1. Introduction
Progress in biopharmaceutical technology has facilitated the creation of antibody-drug conjugates (ADCs), a groundbreaking category of anticancer therapies, heralding a new epoch in cancer treatment with significant accomplishments [1]. An ADC comprises an antibody, a potent cytotoxic drug molecule (payload), and a linker that connects the two components. ADCs function as a hybrid of targeted and chemotherapeutic agents by precisely identifying the target antigen through the antibody and delivering the cytotoxic payload to eradicate the target cells. The diminutive molecules of specific antibody-drug conjugates (ADCs) can infiltrate the cell membrane, consequently inducing apoptosis in adjacent tumor cells, a phenomenon referred to as the bystander effect. Moreover, specific antibody-drug conjugates (ADCs) exhibit antibody-immunizing effects, including complement-dependent cytotoxicity (CDC), antibody-dependent cytotoxicity (ADCC), and antibody-dependent cell phagocytosis (ADCP) [2].
Antibody-drug conjugates (ADCs) are frequently administered with the following chemotherapeutic agents. Conventional antibodies function by obstructing specific signaling pathways or physiological mechanisms that facilitate tumor proliferation. In the domain of antibody-drug conjugates (ADCs), the binding of the antibody to the target antigen on malignant cells enables the cytotoxic payload to exert its anti-tumor effect by either directly inhibiting tumor cell mitosis or disrupting the DNA structure. Consequently, the theoretical lethality efficacy of antibody-drug conjugates surpasses that of conventional antibodies. Furthermore, owing to their target selectivity, ADCs exhibit reduced systemic toxicity compared to traditional chemotherapeutic agents, thereby offering an improved safety profile [3]. Secondly, for patients exhibiting resistance to current targeted therapies, antibody-drug conjugates (ADCs) that target the same receptors may remain effective, thereby broadening their application in cancer treatment [4]. The human epidermal growth factor receptor 2 (HER2) is a significant target in breast carcinoma. Patients with metastatic breast cancer resistant to anti-HER2 therapy may still experience prolonged benefits from the anti-HER2 antibody-drug conjugate Trastuzumab Deruxtecan (Enhertu, DS-8201). Enhertu demonstrated efficacy in breast cancer patients exhibiting low HER2 expression via the bystander effect, thereby redefining the classification of HER2-positive breast cancer. Antibody-drug conjugates (ADCs) can precisely target novel therapeutic sites [5,6].
The concept of drug coupling originated from Paul Ehrlich's "magic bullet" theory in 1913, requiring almost 90 years of enhancement, formulation, and technological advancement prior to the launch of the first antibody-drug conjugate, Gemtuzumab Ozogamicin (Mylotarg), for clinical use [7]. At present, 15 antibody-drug conjugates (ADCs) have received approval, and over 200 have undergone evaluation in clinical trials [8]. Antibody-drug conjugates (ADCs) have emerged as one of the most rapidly expanding categories of anticancer treatments in the last ten years. The advent of ADCs represents a significant advancement, yet it introduces new challenges related to the design and integration of their various components for optimal performance. The in vivo effects and metabolic processes indicate that variations in target selection, drug structure, recognition site, and linker type can result in substantial differences in pharmacological efficacy, toxicity, pharmacokinetics, and controlled release [9]. These characteristics present significant challenges for chemical manufacturing and control (CMC), pre-clinical studies, and subsequent clinical trials. Consequently, antibody design, linker technology, payload and ligation techniques, have become pivotal challenges in the development of ADCs [10].
2. Selection of Antibodies in ADC Drugs
ADC drug antibodies need to have high antigen affinity and a long circulating half-life, which can be specifically enriched at the tumor site. The types of antibodies used to develop ADC drugs are mainly IgG sub-types, which gradually shift from early lgG4 to lgG1 [11]. At present, the selection of antibodies is mainly humanized antibodies, which have low immunogenicity, high specificity and high affinity. According to the half-life, conjugation convenience, structural stability, and immune function of Fc fragments for different IgG isoforms, the IgG1 isoform was mostly selected for antibodies [12].
2.1. Characteristics of Antibodies
The optimal antibody for application in an ADC must exhibit a robust affinity for target binding. It possesses good stability, minimal immunogenicity, effective internalization, and an extended plasma half-life [13]. The high specificity of antibody molecules is crucial for the success of antibody-drug conjugates, with IgG1 being the predominant subtype utilized. Upon selecting the target antigen, it is essential for the antibody to bind selectively to the antigen for accurate guiding; thus, the antibody's high targeting capability and potential immunogenicity must be evaluated [14].
2.1.1. Low immunogenicity
According to the immunogenicity of antibodies, they can be divided into fully humanized antibodies, humanized antibodies, and chimeric antibodies, and more humanized or fully humanized antibodies can be selected to reduce immune response [15]. Immunogenicity is primarily related to the source of the antibody. At the beginning of the development of ADC drugs, murine antibodies were still mainly used, and the strong immunogenicity led to greater side effects and a high failure rate of research and development. With the technological progress of protein engineering and genetic engineering, antibodies are gradually developing in the direction of humanization, and the immunogenicity continues to decrease. At present, it has been able to achieve full humanization, which has improved the efficiency of ADC drug research and development. For the marketed ADCs, only Adcetris uses chimeric antibodies, while other marketed drugs and most ADCs currently in development use humanized or human monoclonal antibodies [16]. There are 4 main categories of therapeutic antibodies based on their source.
The first generation of ADCs
Rodents: problems with immunogenicity, short half-life, and limited penetration at tumor sites
Chimerism: Reduced immunogenicity and extended half-life compared to murine monoclonal antibodies
Second-generation ADCs
Humanized: Reduced immunogenicity compared to chimeric monoclonal antibodies
Third-generation ADCs
Fully humanized: Significantly reduced immunogenicity compared to humanized monoclonal antibodies
2.1.2. Long half-life
The Fc fragment of the antibody can be enhanced or attenuated to modulate the intensity of the immune response mediated, which can also affect its stability and half-life [17]. ADC drugs predominantly utilize the IgG1 molecule within the humanized monoclonal antibody subtype, which offers a high affinity for the target antigen, an extended half-life in circulation, and numerous natural conjugation sites, comprising 60-70% of plasma, significantly surpassing other subtypes [18]. Advancements in technology have facilitated the utilization of IgG4 in the research and development of ADC medicines; nonetheless, IgG4 is less prevalent in plasma, exhibits reduced structural stability compared to IgG1, and is prone to aggregation at low pH levels. Given that various isoforms of IgG has distinct functions, future research and development may concentrate on selecting different isoforms of heavy and light chains for recombination to produce novel monoclonal antibodies. Konitzer et al. reconstructed the IgG2 or IgG4 heavy chain in place of the IgG1 heavy chain, resulting in a more stable profile for the combined antibody [19].
2.1.3. High affinity
The effectiveness of antibody internalization is mostly associated with the antibody's affinity for the antigen. Increased affinity of antibodies utilized in the ADC enhances internalization efficiency; however, antibodies with excessively high antigen affinity diminish the penetration efficiency of solid tumors, thereby impacting the ADC drug's ability to reach the tumor interior [20]. Consequently, antigens must be identified and attached by antibodies with an adequate affinity (Kd≤10 nM) to guarantee fast uptake in target cells [21]. Take into account the molecular weight of the chosen antibody; if the molecular weight is excessively high, it may struggle to traverse the capillary endothelium layer and extracellular space, whereas a molecular weight that is too low could compromise its in vivo half-life. The primary focus of ADC antibody optimization is to enhance endocytosis and osmotic efficacy. The primary hurdles for antibody-drug conjugates (ADCs) are insufficient efficacy and off-target toxicity, largely due to the low number of ADCs that reach the tumor and the restricted rate of internalization [22].
2.2. Target Selection
Appropriate targets markedly affect the pharmacological properties of ADCs. The target antigens of approved antibody-drug conjugates (ADCs) are specific proteins that are overexpressed in common tumor cells, such as HER2, TROP2, and Nectin4 in solid tumors, and CD19, CD22, CD33, and CD30 in hematological malignancies.[23] Recently, the progress of new ADC targets has been robust, and the techniques for selecting target antigens are consistently being improved. A promising strategy entails the identification of mutant proteins in tumors, which generally display increased levels of ubiquitination modifications and are more easily internalized and degraded than wild-type proteins [24]. Targeting antibody-drug conjugates (ADCs) containing oncogenic mutant proteins can improve therapeutic specificity. Recent advancements in fundamental tumor immunology research have expanded the target development of antibody-drug conjugates (ADCs) from traditional tumor antigens to encompass microenvironmental and cancer stem cell antigens[25]. An increasing number of ADCs based on this principle may enter preclinical and clinical trials in the future. Furthermore, the recognition of these new targets will greatly expand the applications for future ADC therapeutics [26].
2.3. Future Development of Antibodies in ADCs
Numerous technology platforms focus on enhancing ADC antibodies, with prospective advancements involving the selection of antibody serotypes, augmentation of antibody humanization, and modification and optimization of antibody structures. Recent proposals for antibody-drug conjugate (ADC) design include conditional antibody ADCs, bispecific ADCs, non-internalized ADCs, and the substitution of antibody vectors with alternative molecular types, which will shape the future trajectory of ADC development [27]. Researchers are presently devising methods to enhance delivery efficiency and internalization speed, including improving penetration and using ADC medicines with bispecific antibodies [28].
2.3.1. Use Bispecific antibodies to enhance affinity and specificity
Bispecific antibodies can concurrently bind to two antigens on a single target cell, a characteristic that may enhance the therapeutic window while minimizing off-target effects on non-target cells [29]. Moreover, employing bispecific antibodies that target two distinct epitope antigens can augment the antibody's affinity for the antigen, thereby enhancing the endocytosis efficiency of the ADC medication [30].
2.3.2. Remove the Fc end to improve permeability
Due to their substantial size, ADC therapeutics must surmount the stromal barrier and penetrate tumor tissue when addressing solid tumors. The current technique primarily aims to reduce the molecular weight of the antibody, as it constitutes a substantial portion of the structure [31]. For instance, scFv (single-chain variable fragment) or sdAb (single-domain antibody) serve as foundational components for antibody production; due to their low molecular weight, the resultant antibodies are smaller than IgG and can penetrate tumors more rapidly [32].
2.3.3. Bystander effect to resolve binding site barriers
A contributing factor to inadequate tumor permeability may be the binding site barrier effect [33]. This phenomenon occurs when antibodies with high affinity for target antigens adjacent to blood arteries exhibit diminished distribution away from these vessels due to the quick and strong binding of antigens, leading to decreased tumor clearance efficacy of antibody-drug conjugates (ADCs). Small-sized ADCs that enhance permeability and dispersion can utilize the "bystander effect" to eliminate neighboring tumor cells and overcome site barriers [34].
3. Current Status of Antibody Development for ADC Drugs
In 1975, Kohler and Milstein developed hybridoma technique and produced the first generation of monoclonal antibodies, namely murine monoclonal antibodies, marking the commencement of a new era in monoclonal antibody research [35]. In clinical treatment, murine monoclonal antibodies exhibit significant immunogenicity, resulting in the production of human anti-mouse antibody (HAMA) reactions. With advancements in antibody preparation technology, the development of monoclonal antibodies has evolved from murine antibodies to human-mouse chimeric antibodies, humanized antibodies, and ultimately fully human antibodies, thereby substantially reducing immunogenicity and enhancing the efficacy of monoclonal antibodies [36]. The immunogenicity of antibodies has significantly diminished, while the safety and efficacy of monoclonal antibodies have been enhanced. In recent years, the swift advancement of humanized and completely human antibodies, characterized by low rejection rates and extended blood half-lives, has emerged as a predominant trend in antibody drug development, comprising over 80% of newly authorized antibody therapeutics by the FDA [37,38]. Currently, the majority of global ADC medicines are created with these antibodies.Monoclonal antibodies utilized as ADC carriers are enumerated below based on the target molecules they identify:
3.1. Antibody drugs targeting HER2
3.1.1. Trastuzumab
Trastuzumab (Herceptin) is an antibody therapy that specifically targets HER2. Hereeptin is a recombinant humanized IgG1 monoclonal antibody targeting HER2. Trastuzumab (Herceptin) is a recombinant humanized anti-HER2 IgG1 monoclonal antibody that received FDA approval in 1998 for the treatment of metastatic breast cancer [39]. In 1998, the FDA sanctioned Hereeptin for the treatment of metastatic breast cancer, establishing it as the inaugural antibody drug approved for oncological therapy. In 2006 and 2010, the FDA sanctioned patulin for the management of postoperative early breast cancer, metastatic gastric cancer, and esophagogastric junction cancer, respectively [40].
3.1.2. Pertuzumab
Pertuzumab (Peb) is a fully recombinant humanized anti-HER2 IgG1 monoclonal antibody. It attaches to domain II of the extracellular region of the HER2 receptor, inhibiting HER2 homo- or heterodimerization. Formation of homo- or heterodimers and the inhibition of downstream signaling pathways [41]. In June 2012, the FDA sanctioned pertuzumab for the treatment of HER2-positive advanced or metastatic breast cancer [42].
3.2. Antibody Drugs Targeting EGFR
3.2.1. Cetuximab
Cetuximab (Erbitux) is a chimeric IgG1 monoclonal antibody that antagonizes endogenous ligands by binding to the extracellular domains of the epidermal growth factor receptor (EGFR). competes with endogenous ligands for binding to the extracellular domain of the EGFR. Cetuximab obtained FDA marketing authorization in 2004 [43]. FDA-approved for the treatment of metastatic colorectal cancer and head and neck squamous cell carcinoma [44].
3.3. Antibody drugs targeting CD20
3.3.1. Rituximab
Rituximab (Rituxan) is a chimeric monoclonal antibody, integrating human and murine components, that specifically targets CD20. In 1997, it obtained U.S. FDA approval for the treatment of B-cell non-Hodgkin's lymphoma, establishing it as the inaugural monoclonal antibody authorized for clinical application by the FDA [45]. In 2010, the European Commission approved it for the maintenance treatment of follicular lymphoma after initial therapy. Moreover, it has been widely employed in the treatment of systemic lupus erythematosus, idiopathic thrombocytopenic purpura, chronic lymphocytic leukemia, multiple myeloma, and autoimmune pancreatitis. [46].
3.3.2. Teimumab
Tibritumomab (ibritumomab, Zevalin) is the first radio-labeled monoclonal antibody in the world. It is a radioisotope-labeled anti-CD20 IgG1 monoclonal antibody, approved for use in the United States in 2002 for the treatment of refractory relapsed B-cell non-Hodgkin's lymphoma. [47].
4. Conclusion and Prospect
Antibody-drug conjugates (ADCs) combine the high specificity of antibodies with the powerful tumor-killing properties of cytotoxic agents, becoming a central focus in anti-tumor antibody research and development. Antibodies, functioning as the carriers for ADC therapeutics, are essential factors in the development of effective ADCs. The development of monoclonal antibodies has advanced through several stages: murine, human-mouse chimeric, humanized, and fully human antibodies. The immunogenicity of monoclonal antibodies has progressively diminished, while their blood half-life has steadily grown, establishing a basis for the advancement of optimal ADC therapeutics. In the advancement of next-generation antibody-drug conjugates, it is essential to evaluate and enhance the target, affinity, immunogenicity, relative molecular mass, and efficacy of the antibody. Furthermore, there is a continuous need to identify novel effective antibody targets and to engineer more potent, smaller relative molecular mass, and more permeable antibodies or antibody fragments for the development of optimized ADC therapeutics.
References
[1]. Parit S, Manchare A, Gholap A D, et al. Antibody-drug conjugates: a promising breakthrough in cancer therapy[J]. International Journal of Pharmaceutics, 2024, 659: 124211.
[2]. Casi G, Neri D. Antibody–drug conjugates and small molecule–drug conjugates: opportunities and challenges for the development of selective anticancer cytotoxic agents: miniperspective[J]. Journal of medicinal chemistry, 2015, 58(22): 8751-8761.
[3]. Ponziani S, Di Vittorio G, Pitari G, et al. Antibody-drug conjugates: the new frontier of chemotherapy[J]. International Journal of Molecular Sciences, 2020, 21(15): 5510.
[4]. Shefet-Carasso L R, Benhar I. Antibody-targeted drugs and drug resistance—challenges and solutions[J]. Drug Resistance Updates, 2015, 18: 36-46.
[5]. Najminejad Z, Dehghani F, Mirzaei Y, et al. Clinical perspective: antibody-drug conjugates for the treatment of HER2-positive breast cancer[J]. Molecular Therapy, 2023, 31(7): 1874-1903.
[6]. Nie T, Blair H A. Trastuzumab deruxtecan: a review in unresectable or metastatic HER2-positive breast cancer[J]. Targeted Oncology, 2023, 18(3): 463-470.
[7]. Sun C, Yang X, Tang L, et al. A pharmacovigilance study on drug-induced liver injury associated with antibody-drug conjugates (ADCs) based on the food and drug administration adverse event reporting system[J]. Expert Opinion on Drug Safety, 2024, 23(8): 1049-1060.
[8]. Liu K, Li M, Li Y, et al. A review of the clinical efficacy of FDA-approved antibody‒drug conjugates in human cancers[J]. Molecular Cancer, 2024, 23(1): 62.
