet needs, a large number of affected patients and a lack of effective treatments.

The availability of well-characterised targets is essential and oncology benefits from extensive research identifying tumour-specific antigens, making it a fertile ground for ADC development. Advances in ADC technology, such as improved linkers and more effective cytotoxic payloads, enable the exploration of non-oncological indications as well, driving investment decisions based on scientific feasibility in these areas.

Oncology often has accelerated approval pathways due to the critical nature of the diseases, attracting R&D investment. Expedited programs like the FDA’s Breakthrough Therapy Designation and Fast Track can shorten development timelines and reduce risk. Clear and supportive regulatory pathways for non-oncological diseases can also encourage investment in those areas.

Intense competition in oncology may lead companies to seek opportunities in less crowded fields, assessing the number of competitors and the status of competing therapies. Therapeutic areas with fewer competitors or novel targets can be attractive, offering companies the chance to establish leadership and differentiate their products.

The possibility of developing treatments that significantly improve survival or quality of life drives investment in oncology. Similarly, the potential to develop transformative treatments in other areas, such as autoimmune or genetic diseases, can attract investment if the therapeutic promise is high.

Companies often focus on areas aligned with their core competencies and existing expertise. A company with a strong oncology focus may prioritise ADCs in cancer, while those with expertise in other fields may invest in those areas. Portfolio diversification is another strategy, with companies investing in multiple therapeutic areas to balance risk and maximise opportunities.

Partnerships and collaborations can influence R&D priorities. Collaborating with academic institutions, biotech companies, and research organisations provides access to new targets, technologies, and expertise, driving investment in specific areas.

Finally, companies evaluate potential return on investment (ROI), considering development costs, time to market, pricing, and reimbursement prospects. Oncology drugs often command high prices, potentially leading to higher ROI. Projects with clear funding pathways and resource availability are more likely to be prioritised.

What are the main challenges associated with the development and manufacturing of ADCs?

The development and manufacturing of ADCs is challenging due to their complex composition, involving a monoclonal antibody, a cytotoxic drug and a linker. Overcoming scientific, technical and regulatory hurdles requires multidisciplinary expertise and robust processes to bring these therapies to market safely and effectively.

Key challenges include selecting and optimising the components, such as identifying antibodies that target specific cells with high specificity, choosing potent cytotoxic drugs with minimal toxicity to healthy cells and developing stable linkers that release the drug inside target cells. Achieving precise and consistent conjugation, ensuring batch consistency and maintaining molecular integrity are crucial for regulatory approval.

Manufacturing ADCs involves scaling up production while maintaining quality, navigating complex processes and implementing rigorous quality control measures. Addressing stability issues and optimising storage conditions are necessary to ensure efficacy and safety over time.

Ensuring safety and efficacy involves minimising off-target effects and balancing the therapeutic window. Navigating complex regulatory pathways, providing comprehensive data, managing high development costs, and securing favourable reimbursement are essential for successfully bringing ADC therapies to market.

What role do contract development manufacturing organisations (CDMOs) play in addressing these challenges?

CDMOs are crucial. They provide pharmaceutical companies with the expertise and infrastructure needed to overcome technical, logistical and regulatory hurdles, ensuring efficient development, high-quality standards and timely market entry.

CDMOs bring specialised knowledge including antibody production, linker technology and cytotoxic drug conjugation. They offer sophisticated process development capabilities, optimising conjugation methods and purification techniques. Advanced manufacturing facilities and comprehensive analytical services ensure the production of high-quality ADCs with consistent drug-to-antibody ratios (DAR), purity, stability and potency.

Regulatory support is another critical service provided by CDMOs, helping navigate the complex regulatory landscape for ADCs and ensuring compliance with global standards. Their assistance in preparing documentation and data for regulatory submissions is invaluable. By outsourcing to CDMOs, pharmaceutical companies can focus on core competencies while leveraging the CDMO’s expertise to optimise resources and accelerate development timelines.

Many CDMOs offer end-to-end services, from early-stage development to commercial-scale manufacturing and packaging, ensuring continuity and reducing technology transfer risks. They invest in cutting-edge technologies and engage in continuous improvement practices, enhancing ADC production efficiency and quality. Dedicated project management helps coordinate various aspects of ADC development, ensuring timelines and milestones are met.

By partnering with CDMOs, pharmaceutical companies can share the risks associated with ADC development and manufacturing. CDMOs often have contingency plans in place to handle unexpected challenges, ensuring continuity in the supply chain and mitigating risks related to process failures, regulatory hurdles and market uncertainties.

What are the key factors that contribute to the successful approval of ADC therapies?

The successful approval of ADC therapies depends on robust preclinical data, including target validation, mechanism of action and a strong safety profile. Clinical efficacy requires significant benefits like improved survival, demonstrated through well designed trials with appropriate endpoints and patient criteria.

Safety and tolerability necessitate effective management of adverse events and a favourable therapeutic window. Manufacturing excellence ensures consistent, high-quality ADCs with scalable production from clinical to commercial scale.

Regulatory strategies involve early engagement with agencies and comprehensive dossiers covering preclinical, clinical and manufacturing data. Biomarker development and companion diagnostics are crucial for patient selection and personalised treatment.

A strong intellectual property portfolio and leveraging regulatory incentives protect market share. A commercial strategy includes market access, reimbursement, pricing and a reliable distribution network for widespread availability.

Post-approval commitments include Phase IV studies and robust pharmacovigilance programs to monitor long-term safety and efficacy. These measures ensure ongoing success and safety of ADC therapies.

What future trends do you foresee in the ADC landscape, especially in terms of market growth?

Innovations in ADC technologies include next-generation linkers and payloads for improved stability and selectivity, as well as bispecific and multi-specific ADCs that target multiple antigens for enhanced efficacy.

Expanding therapeutic applications are on the horizon, with increased exploration into non-oncological diseases and personalised medicine approaches using biomarkers for tailored therapies. Combination therapies and dual-function ADCs, which integrate other treatments or gene-editing tools, are expected to further enhance efficacy.

Manufacturing advancements such as continuous manufacturing and automation will improve scalability and cost-efficiency. Regulatory frameworks are likely to become more adaptive and global harmonisation of standards may streamline the approval process. Increased investments, strategic partnerships and robust intellectual property protections will drive further development and market growth, while sustainable pricing and favourable reimbursement policies aim to ensure patient access and affordability.

 

About the author

Mike WardMike Ward, Head of Thought Leadership for Life Sciences & Healthcare, Clarivate

As both a journalist and analyst, Mike has been writing, analysing and commenting on the life sciences industry for more than 35 years. His actionable insights and analysis support pharma, biotech, healthcare and medtech stakeholders gain a deeper understanding of market news and trends. Mike has held editorial and content leadership roles at various publications and organisations across the industry.

Leave a Reply

Your email address will not be published. Required fields are marked *