Opinion Article - Journal of Cancer Immunology & Therapy (2025) Volume 8, Issue 1
From Lab to Clinic: The Development and Production of Monoclonal Antibodies
Kanna Sridharan *
Department of Pharmacology & Therapeutics, Arabian Gulf University, Bahrain
- *Corresponding Author:
- Kanna Sridharan
Department of Pharmacology & Therapeutics, Arabian Gulf University, Bahrain
E-mail: sri.kann@gmail.com
Received: 03-Feb -2025, Manuscript No. AAJCIT-25-161375; Editor assigned: 04-Feb-2025, PreQC No. AAJCIT-25-161375 (PQ); Reviewed:18-Feb-2025, QC No. AAJCIT-25-161375; Revised:23-Feb-2025, Manuscript No. AAJCIT-25-161375 (R); Published:28-Feb-2025, DOI:10.35841/aajcit-8.1.249
Citation: Sridharan K. From lab to clinic: The development and production of monoclonal antibodies. J Cancer Immunol Ther. 2025;8(1):249
Abstract
Introduction
Monoclonal antibodies (mAbs) have revolutionized modern medicine, providing targeted treatments for various diseases, including cancer, autoimmune disorders, and infectious diseases. Their journey from the laboratory to clinical application involves rigorous research, development, and manufacturing processes to ensure their safety and efficacy. This article explores the stages of monoclonal antibody development, from discovery to production, and their impact on healthcare [1].
The development of monoclonal antibodies begins with antigen selection. Scientists identify specific antigens that play a crucial role in disease progression. These antigens, usually proteins or peptides, are chosen based on their potential as therapeutic targets. Once identified, the next step is to generate monoclonal antibodies that specifically bind to the target antigen [2].
Hybridoma technology, pioneered by Köhler and Milstein in 1975, remains one of the most widely used methods for producing monoclonal antibodies. In this method, B cells from an immunized animal, typically a mouse, are fused with myeloma cells to create hybridomas. These hybridomas are immortalized cell lines that produce large quantities of identical antibodies. Alternative methods, such as recombinant DNA technology and phage display, have since been developed to improve antibody specificity and humanization [3].
Before clinical trials, monoclonal antibodies undergo preclinical testing to assess their safety, efficacy, and pharmacokinetics. In vitro studies are conducted to evaluate their binding affinity, stability, and biological activity. Subsequently, in vivo animal models are used to determine their therapeutic potential and potential side effects. The data from preclinical studies guide the decision to advance the antibody candidate to clinical trials [4].
Clinical trials for monoclonal antibodies follow a phased approach: Involves a small group of healthy volunteers or patients to assess safety, dosage, and pharmacokinetics. Expands to a larger group of patients to evaluate efficacy and monitor side effects [5].
Involves a large-scale study to confirm effectiveness, compare with existing treatments, and identify adverse reactions. Once a monoclonal antibody successfully completes clinical trials, regulatory agencies such as the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA) review the data for approval [6].
The rigorous approval process ensures that only safe and effective therapies reach patients. The large-scale production of monoclonal antibodies requires advanced biotechnological processes. Mammalian cell lines, such as Chinese Hamster Ovary (CHO) cells, are widely used to express recombinant monoclonal antibodies due to their ability to perform post-translational modifications critical for antibody function [7].
The production process involves several key steps: Stable cell lines expressing the desired antibody are generated and optimized. Cell cultures are grown in bioreactors under controlled conditions to maximize antibody yield [8].
Purification steps, including protein A chromatography, ultrafiltration, and viral inactivation, are performed to ensure product purity. Analytical testing ensures batch consistency, potency, and absence of contaminants. Monoclonal antibodies have transformed treatment options across various medical fields. In oncology, drugs like trastuzumab (Herceptin) target HER2-positive breast cancer, significantly improving patient outcomes [9].
In autoimmune diseases, therapies like adalimumab (Humira) inhibit tumor necrosis factor (TNF) to manage conditions such as rheumatoid arthritis. Recent advancements in monoclonal antibody therapy include bispecific antibodies and antibody-drug conjugates (ADCs). Bispecific antibodies simultaneously bind two different antigens, enhancing therapeutic efficacy. ADCs link monoclonal antibodies to cytotoxic drugs, enabling targeted cancer cell destruction with minimal side effects [10].
Conclusion
The journey of monoclonal antibodies from lab to clinic is a testament to scientific innovation and medical advancement. With ongoing research and technological improvements, monoclonal antibodies will continue to shape the future of precision medicine, offering hope to patients with previously untreatable conditions.
References
- Kane MA. Preventing cancer with vaccines: progress in the global control of cancer. Cancer Prev Res. 2012;5(1):24-9.
- Massarweh A, Eliakim-Raz N, Stemmer A, et al., Evaluation of seropositivity following BNT162b2 messenger RNA vaccination for SARS-CoV-2 in patients undergoing treatment for cancer. JAMA Oncol. 2021;7(8):1133-40.
- Stanford MM, McFadden G. Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther. 2007;7(9):1415-25.
- Wen R, Umeano AC, Kou Y, et al., Nanoparticle systems for cancer vaccine. Nanomed. 2019;14(5):627-48.
- Mannino MH, Zhu Z, Xiao H, et al., The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 2015;367(2):103-7.
- Goshen-Lago T, Waldhorn I, Holland R, et al., Serologic status and toxic effects of the SARS-CoV-2 BNT162b2 vaccine in patients undergoing treatment for cancer. JAMA Oncol. 2021;7(10):1507-13.
- Zhou J, Yu G, Huang F. Supramolecular chemotherapy based on host–guest molecular recognition: a novel strategy in the battle against cancer with a bright future. Chem Soc Rev. 2017;46(22):7021-53.
- Stone EG, Morton SC, Hulscher ME, et al., Interventions that increase use of adult immunization and cancer screening services: a meta-analysis. Ann Intern Med. 2002;136(9):641-51.
- Klebanoff CA, Gattinoni L, Restifo NP. CD8+ T?cell memory in tumor immunology and immunotherapy. Immunol Rev. 2006;211(1):214-24.
- Overwijk WW, Lee DS, Surman DR, et al., Vaccination with a recombinant vaccinia virus encoding a “self” antigen induces autoimmune vitiligo and tumor cell destruction in mice: requirement for CD4+ T lymphocytes. Proc Natl Acad Sci. 1999;96(6):2982-7.
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref