Bridging the gap between academic research and real-world solutions
In the pursuit of scientific advancement, the journey from theoretical research to tangible solutions is often fraught with challenges.

Written by
Kristoffer Danielsson
The Evolving Frontier of Antibody-Drug Conjugates
I. Executive Summary & Introduction: A Paradigm in Motion
The field of antibody-drug conjugates has transitioned from a promising therapeutic concept to a validated, cornerstone modality in modern oncology. After decades of incremental progress, the landscape is now characterized by an unprecedented acceleration in both clinical approvals and technological innovation. This dynamism is reflected in surging commercial and scientific interest, with 2024 alone witnessing a 33% increase in new ADC assets and a 23% rise in new clinical trials, underpinned by investment deals totaling $64.3 billion. The successful translation of late-stage assets into clinical practice was underscored by a series of regulatory approvals in early 2025 for candidates like datopotamab deruxtecan and telisotuzumab vedotin, which had been under review in late 2024.
This report will analyze this inflection point, arguing that the evolution of ADCs is advancing on two parallel and increasingly convergent fronts: enhancing targeting precision through multi-specific antibody engineering and expanding the therapeutic arsenal through novel payload modalities. The maturation of the field is evident in a dual-track investment strategy. On one hand, there is a strategic shift toward the creation of versatile, proprietary "platforms."
Companies are no longer just building single assets; they are engineering scalable, repeatable engines for generating next-generation candidates. This "platformization" of ADC development.
Exemplified by Daiichi Sankyo’s DXd technology, Sutro Biopharma’s XpressCF+, Biocytogen’s RenLite system, and Innovent Biologics' SoloTx and DuetTx platforms.
Is accelerating discovery and creating value through broad collaborations. On the other hand, the landscape is equally characterized by a surge in high value transactions centered on promising single assets, where large pharmaceutical companies acquire or license specific, de-risked candidates to rapidly bolster their oncology pipelines.
Major deals, such as Pfizer $43B acquisition of Seagen, AbbVie’s $10.1B acquisition of ImmunoGen for its approved drug Elahere, Johnson & Johnson's $2 billion purchase of Ambrx for its lead candidate ARX517, and Ipsen's $900 million deal for Sutro's STRO-003, underscore the immense value placed on individual, high potential ADCs.
This analysis will first detail the recent landmark approvals that are setting new standards of care in solid tumors. It will then dissect the emerging platforms poised to shape the future of cancer therapy, including bispecific antibody-drug conjugates designed for superior tumor selectivity and dual-payload ADCs engineered to overcome resistance. Finally, the report will explore the most forward-looking innovations that redefine the very nature of the ADC payload, shifting from purely cytotoxic agents to functional biologic tools such as immune-stimulating antibody conjugates and degrader-antibody conjugates.
II. New Standards of Care: Recent ADC Approvals in Solid Tumors
The first half of 2025 saw pivotal FDA approvals that have significantly expanded the role of ADCs, particularly in heavily pre-treated non-small cell lung cancer. These approvals validate new targets and solidify the position of ADCs as a critical therapeutic class for patients who have exhausted conventional options. This success demonstrates a clear and effective market strategy: carving out definitive niches in biomarker defined patient populations with high unmet needs. Rather than competing directly with first-line treatments, ADC developers are proving their value in the challenging setting of treatment resistance, establishing a new tier of therapy after the failure of both targeted agents and chemotherapy. This approach not only addresses a critical clinical gap but also builds a strong foundation of evidence from which to explore moving these potent therapies into earlier lines of treatment.
A. Datopotamab Deruxtecan (Datroway): The First TROP2-Directed Therapy in Lung Cancer
On June 23, 2025, AstraZeneca and Daiichi Sankyo secured accelerated FDA approval for Datroway , establishing it as the first TROP2-directed therapy for lung cancer. The approval covers adult patients with locally advanced or metastatic EGFR-mutated non-small cell lung cancer who have progressed following prior EGFR-directed therapy and platinum-based chemotherapy. This indication is for a patient population with limited options, whose disease has developed resistance to multiple lines of standard treatment.
The approval was based on compelling efficacy data from a pooled analysis of the Phase II TROPION-Lung05 and Phase III TROPION-Lung01 trials. In a cohort of 114 patients with EGFR-mutated NSCLC, Datroway demonstrated a confirmed objective response rate of 45% (95% CI: 35-54) as assessed by blinded independent central review. The responses were durable, with a median duration of response of 6.5 months (95% CI: 4.2-8.4). Datroway’s mechanism leverages Daiichi Sankyo’s proprietary DXd ADC platform, comprising a humanized anti-TROP2 monoclonal antibody covalently linked to a potent topoisomerase I inhibitor payload, deruxtecan.
This lung cancer approval followed a prior full approval on January 17, 2025, for unresectable or metastatic HR-positive, HER2-negative breast cancer. That decision was based on the TROPION-Breast01 trial, which showed that Datroway significantly improved progression-free survival compared to investigator's choice of chemotherapy (median PFS 6.9 vs. 4.9 months; HR 0.63). At the time of analysis, however, overall survival data were not mature and had not shown a statistically significant benefit.
B. Telisotuzumab Vedotin (Emrelis): A First-in-Class c-Met-Targeted ADC
Just a month prior, on May 14, 2025, AbbVie received accelerated FDA approval for Emrelis , a potential FIC ADC targeting the c-Met protein. The indication is for adult patients with locally advanced or metastatic, non-squamous NSCLC with high c-Met protein overexpression who have previously received systemic therapy.
This approval was critically dependent on a companion diagnostic. Patient eligibility is determined by the Roche VENTANA MET (SP44) RxDx Assay, which defines high c-Met overexpression as strong (3+) immunohistochemistry staining in at least 50% of tumor cells. This highlights the necessity of precise biomarker-driven patient selection for this therapeutic strategy.
The regulatory decision was supported by data from the Phase II LUMINOSITY study. In the pivotal cohort of 84 patients with high c-Met overexpression, Emrelis monotherapy achieved an ORR of 35% (95% CI: 24-46) and a median DoR of 7.2 months (95% CI: 4.2-12). Emrelis consists of a c-Met-directed antibody linked to the well-established cytotoxic payload monomethyl auristatin E. Its safety profile includes warnings for peripheral neuropathy, interstitial lung disease or pneumonitis, and ocular adverse reactions, requiring careful patient monitoring. Continued approval is contingent on the results of the ongoing Phase III confirmatory trial, TeliMET NSCLC-01.
Table 1: Comparative Summary of Key 2025 FDA Approvals for ADCs in NSCLC
Drug (Brand/Generic) | Target & Payload | Indication | Patient Selection Criteria | Key Trial(s) | Objective Response Rate | Duration of Response | Key Safety Signals | Manufacturer(s) |
Datroway (datopotamab deruxtecan) | TROP2 & Topoisomerase I Inhibitor (Deruxtecan) | Locally advanced or metastatic EGFR-mutated NSCLC, post-EGFR therapy and platinum-based chemotherapy | History of EGFR mutation; prior EGFR TKI and platinum chemotherapy | TROPION-Lung05, TROPION-Lung01 | 45% | 6.5 months | Interstitial Lung Disease/Pneumonitis, Stomatitis, Ocular Toxicity | AstraZeneca & Daiichi Sankyo |
Emrelis (telisotuzumab vedotin) | c-Met & Microtubule Inhibitor (MMAE) | Locally advanced or metastatic non-squamous NSCLC, post-prior systemic therapy | High c-Met protein overexpression (≥50% of cells with 3+ staining) via companion diagnostic | LUMINOSITY | 35% | 7.2 months | Peripheral Neuropathy, ILD/Pneumonitis, Ocular Toxicity, Infusion-Related Reactions | AbbVie |
III. The Next Wave: Engineering Multi-Specific and Multi-Payload Conjugates
Building on the success of monospecific ADCs, the next wave of innovation is focused on creating more sophisticated constructs designed to overcome the fundamental challenges of tumor heterogeneity and acquired drug resistance. Two dominant strategies are emerging from the pipeline: bispecific antibody-drug conjugates, which enhance targeting precision by engaging two antigens, and dual-payload ADCs, which aim to deliver a synergistic therapeutic punch by combining two distinct warheads.
A. Bispecific ADCs: Enhancing Specificity and Functionality
Bispecific ADCs represent a logical evolution of the ADC concept, leveraging a dual-targeting mechanism to achieve superior outcomes. By requiring the co-expression of two distinct tumor-associated antigens for optimal binding and internalization, BsADCs can significantly improve tumor specificity and reduce the on-target, off-tumor toxicity that can limit the therapeutic window of single-target ADCs. Furthermore, this approach can enable novel synergistic mechanisms by simultaneously modulating two different biological pathways.
Several companies are pioneering this approach with robust platforms:
Biocytogen has emerged as a key player with its RenLite common light chain mouse platform, which facilitates the efficient assembly of bispecific antibodies and has yielded a pipeline of over 20 BsADC assets. Notable candidates include a SEZ6 x B7H3 BsADC for small cell lung cancer and a PTK7 x TROP2 BsADC (BCG033) for triple-negative breast cancer, both of which have demonstrated superior preclinical efficacy compared to their monospecific counterparts. The value of this platform is further highlighted by a collaboration with SOTIO to develop next-generation ADCs for solid tumors.
Innovent Biologics is developing a diverse pipeline of BsADCs with innovative target combinations. Its EGFR/B7H3 BsADC (IBI3001) targets two well-validated cancer antigens. Perhaps more revolutionary is its TROP2/PD-L1 BsADC (IBI3014), which aims to integrate direct, payload-mediated tumor killing with immune checkpoint blockade in a single molecule. This design could simultaneously debulk the tumor and activate an anti-tumor immune response, potentially overcoming resistance to both chemotherapy and immunotherapy.
Alphamab Oncology is also exploring this integrated immuno-oncology approach with JSKN022, a PD-L1 x ITGB6/8 BsADC. This molecule is designed to block the PD-1/PD-L1 immune checkpoint while also delivering a cytotoxic payload to cells expressing integrins associated with metastasis, offering a novel strategy for treating tumors refractory to standard IO therapies.
B. Dual-Payload ADCs: A Strategy of Synergy and Resistance Evasion
A parallel strategy to enhance ADC efficacy involves delivering two distinct payloads to the same cancer cell. This approach is designed to produce synergistic cell killing by targeting multiple, non-overlapping cellular pathways, which can be particularly effective against heterogeneous tumors. By employing two different mechanisms of action, dual-payload ADCs can also pre-emptively address acquired resistance to a single cytotoxic agent, a common mechanism of treatment failure.
The technical challenges of conjugating two different drugs to a single antibody are significant, but several companies are making progress:
Sutro Biopharma has strategically prioritized its dual-payload ADC program, leveraging its XpressCF+ cell-free protein synthesis platform. This technology allows for the precise, site-specific incorporation of non-natural amino acids, enabling the creation of homogeneous ADCs with defined drug-to-antibody ratios for two different warheads. The company plans to file an IND for its first wholly-owned dual-payload ADC in 2027 and is actively collaborating with Astellas on dual-payload immunostimulatory ADCs. Preclinical data presented at AACR 2025 showcased combinations of a topoisomerase I inhibitor with either a microtubule inhibitor or a PARP inhibitor.
Innovent Biologics has established a dedicated dual-payload ADC platform, DuetTx, and has already advanced the world’s first dual-payload ADC into clinical trials: IBI3020, which targets CEACAM5.
Chengdu Kanghong is on the verge of a major milestone with KH815, a TROP2-targeting dual-payload ADC that is expected to be the first such molecule to enter human trials.
The most advanced innovators in the field are beginning to merge these two next-generation strategies. Alphamab Oncology's JSKN021 is a prime example of this convergence. It is not only a bispecific ADC targeting both EGFR and HER3 but also a dual-payload ADC carrying a novel topoisomerase I inhibitor (T01) and the microtubule inhibitor MMAE. This "bispecific dual-payload ADC" represents the current apex of ADC engineering, aiming to solve the problem of tumor heterogeneity with dual targeting while simultaneously tackling drug resistance with two distinct cytotoxic mechanisms. Such molecules, while exceptionally complex to design and manufacture, embody the field's ambition to create the ultimate precision therapeutic for the most challenging cancers.
Table 2: Selected Next-Generation Conjugates in the Pipeline
Candidate | Company | Modality | Target(s) | Payload(s) | Rationale / Key Innovation | Development Stage |
JSKN021 | Alphamab Oncology | BsADC & Dual-Payload | EGFR & HER3 | Topo I Inhibitor (T01) & MMAE | Combines dual targeting to overcome heterogeneity with dual payloads to prevent resistance. | Preclinical |
IBI3014 | Innovent Biologics | BsADC | TROP2 & PD-L1 | Topo I Inhibitor | Integrates direct tumor killing (TROP2) with immune checkpoint blockade (PD-L1) in one molecule. | Preclinical |
BCG033 | Biocytogen | BsADC | PTK7 & TROP2 | Undisclosed | Dual targeting to improve tumor specificity and efficacy in TNBC compared to single-target ADCs. | Preclinical |
IBI3020 | Innovent Biologics | Dual-Payload | CEACAM5 | Undisclosed | First dual-payload ADC to enter clinical trials; designed for high efficacy in resistant tumors. | Clinical (Phase I) |
STRO-00X/Y | Sutro Biopharma | Dual-Payload | HER2 | Topo I Inhibitor & PARP Inhibitor | Leverages XpressCF+ platform for precise, site-specific conjugation of two synergistic payloads. | Preclinical |
KH815 | Chengdu Kanghong | Dual-Payload | TROP2 | RNA Polymerase Inhibitor & Topo I Inhibitor | Poised to be the first dual-payload ADC to enter human trials. | Clinical (Phase I) |
IV. Redefining the "Warhead": The Emergence of Novel Payload Modalities
While innovations in antibody engineering are enhancing targeting precision, a concurrent revolution is occurring at the other end of the molecule: the payload. The field is undergoing a fundamental philosophical shift, moving beyond a singular focus on direct cytotoxicity. The payload is evolving from a simple "warhead" designed to kill a cell into a sophisticated "biologic tool" engineered to manipulate cellular processes. This conceptual leap is giving rise to entirely new classes of antibody-conjugates that can reprogram the tumor microenvironment or hijack cellular machinery, dramatically expanding the therapeutic potential of the ADC platform into immunomodulation and targeted protein degradation.
A conceptual diagram illustrating this architectural evolution would begin with a simple monoclonal antibody. This evolves into a traditional ADC, depicted as an antibody with a single type of cytotoxic payload attached. The structure then advances into more complex forms: the Bispecific ADC, which targets two antigens, and the Dual-Payload ADC, which carries two different payloads. From this advanced stage, the diagram branches further to showcase novel payload classes, including Immune-Stimulating Antibody Conjugates and Degrader-Antibody Conjugates, representing the shift from cytotoxic warheads to functional biologic tools.
A. Immune-Stimulating Antibody Conjugates: Converting "Cold" Tumors to "Hot"
Immune-stimulating antibody conjugates represent a paradigm shift in which the cytotoxic payload is replaced with an immune agonist, such as a Toll-like receptor 7/8 agonist. The primary goal is not to kill the tumor cell directly, but to use the antibody to deliver the immune stimulant precisely to the tumor microenvironment. Once there, the agonist activates innate immune cells like macrophages and dendritic cells, triggering a localized inflammatory response. This process can prime a robust, T-cell-mediated adaptive immune response, effectively converting immunologically "cold," non-responsive tumors into "hot" tumors that are susceptible to immunotherapy, including checkpoint inhibitors.
This targeted approach is crucial because systemic administration of potent immune agonists is often limited by severe toxicities, including cytokine release syndrome. Despite their promise, ISACs are in early clinical development and face significant hurdles. A key challenge is achieving the optimal balance between potent immune activation and manageable safety. Early clinical results for candidates like Bolt Biotherapeutics' BDC-1001, a HER2-targeting ISAC, have been mixed, showing limited response rates and highlighting the difficulty in optimizing the dose, linker stability, and payload release kinetics required for success.
B. Degrader-Antibody Conjugates: Targeting the "Undruggable" Proteome
Degrader-antibody conjugates are another revolutionary modality that conjugates an antibody to a targeted protein degrader, such as a proteolysis-targeting chimera or a molecular glue. Instead of merely inhibiting a target protein, a DAC delivers a payload that hijacks the cell's own ubiquitin-proteasome system to tag the protein for destruction.
This mechanism offers several distinct advantages. First, it can eliminate the entire target protein, providing a more complete and durable effect than transient inhibition. Second, the degrader payload acts catalytically, meaning a single molecule can induce the destruction of multiple target protein molecules, leading to profound efficacy even at low concentrations. Most importantly, this approach can target proteins that lack traditional enzymatic active sites, such as scaffold proteins and transcription factors.
Which have long been considered "undruggable" by conventional small molecules.
The DAC field is nascent, with no FDA-approved therapies to date. However, the first candidates are now entering the clinic, generating significant industry excitement. Orum Therapeutics' ORM-6151, a CD33-targeted DAC designed to degrade the GSPT1 protein in acute myeloid leukemia, was acquired by Bristol Myers Squibb in a major deal and is currently in a Phase I trial. This move by a major pharmaceutical player validates the high potential of the DAC platform.
C. An Expanding Arsenal of Novel Cytotoxics
Alongside the development of non-cytotoxic payloads, researchers are also expanding the library of traditional warheads to overcome resistance to standard agents like auristatins and maytansinoids. The clinical success of the topoisomerase I inhibitor deruxtecan in ADCs like Enhertu and Datroway has spurred intense interest in this class of payload. Other emerging cytotoxic mechanisms include:
Translation Inhibitors: Hexagon Bio is developing HB-06510, a natural product-derived protein translation inhibitor, as a novel ADC payload for multiple myeloma. This payload has demonstrated potent cytotoxicity even in multi-drug resistant cell lines.
