The Rise of ADCs: Unlocking the Power of Precision Cancer Therapy

May 1, 2024
7 min read

Antibody-drug conjugates (ADCs) have emerged as a promising class of targeted cancer therapeutics, offering the potential for precise tumour targeting and reduced systemic toxicity widely used in the treatment of hematologic malignancies and solid tumours. In this article we will discuss the rise of ADCs, elucidating their mechanism of action, the evolution of ADC technology, current clinical applications, challenges, and future prospects.

Cancer, it’s a disease in which some of human body’s cells grow uncontrollably and spread to other parts of the human body. It can start almost anywhere in the body.1 Cancer remains one of the most pressing global health challenges, with its complex and heterogeneous nature posing significant hurdles to effective treatment. Cancer therapies have evolved considerably in recent decades, substantially improving the quality of life and survival of patients with cancer. Traditional chemotherapy, while potent, often lacks specificity, leading to severe side effects and limited therapeutic efficacy. The advent of targeted therapies has heralded a new era in cancer treatment, aiming to selectively eradicate malignant cells while sparing normal tissues.2 Among these targeted approaches, antibody-drug conjugates (ADCs) have emerged as a groundbreaking strategy, harnessing the specificity of monoclonal antibodies to deliver cytotoxic payloads directly to cancer cells.3

Mechanism of Action of ADCs:

Antibody–drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic agents, potentially reducing the severity of side effects by preferentially targeting their payload to the tumour site.4 The mechanism of action underlying ADCs involves a interplay between three key components: the monoclonal antibody (mAb), the cytotoxic payload, and the linker molecule. This intricate system harnesses the specificity of monoclonal antibodies to selectively deliver potent cytotoxic agents directly to cancer cells while sparing healthy tissues, thereby minimizing systemic toxicity and maximizing therapeutic efficacy.4, 5

At the core of ADCs lies the monoclonal antibody, a protein engineered to recognize and bind with high affinity to specific antigens overexpressed on the surface of cancer cells. These antigens, often tumour-associated or tumour-specific, serve as molecular markers that distinguish malignant cells from their normal counterparts. Through advanced techniques such as phage display or hybridoma technology, researchers can generate monoclonal antibodies tailored to target a wide array of cancer antigens, including but not limited to HER2, CD20, and CD30. By exploiting the unique antigenic profile of cancer cells, ADCs achieve tumour selectivity, a critical feature that distinguishes them from conventional chemotherapeutic agents.3-6 Following antigen recognition and binding, the ADC internalizes into the cancer cell through receptor-mediated endocytosis, a process facilitated by the engagement of the antibody with its target antigen. Once inside the cell, the ADC undergoes intracellular trafficking, eventually reaching the lysosomal compartment. Within the lysosome, the acidic environment triggers the cleavage of the linker molecule, a chemical bridge that connects the antibody and the cytotoxic payload. This cleavage event, often mediated by enzymatic or chemical degradation, releases the active drug into the cytoplasm, where it exerts its cytotoxic effects.6

The cytotoxic payload, or the "warhead" of the ADC, represents a diverse array of potent anticancer agents with different mechanisms of action, including microtubule inhibitors, DNA-damaging agents, and DNA alkylating agents. These payloads are carefully selected based on their ability to induce cell death upon intracellular release while demonstrating minimal off-target effects in healthy tissues. Examples of commonly used cytotoxic payloads include auristatins, maytansinoids, and calicheamicins, each with unique mechanisms of action that exploit specific vulnerabilities within cancer cells.3-6

Upon release from the antibody, the cytotoxic payload exerts its pharmacological effects through various mechanisms tailored to disrupt essential cellular processes vital for cancer cell survival. For instance, microtubule inhibitors such as auristatins and maytansinoids disrupt mitotic spindle formation, leading to cell cycle arrest and eventual apoptosis. Similarly, DNA-damaging agents like calicheamicins induce DNA double-strand breaks, triggering apoptotic pathways and preventing cancer cell proliferation. By targeting fundamental cellular structures and processes critical for tumour growth and survival, ADCs deliver a potent cytotoxic payload with remarkable precision, effectively eradicating cancer cells while sparing normal tissues. The effectiveness of ADCs relies not only on the specificity of the monoclonal antibody and the potency of the cytotoxic payload but also on the stability and design of the linker molecule. The linker serves as a molecular bridge that connects the antibody and the cytotoxic agent, ensuring the controlled release of the payload within the target cell. Various linker designs have been developed to optimize drug release kinetics, stability in circulation, and compatibility with different cytotoxic payloads. Cleavable linkers, such as disulfide or peptide-based linkers, enable payload release in the reducing environment of the cytoplasm, whereas non-cleavable linkers, such as thioether or maleimide linkers, ensure payload stability during circulation and trafficking. The choice of linker design depends on factors such as the pharmacokinetic profile of the ADC, the intracellular trafficking route, and the desired release kinetics of the cytotoxic payload. In addition to their direct cytotoxic effects, ADCs can elicit secondary immune responses through various mechanisms, including antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Upon binding to cancer cells, the antibody component of the ADC may engage with Fcγ receptors expressed on immune effector cells, such as natural killer (NK) cells and macrophages, triggering the release of cytotoxic granules and promoting target cell lysis. Similarly, the binding of antibodies to cancer cells can activate the complement cascade, leading to the formation of membrane attack complexes that induce cell lysis. These immune-mediated mechanisms serve to augment the anticancer effects of ADCs, providing an additional layer of therapeutic benefit beyond direct cytotoxicity.3,5,6

