Abstract
Although antibody–drug conjugates (ADCs) revolutionized cancer treatment, especially in breast cancer, resistance remains a major challenge. It may involve target expression and distribution, linker stability, ADC intratumor penetration and intracellular internalization, payload activity but also interaction with the tumor microenvironment. Strategies to overcome resistance under investigation include combination therapies, novel payloads, and next-generation ADC designs. The identification of predictive biomarkers may also help guide treatment selection and prevent resistance.
Subject terms: Biomarkers, Cancer, Drug discovery, Oncology
Introduction
Antibody drug conjugates (ADCs) have revolutionized tumor treatment, but resistance often limits long-term efficacy1. Available data on the full spectrum of resistance mechanisms remain limited, partly due ADCs structural and mechanistic complexity, which enables a wide range of potential resistance pathways. Furthermore, multiple resistance mechanisms can arise simultaneously and interact synergistically. Despite growing research efforts, effective strategies to overcome it are still under investigation.
ADC structure and main mechanisms of action
ADCs present three key components: a humanized monoclonal antibody (mAb) targeting a tumor antigen, a cytotoxic payload, and a linker connecting them2. Their key mechanism of action begins with antigen binding, followed by internalization and payload release intracellularly1. Beyond direct cytotoxicity on the antigen-expressing cells, ADCs could trigger immune responses and exert a “bystander effect”, through payload diffusion to the neighboring antigen-negative cells, particularly relevant in tumors with heterogeneous antigen expression3. Following intravenous administration, ADCs must remain stable in circulation. IgG1 antibodies are commonly used for their long half-life (~21 days), manufacturing ease, and immune effector functions4. The antigen-binding fragments (Fabs) region ensures antigen recognition, while the constant region (Fc) engage immune responses such as complement activation, cytotoxicity, and antibody-dependent cell-mediated cytotoxicity (ADCC)5. Fc modifications can modulate half-life, though may compromise antibody-dependent functions1,6,7. Linkers are cleavable or non-cleavable: the former respond to intracellular cues (e.g., proteases, acidic pH) but may risk premature payload release; the latter require full internalization and lysosomal degradation, enhancing stability7,8.
Payloads currently fall into two main categories: microtubule inhibitors and DNA-damaging agents like topoisomerase I inhibitors (TOP1i)5. The drug-to-antibody ratio (DAR) reflects the number of cytotoxic molecules per antibody, influencing pharmacokinetics, safety, and activity profile5.
Several ADCs are approved for breast cancer (BC) across different countries: trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), sacituzumab govitecan (SG), datopotamab deruxtecan (Dato-DXd), and sacituzumab tirumotecan (Sac-TMT)9,10. T-DM1 and T-DXd share the human epidermal growth factor 2 (HER2)-targeting antibody but differ in payloads, linkers, and DARs. Dato-DXd, SG, and Sac-TMT use TOP1i and target trophoblast cell surface antigen 2 (TROP2), but differ in all components8,10,11.
Possible mechanisms of resistance to ADCs
ADC stability and penetration
ADC stability in circulation is critical for efficacy and safety, depending on both antibody choice and stable linker to prevent premature payload release, which can cause off-target toxicity and reduce tumor delivery, compromising selectivity and therapeutic benefit8,12. Non-cleavable linkers offer greater plasma stability and therapeutic index, though newer cleavable ones also show enhanced stability12.
Intratumoral distribution is also a critical factor in ADC efficacy13 (Fig. 1a). In solid tumors, poor penetration due to abnormal vasculature, hypoxia, elevated interstitial pressure, and large ADC size is an early resistance mechanism14–16. Additional factors like high antigen expression, rapid internalization and binding site barrier (BSB), caused by antibody binding near vasculature, also impair ADC delivery1,17.
Fig. 1. Mechanisms of resistance to ADCs.
a Poor intratumor penetration: physical barriers, blood site barrier (BSB), target-mediated drug disposition (TMDD). b Target-related resistance: reduced antigen expression and heterogeneity in antigen expression. c Altered Internalization and Trafficking: heterodimerization with human epidermal growth factor 3 (HER3) or epidermal growth factor receptor (EGFR), and lysosomal alkalinization. d Payload Resistance: overexpression of drug-efflux pumps, release via extracellular vesicles (EVs), drug target mutations. e Bypass mechanisms related to payloads: activation of alternative survival pathways involving Signal Transducer and Activator of Transcription 3 (STAT 3), Leukemia Inhibitory Factor Receptor (LIFR), Heat Shock Protein Beta-1 (HSPB1), circular RNA (crRNA), Polo-Like Kinase 1(PLK1), Cyclin B1. f Tumor microenvironment (TME): increased cancer-associated fibroblasts (CAFs), CD8 + T cells and Natural Killer (NK) cells, and enrichment of cancer stem cells (CSCs).
Some preliminary clinical evidence also pointed out the relevance of the ADC intratumoral distribution18.
In the ICARUS-BREAST01 trial with patritumab deruxtecan (HER3-DXd), exploratory analysis from on-treatment tumor biopsies suggested that the intratumoral distribution of the ADC may influence response, highlighting the need to optimize ADC delivery19.
Reduction of target expression and localization
Target expression and localization play a key role in enabling adequate ADC tumor distribution through binding to tumor cells5,20 and therefore in determining ADC resistance (Fig. 1b).
Regarding target expression, although T-DM1 is ineffective in HER2-negative or HER2-low BC, newer-generation ADCs like T-DXd have shown activity in tumors with low HER2 expression21,22, likely due to the bystander effect observed with T-DXd but not with T-DM123.
However, the benefit of T-DXd remains associated with HER2 expression levels, as shown in the phase 2 DAISY trial, evaluating this ADC in mBC, which reported objective response rates (ORRs) of 70.6% (95% Confidence Interval [CI], 58.3–81.0) in HER2-overexpressing, 37.5% (95% CI, 26.4–49.7) in HER2-low, and 29.7% (95% CI, 15.9–47.0) in HER2-negative disease, indicating a direct correlation between HER2 expression and efficacy24.
Exploratory biomarker analyses from the ASCENT and TROPiCS-02 phase 3 trials suggested a trend toward improved ORR and median progression-free survival (mPFS) with higher TROP2 expression. In ASCENT (pretreated metastatic triple-negative [mTN]BC), SG showed longer PFS (6.9, 5.6, and 2.7 months, 95% CI) and higher ORR (44%, 38%, and 22%) in patients with high, medium, and low TROP2 expression, respectively, though benefit was observed across all subgroups25.
Similarly, in the TROPiCS-02 trial (pretreated, endocrine-resistant [Hormone receptors] HR-positive/HER2-negative mBC), a post hoc analysis showed improved outcomes with SG versus control regardless of TROP2 expression (histochemical score [H-score]; range, 0–300), with a trend toward longer mPFS in patients with H-score ≥100 vs <100 (6.4 vs 5.3, 95% CI)26.
ADCs can also induce resistance by downregulating antigen expression and promoting tumor cell tolerance27,28. In vitro models of Dato-DXd resistance revealed marked TROP2 downregulation29. Also in clinical settings, TROP2 mutations, like T256R, were associated with impaired membrane localization and drug binding, and ultimately with acquired resistance to SG30. Similarly, the phase 1 U31402-A-U102 trial, evaluating HER3-DXd in non-small cellular lung cancer (NSCLC), identified acquired mutations in the gene codifying for human epidermal growth factor 3 (HER3); leading to reduced HER3 expression and decreased drug efficacy31. In the DAISY trial, 65% of patients (95% CI, 40.8–84.6) showed decreased HER2 expression upon resistance24.
Furthermore, the spatial distribution of the target within the tumor also plays a critical role in shaping the response to therapy, as it can affect the interactions of the ADC with the tumor cells and therefore the drug accessibility. In the DAISY study, non-responder tumors in the HER2-positive cohort exhibited a higher proportion of cluster 6, characterized by low HER2 staining and prevalence of stromal component24.
Similarly, HER2 heterogeneity was linked to resistance to T-DM1 plus pertuzumab and lower pathologic complete response (pCR) rates (55% vs 0%, P < 0.0001)32. Likewise, the KRISTINE trial showed lower pCR in tumors with focal or variable HER2-overexpression, while high HER2 clustering predicted better trastuzumab response13,33.
Altered internalization and intracellular trafficking pathways
Once bound to their target, ADCs promote antigen clustering and internalization, mainly via clathrin-mediated endocytosis, though alternative pathways exist15,22,34,35. Subsequently, the ADC–antigen complex is trafficked through lysosomes, where acidic pH and proteases release the payload22,34,36 (Fig. 1c).
Resistance to T-DM1 may emerge through multiple interconnected internalization-related mechanisms in preclinical data: altered lysosomal trafficking, increased lysosomal pH, and sequestration in Caveolin-1-positive compartments with a neutral pH37–39. Loss of Solute Carrier Family 46 Member A3 (SLC46A3, a lysosomal transporter protein), further impairs payload cytoplasmic release40.
Preclinical studies described that epidermal growth factor receptor (EGFR) can form heterodimers with HER2, altering trafficking and reducing T-DXd internalization, while HER3/HER2 dimerization seemed to affect trastuzumab efficacy35. In addition, in the EMILIA trial high EGFR expression correlated with poorer outcomes with T-DM141.
Heregulin-induced HER2/HER3/HER4 dimerization also showed to reduce T-DM1 cytotoxicity in both preclinical and early-phase clinical studies42,43.
Target internalization capability can also exert a key role; prior preclinical data pointed out that TROP2 cellular localization and endocytosis may vary across different tumor types44,45.
This is in line with the recent findings of an exploratory analysis of TROPION-Lung01 trial, showing that patients with TROP2 QCS-positive tumors (reflecting higher TROP2 internalization) had improved outcomes with Dato-DXd versus docetaxel (mPFS: 6.9 vs 4.1 months; Hazard Ratio [HR] 0.57, 95% CI, 0.41–0.79)46.
Payload-associated resistance
Several resistance mechanisms to ADC payloads have been described, involving both reduced intracellular drug concentration and impaired payload activity.
Efflux transporter upregulation
Many payload-related mechanisms of resistance mirror classic chemotherapy resistance, such as the upregulation of ATP-binding cassette (ABC) transporters47,48 (Fig. 1d). Preclinical studies have linked Multi-Drug Resistance 1 (MDR1)-mediated efflux to T-DM1 resistance28,49.
One potential strategy to overcome this type of resistance involves the use of ADCs less susceptible to efflux pumps: in HER2-positive resistant mouse models, disitamab vedotin (DV) outperformed T-DM1 and T-DXd, suggesting a role for efflux pumps, although mechanisms remain unclear50. This resistance mechanism was also observed clinically: ABCC1 expression in post-T-DXd samples correlated with poorer T-DXd-associated overall survival (OS)51.
Payload target mutations
Mutations in genes involved in payload activity may also determine ADC resistance (Fig. 1d). Retinoblastoma 1 (RB1) mutations have been associated with T-DXd resistance in TP53-/HER2-mutated NSCLC52.
With respect to BC data, in the DAISY trial, SLX4 mutations, a gene encoding for a DNA repair protein that might have a role in resistance to TOP1i, were more frequent at progression to T-DXd than at baseline, and SLX4 knockdown in cell lines induced resistance24. TOP1 mutations (e.g., E418K and frameshift) were detected in tissue samples after SG progression30. Abelman et al. reported TOP1 mutations (E418K, R364H, W401C) in circulating tumor DNA (ctDNA) of patients progressing on a TOP1i-based ADC, correlating with cross-resistance and shorter duration on a second TOP1i-ADC (52 vs 455 days)53.
Extracellular vesicles (EVs) mediated resistance
Extracellular vesicles (EVs) also influence ADC response in preclinical models (Fig. 1d). HER2-positive EVs can enhance the bystander effect by delivering payloads to neighboring cells, but may also promote resistance by acting as drug decoys54,55.
