Antibody art image

Enhanced specificity and synergistic function of bispecific and multispecific antibodies

Monoclonal antibodies have emerged as powerful therapeutic modalities. Understanding of antibodies, and how to change specific characteristics through antibody engineering to influence their pharmacokinetic and physiochemical properties has progressed dramatically in recent years [1]. Now considered a mainstream therapeutic, monoclonal antibodies have provided the foundational knowledge for the progression to increasingly complex antibodies and antibody fusion molecules. As of 2022, 27 of these complex antibody molecules had been approved by regulatory agencies around the world [2]. Further, preclinical and clinical pipelines have become rich with engineered complex antibody formats that have the potential to enhance therapeutic efficacy over their monospecific counterparts in complex diseases such as cancer [3,4].

Generated through antibody engineering methodologies, these complex antibody and antibody fusion molecules consist of bispecifics, multispecifics, and Fc-fusion proteins. Bispecific antibodies allow a single antibody-like molecule to simultaneously engage 2 different targets, or antigens [5]. Similarly, multispecific antibodies consist of multiple antigen targeting domains within a single molecule. When compared to monospecific antibodies, therapeutic efficacy of these bi and multispecific constructs are enhanced through this multiple and simultaneous target engagement [6]. These molecules can further be engineered and differentiated based on their intended effect after binding to their target, which can include target antigen neutralization, recruitment/activation of desired immune cells, and labeling of desired antigens for subsequent effector functions [5].

An example of a bispecific antibody on the market is Blinatumomab. Consisting of a fragment-based bispecific, Blinatumomab functions by ligating CD19+ target cells with T cells through the dual targeting of CD3 and CD19 in leukemias and lymphomas [5]. Another example molecule is known as a TriKE, which is a trispecific killer engager. These function by linkage of target cancer cells to effector cells that have been primed by the TriKE for enhanced cytotoxicity against the target cell [7].

Fc-fusion proteins are engineered biologics that contain the IgG Fc domain that is fused to various effector molecules [8]. Multiple advantages can be conferred through this fusion, such as enhanced in vivo stability, ultimately leading to better therapeutic outcomes [8]. Etanercept, Abatacept, and Belatacept are all commercially approved Fc fusion proteins that have proved to be indispensable in the autoimmunity and transplant space [9]. Together, these antibody derivative formats have demonstrated significant promise in the clinic and see continued investments in development. Further research will progress our understanding of these molecules.


Benefits of a complete gene to protein workflow

As understanding of engineered antibody constructs and their antigen targets progresses, the need to explore new ideas and additional constructs is inevitable. Novel biomarkers of disease progression will be identified, necessitating innovative combinatorial antibody or Fc-fusion constructs to be developed for evaluation of potential therapeutic synergy. Optimizing the affinity for a specific antigen target and enhancing the in vivo stability of these constructs will become a top priority to advance the applied use of these molecules. Such optimizations will require both molecular engineering and subsequent evaluation of the biological response to these changes. Workflows in therapeutic antibody and Fc-fusion proteins typically involves screening several, and in some cases hundreds, potential constructs through various technologies to identify top performing candidates. Poor or inconsistent expression and long lead times of candidate constructs can slow progress by necessitating scale up to generate enough protein for evaluation studies, oftentimes only to potentially be discontinued due to suboptimal results. Thus, this screening process to identify optimal constructs can be both extremely time consuming and heavily resource dependent.

Using the complete gene-to-protein workflow, Thermo Fisher Scientific can facilitate novel protein and antibody development from early discovery to preclinical phase. For GeneArt expression and purification services, Thermo Fisher Scientific leverages transient, high-yield protein expression systems and gene optimization to cut development timelines, saving valuable resources. Saving time in the discovery process and accelerating the identification of lead candidates can pay off with downstream activities such as additional analytical studies, moving towards final candidate selection faster.