[9]. Muro S. Challenges in design and characterization of ligand-targeted drug delivery systems[J]. Journal of Controlled Release, 2012, 164(2): 125-137.
[10]. Leung D, Wurst J M, Liu T, et al. Antibody conjugates-recent advances and future innovations[J]. Antibodies, 2020, 9(1): 2.
[11]. Biological drug products: development and strategies[J]. 2013.
[12]. Husain B, Ellerman D. Expanding the boundaries of biotherapeutics with bispecific antibodies[J]. BioDrugs, 2018, 32(5): 441-464.
[13]. Leung D, Wurst J M, Liu T, et al. Antibody conjugates-recent advances and future innovations[J]. Antibodies, 2020, 9(1): 2.
[14]. Ackaert C, Smiejkowska N, Xavier C, et al. Immunogenicity risk profile of nanobodies[J]. Frontiers in immunology, 2021, 12: 632687.
[15]. Rossotti M A, Bélanger K, Henry K A, et al. Immunogenicity and humanization of single‐domain antibodies[J]. The FEBS Journal, 2022, 289(14): 4304-4327.
[16]. Hasan M M, Laws M, Jin P, et al. Factors influencing the choice of monoclonal antibodies for antibody–drug conjugates[J]. Drug Discovery Today, 2022, 27(1): 354-361.
[17]. Jazayeri J A, Carroll G J. Fc-based cytokines: prospects for engineering superior therapeutics[J]. BioDrugs, 2008, 22: 11-26.
[18]. Damelin M, Zhong W, Myers J, et al. Evolving strategies for target selection for antibody-drug conjugates[J]. Pharmaceutical research, 2015, 32: 3494-3507.
[19]. Könitzer J D, Sieron A, Wacker A, et al. Reformatting rituximab into human IgG2 and IgG4 isotypes dramatically improves apoptosis induction in vitro[J]. PLoS One, 2015, 10(12): e0145633.
[20]. Kim E G, Kim K M. Strategies and advancement in antibody-drug conjugate optimization for targeted cancer therapeutics[J]. Biomolecules & therapeutics, 2015, 23(6): 493.
[21]. Leelawattanachai J, Kwon K W, Michael P, et al. Side-by-side comparison of commonly used biomolecules that differ in size and affinity on tumor uptake and internalization[J]. PloS one, 2015, 10(4): e0124440.
[22]. Kim E G, Kim K M. Strategies and advancement in antibody-drug conjugate optimization for targeted cancer therapeutics[J]. Biomolecules & therapeutics, 2015, 23(6): 493.
[23]. Blakeley J O N, Plotkin S R, Gilbert M R, et al. Rare Tumors of the Central Nervous System: More Similar than Different[J]. American Society of Clinical Oncology Educational Book.
[24]. Dale B, Cheng M, Park K S, et al. Advancing targeted protein degradation for cancer therapy[J]. Nature Reviews Cancer, 2021, 21(10): 638-654.
[25]. Aggarwal D, Yang J, Salam M A, et al. Antibody-drug conjugates: the paradigm shifts in the targeted cancer therapy[J]. Frontiers in immunology, 2023, 14: 1203073.
[26]. Kuhlmann L, Cummins E, Samudio I, et al. Cell-surface proteomics for the identification of novel therapeutic targets in cancer[J]. Expert review of proteomics, 2018, 15(3): 259-275.
[27]. De Cecco M, Galbraith D N, McDermott L L. What makes a good antibody–drug conjugate?[J]. Expert Opinion on Biological Therapy, 2021, 21(7): 841-847.
[28]. Gu Y, Wang Z, Wang Y. Bispecific antibody drug conjugates: Making 1+ 1> 2[J]. Acta Pharmaceutica Sinica B, 2024.
[29]. Cao Y, Lam L. Bispecific antibody conjugates in therapeutics[J]. Advanced drug delivery reviews, 2003, 55(2): 171-197.
[30]. Lim S I. Fine-tuning bispecific therapeutics[J]. Pharmacology & Therapeutics, 2020, 212: 107582.
[31]. Chiu M L, Goulet D R, Teplyakov A, et al. Antibody structure and function: the basis for engineering therapeutics[J]. Antibodies, 2019, 8(4): 55.
[32]. Pauling L. A theory of the structure and process of formation of antibodies[J]. Journal of the American Chemical Society, 1940, 62(10): 2643-2657.
[33]. Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment[J]. Molecular Cancer Research, 2006, 4(2): 61-70.
