Regulatory Pathways for Antibody Therapeutics

Ana Reis, PhD Ana Reis, PhD 10 min read

Since they were first commercialized in 1986, antibody therapeutics have emerged as powerful tools in combating a myriad of human diseases. From infections and inflammatory conditions to autoimmune disorders and solid tumors, monoclonal antibodies are engineered to target specific proteins or cells in the body and offer a targeted approach, tapping into the body's immune response to tackle diseases.

But how do these cutting-edge treatments navigate the intricate web of regulatory pathways to reach patients in need? Today, we’ll explore the regulatory pathways that guide the development and approval of antibody-based treatments.

Understanding Antibody Therapeutics

Before discussing regulatory procedures, let’s review the fundamentals. Antibodies are Y-shaped proteins produced by white blood cells called B cells. They play a crucial role in defending against infections by binding to foreign substances (antigens).

A single mature B cell produces significant quantities of a distinctive antibody tailored to bind exclusively to one region, known as an epitope, on a molecule or antigen. Antibody production techniques, therefore, isolate the individual B cell and generate clones (hybridoma method) or isolate the genetic sequence of the antibody (antibody phage display and B cell sorting) from the B cell. Antibodies that are derived from a single (mono) B cell clone (clonal) are referred to as monoclonal antibodies (mAbs).

Custom monoclonal antibodies are designed to bind to specific antigens ranging from pathogens to cancer cells. With their precise targeting capabilities, mAbs have gained considerable traction in clinical development for various diseases.

The Power of Therapeutic Antibodies

Antibodies can treat various conditions, including infectious diseases, cancer, and autoimmune disorders.

  • Targeted Therapy: Unlike traditional cancer treatments like chemotherapy, which can kill healthy tissues, monoclonal antibody immunotherapies to treat cancer offer targeted treatments that minimize collateral damage to healthy tissues. Therapeutic antibodies can also be tailored to individual patients based on their specific disease characteristics. By identifying unique biomarkers of disease, therapeutic antibodies can precisely target molecules that contribute to the pathology of disease.
  • First-Line Treatment: In many cases, mAbs have become the first line of defense against exposure to toxins, venoms, and select pathogens. Administration of anti-toxin, anti-venoms, or antibodies that neutralize pathogens offers patients quick and effective treatment strategies.
  • Tumor Microenvironment: In the realm of cancer treatment, mAbs can also modulate the tumor microenvironment (TME), which consists of various cell types, including tumor cells, immune cells, stromal cells, and the extracellular matrix. For example, checkpoint inhibitors cut the immunological brakes of immune cells in the TME, enhancing the immune response against solid tumors.
  • Prolonged Duration of Action: Antibody therapies have extended half-lives, unlike drugs that are metabolized by the liver. This means they remain active in the body for longer periods, reducing the frequency of administration. Patients benefit from fewer injections (or infusions), leading to improved convenience, compliance, and efficacy.

Boost your yields! Get your free transient expression handbook to learn how!

Get it Now

Regulatory Agencies and Their Role

The development and approval of therapeutic antibodies involve rigorous evaluation by regulatory agencies worldwide. These agencies play a critical role in evaluating the safety, efficacy, and quality of new therapies, including antibody-based treatments.

They ensure that therapeutic antibodies meet rigorous standards before reaching patients. Let’s delve into their functions and how they contribute to the global landscape of approved antibody therapies.

1. U.S. Food and Drug Administration (FDA)

In the United States, the U.S. Food and Drug Administration (FDA) handles drug approval and the regulatory process for antibody therapies. Here’s what they do:

  • Review and Approval: The FDA evaluates Biologics License Applications (BLAs) submitted by pharmaceutical companies. These applications provide comprehensive clinical data on the antibody’s safety, efficacy, manufacturing processes, and labeling that the FDA goes through.
  • Balancing Benefits and Risks: The FDA carefully weighs the benefits of antibody therapies against potential risks. Their decisions impact patient safety and public health.

2. European Medicines Agency (EMA)

The EMA oversees the approval process for antibody therapies in the European Union.
Their responsibilities include:

  • Marketing Authorization: Similar to the FDA, the EMA assesses applications for marketing authorization. It ensures compliance with EU regulations and evaluates clinical data. 
  • Harmonization: The EMA collaborates with other regulatory agencies globally to harmonize standards and streamline approvals.

Antibody Therapies Approved Worldwide

Let’s explore the global picture. According to the Umabs Antibody Therapies Database, as of June 2022, 165 antibody therapies have received approval from at least one regulatory agency worldwide. Here’s the breakdown:

  • United States: The US leads the way with 122 approved antibody therapies.
  • Europe: Europe closely follows, with 114 approvals.
  • Japan and China: Rapid advancements in the past decade have propelled Japan (82 approvals) and China (73 approvals) into prominent positions.

Types of Approved Antibodies

What about the current tally of approved antibodies? Here’s a breakdown of the numbers to reveal the distribution across different antibody types.

  • Canonical Antibodies (115): These are the traditional monoclonal antibodies widely used for various indications, such as Nebacumab (Centoxin), Daclizumab (Zenapax, Zinbryta), Rituximab (Rituxan, MabThera), etc.
  • Antibody-Drug Conjugates (14): Combining antibodies with chemotherapy agents, these therapies deliver targeted cytotoxic cargo to cells. Examples include Gemtuzumab ozogamicin (Mylotarg), Brentuximab vedotin (Adcetris), and more.
  • Bispecific Antibodies (7): These innovative molecules simultaneously target two different antigens, enhancing treatment precision. Approved bispecific antibodies include Blinatumomab (Blincyto), Catumaxomab (Removab), Emicizumab (Hemlibra), etc.
  • Antibody Fragments (8): Truncated pieces of antibodies that retain antigen-binding specificity, often used in diagnostics, research, and therapy due to their compact size and diverse applications, such as Abciximab (ReoPro, Clotinab), Ranibizumab (Lucentis), and Certolizumab pegol (Cimzia).
  • Radiolabeled Antibodies (3): These antibodies carrying radioactive isotopes are used in diagnostic imaging techniques and targeted radiotherapy. The three approved radiolabeled antibodies are Iodine 131 tositumomab (Bexxar), Iodine 131 derlotuximab biotin (Cotara, Vivatuxin), and Iodine 131 metuximab (Licartin).
  • Fc-Fusion Proteins (12): A protein fused to the Fc region of an antibody having an extend half-life and enhance stability, such as Rilonacept (Arcalyst), Etanercept (Enbrel), Alefacept (Amevive), and more.

PD-1 is the most popular target, with 14 approved antibody-based cancer treatments globally. Antibody therapeutics approved outside the EU or US include Sotrovimab, an anti-SARS-CoV-2 antibody (approved in Japan in July 2021), and Prolgolimab (Forteca), an anti-PD-1 mAb for melanoma (approved in Russia in 2020), among others.

Although the specific antibody production techniques for these approved monoclonal antibodies are not widely disclosed, it is understood that most of them were developed using hybridoma technology, phage display, or transgenic mice expressing human immunoglobulins.

Clinical Development and Approval: Navigating the Regulatory Process

Regulation of clinical development and approval is a complex journey crucial for bringing innovative therapies to the forefront of the treatment of patients. Central to this process is the establishment of primary endpoints and the exploration of the antibody’s therapeutic potential by drug regulatory agencies.

In this context, adherence to regulatory standards is paramount, ensuring that each step taken brings us closer to delivering safe and effective treatments to those who need them most.

1. Discovery and Preclinical Research

Before human trials, extensive preclinical studies are conducted, often utilizing transgenic animals to assess safety and efficacy. Scientists identify targets that are diagnostic or therapeutic candidates, such as cancer biomarkers or viral proteins indispensable for host cell invasion. Once targets are identified, extensive laboratory testing is conducted to develop and characterize candidate antibodies.

Extensive preclinical studies assess the safety, efficacy, and pharmacokinetics of the candidate antibodies in cell cultures and animal models before advancing to human trials.

2. Investigational New Drug (IND) Application

With promising preclinical data in hand, sponsors submit an Investigational New Drug (IND) application to regulatory agencies like the FDA, outlining plans for human trials. Regulatory agencies meticulously review the IND application to ensure that proposed clinical trials meet ethical and safety standards.

3. Clinical Trials

Divided into phases, clinical trials evaluate the safety and effectiveness of the therapy in humans. This process involves rigorous monitoring and data collection. Each phase of clinical development follows a carefully designed protocol outlining study objectives, patient eligibility criteria, treatment regimens, and endpoints.

  • Phase I: Small-scale trials in healthy volunteers or patients evaluate safety, dosing/tolerability, and pharmacokinetics. Researchers determine the optimal dose and assess any adverse effects.
  • Phase II: Phase II trials expand the patient population to further assess safety and begin to explore efficacy in specific disease indications involving patients with the target disease.
  • Phase III: Phase III trials involve large patient cohorts with hundreds or thousands of patients to evaluate the efficacy, safety, and optimal dosing regimens. It also involves comparing the antibody therapy to existing treatments (if any).

4. Regulatory Submission

Upon successful completion of clinical trials, developers compile data from all phases. Biologics License Application (BLA) or a New Drug Application (NDA) to the FDA (or other regulatory agencies) for approval. While rapid development is crucial, patient safety remains paramount. Regulatory agencies employ stringent measures to uphold these standards.

Regulatory agencies decide whether to approve the therapeutic antibody based on the review of clinical data, manufacturing processes, and risk-benefit assessment. Some antibodies are approved as first-line treatments, while others as maintenance therapy or to complement existing therapies.