Despite their remarkable promise, ADCs face several challenges that must be addressed to maximize their clinical utility. One such challenge is the heterogeneity of antigen expression within tumours, which can limit the efficacy of ADCs in patients with heterogeneous or rapidly evolving tumours. Strategies to overcome antigen heterogeneity include the development of multi-targeted ADCs capable of recognizing multiple antigens simultaneously or the combination of ADCs with other targeted therapies or immunotherapies to broaden their therapeutic scope. Furthermore, the development of resistance to ADCs, either through decreased antigen expression or intracellular drug resistance mechanisms, poses a significant obstacle to their long-term efficacy. Addressing these challenges requires a deeper understanding of the molecular mechanisms underlying ADC resistance and the development of strategies to circumvent or overcome resistance mechanisms.5

In conclusion, the mechanism of action of ADCs embodies a multifaceted approach that exploits the specificity of monoclonal antibodies, the potency of cytotoxic payloads, and the precision of linker chemistry to selectively deliver potent anticancer agents to tumour cells while minimizing systemic toxicity. By combining the targeting capabilities of antibodies with the cytotoxic potential of small-molecule drugs, ADCs represent a paradigm shift in cancer therapy, offering the promise of improved clinical outcomes and enhanced quality of life for patients with cancer. Continued research and innovation in the field of ADCs hold the potential to further expand their therapeutic utility and address the challenges that limit their widespread adoption in clinical practice.4,6

History and Evolution of ADC Technology:

The idea of targeted chemotherapy was conceptualized by a German scientist, Paul Ehrlich, over a century ago. His chemotherapy would target a cytotoxin to intended structures in unwanted cells but spare healthy tissues.7 The first ADC trials underway in the 1980s. The first approved ADC was gemtuzumab ozogamicin in 2000, it’s a CD33 antibody is conjugated to an antitumor antibiotic, calicheamicin.8 The development of Antibody-drug conjugates has undergone significant evolution since their inception, driven by advances in antibody engineering, linker chemistry, and payload selection. Early-generation ADCs faced challenges such as limited stability, off-target toxicity, and suboptimal potency, necessitating continuous refinement of design principles.9,10 Key milestones in ADC technology include the optimization of antibody formats to enhance tissue penetration and antigen binding affinity, the design of cleavable and non-cleavable linkers to achieve controlled payload release, and the identification of novel cytotoxic payloads with improved efficacy and tolerability profiles. Furthermore, the integration of conjugation technologies such as site-specific conjugation and engineered cysteine residues has facilitated the precise assembly of ADC components, ensuring reproducible drug-to-antibody ratios and homogeneous product quality.9-12

Clinical Applications:

The clinical success of ADCs has been demonstrated across a spectrum of cancer types, with several agents receiving regulatory approval for the treatment of hematologic malignancies and solid tumours. After decades of preclinical and clinical studies, a series of ADCs have been widely used for treating specific tumour. Notable examples include tbrentuximab vedotin (Adcetris®) for Hodgkin's lymphoma and systemic anaplastic large cell lymphoma, gemtuzumab ozogamicin (Mylotarg®) for acute myeloid leukemia, ado-trastuzumab emtansine (Kadcyla®) for HER2-positive metastatic breast cancer, inotuzumab ozogamicin (Besponsa®) and most recently polatuzumab vedotin-piiq (Polivy®) for B cell malignancies.13 These approvals underscore the therapeutic potential of ADCs in addressing unmet medical needs and improving patient outcomes. Ongoing clinical trials are evaluating ADCs in various settings, including combination therapy approaches, neoadjuvant/adjuvant settings, and rare cancer subtypes, with the aim of further expanding their therapeutic utility.14, 15

Challenges for Clinical Applications of ADCs:

Despite the considerable progress made in ADC development, several challenges remain to be addressed to fully realize their clinical potential. Up to now, more than 80 ADCs were examined in a wide variety of clinical trials. However, more than 55 ADCs clinical trials have been terminated.16 There are many challenges for the clinical applications of ADCs, some of the challenges are discussed here. One major challenge is the identification of suitable target antigens that are selectively expressed on cancer cells to minimize off-target toxicity. Additionally, optimizing the pharmacokinetic properties of ADCs to achieve adequate tumour penetration and payload delivery remains a critical consideration. Furthermore, resistance mechanisms such as antigen loss, heterogeneous antigen expression, and intratumoral heterogeneity pose significant obstacles to treatment durability and efficacy.17-20

Future Directions of Antibody Drug Conjugates

ADCs signify a speedily increasing field in cancer therapy. Several ADCs developed over the past decade. ADCs have created a huge variety of possibilities for designing new ADCs.21 Future directions in ADC research include the exploration of novel antibody formats, next-generation linker technologies, and innovative payload modalities to overcome these challenges and enhance therapeutic outcomes. Moreover, the integration of predictive biomarkers and patient stratification strategies holds promise for tailoring ADC therapy to individual tumour characteristics, thereby advancing the paradigm of precision medicine in oncology. Although there remain many complications to overcome, the development of new ADCs provides incredible opportunities for future cancer treatment.13, 17, 19

Conclusion:

The rise of ADCs represents a significant milestone in cancer therapy, offering the potential for precise tumour targeting and improved therapeutic outcomes. Through the convergence of innovative drug delivery systems and molecular targeting strategies, ADCs have transformed the treatment strategy, providing doctors with a powerful tool to combat cancer with improved efficacy and reduced toxicity.

Reference:

  1. Piña-Sánchez P, Chávez-González A, Ruiz-Tachiquín M, Vadillo E, Monroy-García A, Montesinos JJ, Grajales R, Gutiérrez de la Barrera M, Mayani H. Cancer Biology, Epidemiology, and Treatment in the 21st Century: Current Status and Future Challenges from a Biomedical Perspective. Cancer Control (2021) 28:10732748211038735 
  2. Advancing Cancer Therapy. Nat Cancer 2, 245–246 (2021)
  3. Drago JZ, Modi S, Chandarlapaty S. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol (2021) :327-344
  4. Dumontet C, Reichert JM, Senter PD, Antibody–drug conjugates come of age in oncology. Nat Rev Drug Discovery (2023) 22:641–661
  5. Joubert N, Beck A, Dumontet C, Denevault-Sabourin C, Antibody–drug conjugates: the last decade. Pharmaceuticals (2020)13: 245 
  6. Colombo R. & Rich JR. The therapeutic window of antibody drug conjugates: a dogma in need of revision. Cancer Cell (2022)40: 1255–1263 
  7. Strebhardt K, Ullrich A, Paul Ehrlich's magic bullet concept: 100 years of progress. Nat Rev Cancer (2008) 8:473-480
  8. Mythili S, Rise of Antibody-Drug Conjugates: The Present and Future. Am Soc Clin Oncol Educ Book 43 (2023)
  9. Mack F, Ritchie M, Sapra P. The next generation of antibody drug conjugates. Semin Oncol Epub (2014):637-52
  10. Fatima SW, Khare SK. Benefits and challenges of antibody drug conjugates as novel form of chemotherapy. J Control Release, Epub (2022) 341:555-565
  11. Beck A, Cutting-edge multi-level analytical and structural characterization of antibody–drug conjugates: present and future. Expert. Rev. Proteom. (2019)16: 337–362 
  12. Li M, Zhao X, Yu C, Wang L. Antibody-Drug Conjugate Overview: a State-of-the-art Manufacturing Process and Control Strategy. Pharm Res. (2024):419-440
  13. Zhao P, Zhang Y, Li W, Jeanty C, Xiang G, Dong Y. Recent advances of antibody drug conjugates for clinical applications. Acta Pharm Sin B (2020):1589-1600
  14. Tsuchikama K, An Z. Antibody–drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell (2018) 9:33–46
  15. Diamantis N, Banerji U. Antibody–drug conjugates—an emerging class of cancer treatment. Br J Cancer (2016) 114:362–367.
  16. Coats S, Williams M., Kebble B, Dixit R, Tseng L, Yao NS, Antibody drug conjugates: future directions in clinical and translational strategies to improve the therapeutic index. Clin Cancer Res. (2019) 25:5441–5448
  17. Bergstralh DT, Sekelsky J, Interstrand crosslink repair: can XPF-ERCC1 be let off the hook?. Trends Genet. (2008) 24:70–76
  18. Deans AJ, West SC, DNA interstrand crosslink repair and cancer. Nat Rev Cancer. (2011) 11:467–480
  19. Raja R, Kuziora M, Brohawn PZ, Higgs BW, Gupta A, Dennis PA, Early reduction in ctDNA predicts survival in patients with lung and bladder cancer treated with durvalumab. Clin Cancer Res. (2018) 24:6212–6222
  20. Emens L, Esteva F, Beresford M. Results from KATE2, a randomized phase 2 study of atezolizumab (atezo)+trastuzumab emtansine (T-DM1) vs placebo (pbo)+T-DM1 in previously treated HER2+ advanced breast cancer (BC) Cancer Res. (2018): 79 
  21. Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat Rev Drug Discov. (2017) 16:315–337

Kazi Habib
A seasoned professional with over 15 years of experience in the pharmaceutical field, brings a wealth of knowledge to the world of science

His journey spans across pharmaceuticals, Contract Development and Manufacturing Organizations (CDMOs), and biotechnology companies.

Beyond the corporate landscape, Kazi’s true passion lies in translating complex scientific concepts into accessible and engaging content. With a keen eye for detail and unwavering commitment to excellence, he crafts thought-provoking articles that bridge the gap between science and curious minds.

Join Kazi on an enriching journey through the captivating world of chemistry. Here, insightful analysis meets a fervor for sharing knowledge, creating a space where scientific exploration and engaging storytelling converge.