Protein-based Therapeutics

At BioNTech, we use our expertise to harness different biological pathways of the immune system. Protein-based therapeutics have become a backbone approach in modern medicine due to their fundamental role in all biological processes and the technological advances in synthetic protein production. Based on our proprietary and our collaborators' technology and know-how, we utilize a variety of next-generation protein-based technologies, including antibodies, antibody-drug conjugate therapeutics and synthetic lysins. Our aim is to expand the application of protein-based therapeutics to broader patient populations and novel indications in the field of oncology and infectious disease.

Antibodies

We aim to develop the next generation of immunomodulatory antibodies that activate the immune system against cancer.

Antibodies are part of our body’s natural search-and-destroy army. Different antibodies can be designed to target different types of cancer, enhancing, modifying, or mimicking the immune system’s attack on cells. 

At BioNTech, we use our expertise to harness different biological pathways of the immune system and design pioneering cancer immunotherapeutics. Based on our and our collaborators' technology know-how, we are utilizing a variety of antibody technologies including bispecific antibodies in our Next Generation Checkpoint Immunomodulators platform and single monoclonal antibodies in our Targeted Cancer Antibody platform.

At a glance: Antibodies as a therapeutic drug class

  • Proven technology that has fundamentally changed cancer care improving treatment outcomes and quality of life by engaging the immune system
  • Natural molecules that are the immune system’s first-line defense mechanism
  • Flexible drug class that can be engineered to address one or more targets of choice
  • Established manufacturing for off-the shelf treatment

We apply multiple antibody formats with the goal of developing highly effective therapeutics for various cancer indications:

Bi-specific monoclonal antibodies represent a powerful new therapeutic treatment option as they can bind different targets (e. g. cancer cell proteins or immune cell proteins) simultaneously. This property can be relevant in complex cancer indications where tumor cell recognition by immune cells is impaired or where more than one key molecule should be targeted.

BioNTech develops specifically formatted bispecific antibody candidates with Genmab that have the ability to bind two targets at the same time. As a new class of checkpoint immunomodulators, they are designed to simultaneously block targets that cancer cells use to silence the immune system as well as conditionally stimulate immune cell functions. This approach has the potential to enhance cytotoxic properties at the tumor site leading to the destruction of cancer cells.

Monoclonal antibodies, or mAbs, represent a specialized laboratory-produced class of antibody therapeutics that are engineered to recognize and bind specific proteins on cancer or immune cells. These targeted antibodies can be used to bind to a particular target structure on the surface of the cancer cell, thereby flagging it for subsequent destruction by immune cells. In addition, they can also be designed to bind to surface proteins on immune cells, thereby modulating their activity, e.g., by stimulating or inhibiting certain pathways.

In a proprietary program, we develop monoclonal antibodies against a novel target expressed specifically in pancreatic and other gastrointestinal cancers. This target plays a role in metastasis formation and is a marker of an aggressive form of cancer. Directly blocking this target with a monoclonal antibody offers the possibility to interfere with cancer cells migrating to other tissues, thereby inhibiting metastasis formation.

In collaboration with Genmab, we develop a monoclonal antibody candidate that binds and stimulates T cells, thereby aiming to (re)activate and increase antitumor immunity. Together with OncoC4, we advance a monoclonal antibody candidate that is designed to bind and inhibit immunosuppressive regulatory T cells in the tumor microenvironment. The goal of this approach is to prevent the inhibition of T cell activity and therefore enhance anti-tumor effects.

Our antibody discovery engine enables rapid and efficient production of new mAb candidates, aiming to change the cancer treatment landscape

Our antibody discovery platform can generate and develop highly specific, functional monoclonal antibodies (mAbs). These antibodies are well known for their exceptional and unique target specificity and high affinity while showing a reduced risk of being immunogenic. This corresponds with a better safety profile. This proprietary antibody platform is also used for the identification of alternative antibody formats. With the use of the antibody discovery platform, tumor- and immune cell-specific binders are thus made available for our drug classes.

Combining our antibody knowledge and mRNA expertise, we aim to establish a truly revolutionary class of antibody therapeutics which could be directly produced in the patient’s body. Learn more about our mRNA-encoding antibody platform RiboMab here.

Combining our antibody knowledge and mRNA expertise, we aim to establish a truly revolutionary class of antibody therapeutics, directly produced in the patient’s body. Learn more about our mRNA-encoding antibody platform RiboMabs here

Antibody-drug conjugates

We aim to engineer innovative antibody-drug conjugates as broadly applicable precision medicine therapeutics that we believe could become an alternative to traditional chemotherapy.

Antibody-drug conjugates (ADCs) are a class of potent cancer therapies combining the selectivity of antibodies with the cell-killing properties of anti-cancer chemotherapies (“payload”). Unlike traditional chemotherapeutic approaches, ADCs target tumor-specific surface proteins, thereby potentially limiting the impact of the payload on surrounding tissues and avoiding off-target toxicity.

A critical aspect of the efficacy of ADCs is the stability of the region linking the antibody and the payload, as it largely determines the payload delivery to the target cancer cell. Based on our collaborator’s technology know-how, we aim to design highly stable antibody-payload therapeutic candidates with improved delivery properties. The ADC platform also enables us to combine previously incompatible antibody-payload combinations. These features underline the potential of our ADC candidates as part of a broadly applicable precision medicine drug class that we believe could offer an alternative to traditional chemotherapy.

At a glance: Antibody-drug conjugates as a drug class in precision oncology

  • Combination of an antibody that targets tumor-specific structures and cytotoxic drug compound (“payload”) to ensure targeted delivery of the payload to cancer cells  
  • Antibody and payload are connected by a linker region that determines the stability of the bond, aiming to ensure delivery of the cytotoxic compound without damaging healthy tissue
  • Based on our collaborator's technology platform, we aim to develop highly stable ADC candidates with the potential, if approved, to address a range of solid tumors

Synthetic lysins

Mimicking nature’s evolution to address bacterial infections

Lysins are antibacterial proteins that degrade the bacterial cell wall and that are specific to individual types of bacteria . In nature, lysins are typically produced by bacteriophages, viruses that specifically infect bacteria for replication. Bacteriophages utilize lysins to break open the bacterial cell wall in the final part of their replication cycle to further spread their viral copies.

Based on our LysinBuilder™ platform we aim to design optimized lysins that build on nature’s evolution but have improved pharmaceutical properties. Our next-generation synthetic lysin candidates are designed to degrade the cell wall of specific bacteria  while preserving the natural microbiome of the body. We believe synthetic lysins may have the potential to overcome antibiotic resistance and to improve the standard of care across many types of bacterial infections.

At a glance: Synthetic lysins as a new class of precision anti-bacterials

  • Engineered cell-wall degrading proteins with improved pharmaceutical properties aimed to target and eliminate bacteria
  • Potential novel therapeutic approach to address specific types of bacteria while preserving the natural microbiome of the body
  • Potential to overcome challenges of antibiotic resistance and biofilms 
  • Laboratory studies have shown high efficacy and stability of our synthetic lysin candidates against specific types of bacteria

Read also