Bypass mechanisms related to payloads
A possible alternative mechanism of resistance to the payload is the activation of compensatory survival pathways (Fig. 1e). In particular, resistance to T-DM1 has been associated in preclinical studies with dysregulation of cell cycle and apoptotic signaling: loss of cyclin B1 and downregulation of Polo-Like Kinase 1 (PLK1) impair mitotic arrest, reducing T-DM1 efficacy. Notably, restoring their activity partially re-sensitizes resistant cells56,57. Moreover, overexpression of the Leukemia inhibitory factor receptor (LIFR) activates signal transducer and activator of transcription 3 (STAT3) signaling and increases anti-apoptotic proteins, further promoting resistance to T-DM158. T-DM1-resistant cell lines have also shown alterations in adhesion and prostaglandin-related genes, potentially affecting microtubule binding and supporting payload-specific resistance mechanisms59.
Regarding data on T-DXd, studies in resistant models have shown that a VDAC3-derived circular RNA (crRNA) inhibits Heat Shock Protein (HSP) Beta-1 ubiquitination, thereby contributing to resistance60.
ADC cross-resistance
Finally, also real-world data indirectly showed resistance pattern linked to the payload, by reporting limited PFS benefit when ADCs with the same payload class (e.g., TOP1i) are used sequentially53,61–65.
Conversely, sequencing HER2-targeted ADCs with different payloads appears more effective. In the phase 3 DESTINY-Breast02 trial, T-DXd significantly improved mPFS versus physician’s choice in HER2-positive mBC patients after T-DM1 (17.8 vs 6.9 months; HR 0.36, 95% CI, P < 0.0001)66. However, we cannot exclude that these results are likely to be influenced also by other key differences between the two ADCs, such as the linker characteristics, the DAR, and the bystander effect. Interestingly, a recent post hoc exploratory analysis of DESTINY-Breast02 and DESTINY-Breast03 (phase 3 trial comparing T-DXd with T-DM1 in pretreated HER2-positive mBC) suggests that T-DM1 can be an effective treatment option after T-DXd, showing a median treatment duration of 7.6 months (range 0.0–42.6) in patients receiving T-DM1 after progression on T-DXd67. These findings support payload diversification as a strategy to overcome resistance24,49. Several phase 2 trials are ongoing to further explore these approaches (Table 1).
Table 1.
Ongoing clinical trials evaluating sequencing strategies after prior ADC exposure in BC
| Clinical trial (name/NCT ID) | Phase | Type of sequencing | Setting |
|---|---|---|---|
| SATEEN/NCT06100874 | 2 | SG + Trastuzumab after PD on T-DXd | HER2+ mBC |
| TRADE-DXd/NCT06533826 | 2 | Dato-DXd or T-DXd after PD on prior ADC | HER2- (HER2-low or HER2-0) mBC |
| SERIES/NCT06263543 | 2 | SG after PD on T-DXd | HR+/HER2-low mBC |
| ACE-Breast-03/NCT04829604 | 2 | ARX788 after PD on T-DXd | HER2+ mBC |
| ICARUSBREAST02/NCT06298084 | 1b/2 | HER3-DXd + Olaparib after PD on T-DXd | HER2-low mBC |
ADC antibody–drug conjugate, mBC metastatic breast cancer, TNBC triple-negative breast cancer, HER2 human epidermal growth factor receptor 2, + positive, - negative, HR hormone receptor, SG sacituzumab govitecan, Dato-DXd datopotamab deruxtecan, T-DXd trastuzumab deruxtecan, HER3-DXd patritumab deruxtecan, PD progression disease.
This is a non-exhaustive list intended to illustrate examples.
Role of the tumor microenvironment (TME)
Beyond intracellular mechanisms, the TME plays a crucial role in ADC response and resistance (Fig. 1f). The TME can affect the ADC intratumoral distribution and intervene in the ADC degradation and payload release. On the one side, cancer-associated fibroblasts (CAFs) hinder ADC penetration by producing extracellular matrix proteins, and releasing transforming growth factor β, and also induce an immunosuppressive TME. On the other hand, recent preclinical data indicate that extracellular proteases, like cathepsin L (CTSL), in the TME contribute to T-DXd efficacy in HER2-low and HER2-negative BC, independently of HER2 expression or ADC internalization. High CTSL expression in tumor and stromal compartments enables efficient linker cleavage and payload release, promoting cytotoxicity68.
The immune cells present in the TME may also have a major role in determining the ADC activity. Among the various forms of regulated cell death, immunogenic cell death (ICD) has emerged as a key process in promoting tumor antigen-specific immunity and enhancing responses to anticancer therapies. Comparative studies indicate that the DXd payload induces stronger hallmarks of ICD than DM1, including higher HMGB1 release and increased calreticulin surface exposure on dying tumor cells, suggesting a more potent capacity to stimulate antitumor immune activation68. Consistently, RNA-seq analysis of co-cultured macrophages reveals marked upregulation of antigen-presentation and chemokine genes with both HER2 ADCs, with T-DXd demonstrating more potent immune-stimulatory effects than T-DM1. In HER2-positive BC cell lines, in addition, both T-DXd and T-DM1 show the ability to promote antibody-dependent cellular phagocytosis (ADCP) via Fcγ-receptor engagement. Notably, T-DXd induces markedly stronger immune activation than T-DM1 or trastuzumab, with enhanced T-cell proliferation and CD44 expression68. However, it also upregulates tumor CD47, limiting immune activation. Combining T-DXd with CD47 blockade enhances T-DXd efficacy and induces durable CD8 + T cell memory68.
Other prior preclinical studies consistently showed that ADCs reduce TME immunosuppression, activate dendritic cells (DC) and CD8 + T cells, and upregulate Programmed Death-Ligand 1 (PD-L1) and Major Histocompatibility Complex class I (MHC-I) expression on tumor cells69–71.
Emerging clinical trials are starting to validate some of this evidence. In the ICARUS-BREAST01 clinical trial, response to HER3-DXd was associated with upregulation of immune-related pathways such as interferon-alpha and gamma pathways19.
Finally, ADCs can also modulate cancer stem cells (CSCs), known for their chemo-resistance and stem-like properties, by promoting receptor tyrosine kinase-like orphan receptor 1(ROR1) signaling72–74. In BC models, T-DM1 increases CSCs via Yes-associated protein 1 (YAP1)-mediated ROR1 upregulation75. Therefore, co-inhibition of YAP1 and ROR1 with HER2-targeted therapies has shown promise in preclinical studies20,75.
Possible strategies to overcome ADC resistance
ADC combination
Combining ADCs with other therapies may overcome resistance and enhance efficacy, especially in heterogeneous tumors. However, careful patient selection and clinical context are crucial to balance benefits with the risk of increased adverse events (AEs) (Fig. 2a). Ongoing clinical trials are listed in Table 2.
Fig. 2. Strategies to overcome ADC resistance.
a Combination therapies. ADCs combined with immune checkpoint inhibitors (ICIs), DNA-damage response inhibitors (DDRinh), tyrosine kinase inhibitors (TKIs), other ADCs, antiangiogenic antibodies (antiVEGF Abs), Heat Shock Protein (HSP)90 inhibitors, anti-trastuzumab Abs, Signal Transducer and Activator of Transcription 3 (STAT 3) inhibitors, Phosphoinositide 3-Kinase (PI3K) and anti-epidermal growth factor Receptor (EGFR) Abs. b New-generation payloads: use of next-generation cytotoxic agents, degrader–antibody conjugates (DACs), glue–antibody conjugates (MACs), immune stimulator antibody conjugates (ISACs), antibody–oligonucleotide conjugates (AOCs), and ADCs with multiple payloads. c Innovative ADC structures: small-format drug conjugates, bispecific ADCs (BsADCs), probody–drug conjugates (PDCs), enhanced bystander effect, innovative linker technologies, and conjugation strategies.
Table 2.
Ongoing clinical trials evaluating ADCs in combination strategies for breast cancer
| Clinical trial identifier | Phase | Investigational combination | Setting | Study name |
|---|---|---|---|---|
| ADC + ICI | ||||
| NCT06508216 | 1b/2 | Dato-DXd + durvalumab | Early-relapsing mTNBC | COMPASS-TNBC |
| NCT03742102 | 1b/2 | Dato-DXd + durvalumab | PD-L1 + LA/mTNBC | BEGONIA (Arm 8) |
| NCT05353361 | 1b/2 | SHR-A1811 + adebrelimab (or pyrotinib or pertuzumab) | LA/mTNBC | — |
| NCT01042379 | 2 | Neoadjuvant Dato-DXd + durvalumab | HER2-/Immune-positive early BC | I-SPY2.2 |
| NCT05629585 | 3 | Adjuvant Dato-DXd ± durvalumab | TNBC with residual disease post-NAST | TROPION-Breast03 |
| NCT04873362 | 3 | Adjuvant T-DM1 + atezolizumab | Early HER2 + BC with residual disease | ASTEFANIA |
| NCT05633654 | 3 | Adjuvant SG + pembrolizumab | TNBC with residual disease post-NAST | ASCENT-05/ OptimICE-RD |
| NCT04042701 | 3 | Dato-DXd ± durvalumab first-line | PD-L1 + LA/mTNBC | TROPION-Breast05 |
| NCT06112379 | 3 | Neoadjuvant Dato-DXd + Durva (Adj. Durva) | Early-stage TNBC or HR-low/HER2- BC | TROPION-Breast04 |
| NCT04740918 | 3 | T-DM1 + atezolizumab | HER2+ and PD-L1 + LA/mBC | KATE 3 |
| NCT06312176 | 3 | Sacituzumab tirumotecan± pembrolizumab | HR + / HER2- LA/mBC | TroFuse-010 |
| NCT06841354 | 3 | Sacituzumab tirumotecan± pembrolizumab | PD-L1 < 10 LA/mTNBC | TroFuse-011 |
| ADC + rDDRinh | ||||
| NCT04039230 | 1b/2 | SG + talazoparib | mBC | — |
| NCT06298084 | 1b/2 | HER3-DXd + olaparib | HER2-low mBC post-T-DXd | ICARUSBREAST02 |
| ADC + targeted therapy | ||||
| NCT06331169 | 1 | T-DXd + anlotinib | HER2-low LA/mBC | ALTER-BC-Ib-01 |
| NCT05575804 | 1b/2 | GQ1001 + pyrotinib | HER2+ mBC | — |
| NCT06157892 | 1b/2 | DV + tucatinib | HER2 + LA/mBC or gastric/GEJC | — |
| NCT04983121 | 2 | Neoadjuvant ARX788 + pyrotinib | HER2 + BC | — |
| NCT04539938 | 2 | T-DXd + tucatinib | HER2+ mBC ± active brain metastasis | HER2CLIMB-04 |
| NCT06000033 | 2 | DV + anlotinib | HR–, HER2-low mBC | — |
| NCT06015113 | NA | DV + pyrotinib or neratinib | HER2+ mBC + brain metastasis | — |
| NCT06278870 | 3 | DV + pyrotinib first-line | HER2+ mBC | — |
| NCT06927180 | 2 | Neoadjuvant SHR-A1811 + pertuzumab | HER2 + BC | — |
ADC antibody–drug conjugate, ICI immune checkpoint inhibitor, rDDRinh replication DNA damage response inhibitor, mBC metastatic breast cancer, TNBC triple-negative breast cancer, LA locally advanced, HER2 human epidermal growth factor receptor 2, HR hormone receptor, PD-L1 programmed death-ligand 1, SCLC small cell lung cancer, SCNEC small cell neuroendocrine cancer, GEJC gastroesophageal junction cancer, DV disitamab vedotin, SG sacituzumab govitecan, Dato-DXd datopotamab deruxtecan, T-DM1 trastuzumab emtansine, T-DXd trastuzumab deruxtecan, HER3-DXd patritumab deruxtecan.
This is a non-exhaustive list intended to illustrate examples.
ADC+ immune checkpoint inhibitor (ICI)
The rationale for this combination is based on data showing that combining ADCs with ICIs enhances CD8 + T cell infiltration and activity70,76,77. In the phase 2 KATE2 trial, T-DM1 plus atezolizumab did not improve PFS and showed more serious AEs than T-DM1 alone in HER2-positive pretreated mBC. However, a potential benefit in PD-L1–positive tumors prompted the ongoing KATE3 trial78,79. In the BEGONIA platform trial (first-line mTNBC), Dato-DXd plus durvalumab (Arm 7) showed a 79% ORR (95% CI, 67–88) and 13.8-month mPFS (95% CI, 11–not calculable [NC])80; T-DXd plus durvalumab (Arm 6) achieved a 57% ORR (95% CI, 41–71) and 12.6 months PFS (95% CI, 8–not reached)71,81. Responses occurred regardless of PD-L1 status. Arm 8 is focusing on PD-L1–positive patients receiving Dato-DXd +durvalumab82 (Table 2).
In DS8201-A-U105 phase 1b trial, T-DXd plus nivolumab yielded ORRs of 65.6% (95% CI, 46.8–81.4%) and 50.0% (95% CI, 24.7–75.3%) for pretreated HER2-positive and HER2-low mBC, regardless of PD-L1 status, though limited by small sample sizes83.
The MORPHEUS-panBC umbrella study in untreated PD-L1-positive mTNBC reported a 76.7% ORR for SG plus atezolizumab vs 66.7% in the control arm, with responses linked to TROP2 and stromal Tumor-Infiltrating Lymphocytes (TILs)84.
In the Phase 3 ASCENT-04/KEYNOTE-D19 trial (treatment-naïve, PD-L1–positive locally advanced [LA]/mTNBC), SG plus pembrolizumab improved PFS over chemo-pembrolizumab (HR 0.65; 95% CI 0.51–0.84, P = 0.0009), with a median duration of response of 16.5 versus 9.2 months and an early positive trend in OS85.
Encouraging response rates were also reported in the early BC setting; in early TNBC, Arm A2 of the phase 2 NeoSTAR trial tested neoadjuvant SG plus pembrolizumab, achieving a protocol-defined pCR of 34% (16/50; 95% CI, 19.5–46.7)86. Both Dato-DXd as a single agent and in combination with durvalumab have been evaluated in the I-SPY2.2, a phase 2 neoadjuvant study, as part of sequential treatments in patients with HER2-negative stage II-III BC. Particularly, in the subtype defined as immune-positive according to the I-SPY classification system, the combination of Dato-DXd + durvalumab for 4 cycles showed a pCR of 54% that reached 92% after the sequential treatment with taxanes87 (Table 2).
ADC + DNA-damage response inhibitor (DDRinh)
Combining ADC with a DDRinh offers a promising strategy to overcome resistance to TOP1i payloads through “synthetic lethality”. TOP1i cause DNA damage, which is normally repaired via Poly(ADP-ribose) Polymerase (PARP)-dependent pathways. PARP inhibition blocks this repair, determining DNA breaks accumulation and enhanced cell death1,34,88,89. Preclinical models showed synergy between SG and talazoparib or olaparib, with significant tumor growth delay (P < 0.0017)89.
However, tolerability, especially myelosuppression, remains a key limitation34,90,91. In the phase 1b SEASTAR trial, rucaparib plus SG, showed activity, even post-DDRinh, but with high hematologic toxicity91.
In the NCT04039230 phase 1b study, concurrent SG-talazoparib caused high dose-limiting toxicities (DLTs-71.4%), while sequential administration reduced toxicity and maintained efficacy, enabling phase 2 dose selection90. Trials are ongoing to refine scheduling (Table 2).
ADC + targeted therapy
Incorporating tyrosine kinase inhibitors (TKIs) into dual-targeted approaches may enhance ADC efficacy and selectivity, though data on resistance are still limited1. In EGFR-mutant NSCLC models, osimertinib plus T-DM1 showed synergy, with T-DM1 delaying osimertinib resistance92. Furthermore, EGFR inhibition may upregulate HER3, potentially enhancing HER3-DXd uptake in preclinical models93.
Similarly, HER2-targeting TKIs also enhance T-DM1 efficacy: lapatinib increases HER2 expression, neratinib promotes internalization, while tucatinib’s impact is less defined1,94–97. The phase 2 TEAL study found improved responses with T-DM1+ lapatinib + nab-paclitaxel as neoadjuvant treatment in HER2-positive BC98. In pretreated HER2-positive mBC, the phase 1b NCT05575804 trial of GQ1001 plus pyrotinib showed a 71.4% ORR but high-grade 3–4 AEs (73.3%)99. Another phase 2 trial combining pyrotinib with DV determined a 31.6% ORR and mPFS of 7.1 vs 4.6 months with DV alone, without new safety signals100.
In HER2CLIMB-02 phase 3 trial, tucatinib plus T-DM1 improved PFS (HR 0.76, 95% CI, P = 0.0163) in pretreated HER2-positive LA/mBC, with OS data pending101,102.
Modulating tumor vasculature through antiangiogenic agents may improve ADC delivery1. In platinum-resistant epithelial ovarian cancer models, bevacizumab enhanced tumor necrosis and vascular disruption when combined with mirvetuximab soravtansine or anetumab ravtansine103,104. Clinical data support the safety and efficacy of the mirvetuximab–bevacizumab, though optimal dosing remains under investigation105.
Combining T-DXd with EGFR mAbs (cetuximab or panitumumab) may overcome resistance by enhancing endocytosis of HER2–EGFR heterodimers and target-bound T-DXd, thereby improving its efficacy35.
Dual HER2 inhibition, like T-DM1 plus pertuzumab, may improve ADC tumor distribution43,106,107.
In the phase 3 MARIANNE trial, T-DM1 ± pertuzumab was noninferior to the control arm (mOS 51.8 vs 50.9, HR 0.86, 97.5% CI, 0.67–1.11) in first-line HER2-positive mBC108. KRISTINE phase 3 trial, evaluating neoadjuvant therapy with TDM1 plus pertuzumab in early HER2-positive BC, showed lower pCR with the combination, but similar DFS (HR 1.11; 95% CI, 0.52–2.40)109. In the DESTINY-Breast09 phase 3 trial, T-DXd plus pertuzumab improved mPFS versus the control arm (HR 0.56; 95% CI; P < 0.00001), in first-line HER2-positive mBC110.