[34]. Kim S M, Faix P H, Schnitzer J E. Overcoming key biological barriers to cancer drug delivery and efficacy[J]. Journal of Controlled Release, 2017, 267: 15-30.
[35]. Pandey S. Hybridoma technology for production of monoclonal antibodies[J]. Hybridoma, 2010, 1(2): 017.
[36]. Gao S H, Huang K, Tu H, et al. Monoclonal antibody humanness score and its applications[J]. BMC biotechnology, 2013, 13: 1-12.
[37]. Weiner L M. Fully human therapeutic monoclonal antibodies[J]. Journal of immunotherapy, 2006, 29(1): 1-9.
[38]. Roskos L K, Davis C G, Schwab G M. The clinical pharmacology of therapeutic monoclonal antibodies[J]. Drug Development Research, 2004, 61(3): 108-120.
[39]. McKeage K, Perry C M. Trastuzumab: a review of its use in the treatment of metastatic breast cancer overexpressing HER2[J]. Drugs, 2002, 62(1): 209-243.
[40]. Emens L A. Trastuzumab: targeted therapy for the management of HER-2/neu-overexpressing metastatic breast cancer[J]. American journal of therapeutics, 2005, 12(3): 243-253.
[41]. Cao L, Li Q, Tong Z, et al. HER2-specific immunotoxins constructed based on single-domain antibodies and the improved toxin PE24X7[J]. International Journal of Pharmaceutics, 2020, 574: 118939.
[42]. Lamond N W D, Younis T. Pertuzumab in human epidermal growth-factor receptor 2-positive breast cancer: clinical and economic considerations[J]. International journal of women's health, 2014: 509-521.
[43]. Galizia G, Lieto E, De Vita F, et al. Cetuximab, a chimeric human mouse anti-epidermal growth factor receptor monoclonal antibody, in the treatment of human colorectal cancer[J]. Oncogene, 2007, 26(25): 3654-3660.
[44]. da Silva Santos E, Nogueira K A B, Fernandes L C C, et al. EGFR targeting for cancer therapy: Pharmacology and immunoconjugates with drugs and nanoparticles[J]. International journal of pharmaceutics, 2021, 592: 120082.
[45]. Boye J, Elter T, Engert A. An overview of the current clinical use of the anti-CD20 monoclonal antibody rituximab[J]. Annals of Oncology, 2003, 14(4): 520-535.
[46]. Lim S H, Beers S A, French R R, et al. Anti-CD20 monoclonal antibodies: historical and future perspectives[J]. haematologica, 2010, 95(1): 135.
[47]. Fietz T, Thiel E. Antibody therapy in non-Hodgkin’s lymphoma: the role of rituximab, 90Y-ibritumomab tiuxetan, and alemtuzumab[J]. Targeted Therapies in Cancer, 2007: 153-163.
Cite this article
Liang,Z. (2025). The Development Process and Characteristics of Antibodies in Antibody-drug Conjugate. Theoretical and Natural Science,76,12-19.
Data availability
The datasets used and/or analyzed during the current study will be available from the authors upon reasonable request.
Disclaimer/Publisher's Note
The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of EWA Publishing and/or the editor(s). EWA Publishing and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
About volume
Volume title: Proceedings of the 4th International Conference on Biological Engineering and Medical Science
© 2024 by the author(s). Licensee EWA Publishing, Oxford, UK. This article is an open access article distributed under the terms and
conditions of the Creative Commons Attribution (CC BY) license. Authors who
publish this series agree to the following terms:
1. Authors retain copyright and grant the series right of first publication with the work simultaneously licensed under a Creative Commons
Attribution License that allows others to share the work with an acknowledgment of the work's authorship and initial publication in this
series.
2. Authors are able to enter into separate, additional contractual arrangements for the non-exclusive distribution of the series's published
version of the work (e.g., post it to an institutional repository or publish it in a book), with an acknowledgment of its initial
publication in this series.
3. Authors are permitted and encouraged to post their work online (e.g., in institutional repositories or on their website) prior to and
during the submission process, as it can lead to productive exchanges, as well as earlier and greater citation of published work (See
Open access policy for details).
References
[1]. Parit S, Manchare A, Gholap A D, et al. Antibody-drug conjugates: a promising breakthrough in cancer therapy[J]. International Journal of Pharmaceutics, 2024, 659: 124211.
