Appearance
This report is written by MaltSci based on the latest literature and research findings
How do antibody-drug conjugates target cancer?
Abstract
Antibody-drug conjugates (ADCs) have emerged as a transformative class of targeted therapies in oncology, combining the specificity of monoclonal antibodies with potent cytotoxic agents to deliver drugs directly to cancer cells. This review discusses the mechanisms by which ADCs target cancer, focusing on their ability to selectively bind to tumor-associated antigens (TAAs), leading to internalization and release of cytotoxic payloads within the cancer cells. The review highlights the critical components of ADCs, including antibody selection, linker chemistry, and cytotoxic payloads, which are essential for optimizing therapeutic efficacy. The clinical landscape of ADCs is rapidly evolving, with several ADCs approved for various malignancies and many others in clinical trials. However, challenges such as tumor heterogeneity and resistance mechanisms must be addressed to enhance the effectiveness of ADCs. Future directions in ADC research include the development of next-generation ADCs with improved specificity and efficacy, exploring novel combinations with other therapeutic modalities, and addressing resistance mechanisms. Understanding these aspects is crucial for advancing ADCs as a cornerstone of precision oncology and improving treatment outcomes for cancer patients.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Action of Antibody-Drug Conjugates
- 2.1 Targeting Tumor-Associated Antigens
- 2.2 Internalization and Drug Release Mechanisms
- 3 Components of Antibody-Drug Conjugates
- 3.1 Antibody Selection and Engineering
- 3.2 Linkers: Types and Their Importance
- 3.3 Cytotoxic Payloads: Types and Mechanisms
- 4 Clinical Applications and Current Landscape
- 4.1 Approved ADCs and Their Indications
- 4.2 Emerging ADCs in Clinical Trials
- 5 Challenges and Future Directions
- 5.1 Resistance Mechanisms in Cancer Cells
- 5.2 Strategies for Enhancing ADC Efficacy
- 5.3 Future Perspectives in ADC Development
- 6 Summary
1 Introduction
Antibody-drug conjugates (ADCs) have emerged as a transformative class of targeted therapies in oncology, embodying the long-held aspiration of achieving a "magic bullet" for cancer treatment, as envisioned by Paul Ehrlich over a century ago. By combining the specificity of monoclonal antibodies with the potent cytotoxic effects of chemotherapeutic agents, ADCs facilitate the targeted delivery of drugs directly to cancer cells while sparing healthy tissues from the damaging effects of conventional chemotherapy. This unique mechanism not only enhances therapeutic efficacy but also significantly reduces systemic toxicity, making ADCs a promising avenue for cancer therapy [1][2].
The significance of ADCs in modern oncology cannot be overstated. They represent a sophisticated approach to combatting cancer, addressing some of the critical limitations associated with traditional treatments, such as poor specificity and resultant off-target effects. ADCs consist of three main components: a monoclonal antibody that targets specific tumor-associated antigens, a linker that connects the antibody to a cytotoxic payload, and the cytotoxic agent itself, which induces cell death upon internalization [2][3]. The design and optimization of these components are pivotal for the success of ADCs, as they must work synergistically to ensure effective delivery and therapeutic outcomes.
The current landscape of ADC research and development is marked by rapid advancements and increasing clinical applications. Several ADCs have received regulatory approval, such as brentuximab vedotin for Hodgkin's lymphoma and trastuzumab emtansine for HER2-positive breast cancer, showcasing the potential of this therapeutic strategy [4][5]. Moreover, numerous ADCs are currently in various stages of clinical trials, with an emphasis on solid tumors, highlighting the ongoing efforts to expand their utility beyond hematologic malignancies [6][7].
Despite their promise, the development and clinical application of ADCs are not without challenges. Issues such as tumor heterogeneity, the emergence of resistance mechanisms, and the complexities of linker chemistry and payload selection pose significant hurdles [8][9]. Recent studies have begun to address these challenges, focusing on enhancing the specificity and efficacy of ADCs through innovative design strategies, including the use of novel linkers and payloads, as well as combination therapies that integrate ADCs with other treatment modalities [2][3].
This review is organized into several key sections that will provide a comprehensive overview of the mechanisms by which ADCs target cancer cells, the critical components that constitute these therapeutic agents, and the current state of clinical applications. In the first section, we will explore the mechanisms of action of ADCs, detailing how they target tumor-associated antigens and the processes involved in internalization and drug release. The second section will focus on the various components of ADCs, including antibody selection and engineering, linker types and their significance, and the diverse range of cytotoxic payloads used. Following this, we will discuss the clinical landscape of ADCs, highlighting both approved therapies and those in clinical trials. Finally, we will address the challenges faced in ADC development and outline future directions for research and innovation in this exciting field. Understanding these elements is crucial for optimizing ADC design and improving therapeutic outcomes for cancer patients, thereby advancing the fight against this complex and heterogeneous disease [2][3].
2 Mechanisms of Action of Antibody-Drug Conjugates
2.1 Targeting Tumor-Associated Antigens
Antibody-drug conjugates (ADCs) are an innovative class of targeted cancer therapies that combine the specificity of monoclonal antibodies with potent cytotoxic agents. The mechanism of action of ADCs is primarily based on their ability to selectively target tumor-associated antigens (TAAs) expressed on the surface of cancer cells. This targeting mechanism involves several critical steps:
Binding to Tumor-Associated Antigens: ADCs are designed to recognize and bind to specific TAAs that are overexpressed on malignant cells compared to normal tissues. This selective binding is facilitated by the monoclonal antibody component of the ADC, which has high affinity for the target antigen. The binding of the ADC to the TAA is the first step in the therapeutic process, ensuring that the cytotoxic agent is delivered specifically to cancer cells, thereby minimizing off-target effects and systemic toxicity [3].
Internalization: Following the binding of the ADC to the TAA, the complex is internalized by the tumor cell through receptor-mediated endocytosis. This process allows the ADC to enter the cell, where it can exert its cytotoxic effects. The internalization is a crucial step as it facilitates the release of the cytotoxic payload within the cancer cell [10].
Payload Release: Once inside the tumor cell, the cytotoxic agent is released from the ADC. This release can occur through various mechanisms, such as enzymatic cleavage of the linker that connects the antibody to the cytotoxic drug. The stability of the linker is essential for ensuring that the payload is released in the intracellular environment, where it can exert its lethal effects on the cancer cell [11].
Induction of Cell Death: The cytotoxic payload, which is often a potent chemotherapy agent, induces cell death through mechanisms such as apoptosis or necrosis. The selective delivery of these cytotoxic agents to cancer cells enhances their therapeutic efficacy while reducing the adverse effects commonly associated with traditional chemotherapy [12].
Bystander Effect: An additional advantage of ADCs is the potential for a 'bystander effect,' where the released cytotoxic agent can affect neighboring tumor cells that may not express the target antigen. This phenomenon is particularly important in tumors with heterogeneous antigen expression, as it can lead to broader therapeutic efficacy [10].
The targeting strategy of ADCs not only enhances their efficacy but also represents a significant advancement in precision oncology. By leveraging the specificity of antibodies for TAAs, ADCs aim to improve the therapeutic index, allowing for higher doses of cytotoxic agents to be administered while minimizing damage to normal tissues [5].
In summary, the mechanism of action of antibody-drug conjugates is fundamentally based on their ability to selectively target tumor-associated antigens, facilitating the internalization and release of cytotoxic agents that induce cell death in cancer cells. This targeted approach holds promise for improving outcomes in cancer therapy, particularly in challenging malignancies characterized by resistance to conventional treatments.
2.2 Internalization and Drug Release Mechanisms
Antibody-drug conjugates (ADCs) represent a sophisticated class of targeted cancer therapies that leverage the specificity of monoclonal antibodies to deliver cytotoxic agents directly to tumor cells. The mechanisms of action of ADCs primarily involve the binding of the antibody component to specific antigens expressed on the surface of cancer cells, followed by internalization and subsequent release of the cytotoxic payload within the target cells.
The targeting mechanism begins with the ADC binding to its specific tumor-associated antigen through the antibody. This binding event facilitates the internalization of the ADC into the cancer cell via receptor-mediated endocytosis. Upon internalization, the ADC is trafficked through the endosomal and lysosomal compartments of the cell, where the linker that connects the antibody to the cytotoxic drug is cleaved, typically by lysosomal enzymes. This cleavage releases the cytotoxic payload inside the cell, allowing it to exert its therapeutic effects, such as disrupting DNA or interfering with the microtubule network, leading to cell death (Sweeney-Lasch et al. 2025; Xu 2015).
Moreover, the design of ADCs incorporates various linker chemistries that can be cleavable or non-cleavable, impacting the release of the cytotoxic drug. Cleavable linkers are specifically designed to be sensitive to the enzymatic environment within the lysosomes, ensuring that the drug is released precisely where it is needed, while non-cleavable linkers might allow for the drug to be released in a more controlled manner outside the target cell, which can also facilitate a bystander effect where neighboring cells that do not express the target antigen can be affected by the released drug (Fucà et al. 2024; Jose et al. 2025).
In addition to the internalization and drug release processes, the efficacy of ADCs is also influenced by factors such as the density of the target antigen on the tumor cell surface and the overall pharmacokinetics of the ADC, which include distribution, metabolism, and excretion. These pharmacological parameters are critical for optimizing the therapeutic window of ADCs, ensuring that they are effective against tumors while minimizing systemic toxicity (Polakis 2016; Kalim et al. 2017).
Overall, the internalization and drug release mechanisms of ADCs are central to their action, providing a targeted approach to cancer therapy that capitalizes on the unique properties of both antibodies and cytotoxic agents. By combining these elements, ADCs have shown promise in treating various malignancies, including those characterized by heterogeneous biomarker expression, such as endometrial cancer (Fucà et al. 2024). The continuous exploration of these mechanisms will further enhance the development of ADCs, addressing challenges such as resistance and improving treatment outcomes.
3 Components of Antibody-Drug Conjugates
3.1 Antibody Selection and Engineering
Antibody-drug conjugates (ADCs) represent a sophisticated class of targeted cancer therapies that leverage the specificity of monoclonal antibodies (mAbs) to selectively deliver potent cytotoxic agents to tumor cells while minimizing damage to normal tissues. The targeting mechanism of ADCs is fundamentally based on the unique interaction between the antibody component and specific antigens expressed on the surface of cancer cells.
The primary components of ADCs include the antibody, the cytotoxic payload, and a chemical linker that connects the two. The selection of the antibody is critical, as it must demonstrate high specificity for antigens that are predominantly expressed on malignant cells, ensuring that the cytotoxic effects are confined to the tumor environment. The most common backbone for ADCs is humanized or fully human immunoglobulin G1 (IgG1) antibodies due to their favorable properties, such as stability in systemic circulation, robust engagement with Fcγ receptors for immune effector functions, and reduced immunogenicity [13].
When selecting an antibody for ADC development, several factors must be considered to optimize its efficacy. These include the antibody's binding affinity for the target antigen, the internalization rate of the antibody-antigen complex, and the potential for overcoming barriers to tissue penetration, such as the binding-site barrier effect. Emerging strategies involve the use of tumor-specific antigen variants or unique post-translational modifications to enhance selectivity and improve the therapeutic index [14].
Advancements in antibody engineering have further refined ADC design. Innovations include the development of site-specific conjugation techniques that allow for the precise attachment of the cytotoxic payload to predetermined sites on the antibody. This not only improves the homogeneity of the ADC but also enhances its pharmacokinetic properties and clinical predictability [15]. The conjugation strategies may involve lysine- and cysteine-based chemistries, enzymatic tagging, glycan remodeling, and the incorporation of non-canonical amino acids [13].
Moreover, the optimization of the drug-to-antibody ratio (DAR) is essential in ADC design, as it influences the therapeutic efficacy and safety profile. A balanced DAR ensures that sufficient cytotoxic agents are delivered to effectively kill cancer cells without causing excessive off-target toxicity [2].
In summary, the targeting of cancer by ADCs hinges on the careful selection and engineering of antibodies that exhibit high specificity for tumor-associated antigens, combined with innovations in linker chemistry and payload design. These strategies aim to maximize the therapeutic index while minimizing adverse effects, ultimately enhancing the efficacy of cancer treatments [2][13][14].
3.2 Linkers: Types and Their Importance
Antibody-drug conjugates (ADCs) represent a sophisticated approach to cancer therapy, integrating the specificity of monoclonal antibodies with the potent cytotoxicity of small-molecule drugs. The primary components of ADCs include a monoclonal antibody, a cytotoxic payload, and a linker that connects the two. The efficacy of ADCs in targeting cancer cells hinges significantly on these components, particularly the linkers, which play a crucial role in the stability and release of the cytotoxic drug.
The mechanism by which ADCs target cancer cells begins with the monoclonal antibody component, which binds specifically to tumor-associated antigens on the surface of cancer cells. This binding facilitates the internalization of the ADC through receptor-mediated endocytosis, leading to the formation of ADC-antigen complexes. Once inside the cell, the ADC is trafficked through the endosomal-lysosomal pathway, where the linker is cleaved, resulting in the release of the cytotoxic payload that induces apoptosis in the tumor cells[16].
Linkers are essential for the overall performance of ADCs, as they determine the stability of the conjugate in circulation and the controlled release of the cytotoxic agent at the tumor site. Linkers can be classified into two main categories: cleavable and non-cleavable. Cleavable linkers are designed to be stable in the bloodstream but are activated in the tumor microenvironment or within cancer cells, allowing for selective drug release. Common types of cleavable linkers include hydrazone, disulfide, and peptide linkers, each engineered to respond to specific intracellular conditions, such as pH changes or enzymatic activity, facilitating the release of the cytotoxic drug precisely where it is needed[17].
The design of the linker is a critical challenge in ADC development, as it must balance stability during circulation with the ability to release the drug at the tumor site. For instance, some linkers are sensitive to the acidic environment within endosomes or to specific enzymes that are overexpressed in tumor tissues, such as cathepsins or neutrophil elastase[16][18]. Innovations in linker technology have focused on improving pharmacokinetics and reducing off-target toxicity, which remains a significant concern in ADC therapy[13].
Recent advancements have introduced novel linker designs, such as photoremovable linkers that can be activated by light, providing an additional layer of control over drug release. These linkers offer spatiotemporal control, potentially improving the therapeutic index of ADCs by minimizing systemic exposure to the cytotoxic payload[19]. Additionally, the development of site-specific conjugation strategies has enhanced the homogeneity and stability of ADCs, further contributing to their clinical efficacy[20].
In summary, the targeting of cancer by ADCs is a multifaceted process that relies heavily on the strategic design of their components, particularly the linkers. These linkers not only serve as connectors between the antibody and the drug but also play a pivotal role in ensuring that the cytotoxic agent is delivered effectively to the cancer cells while minimizing damage to normal tissues. As research continues to evolve, innovations in linker chemistry and design are expected to enhance the therapeutic potential of ADCs, making them a cornerstone of targeted cancer therapies[16][17].
3.3 Cytotoxic Payloads: Types and Mechanisms
Antibody-drug conjugates (ADCs) are innovative therapeutic agents designed to deliver cytotoxic drugs specifically to cancer cells, thus minimizing damage to normal tissues. The fundamental components of ADCs include monoclonal antibodies, linkers, and cytotoxic payloads, each playing a crucial role in the mechanism of targeting and efficacy of these therapies.
The targeting mechanism of ADCs begins with the monoclonal antibodies, which are engineered to bind selectively to tumor-associated antigens expressed on the surface of cancer cells. This specificity is critical as it ensures that the cytotoxic payload is delivered predominantly to malignant cells, thereby enhancing the therapeutic index of the treatment. Once the antibody binds to its target antigen, the ADC is internalized into the cancer cell through receptor-mediated endocytosis. This process leads to the formation of ADC-antigen complexes that are trafficked through the endosomal-lysosomal pathway, where the linker is cleaved, resulting in the release of the cytotoxic payload directly within the tumor cell [16][21].
The cytotoxic payloads used in ADCs can vary widely in their mechanisms of action and types. These payloads are typically highly potent cytotoxic agents that would be too toxic for systemic administration on their own. Recent advancements have introduced novel payloads that not only induce apoptosis through traditional mechanisms but also employ alternative pathways. For instance, some payloads include microtubule inhibitors, spliceosome modulators, and RNA polymerase inhibitors, which target different cellular processes to exert their cytotoxic effects [21].
The design of the linker connecting the antibody and the cytotoxic payload is equally important, as it influences the stability and release of the drug within the target cells. Innovations in linker chemistry have led to the development of more stable and effective ADCs, which can better withstand circulation in the bloodstream before reaching the tumor [13]. Furthermore, the optimization of drug-to-antibody ratios (DARs) is crucial for enhancing the efficacy and safety profiles of ADCs [13].
In summary, ADCs leverage the specificity of monoclonal antibodies to deliver potent cytotoxic payloads directly to cancer cells, thereby improving treatment efficacy while minimizing systemic toxicity. The ongoing research in the field focuses on refining the components of ADCs, including antibody selection, linker chemistry, and the development of innovative cytotoxic payloads, which collectively enhance the therapeutic potential of these targeted therapies [3][13].
4 Clinical Applications and Current Landscape
4.1 Approved ADCs and Their Indications
Antibody-drug conjugates (ADCs) represent a significant advancement in targeted cancer therapy, utilizing the specificity of monoclonal antibodies to selectively deliver cytotoxic agents to tumor cells. The mechanism of action of ADCs involves the conjugation of a monoclonal antibody, which targets specific antigens expressed on the surface of cancer cells, to a potent cytotoxic drug through a stable linker. This design allows for the targeted killing of cancer cells while minimizing damage to normal tissues, addressing one of the major limitations of conventional chemotherapy.
The targeting mechanism begins when the ADC binds to its specific tumor-associated antigen on the cancer cell surface. Following binding, the ADC is internalized via receptor-mediated endocytosis, leading to the release of the cytotoxic payload within the tumor cell's lysosome. This process not only affects the targeted cancer cell but can also induce a bystander effect, impacting neighboring tumor cells that may not express the antigen but are affected by the released cytotoxic agent[22].
Over the years, significant progress has been made in the development and clinical application of ADCs. As of now, twelve ADCs have been approved by the U.S. Food and Drug Administration (FDA) for various hematological and solid tumors. Notably, ADCs have demonstrated wider therapeutic indexes compared to traditional chemotherapy due to their selective delivery of toxic payloads, thereby reducing off-target effects and improving overall treatment outcomes[23].
For instance, ADCs targeting the human epidermal growth factor receptor 2 (HER2) have been particularly successful in treating HER2-positive breast cancer. The approved ADCs, such as trastuzumab emtansine (T-DM1), utilize the targeting capability of trastuzumab to deliver the cytotoxic agent directly to HER2-expressing tumor cells, significantly improving patient prognosis and providing effective treatment options for those resistant to previous therapies[24].
Moreover, ADCs are being explored for their potential in combination therapies, enhancing the efficacy of existing treatments. The integration of novel markers, linkers, and payloads is also under investigation to further improve the specificity and potency of these therapeutic agents[22].
The landscape of ADCs continues to evolve, with ongoing clinical trials evaluating around 80 ADCs targeting various antigens for the treatment of both hematological and solid malignancies[25]. This expansion signifies a growing recognition of ADCs as a transformative approach in oncology, providing new avenues for personalized cancer treatment strategies and potentially reshaping treatment paradigms across different cancer types. The continuous refinement of ADC design, targeting strategies, and understanding of resistance mechanisms will likely enhance their clinical efficacy and broaden their indications in the future[11].
In conclusion, ADCs exemplify a novel therapeutic strategy that harnesses the specificity of antibodies to deliver cytotoxic drugs directly to cancer cells, significantly impacting clinical practice in oncology and offering hope for improved outcomes in cancer treatment.
4.2 Emerging ADCs in Clinical Trials
Antibody-drug conjugates (ADCs) represent a significant advancement in targeted cancer therapy, effectively combining the specificity of monoclonal antibodies with the potent cytotoxic effects of chemotherapy. This innovative approach allows for selective targeting of tumor cells while minimizing damage to healthy tissues, thereby enhancing the therapeutic index.
ADCs operate by utilizing antibodies that bind to specific antigens expressed on the surface of tumor cells. Once bound, the ADC is internalized through receptor-mediated endocytosis, leading to the release of the cytotoxic payload within the tumor cell. This mechanism not only reduces systemic toxicity but also enhances the efficacy of the therapeutic agents by delivering them directly to the cancerous cells [3].
The clinical landscape of ADCs is rapidly evolving, with 11 products currently approved by the U.S. Food and Drug Administration (FDA) and over 500 active clinical trials investigating new ADCs [7]. These ADCs have demonstrated significant efficacy across various cancer types, including breast cancer, where they have transformed treatment paradigms by selectively delivering cytotoxic agents to malignant cells while sparing normal tissues [26].
Emerging ADCs in clinical trials are focusing on innovative designs to overcome challenges such as drug resistance, tumor heterogeneity, and treatment-related adverse events. For instance, advancements in antibody engineering and linker design have been pivotal in enhancing the stability and therapeutic efficacy of ADCs [27]. Additionally, the integration of bispecific antibodies into ADC designs is showing promise, as these constructs can exhibit superior internalization and selectivity, potentially leading to increased safety and therapeutic efficacy [28].
Furthermore, combination therapies involving ADCs and other modalities, such as immune checkpoint inhibitors and small-molecule therapies, are being explored to enhance treatment outcomes. Clinical studies indicate that such combinations can significantly improve response rates and progression-free survival across various cancers [9].
In summary, the targeting mechanism of ADCs relies on the specificity of monoclonal antibodies to deliver cytotoxic agents directly to tumor cells, which is a cornerstone of their clinical application. The ongoing research and clinical trials are poised to further refine ADC technology, expanding their utility in cancer therapy and addressing existing challenges to maximize their therapeutic potential [7][27][29].
5 Challenges and Future Directions
5.1 Resistance Mechanisms in Cancer Cells
Antibody-drug conjugates (ADCs) represent a sophisticated therapeutic approach that combines the specificity of monoclonal antibodies with potent cytotoxic agents, enabling targeted delivery of these agents to cancer cells. This targeted delivery is predicated on the ability of antibodies to recognize and bind to specific antigens expressed on the surface of tumor cells, which ideally minimizes damage to normal tissues and enhances the therapeutic index of the cytotoxic payloads.
ADCs function by linking a cytotoxic drug to a monoclonal antibody through a stable chemical linker. The mechanism begins when the ADC binds to its target antigen on the cancer cell surface, leading to internalization of the conjugate. Once inside the cell, the linker is cleaved, releasing the cytotoxic drug, which then exerts its lethal effects on the tumor cell. This mechanism not only allows for localized treatment but also capitalizes on the tumor's unique antigenic profile, thereby increasing efficacy while reducing systemic exposure and associated side effects [30][31][32].
However, the effectiveness of ADCs is often compromised by various resistance mechanisms that can emerge in cancer cells. These mechanisms can be categorized into several key areas:
Alteration in Antigen Expression: Tumor cells may downregulate or completely lose the expression of the target antigen, rendering the ADC ineffective. This can occur through genetic mutations or epigenetic modifications that affect the expression of surface antigens [11][30].
Changes in ADC Processing: The internalization and processing of the ADC within the cancer cell can be disrupted. This includes alterations in endosomal trafficking or impaired lysosomal degradation, which prevent the release of the cytotoxic payload [11][31].
Efflux Mechanisms: Cancer cells can develop or enhance efflux mechanisms that actively pump the cytotoxic drug out of the cell, thus reducing its intracellular concentration and effectiveness. This is often mediated by transport proteins such as ATP-binding cassette (ABC) transporters [30][31].
Intrinsic Tumor Cell Dynamics: Variability in tumor cell signaling pathways and dynamics can influence how cells respond to the cytotoxic effects of ADCs. Factors such as apoptosis resistance or altered signaling cascades can lead to reduced sensitivity to the payload [11][12].
To address these challenges, researchers are exploring several strategies aimed at overcoming resistance mechanisms. These include:
Development of Next-Generation ADCs: This involves creating ADCs that target multiple antigens or utilize different payloads to circumvent resistance associated with a single target [31][33].
Combination Therapies: Combining ADCs with other therapeutic modalities, such as immune checkpoint inhibitors or conventional chemotherapy, can enhance anti-tumor efficacy and mitigate resistance. This approach aims to exploit synergistic effects that may overcome the inherent resistance of cancer cells [9][33].
Improving Linker Technology: Transitioning from non-cleavable to cleavable linkers can improve the delivery of cytotoxic agents directly into the tumor cells, thus enhancing the overall therapeutic effect [11][31].
In summary, while ADCs offer a promising avenue for targeted cancer therapy, the emergence of resistance mechanisms presents significant challenges. Continued research into understanding these mechanisms and developing innovative strategies to overcome them is essential for enhancing the clinical efficacy of ADCs and improving patient outcomes in cancer treatment.
5.2 Strategies for Enhancing ADC Efficacy
Antibody-drug conjugates (ADCs) are a class of biopharmaceutical drugs that combine the targeting capability of monoclonal antibodies with the cytotoxic effects of chemotherapy agents. This combination allows ADCs to selectively deliver potent drugs to cancer cells while minimizing damage to normal tissues. The targeting mechanism relies on the ability of antibodies to bind specifically to antigens that are overexpressed on the surface of tumor cells. Upon binding, ADCs are internalized via receptor-mediated endocytosis, leading to the release of the cytotoxic payload within the tumor cell, thereby inducing cell death. This targeted delivery mechanism is a significant advancement over traditional chemotherapy, which lacks specificity and often results in systemic toxicity[22][34][35].
Despite their promise, ADCs face several challenges that can limit their efficacy. One of the primary issues is the development of resistance, which can arise from various factors, including the expression levels of target antigens, alterations in the internalization and trafficking of the ADC, and the inherent characteristics of the tumor cells themselves. Resistance mechanisms may involve changes in antigen expression, impaired internalization pathways, and alterations in the cellular response to the cytotoxic payload[11][12][29].
To enhance the efficacy of ADCs, several strategies are being explored. One approach is the development of novel payloads that can overcome resistance mechanisms and improve therapeutic outcomes. For instance, integrating immune-stimulating agents, natural toxins, or even radionuclides as payloads may increase the specificity and potency of ADCs, thereby enhancing their effectiveness[29][36]. Another strategy involves the use of combination therapies, where ADCs are administered alongside immune checkpoint inhibitors or other targeted therapies. This combination can potentially amplify the anti-tumor immune response and mitigate resistance[12][33].
Furthermore, advancements in antibody engineering and linker design are crucial for improving the stability and pharmacokinetics of ADCs. Innovations in these areas can lead to better-targeted delivery and reduced off-target effects, ultimately enhancing the therapeutic index of ADCs[27][29]. Personalized medicine approaches, where treatment is tailored based on the molecular profile of individual tumors, also hold promise in optimizing ADC efficacy and minimizing adverse effects[34][35].
In summary, while ADCs represent a significant advancement in targeted cancer therapy, addressing the challenges of resistance and optimizing their efficacy through innovative strategies is essential for their continued success in clinical applications. The ongoing research into novel payloads, combination therapies, and improved design methodologies is likely to broaden the therapeutic potential of ADCs and improve patient outcomes in cancer treatment[11][22][37].
5.3 Future Perspectives in ADC Development
Antibody-drug conjugates (ADCs) represent a sophisticated approach to targeted cancer therapy, combining the specificity of monoclonal antibodies with the potency of cytotoxic drugs. The mechanism of action involves the binding of ADCs to specific antigens expressed on tumor cells, followed by internalization through receptor-mediated endocytosis. Once internalized, the cytotoxic payload is released intracellularly, leading to selective tumor cell death while minimizing damage to normal tissues [22][35].
The targeting capability of ADCs is primarily determined by the monoclonal antibody component, which is designed to recognize and bind to specific antigens that are overexpressed on cancer cells compared to normal cells. This selective binding is critical as it enhances the therapeutic index of the treatment, allowing for more effective tumor eradication with reduced systemic toxicity [26][38]. However, the development of ADCs is not without challenges.
One significant challenge is the development of resistance mechanisms. Cancer cells may alter the expression of target antigens, engage in drug efflux, or activate alternative signaling pathways that circumvent the cytotoxic effects of the ADC [11][11]. Additionally, the stability of the linker used to attach the drug to the antibody is crucial, as instability can lead to premature release of the drug in circulation, potentially increasing toxicity [29].
Looking towards the future, the development of next-generation ADCs aims to address these challenges through several innovative strategies. These include the exploration of novel target antigens that are more specific to cancer cells, improving linker technology to enhance stability and reduce off-target effects, and the incorporation of bispecific antibodies which can simultaneously target multiple antigens or pathways [27][28]. Furthermore, advancements in antibody engineering, such as the use of humanized antibodies and optimized conjugation techniques, are expected to enhance the efficacy and safety profiles of ADCs [39][40].
In summary, while ADCs have demonstrated significant promise in targeting cancer through precise delivery of cytotoxic agents, ongoing research is crucial to overcome existing challenges such as resistance mechanisms and linker stability. Future directions will likely focus on enhancing the specificity and efficacy of ADCs, potentially expanding their application across a broader range of malignancies and improving outcomes for patients. The integration of innovative technologies and therapeutic strategies will be essential in shaping the next generation of ADCs, making them a cornerstone of precision oncology [7][40].
6 Conclusion
The review highlights the significant advancements in antibody-drug conjugates (ADCs) as a targeted cancer therapy, emphasizing their unique mechanism of action that combines monoclonal antibodies with cytotoxic agents to selectively kill cancer cells while minimizing harm to healthy tissues. The main findings indicate that ADCs effectively utilize tumor-associated antigens for precise targeting, facilitating internalization and release of the cytotoxic payload within tumor cells. This targeted approach has led to the approval of several ADCs for various cancers, demonstrating improved therapeutic indices compared to traditional chemotherapies. However, challenges such as tumor heterogeneity, resistance mechanisms, and the complexities of linker chemistry and payload selection persist. Future research directions are focused on developing next-generation ADCs with enhanced specificity and efficacy, exploring novel linkers and payloads, and integrating ADCs with other therapeutic modalities to overcome resistance and improve patient outcomes. The continuous evolution of ADC technology is expected to expand their clinical applications, offering new hope in the fight against cancer.
References
- [1] Yiming Jin;Shahab Edalatian Zakeri;Raman Bahal;Andrew J Wiemer. New Technologies Bloom Together for Bettering Cancer Drug Conjugates.. Pharmacological reviews(IF=17.3). 2022. PMID:35710136. DOI: 10.1124/pharmrev.121.000499.
- [2] Swapnali Parit;Ajit Manchare;Amol D Gholap;Prashant Mundhe;Navnath Hatvate;Satish Rojekar;Vandana Patravale. Antibody-Drug Conjugates: A promising breakthrough in cancer therapy.. International journal of pharmaceutics(IF=5.2). 2024. PMID:38750981. DOI: 10.1016/j.ijpharm.2024.124211.
- [3] Jiya Jose;John D Hooper;Fernando Souza-Fonseca-Guimaraes. Highlights of 2024. Broadening anti-cancer immunotherapy modalities with antibody-drug conjugates: emerging insights from clinical studies.. Immunology and cell biology(IF=3.0). 2025. PMID:40064476. DOI: 10.1111/imcb.70017.
- [4] Aiko Nagayama;Leif W Ellisen;Bruce Chabner;Aditya Bardia. Antibody-Drug Conjugates for the Treatment of Solid Tumors: Clinical Experience and Latest Developments.. Targeted oncology(IF=4.0). 2017. PMID:29116596. DOI: 10.1007/s11523-017-0535-0.
- [5] Anish Thomas;Beverly A Teicher;Raffit Hassan. Antibody-drug conjugates for cancer therapy.. The Lancet. Oncology(IF=35.9). 2016. PMID:27299281. DOI: 10.1016/S1470-2045(16)30030-4.
- [6] Hao Xie;Alex A Adjei. Antibody-Drug Conjugates for the Therapy of Thoracic Malignancies.. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer(IF=20.8). 2019. PMID:30599202. DOI: 10.1016/j.jtho.2018.11.034.
- [7] Edidiong Udofa;Disha Sankholkar;Samir Mitragotri;Zongmin Zhao. Antibody drug conjugates in the clinic.. Bioengineering & translational medicine(IF=5.7). 2024. PMID:39545074. DOI: 10.1002/btm2.10677.
- [8] Beverly A Teicher;Ravi V J Chari. Antibody conjugate therapeutics: challenges and potential.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2011. PMID:22003066. DOI: 10.1158/1078-0432.CCR-11-1417.
- [9] Xianglong Shi;Kai Tang;Quanbin Zhang;Qingkun Han;Lin Quan;Yijing Li;Jianqiao Cui;Nuan Feng;Jianbao Gong;Baoxin Shang;Xuwen Li. Antibody-drug conjugate combinations in cancer treatment: clinical efficacy and clinical study perspectives.. Frontiers in pharmacology(IF=4.8). 2025. PMID:40061961. DOI: 10.3389/fphar.2025.1556245.