Targeting resistance pathways is promising in preclinical studies. STAT3 inhibition (napabucasin), HSP90 inhibition (geldanamycin), and Phosphoinositide 3-Kinase ([PI3K] CDC-0941) enhanced T-DM1 efficacy in HER2-positive models, though toxicity remains a concern58,111–113. Several trials are ongoing (Table 2).
ADC + ADC
Combining ADCs with non-overlapping toxicities may enhance efficacy. In the phase 1 DAD trial, SG plus enfortumab vedotin showed a 70% ORR (95% CI, 47–87%) in metastatic urothelial carcinoma114. Currently, no clinical trials are assessing ADC combinations in BC, their feasibility would depend on manageable toxicity profiles.
New-generation payloads
Cytotoxic payloads
To overcome ADC resistance, one strategy is using payloads less susceptible to efflux or resistance mechanisms (Fig. 2b). PNU-159682, a highly potent anthracycline (~100× doxorubicin) not affected by efflux pumps, showed preclinical efficacy where monomethyl auristatin (MMA)E or DM1 failed115. Another approach uses payloads like eribulin, which penetrates cells efficiently due to its balanced hydrophilicity/lipophilicity116. BB-1701, an eribulin-based HER2-targeting ADC, outperformed T-DM1 and T-DXd in HER2-expressing and HER2-low models, with strong bystander and immune-modulating effects (e.g., TILs recruitment and induction of immunogenic cell death)117. Preliminary phase 1 data suggest antitumor activity and manageable safety in HER2-low BC, including patients pretreated with anti-HER2 ADCs118 (Table 3).
Table 3.
Active clinical trials of innovative ADC platforms in breast cancer
| Clinical trial identifier | Phase | Investigational agent | Setting | Platform type |
|---|---|---|---|---|
| NCT05511844 | 1 | ORM-5029 | HER2+ advanced solid tumors | MAC |
| NCT05399654 | 2 | TAC-001 | Advanced solid tumor | ISAC |
| NCT05070247 | 1/2 | TAK-500 ± Pembrolizumab | LA/m solid tumor | ISAC |
| NCT05514717 | 1 | XMT-2056 | HER2+ advanced/recurrent Solid tumors | ISAC |
| NCT04561362 | 1/2 | BT8009 ± Pembrolizumab | Nectin-4+ advanced malignancies | Small format |
| NCT04180371 | 1/2 | BT5528 ± Nivolumab | EphA2+ advanced solid tumors | Small format |
| NCT06042894 | 2 | SI-B003 ± BL-B01D1 | HER2– LA/mBC | BsADC |
| NCT06382142 | 3 | BL-B01D1 + Chemotherapy | LA/mTNBC | BsADC |
| NCT05470348 | 1 | BL-B01D1 | LA/mBC and solid tumors | BsADC |
| NCT06471205 | 2 | BL-B01D1 + anti-PD-1 | LA/mTNBC | BsADC |
| NCT06343948 | 3 | BL-B01D1 + Chemotherapy | HR+/ HER2– LA/mBC | BsADC |
| NCT06846437 | 3 | JSKN003 | Pretreated HER2 + LA/mBC | BsADC |
| NCT06079983 | 3 | JSKN003 | Pretreated LA/mBC | BsADC |
| NCT06592417 | 1 | JSKN016 | Advanced solid tumors | BsADC |
| NCT05735496 | 1 | TQB2102 | Advanced solid tumors | BsADC |
| NCT06452706 | 2 | TQB2102 | HER2– mBC | BsADC |
| NCT07008976 | 3 | TQB2102 | HER2+ mBC | BsADC |
| NCT06198751 | 2 | TQB2102 neoadjuvant | Early HER2 + BC | BsADC |
| NCT06561607 | 3 | TQB2102 | HER2-low BC | BsADC |
| NCT07003074 | 3 | TQB2102 | HER2+ recurrent/mBC | BsADC |
| NCT06942234 | 1/2 | JSKN016 + combination therapy | HER2– LA/mBC | BsADC |
| NCT06349408 | 1 | IBI3001 | Solid tumors | BsADC |
| NCT06418061 | 1 | IBI3005 | Solid tumors | BsADC |
| NCT06493864 | 1 | BL-B16D1 | BC and other solid tumors | BsADC |
| NCT06475937 | 1 | DM001 | Advanced solid tumors | BsADC |
| NCT06554795 | 1/2 | DB-1419 | Advanced/m solid tumors | BsADC |
| NCT06725381 | 1 | SKB571 | Advanced solid tumors | BsADC |
| NCT06707610 | 1 | ALK202 | Advanced solid tumors | BsADC |
| NCT06685068 | 1/2 | GEN1286 | Advanced solid tumors | BsADC |
| NCT06751329 | 1 | DM002 | Advanced solid tumors | BsADC |
| NCT05785039 | 2 | BL-B01D1 | Urinary system and other solid tumors | New Linker/Conj. |
| NCT04829604 | 2 | ARX788 | HER2+ mBC | New Linker/Conj. |
| NCT05377996 | 1 | XMT-1660 | Solid tumors | New Linker/Conj. |
| NCT 04257110 | 1 | BB-1701 | LA/m HER2+ Solid Tumors | Bystander effect |
| NCT06188559 | 2 | BB-1701 | HER2 + / HER2-low mBC | Bystander effect |
ADC Antibody–drug Conjugate, MAC Glue–antibody Conjugate, ISAC immune-stimulating antibody conjugate, LA locally advance, m metastatic, BC breast cancer, Small format small format antibody–drug conjugate, BsADC bispecific antibody–drug conjugate, TNBC triple-negative breast cancer, HR+ hormone receptor-positive, HER2 human epidermal growth factor receptor 2, PD-1 programmed cell death protein 1, HNSCC head and neck squamous cell carcinoma, Conj. conjugation.
This is a non-exhaustive list intended to illustrate examples.
Trastuzumab rezetecan (SHR-A1811), an HER2-targeting ADC carrying a TOP1i payload, showed promising results in a phase 1 trial with ORRs in HER2-positive BC (76.3%), in HER2-low BC (60.4%), and non-breast tumors (45.9%)119. Notably, responses persisted in patients previously treated with T-DM1 (82.4%) and other HER2 ADCs, including T-DXd120.