[2]. Casi G, Neri D. Antibody–drug conjugates and small molecule–drug conjugates: opportunities and challenges for the development of selective anticancer cytotoxic agents: miniperspective[J]. Journal of medicinal chemistry, 2015, 58(22): 8751-8761.
[3]. Ponziani S, Di Vittorio G, Pitari G, et al. Antibody-drug conjugates: the new frontier of chemotherapy[J]. International Journal of Molecular Sciences, 2020, 21(15): 5510.
[4]. Shefet-Carasso L R, Benhar I. Antibody-targeted drugs and drug resistance—challenges and solutions[J]. Drug Resistance Updates, 2015, 18: 36-46.
[5]. Najminejad Z, Dehghani F, Mirzaei Y, et al. Clinical perspective: antibody-drug conjugates for the treatment of HER2-positive breast cancer[J]. Molecular Therapy, 2023, 31(7): 1874-1903.
[6]. Nie T, Blair H A. Trastuzumab deruxtecan: a review in unresectable or metastatic HER2-positive breast cancer[J]. Targeted Oncology, 2023, 18(3): 463-470.
[7]. Sun C, Yang X, Tang L, et al. A pharmacovigilance study on drug-induced liver injury associated with antibody-drug conjugates (ADCs) based on the food and drug administration adverse event reporting system[J]. Expert Opinion on Drug Safety, 2024, 23(8): 1049-1060.
[8]. Liu K, Li M, Li Y, et al. A review of the clinical efficacy of FDA-approved antibody‒drug conjugates in human cancers[J]. Molecular Cancer, 2024, 23(1): 62.
[9]. Muro S. Challenges in design and characterization of ligand-targeted drug delivery systems[J]. Journal of Controlled Release, 2012, 164(2): 125-137.
[10]. Leung D, Wurst J M, Liu T, et al. Antibody conjugates-recent advances and future innovations[J]. Antibodies, 2020, 9(1): 2.
[11]. Biological drug products: development and strategies[J]. 2013.
[12]. Husain B, Ellerman D. Expanding the boundaries of biotherapeutics with bispecific antibodies[J]. BioDrugs, 2018, 32(5): 441-464.
[13]. Leung D, Wurst J M, Liu T, et al. Antibody conjugates-recent advances and future innovations[J]. Antibodies, 2020, 9(1): 2.
[14]. Ackaert C, Smiejkowska N, Xavier C, et al. Immunogenicity risk profile of nanobodies[J]. Frontiers in immunology, 2021, 12: 632687.
[15]. Rossotti M A, Bélanger K, Henry K A, et al. Immunogenicity and humanization of single‐domain antibodies[J]. The FEBS Journal, 2022, 289(14): 4304-4327.
[16]. Hasan M M, Laws M, Jin P, et al. Factors influencing the choice of monoclonal antibodies for antibody–drug conjugates[J]. Drug Discovery Today, 2022, 27(1): 354-361.
[17]. Jazayeri J A, Carroll G J. Fc-based cytokines: prospects for engineering superior therapeutics[J]. BioDrugs, 2008, 22: 11-26.
[18]. Damelin M, Zhong W, Myers J, et al. Evolving strategies for target selection for antibody-drug conjugates[J]. Pharmaceutical research, 2015, 32: 3494-3507.
[19]. Könitzer J D, Sieron A, Wacker A, et al. Reformatting rituximab into human IgG2 and IgG4 isotypes dramatically improves apoptosis induction in vitro[J]. PLoS One, 2015, 10(12): e0145633.
[20]. Kim E G, Kim K M. Strategies and advancement in antibody-drug conjugate optimization for targeted cancer therapeutics[J]. Biomolecules & therapeutics, 2015, 23(6): 493.
[21]. Leelawattanachai J, Kwon K W, Michael P, et al. Side-by-side comparison of commonly used biomolecules that differ in size and affinity on tumor uptake and internalization[J]. PloS one, 2015, 10(4): e0124440.
[22]. Kim E G, Kim K M. Strategies and advancement in antibody-drug conjugate optimization for targeted cancer therapeutics[J]. Biomolecules & therapeutics, 2015, 23(6): 493.
[23]. Blakeley J O N, Plotkin S R, Gilbert M R, et al. Rare Tumors of the Central Nervous System: More Similar than Different[J]. American Society of Clinical Oncology Educational Book.
[24]. Dale B, Cheng M, Park K S, et al. Advancing targeted protein degradation for cancer therapy[J]. Nature Reviews Cancer, 2021, 21(10): 638-654.