- [10] Giovanni Fucà;Ilaria Sabatucci;Mariachiara Paderno;Domenica Lorusso. The clinical landscape of antibody-drug conjugates in endometrial cancer.. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society(IF=4.7). 2024. PMID:40229038. DOI: 10.1136/ijgc-2024-005607.
- [11] Mingxia Jiang;Qiao Li;Binghe Xu. Spotlight on ideal target antigens and resistance in antibody-drug conjugates: Strategies for competitive advancement.. Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy(IF=21.7). 2024. PMID:38677200. DOI: 10.1016/j.drup.2024.101086.
- [12] Gloria Lalli;Ilaria Sabatucci;Mariachiara Paderno;Fabio Martinelli;Mauro Signorelli;Matteo Maruccio;Giampaolo Di Martino;Giovanni Fucà;Domenica Lorusso. Navigating the Landscape of Resistance Mechanisms in Antibody-Drug Conjugates for Cancer Treatment.. Targeted oncology(IF=4.0). 2025. PMID:40234302. DOI: 10.1007/s11523-025-01140-w.
- [13] Ibrahim A Alradwan;Meshal K Alnefaie;Nojoud Al Fayez;Alhassan H Aodah;Majed A Majrashi;Meshael Alturki;Mohannad M Fallatah;Fahad A Almughem;Essam A Tawfik;Abdullah A Alshehri. Strategic and Chemical Advances in Antibody-Drug Conjugates.. Pharmaceutics(IF=5.5). 2025. PMID:41012501. DOI: 10.3390/pharmaceutics17091164.
- [14] Han Liu;Hongye Zeng;Xiaojing Qin;Wenjing Ning;Lin Xu;Shiting Yang;Xue Liu;Wenxin Luo;Ningshao Xia. The Icarian flight of antibody-drug conjugates: target selection amidst complexity and tackling adverse impacts.. Protein & cell(IF=12.8). 2025. PMID:39813112. DOI: 10.1093/procel/pwaf002.
- [15] Qun Zhou. Site-Specific Antibody Conjugation with Payloads beyond Cytotoxins.. Molecules (Basel, Switzerland)(IF=4.6). 2023. PMID:36770585. DOI: 10.3390/molecules28030917.
- [16] Xiaoyang Yao;Yingying Lu;Shuo Shi;Yu Wang. Antibody-drug conjugates (ADCs): "Smart Missiles" targeting cancer therapy.. Colloids and surfaces. B, Biointerfaces(IF=5.6). 2026. PMID:41015828. DOI: 10.1016/j.colsurfb.2025.115161.
- [17] Rotimi Sheyi;Beatriz G de la Torre;Fernando Albericio. Linkers: An Assurance for Controlled Delivery of Antibody-Drug Conjugate.. Pharmaceutics(IF=5.5). 2022. PMID:35214128. DOI: 10.3390/pharmaceutics14020396.
- [18] Jun Lu;Feng Jiang;Aiping Lu;Ge Zhang. Linkers Having a Crucial Role in Antibody-Drug Conjugates.. International journal of molecular sciences(IF=4.9). 2016. PMID:27089329. DOI: .
- [19] Audrey Nathania Johan;Yi Li. Development of Photoremovable Linkers as a Novel Strategy to Improve the Pharmacokinetics of Drug Conjugates and Their Potential Application in Antibody-Drug Conjugates for Cancer Therapy.. Pharmaceuticals (Basel, Switzerland)(IF=4.8). 2022. PMID:35745573. DOI: 10.3390/ph15060655.
- [20] Yaxin Lei;Minglei Zheng;Peng Chen;Chen Seng Ng;Teck Peng Loh;Huitao Liu. Linker Design for the Antibody Drug Conjugates: A Comprehensive Review.. ChemMedChem(IF=3.4). 2025. PMID:40432256. DOI: 10.1002/cmdc.202500262.
- [21] Fatima Akram;Amna Murrawat Ali;Muhammad Tayyab Akhtar;Taseer Fatima;Ifrah Shabbir;Ikram Ul Haq. The journey of antibody-drug conjugates for revolutionizing cancer therapy: A review.. Bioorganic & medicinal chemistry(IF=3.0). 2025. PMID:39586174. DOI: 10.1016/j.bmc.2024.118010.
- [22] Jun He;Xianghua Zeng;Chunmei Wang;Enwen Wang;Yongsheng Li. Antibody-drug conjugates in cancer therapy: mechanisms and clinical studies.. MedComm(IF=10.7). 2024. PMID:39070179. DOI: 10.1002/mco2.671.
- [23] Benjamina Esapa;Jiexuan Jiang;Anthony Cheung;Alicia Chenoweth;David E Thurston;Sophia N Karagiannis. Target Antigen Attributes and Their Contributions to Clinically Approved Antibody-Drug Conjugates (ADCs) in Haematopoietic and Solid Cancers.. Cancers(IF=4.4). 2023. PMID:36980732. DOI: 10.3390/cancers15061845.
- [24] Lixi Li;Di Zhang;Binliang Liu;Dan Lv;Jingtong Zhai;Xiuwen Guan;Zongbi Yi;Fei Ma. Antibody-drug conjugates in HER2-positive breast cancer.. Chinese medical journal(IF=7.3). 2021. PMID:34935688. DOI: 10.1097/CM9.0000000000001932.
- [25] Haiyong Peng. Perspectives on the development of antibody-drug conjugates targeting ROR1 for hematological and solid cancers.. Antibody therapeutics(IF=4.5). 2021. PMID:34805745. DOI: 10.1093/abt/tbab023.