ADC with multi-payloads
Dual-drug ADCs aim to overcome tumor heterogeneity by enhancing antitumor activity and accumulation, while avoiding binding competition seen with co-administered single-drug ADCs121 (Fig. 2b). However, ensuring consistent duo-payload conjugation with uniform DARs remains a challenge. Advances in linker technology, such as cysteine/selenocysteine engineering and hetero-trifunctional linkers, enable site-specific, stable attachment of dual payloads like PNU-159682 and MMAF, enhancing cytotoxicity in HER2-positive models122–124. However, all related clinical trials have been discontinued to date.
Degrader–antibody conjugates (DACs)
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules that induce targeted protein degradation by recruiting E3 ligases to specific intracellular proteins125. Though limited cell permeability, their conjugation to mAbs enables targeted delivery, leading to the development of DACs115,126,127 (Fig. 2b).
Structurally similar to ADCs, DACs carry higher DARs due to PROTACs’ lower potency. After receptor-mediated endocytosis, PROTACs are released into the cytoplasm to trigger degradation128. Maneiro et al. developed a trastuzumab-PROTAC conjugate to selectively degrade bromodomain-containing protein 4 (BRD4) in HER2-positive BC cell lines, without affecting HER2-negative cells129. To date, no clinical trials are ongoing.
Molecular glues and glue–antibody conjugates (MACs)
MACs represent a novel ADC class using monovalent degraders (molecular glues) as payloads that recruit E3 ligases like cereblon to trigger proteasomal degradation of neo substrates proteins, such as G1 to S phase transition 1 (GSPT1), often overexpressed in cancer, including BC130,131 (Fig. 2b). ORM-5029, an AnDC™ (Antibody neoDegrader Conjugate) combining the GSPT1 degrader SMol006 with pertuzumab via a Val-Cit PABc linker, shows greater cytotoxicity than other GSPT1 degraders and comparable efficacy to T-DXd in HER2-positive models132. A phase 1 trial is ongoing133 (Table 3).
Immune-stimulating antibody conjugates (ISACs)
ISACs combine a tumor-targeting antibody, non-cleavable linker, and immunostimulatory payload to boost anti-tumor immunity by activating innate and adaptive immune responses within TME, overcoming some limitations of conventional ADCs (Fig. 2b). However, systemic immune activation and cytokine release syndrome remain safety concerns. Development focuses on Toll-Like Receptors (TLRs), particularly TLR7/8/9 and Stimulator of Interferon Genes (STING) agonists134. STING activation triggers type I interferon responses134,135. IMSA1729, an EGFR-targeting STING-agonist ADC, showed strong tumor control and synergy with anti-PD-L1 in preclinical melanoma models136. XMT-2056, a HER2-targeting STING-agonist ISAC, demonstrated broad efficacy, including in HER2-low models, with manageable cytokine induction137; a phase 1 trial is ongoing (Table 3).
TLRs recognize pathogen-associated patterns to initiate immune responses134. SBT6050 (HER2-targeting TLR8-agonist) showed localized immune activation preclinically and entered early-phase trials, alone or with ICIs and HER2-targeted therapies (NCT04460456, NCT05091528), but both were discontinued138,139, other trials are ongoing (Table 3).
Antibody–oligonucleotide conjugates (AOCs)
AOCs combine the gene-silencing ability of oligonucleotides, like small interfering RNA (siRNA) and anti-sense oligonucleotides, with the target deliverability of antibodies, with the goal of improving serum stability, membrane permeability, and tissue specificity (Fig. 2b). However, challenges remain, particularly in the conjugation of oligonucleotides to nanoparticles, due to their size and charge140. A preclinical HER2-targeted AOCs using PLK1 siRNAs and HER2-ScFv-protamine fusion protein (F5-P) showed effective PLK1 inhibition, apoptosis induction, and tumor suppression in HER2-positive BC models141.
Innovative ADC structures
Small-format drug conjugates
Smaller antibody formats, like single-chain variable fragment (scFvs) and Fabs, improve tumor penetration and internalization, due to their reduced size compared to full-length immunoglobulins142 (Fig. 2c). Moreover, they present a lower off-target toxicity from lacking an Fc domain13. However, these formats also face limitations, including rapid systemic clearance142. Bicycle peptides offer a promising alternative, combining small size with efficient uptake115 (Table 3).
Bispecific antibody–drug conjugates (BsADCs)
BsADCs are bispecific antibody conjugates targeting two distinct epitopes on the same or different antigens, improving tumor selectivity and internalization in heterogeneous or resistant tumors143 (Fig. 2c). Biparatopic BsADCs, bind two non-overlapping epitopes on the same antigen like HER2, enhancing receptor clustering and degradation, showing activity in T-DM1-resistant and HER2-low settings144,145.
Andreev et al. developed a HER2/ Prolactin Receptor (PRLR) BsADC that enhanced HER2 degradation and outperformed HER2-ADCs in dual-expressing tumors146. Other preclinical BsADCs, including HER2/HER3, TROP2/HER2, demonstrated superior antitumor activity versus monospecific ADCs13,147.
BL-B01D1, targeting EGFR/HER3 with a camptothecin payload, showed promising activity in early trials148,149 (Table 3). In HER2-negative or HER2-low mBC, it showed an ORR of 42.1% and a mPFS of 6.9 months149.
TQB2102, a BsADC targeting HER2 extracellular domains II and IV, showed preliminary antitumor activity in a phase 1 study (NCT05735496) in advanced solid tumors, including mBC, with an ORR of 41.2% and good tolerability. Notably, 31% of HER2-expressing mBC patients had received prior anti-HER2 ADCs, including T-DM1 and T-DXd150.
JSKN016, a TROP2/HER3-targeting BsADC, showed preliminary antitumor activity (ORR 80%) and a manageable safety profile in pretreated mTNBC in an ongoing phase 1 trial (NCT06592417)151.
A list of ongoing trials is provided in Table 3.