If approved, labeling information detailing indications, dosing, and safety information is finalized. Additionally, post-market commitments may be required for further monitoring or studies.

5. Post-Approval Surveillance

Even after approval, regulatory agencies continue to monitor the safety and efficacy of therapeutic antibodies through pharmacovigilance programs to detect and address any unforeseen safety concerns that may arise in real-world settings.

Healthcare professionals and patients are encouraged to report any adverse events or side effects associated with the use of the therapeutic antibody, facilitating ongoing post-market surveillance and safety monitoring.

Accelerating Therapeutic Antibody Development with Regulatory Strategies

To hasten the availability of treatment for patients, particularly in domains concerning severe and life-threatening illnesses with unaddressed medical requirements, numerous regulatory bodies have implemented pathways aimed at expediting the process of drug development and approval.

For example, the FDA has implemented expedited pathways to streamline the approval process for promising candidates and speed up approval rates for antibody therapeutics:

  • Priority Review: Expedited review is granted to therapies that offer significant advancements in treatment of diseases, shortening the review timeline to 6 months.
  • Breakthrough Therapy: A procedure designed to accelerate the progression and evaluation of therapies targeting a severe condition, with initial clinical data suggesting potential significant advancements compared to existing therapies on clinically relevant endpoints.
  • Accelerated Approval: These guidelines permit medications addressing severe conditions with unmet medical requirements to gain approval relying on a surrogate endpoint.
  • Fast-Track Designation: Designed for therapies addressing unmet medical needs, fast-track designation expedites development and review processes.

Approval Success Rates

Approval success rates reflect the likelihood of an antibody therapeutic progressing through clinical trials and ultimately receiving regulatory approval. These rates are crucial for resource allocation and decision-making within the biopharmaceutical industry.

Antibodies developed between 2010 and 2019 demonstrated higher success rates for every phase transition compared to earlier years, leading to substantially improved overall approval success rates. In recent years, well-validated targets (such as CD20, HER2, PD-1, and PD-L1) and intensive research on innovative antibody formats contributed to even higher success rates.

Antibody therapeutics have a global approval success rate in the range of 14–32%, while the US/EU approval success rates are within 28% of therapeutic antibodies achieving approval.
In general, antibodies developed for non-cancer indications tend to have higher success rates.

Types of Antibodies and Their Considerations

Not all therapeutic antibodies are created equal. Variations in their structure and origin can also impact their therapeutic potential and regulatory pathway.

  • Human vs. Murine Antibodies:  Fully human antibodies (usually obtained by isolating antigen-specific B cells from the blood) tend to have lower immunogenicity, reducing the likelihood of adverse immune reactions. While effective, murine antibodies derived from mice can trigger immune responses in humans due to immunogenicity, necessitating additional steps to mitigate these risks.
  • Chimeric Antibodies: A type of engineered antibody that combines components from different species. These antibodies are designed to harness the beneficial properties of both human and non-human antibodies, offering advantages in therapeutic applications.
  • Constant and Variable Regions: Antibodies have constant (Fc) and variable (Fab) regions. An antibody’s constant region plays a crucial role in its pharmacokinetics and effector functions, influencing its therapeutic profile. These regions determine the antibody’s specificity, dictating which targets it can bind to with precision.
  • Transgenic Animals and Rapid Development: Transgenic mice produce human antibodies, allowing scientists to make human antibodies by immunizing mice, expediting therapeutic antibody development. Rapid development is crucial for treating infectious diseases or emerging threats.

When it comes to regulatory pathways for antibody therapeutics, it’s evident that innovation and safety go hand in hand. Antibody therapeutics hold immense promise in treating a wide range of human diseases, and regulatory agencies play a pivotal role in ensuring their safety and efficacy. From the laboratories where they are crafted to the regulatory agencies tasked with safeguarding public health, collaboration and diligence pave the way forward.

With each milestone achieved, we inch closer to realizing the full potential of antibody therapeutics in treating patients and combating various diseases. As research continues, we can expect more innovative antibody-based treatments to emerge and get approved, benefiting patients worldwide.

Let’s continue this journey with unwavering dedication fueled by the promise of a healthier tomorrow. As a leading contract research organization, ProteoGenix boasts a wealth of expertise in crafting tailored therapeutic monoclonal antibodies. With our comprehensive therapeutic antibody services, we’re equipped to guide you through every step, from pinpointing targets to nurturing clinical candidates.

Partner with us and accelerate your journey toward groundbreaking treatments. Don’t hesitate to reach out – we’re here for you.

 

 


Author Author: Ana Reis, PhD

Ana Reis is a scientific content manager at ProteoGenix. She has a PhD degree in biotechnology and a master's degree in science communication. Throughout her academic career, she has co-authored dozens of scientific articles on topics ranging from bioinformatics to chemical engineering.