- [26] Mariana Ribeiro Monteiro;Natalia Cristina Cardoso Nunes;Aumilto Augusto da Silva Junior;Angelo Bezerra de Souza Fêde;Gustavo de Oliveira Bretas;Cristiano de Pádua Souza;Max Mano;Jesse Lopes da Silva. Antibody-Drug Conjugates in Breast Cancer: A Comprehensive Review of How to Selectively Deliver Payloads.. Breast cancer (Dove Medical Press)(IF=3.4). 2024. PMID:38434801. DOI: 10.2147/BCTT.S448191.
- [27] Jung Yin Fong;Zhixin Phuna;Di Yang Chong;Christophorus Manuel Heryanto;Yu Shyan Low;Khang Chiang Oh;Yan Huen Lee;Allan Wee Ren Ng;Lionel Lian Aun In;Michelle Yee Mun Teo. Advancements in antibody-drug conjugates as cancer therapeutics.. Journal of the National Cancer Center(IF=9.4). 2025. PMID:40814440. DOI: 10.1016/j.jncc.2025.01.007.
- [28] Hongye Zeng;Wenjing Ning;Xue Liu;Wenxin Luo;Ningshao Xia. Unlocking the potential of bispecific ADCs for targeted cancer therapy.. Frontiers of medicine(IF=3.5). 2024. PMID:39039315. DOI: 10.1007/s11684-024-1072-8.
- [29] Victor S Goldmacher. Antibody-drug conjugates in cancer and beyond: progress, promise, and perspectives.. Antibody therapeutics(IF=4.5). 2025. PMID:40860900. DOI: 10.1093/abt/tbaf013.
- [30] Rachel Occhiogrosso Abelman;Bogang Wu;Laura M Spring;Leif W Ellisen;Aditya Bardia. Mechanisms of Resistance to Antibody-Drug Conjugates.. Cancers(IF=4.4). 2023. PMID:36831621. DOI: 10.3390/cancers15041278.
- [31] Sijia Li;Xinyu Zhao;Kai Fu;Shuangli Zhu;Can Pan;Chuan Yang;Fang Wang;Kenneth K W To;Liwu Fu. Resistance to antibody-drug conjugates: A review.. Acta pharmaceutica Sinica. B(IF=14.6). 2025. PMID:40177568. DOI: 10.1016/j.apsb.2024.12.036.
- [32] Gang Li;Dengqin He;Xiaojia Cai;Wen Guan;Yali Zhang;Jia-Qiang Wu;Hongliang Yao. Advances in the development of phosphodiesterase-4 inhibitors.. European journal of medicinal chemistry(IF=5.9). 2023. PMID:36809706. DOI: 10.1016/j.ejmech.2023.115195.
- [33] Yahui Lv;Xiaoran Cui;Tao Li;Chang Liu;An Wang;Ting Wang;Xin Zhou;Ruixin Li;Fan Zhang;Yi Hu;Tong Zhang;Zhefeng Liu. Mechanism of action and future perspectives of ADCs in combination with immune checkpoint inhibitors for solid tumors.. Clinical and experimental medicine(IF=3.5). 2025. PMID:40319436. DOI: 10.1007/s10238-025-01655-6.
- [34] Cynthia Mark;Jin Sun Lee;Xiaojiang Cui;Yuan Yuan. Antibody-Drug Conjugates in Breast Cancer: Current Status and Future Directions.. International journal of molecular sciences(IF=4.9). 2023. PMID:37762027. DOI: 10.3390/ijms241813726.
- [35] Roberto Paz-Manrique;Joseph A Pinto;Henry L Gomez Moreno. Antibody-Drug Conjugates in Breast Cancer: Toward a Molecular Perspective Into Clinical Practice.. JCO precision oncology(IF=5.6). 2024. PMID:39475662. DOI: 10.1200/PO.24.00173.
- [36] Davide Izzo;Liliana Ascione;Lorenzo Guidi;Renato Maria Marsicano;Chrysanthi Koukoutzeli;Dario Trapani;Giuseppe Curigliano. Innovative payloads for ADCs in cancer treatment: moving beyond the selective delivery of chemotherapy.. Therapeutic advances in medical oncology(IF=4.2). 2025. PMID:39759830. DOI: 10.1177/17588359241309461.
- [37] Stefano Sganga;Silvia Riondino;Giovanni Maria Iannantuono;Roberto Rosenfeld;Mario Roselli;Francesco Torino. Antibody-Drug Conjugates for the Treatment of Renal Cancer: A Scoping Review on Current Evidence and Clinical Perspectives.. Journal of personalized medicine(IF=3.0). 2023. PMID:37763107. DOI: 10.3390/jpm13091339.
- [38] Umbreen Hafeez;Sagun Parakh;Hui K Gan;Andrew M Scott. Antibody-Drug Conjugates for Cancer Therapy.. Molecules (Basel, Switzerland)(IF=4.6). 2020. PMID:33081383. DOI: 10.3390/molecules25204764.
- [39] Irene Sevilla-Carrillo;Eloína García-Tercero;Carlos Alonso-Moreno;Carmen Moya-Lopez. Antibody Drug Conjugates (ADCs): Shaping the Future of Precision Oncology.. Anti-cancer agents in medicinal chemistry(IF=3.0). 2025. PMID:39901681. DOI: 10.2174/0118715206348204241128063329.
- [40] Devesh Aggarwal;Jie Yang;Md Abdus Salam;Sagnik Sengupta;Md Yusuf Al-Amin;Saad Mustafa;Mohammad Aasif Khan;Xun Huang;Jogendra Singh Pawar. Antibody-drug conjugates: the paradigm shifts in the targeted cancer therapy.. Frontiers in immunology(IF=5.9). 2023. PMID:37671162. DOI: 10.3389/fimmu.2023.1203073.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Antibody-drug conjugates · Tumor associated antigens · Internalization · Cytotoxic payloads · Precision oncology
© 2025 MaltSci