[25]. Aggarwal D, Yang J, Salam M A, et al. Antibody-drug conjugates: the paradigm shifts in the targeted cancer therapy[J]. Frontiers in immunology, 2023, 14: 1203073.
[26]. Kuhlmann L, Cummins E, Samudio I, et al. Cell-surface proteomics for the identification of novel therapeutic targets in cancer[J]. Expert review of proteomics, 2018, 15(3): 259-275.
[27]. De Cecco M, Galbraith D N, McDermott L L. What makes a good antibody–drug conjugate?[J]. Expert Opinion on Biological Therapy, 2021, 21(7): 841-847.
[28]. Gu Y, Wang Z, Wang Y. Bispecific antibody drug conjugates: Making 1+ 1> 2[J]. Acta Pharmaceutica Sinica B, 2024.
[29]. Cao Y, Lam L. Bispecific antibody conjugates in therapeutics[J]. Advanced drug delivery reviews, 2003, 55(2): 171-197.
[30]. Lim S I. Fine-tuning bispecific therapeutics[J]. Pharmacology & Therapeutics, 2020, 212: 107582.
[31]. Chiu M L, Goulet D R, Teplyakov A, et al. Antibody structure and function: the basis for engineering therapeutics[J]. Antibodies, 2019, 8(4): 55.
[32]. Pauling L. A theory of the structure and process of formation of antibodies[J]. Journal of the American Chemical Society, 1940, 62(10): 2643-2657.
[33]. Cairns R, Papandreou I, Denko N. Overcoming physiologic barriers to cancer treatment by molecularly targeting the tumor microenvironment[J]. Molecular Cancer Research, 2006, 4(2): 61-70.
[34]. Kim S M, Faix P H, Schnitzer J E. Overcoming key biological barriers to cancer drug delivery and efficacy[J]. Journal of Controlled Release, 2017, 267: 15-30.
[35]. Pandey S. Hybridoma technology for production of monoclonal antibodies[J]. Hybridoma, 2010, 1(2): 017.
[36]. Gao S H, Huang K, Tu H, et al. Monoclonal antibody humanness score and its applications[J]. BMC biotechnology, 2013, 13: 1-12.
[37]. Weiner L M. Fully human therapeutic monoclonal antibodies[J]. Journal of immunotherapy, 2006, 29(1): 1-9.
[38]. Roskos L K, Davis C G, Schwab G M. The clinical pharmacology of therapeutic monoclonal antibodies[J]. Drug Development Research, 2004, 61(3): 108-120.
[39]. McKeage K, Perry C M. Trastuzumab: a review of its use in the treatment of metastatic breast cancer overexpressing HER2[J]. Drugs, 2002, 62(1): 209-243.
[40]. Emens L A. Trastuzumab: targeted therapy for the management of HER-2/neu-overexpressing metastatic breast cancer[J]. American journal of therapeutics, 2005, 12(3): 243-253.
[41]. Cao L, Li Q, Tong Z, et al. HER2-specific immunotoxins constructed based on single-domain antibodies and the improved toxin PE24X7[J]. International Journal of Pharmaceutics, 2020, 574: 118939.
[42]. Lamond N W D, Younis T. Pertuzumab in human epidermal growth-factor receptor 2-positive breast cancer: clinical and economic considerations[J]. International journal of women's health, 2014: 509-521.
[43]. Galizia G, Lieto E, De Vita F, et al. Cetuximab, a chimeric human mouse anti-epidermal growth factor receptor monoclonal antibody, in the treatment of human colorectal cancer[J]. Oncogene, 2007, 26(25): 3654-3660.
[44]. da Silva Santos E, Nogueira K A B, Fernandes L C C, et al. EGFR targeting for cancer therapy: Pharmacology and immunoconjugates with drugs and nanoparticles[J]. International journal of pharmaceutics, 2021, 592: 120082.
[45]. Boye J, Elter T, Engert A. An overview of the current clinical use of the anti-CD20 monoclonal antibody rituximab[J]. Annals of Oncology, 2003, 14(4): 520-535.
[46]. Lim S H, Beers S A, French R R, et al. Anti-CD20 monoclonal antibodies: historical and future perspectives[J]. haematologica, 2010, 95(1): 135.
[47]. Fietz T, Thiel E. Antibody therapy in non-Hodgkin’s lymphoma: the role of rituximab, 90Y-ibritumomab tiuxetan, and alemtuzumab[J]. Targeted Therapies in Cancer, 2007: 153-163.