At the heart of this platform is the GeneOptimizer algorithm, which maximizes RNA and protein expression of each construct. The algorithm optimizes candidate construct sequences against 20 different parameters related to transcription, splicing, translation, and mRNA degradation to improve efficiency of expression. These optimized constructs are then paired with the high performance Expi expression systems, Expi293 and ExpiCHO, to maximize candidate construct protein yields. Thermo Fisher Scientific provides customers with the ultimate flexibility, offering from 2.5 mL culture volumes for discovery and optimization, up to 100 L scale for early preclinical trials. This proven platform ensures reliable and rapid turnaround times, with delivery of the first protein candidates in as few as 21 days.

For an in-depth customer example, watch our webinar on Bifunctional Fc-based fusion protein therapies

Facilitating the discovery and development process of novel protein constructs

The understanding of antibodies and antibody engineering has progressed rapidly over the past several years. As a result, more sophisticated and elegantly engineered therapeutic proteins, such as bispecific, multispecific, and novel Fc-fusion proteins, are emerging. Combined with an ever-growing knowledge of the biological etiology of specific diseases, discovery and development of novel antibody-based therapeutics has accelerated to an unprecedented pace. New ideas and novel constructs require rapid experimentation to quickly identify and isolate optimal therapeutic candidates for subsequent scale up and further evaluation.

GeneArt protein expression and purification services facilitate novel antibody and antibody-based protein discovery through construct gene optimization and subsequent transient expression services that provide maximized yields of recombinant proteins at a variety of scales, all with abbreviated timelines. These services enable rapid evaluation of candidate constructs while saving time and resources by avoiding costly scale up efforts to generate sufficient material for characterization studies. When you partner with Thermo Fisher Scientific, you access years of gene optimization and proven protein expression experience that can help progress your protein therapeutic pipeline.


References
  1. Suurs F V., Lub-de Hooge MN, de Vries EGE, de Groot DJA. A review of bispecific antibodies and antibody constructs in oncology and clinical challenges. Vol. 201, Pharmacology and Therapeutics. Elsevier Inc.; 2019. p. 103–19. DOI: 10.1016/j.pharmthera.2019.04.006
  2. Lyu X, Zhao Q, Hui J, Wang T, Lin M, Wang K, et al. The global landscape of approved antibody therapies. Antib Ther. 2022 Oct 1;5(4):233–57. DOI: 10.1093/abt/tbac021
  3. Labrijn AF, Janmaat ML, Reichert JM, Parren PWHI. Bispecific antibodies: a mechanistic review of the pipeline. Vol. 18, Nature Reviews Drug Discovery. Nature Publishing Group; 2019. p. 585–608. DOI: 10.1038/s41573-019-0028-1
  4. Jin S, Sun Y, Liang X, Gu X, Ning J, Xu Y, et al. Emerging new therapeutic antibody derivatives for cancer treatment. Vol. 7, Signal Transduction and Targeted Therapy. Springer Nature; 2022. DOI: 10.1038/s41392-021-00868-x
  5. Shim H. Bispecific antibodies and antibody–drug conjugates for cancer therapy: Technological considerations. Vol. 10, Biomolecules. MDPI AG; 2020. DOI: 10.3390/biom10030360
  6. Elshiaty M, Schindler H, Christopoulos P. Principles and current clinical landscape of multispecific antibodies against cancer. Vol. 22, International Journal of Molecular Sciences. MDPI; 2021. DOI:10.3390/ijms22115632
  7. Vallera DA, Felices M, McElmurry R, McCullar V, Zhou X, Schmohl JU, et al. IL15 Trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo Expansion, and Enhanced Function. Clinical Cancer Research. 2016 Jul 15;22(14):3440–50. DOI: 10.1158/1078-0432.CCR-15-2710
  8. Rath T, Baker K, Dumont JA, Peters RT, Jiang H, Qiao SW, et al. Fc-fusion proteins and FcRn: Structural insights for longer-lasting and more effective therapeutics. Vol. 35, Critical Reviews in Biotechnology. Informa Healthcare; 2015. p. 235–54. DOI: 10.3109/07388551.2013.834293
  9. Dimitrov DS. Therapeutic proteins. Methods in Molecular Biology. 2012;899:1–26. DOI: 10.1007/978-1-61779-921-1_1