top of page

Antibody-Drug Conjugates: Unlocking the Future in Cancer Therapies

  • Writer: Fay
    Fay
  • 7 hours ago
  • 4 min read
ree

Introduction


Cancer is a major global public health challenge, claiming approximately 10 million lives in 2020 alone. Over the past decades, cytotoxic chemotherapy has been the cornerstone of cancer treatment, featuring common drugs such as cisplatin, paclitaxel, and methotrexate. While these agents effectively kill tumor cells, their lack of specificity inevitably damages normal tissues, leading to severe side effects that limit clinical application. Balancing therapeutic efficacy with reduced toxicity remains a critical focus in cancer treatment research.


As early as the beginning of the 20th century, German scientist Paul Ehrlich envisioned the “magic bullet” concept—an ideal drug that could precisely identify and eliminate diseased cells without harming healthy tissues. With the advent of hybridoma technology in 1975, this vision gradually became reality—the development of monoclonal antibodies (mAbs) ushered in a new era for cancer therapy. By recognizing specific or highly expressed antigens on cancer cell surfaces (such as HER2 in breast cancer or CD20 in lymphoma), mAbs enable targeted tumor attacks and have already spawned numerous successful anticancer drugs. However, monoclonal antibodies alone often exhibit limited efficacy and cannot fully replace traditional chemotherapy. To strike a balance between precision targeting and potent killing, researchers proposed the concept of antibody-drug conjugates (ADCs). By combining the precise delivery capability of mAbs with the powerful cytotoxic effects of drugs, ADCs have opened new possibilities for cancer treatment. This article will introduce the biological mechanisms and development process of ADCs.


Key components of ADC


The science behind ADCs relies on three essential components: the antibody, the linker, and the payload. The antibody acts as a homing device, seeking out specific proteins (antigens) that are abundant in cancer cells but scarce in normal tissue. Once the antibody binds to its target, the ADC is absorbed into the cancer cell. At this point, the linker—an engineered chemical bridge—breaks down in a controlled manner, releasing the cytotoxic payload inside the cell. The payload, often a highly potent chemotherapy drug, then disrupts critical cellular processes, leading to cancer cell death.


What makes ADCs so remarkable is the fine-tuned balance of these three parts. The antibody must be highly selective to avoid off-target effects, the linker must be stable enough to prevent premature drug release in the bloodstream, and the payload must be potent enough to kill the cancer cell with minimal exposure. Advances in each of these areas over the past two decades have transformed ADCs from early experimental concepts into viable, approved treatments for various cancers, such as breast cancer and lymphomas.



Mechanism of ADC


ADC are dubbed “biological missiles” precisely because they combine “precision targeting” with “highly efficient killing.” Unlike traditional chemotherapy, ADCs do not randomly release toxic drugs throughout the body. Instead, they deliver the “bomb” to cancer cells via antibodies, then detonate it precisely within the cells. This approach enhances therapeutic efficacy while minimizing damage to healthy tissues.


This process begins when the antibody component of the ADC recognizes specific antigens on the surface of cancer cells. Upon binding to its target, the ADC is actively engulfed by the cancer cell and transported into an intracellular vesicle (endosome). As the endosome matures and fuses with a lysosome, the ADC's “transport shell” is degraded, releasing the potent cytotoxic drug (payload) it carries. The released drug precisely targets critical cancer cell functions—such as disrupting DNA or interfering with microtubule structures—forcing cells to halt division and ultimately die.


More intriguingly, some payloads exhibit permeability, diffusing out of dead cancer cells to continue affecting surrounding tumor cells. This phenomenon, known as the “bystander effect,” not only broadens the killing range but may also indirectly enhance overall therapeutic efficacy by altering the tumor microenvironment.


The anti-cancer mechanisms of ADCs extend beyond this. Their antibody components can activate immune responses, such as antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC). These mechanisms mobilize immune cells like natural killer cells and macrophages to collectively attack tumors. Furthermore, the antibodies in certain ADCs directly interfere with cancer cell signaling pathways. For example, trastuzumab in T-DM1 not only delivers the drug into HER2-positive cancer cells but also blocks interactions between the HER2 receptor and other receptors. This disrupts key signaling pathways crucial for cell proliferation and survival (such as PI3K or MAPK pathways), delivering a dual strike that induces cancer cell apoptosis.


In essence, the mechanism of ADCs constitutes a comprehensive “multi-layered defense + precision strike” system. It combines targeted delivery and potent toxic effects with the ability to mobilize the immune system and reshape the tumor microenvironment, demonstrating greater overall anticancer potential than monotherapy.


Progress in ADC Development


Looking back at the evolution of ADCs, we see a journey from concept to clinical application, marked by continuous technological iteration alongside persistent challenges and breakthroughs. The advent of first-generation ADCs demonstrated the feasibility of the novel “antibody-toxin” model. However, their efficacy and safety were suboptimal due to limitations in linker stability, pharmacokinetic properties, and the heterogeneity resulting from random conjugation. Second-generation ADCs achieved enhanced targeting and improved plasma stability through optimizations in humanized antibodies, linkers, and payload selection, paving the way for successful products like brentuximab vedotin and ado-trastuzumab emtansine. However, off-target toxicity and DAR (drug-to-antibody ratio) heterogeneity remained limiting factors.


The advent of third-generation ADCs marks a qualitative leap in technology. Site-specific conjugation ensures uniform payload distribution, while more potent and diverse cytotoxic drugs significantly enhance killing capacity. Fully humanized antibodies and more hydrophilic linkers further improve safety and pharmacokinetic performance. The successive approvals of products like polatuzumab vedotin, enfortumab vedotin, and fam-trastuzumab deruxtecan signify that ADCs have transitioned from initial experimental exploration to a mature and expanding phase of clinical application.


Overall, the three-generation evolution of ADCs clearly demonstrates a progression from “proof-of-concept” to “technical optimization” and ultimately to “stable efficacy.” With the continuous expansion of targets and ongoing innovation in payloads and linkers, ADCs are gradually becoming a key pillar driving precision oncology. They represent not only an innovation in drug formulation but also foreshadow potentially profound transformations in the future landscape of cancer treatment.


Source



Assessed and Endorsed by the MedReport Medical Review Board



 
 

©2025 by The MedReport Foundation, a Washington state non-profit organization operating under the UBI 605-019-306

 

​​The information provided by the MedReport Foundation is not intended or implied to be a substitute for professional medical advice, diagnosis, or treatment. The MedReport Foundation's resources are solely for informational, educational, and entertainment purposes. Always seek professional care from a licensed provider for any emergency or medical condition. 
 

bottom of page