Ultra-Potent Toxins: Extremely potent natural compounds like the enediyne Lidamycin, which is orders of magnitude more toxic than conventional agents, are being explored as payloads. Their extreme potency allows for effective cell killing even with very low levels of drug delivery to the tumor.
Table 3: Overview of Emerging Payload Technologies
Modality | Mechanism of Action | Key Advantage | Primary Challenge | Lead Candidate/Company |
Immune-Stimulating Conjugate | Delivers an immune agonist (e.g., a Toll-like receptor 7/8 agonist) to the tumor microenvironment to activate innate and adaptive immunity. | Can convert immunologically "cold" tumors to "hot," potentially overcoming resistance to immunotherapy. | Balancing potent immune activation with systemic toxicity (e.g., cytokine release syndrome). | BDC-1001 (Bolt Biotherapeutics) |
Degrader-Antibody Conjugate | Delivers a protein degrader (e.g., a proteolysis-targeting chimera) to hijack the cell's proteasome system and destroy a target protein. | Can catalytically eliminate "undruggable" targets and may provide a more durable response than inhibitors. | Complex chemistry, optimizing intracellular release and degrader activity, managing on-target toxicities. | ORM-6151 (Orum Therapeutics / Bristol Myers Squibb) |
Novel Cytotoxic | Employs warheads with new mechanisms of action, such as translation inhibition or ultra-potent DNA damage. | Can overcome acquired resistance to standard ADC payloads like microtubule or topoisomerase inhibitors. | Identifying payloads with favorable properties (potency, stability, conjugation chemistry) and manageable toxicity. | HB-06510 (Hexagon Bio) |
V. Synthesis and Future Outlook
The antibody-drug conjugate field is in a period of unprecedented expansion and innovation, solidifying its role as a central pillar of precision oncology. The developments of 2024 and 2025 have not only expanded the clinical utility of ADCs but have also laid the groundwork for a new generation of therapeutics with fundamentally enhanced capabilities.
The key trends shaping the landscape are clear. First, clinical validation and expansion are proceeding at a rapid pace. Recent approvals have firmly established ADCs as a standard of care in biomarker-selected, heavily pre-treated patient populations, and ambitious clinical trial programs are now underway to move these effective agents into earlier lines of therapy. Second, the pipeline is defined by a rapid progression toward
increasing structural complexity. The development of bispecific and dual-payload ADCs is a direct response to the challenges of tumor heterogeneity and drug resistance, with innovators combining multiple targeting and payload strategies into single, highly engineered molecules. Third, a profound
payload diversification is underway. The conceptual shift from cytotoxic "warheads" to "biologic tools" like immune agonists and protein degraders is unlocking the potential of the ADC platform to modulate complex biology in ways previously unimaginable.
Looking forward, several themes will dominate the next phase of ADC development. The synergy between ADCs and immunotherapy is a particularly powerful emerging strategy. This is being explored both through combination trials of separate agents, such as the promising results from the ASCENT-04 study of sacituzumab govitecan plus pembrolizumab , and through integrated single-molecule approaches like Innovent’s TROP2/PD-L1 BsADC. As these more complex molecules advance, the challenge of
managing novel toxicities will become paramount. The interstitial lung disease associated with certain topoisomerase I inhibitor payloads serves as a critical reminder that new mechanisms bring new safety considerations that will require proactive monitoring and management strategies. Finally, the ADC landscape is witnessing a notable geopolitical shift in innovation. A significant portion of the most advanced work in bispecific, dual-payload, and degrader conjugates is being driven by highly innovative companies in China and South Korea, signaling a more globally distributed R&D ecosystem.
In conclusion, the ADC field is realizing its long-held promise. The convergence of multi-specific targeting, multi-payload delivery, and novel biological mechanisms is creating a new generation of therapeutics with the potential to transform outcomes for patients with cancer and, eventually, other complex diseases. The primary hurdles ahead lie in navigating the manufacturing and clinical development complexity of these sophisticated molecules, understanding and mitigating their unique safety profiles, and continuing to refine patient selection to ensure the right drug reaches the right patient. The remarkable pace of progress suggests that the "magic bullet" concept, once a distant aspiration, is rapidly evolving into a clinical reality.
——————
Author: Kristoffer Danielsson
Founder & CEO
Jarlen Capital
Email: Kristoffer@jarlencapital.com