Prodrug-conjugated antibodies (PDCs)
Emerging ADCs aim to mitigate on-target, off-tumor toxicity, especially when targeting antigens like EGFR or TROP2, also found on normal tissues7,8. PDCs use inactive payloads or self-masking domains that are selectively activated in the tumor TME by proteases or low pH, enhancing tumor specificity and minimizing systemic damage (Fig. 2c). However, precise tumor profiling is needed to match masking strategies effectively7,152.
New linkers and novel conjugation strategies
Increasing linker hydrophilicity is a key strategy to enhance ADC efficacy and overcome resistance (Fig. 2c).
Hydrophilic linkers, like polyethylene glycol-4 Maleimide (PEG4Mal), improve antitumor activity and reduce clearance, especially in MDR-expressing tumors, outperforming hydrophobic linkers47. Other hydrophilic linkers (sulfonate, PEG, polysarcosine, DNA-based linkers) improve stability, pharmacokinetics, and reduce toxicity115. Traditional stochastic conjugation yields heterogeneous ADCs with variable DARs7, while site-specific conjugation (e.g., engineered cysteines, unnatural amino acids [UAAs], enzymatic ligation) improve homogeneity and therapeutic index115,153.
GQ1001, a trastuzumab–DM1 ADC using ligase-dependent conjugation, reduces free DM1 toxicity and showed 40% ORR with manageable side effects in a phase 1 trial154,155. ARX788, a site-specific HER2-targeted ADC using UAA-MMAF conjugation, demonstrated activity in HER2-positive and HER2-low mBC, including pretreated T-DXd patients, a phase 2 trial is ongoing156 (Table 3). In ACE-Breast-02 phase 3 trial, ARX788 improved PFS (HR 0.64, 95% CI P = 0.0006) versus lapatinib-capecitabine with low discontinuation157.
Emlitatug ledadotin, a site-specific B7-H4 ADC, showed promising activity (ORR 23%; 95% CI, 5–54%) in heavily pretreated B7-H4–high mTNBC158.
In addition, branched linkers, such as Kumar’s hetero-trifunctional linker, enable higher DARs or dual-payload designs, further enhancing ADC potency124. Some clinical trials are ongoing (Table 3).
Conclusion
Understanding resistance mechanisms to ADCs is essential for developing new agents capable of overcoming them. Although many have been identified in preclinical studies, further research is needed to clarify their clinical relevance. To address resistance in clinical practice, several strategies are currently under development. These include combination therapies with other agents, such as ICI, DDRinh, and TKI, as well as the use of new-generation payloads and alternative formats, such as DACs, MACs, ISACs, AOCs, or multi-payload ADCs. Another promising approach involves moving beyond the traditional ADC structure, with the development of small-format drug conjugates, BsADCs, PDCs, and optimization of the bystander effect, linker technologies, and conjugation strategies. Multiple clinical trials are currently ongoing to assess the efficacy and safety of these approaches. In addition, the identification of predictive dynamic biomarkers, such as ctDNA, plasma copy number variations, and artificial intelligence-based analyses, may enable real-time monitoring of ADC efficacy and resistance development.
Author contributions
I.V. and T.G.: conceptualization, design, and initial drafting of the manuscript. B.P.: conceptualization, design, and critical revision of the manuscript. T.G., L.A., and B.P.: critical revision of the manuscript. All authors reviewed and approved the final version of the manuscript.
Data availability
No datasets were generated or analyzed during the current study.
Competing interests
I.V. declares no competing interests. T.G. reports having received travel support from AstraZeneca, Gilead, and Pfizer; has served in a consulting/advisory role for AstraZeneca and Cancerologie-Pratique; and has received research funding from Amgen. L.A. reports relationships with Amgen, Roche, AstraZeneca, and Novartis involving royalties, and has served in an advisory role for Merck Serono. B.P. has received consulting fees (institutional) from AstraZeneca, Seagen, Gilead, Novartis, Lilly, MSD, and Pierre Fabre; personal consulting fees from Pierre Fabre and Daiichi-Sankyo; research funding (institutional) from AstraZeneca, Daiichi-Sankyo, Gilead, Seagen, and MSD; and travel support from AstraZeneca, Pierre Fabre, Lilly, Daiichi-Sankyo, and MSD.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Wei, Q. et al. The promise and challenges of combination therapies with antibody-drug conjugates in solid tumors. J. Hematol. Oncol. J. Hematol. Oncol.17, 1 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chau, C. H., Steeg, P. S. & Figg, W. D. Antibody-drug conjugates for cancer. Lancet394, 793–804 (2019). [DOI] [PubMed] [Google Scholar]
- 3.Pommier, Y. & Thomas, A. New life of topoisomerase I inhibitors as antibody-drug conjugate warheads. Clin. Cancer Res. J. Am. Assoc. Cancer Res.29, 991–993 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ko, S. et al. An Fc variant with two mutations confers prolonged serum half-life and enhanced effector functions on IgG antibodies. Exp. Mol. Med.54, 1850–1861 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Khoury, R. et al. Mechanisms of resistance to antibody-drug conjugates. Int. J. Mol. Sci.24, 9674 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Criscitiello, C., Morganti, S. & Curigliano, G. Antibody-drug conjugates in solid tumors: a look into novel targets. J. Hematol. Oncol.14, 20 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tsuchikama, K., Anami, Y., Ha, S. Y. Y. & Yamazaki, C. M. Exploring the next generation of antibody-drug conjugates. Nat. Rev. Clin. Oncol.21, 203–223 (2024). [DOI] [PubMed] [Google Scholar]
- 8.Colombo, R., Tarantino, P., Rich, J. R., LoRusso, P. M. & de Vries, E. G. E. The journey of antibody-drug conjugates: lessons learned from 40 years of development. Cancer Discov.14, 2089–2108 (2024). [DOI] [PubMed] [Google Scholar]
- 9.Saleh, É. A. et al. Mechanisms of action and resistance to anti-HER2 antibody-drug conjugates in breast cancer. Cancer Drug Resist.7, 22 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Larose, ÉA. et al. Antibody-drug conjugates in breast cancer treatment: resistance mechanisms and the role of therapeutic sequencing. Cancer Drug Resist.8, 11 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yin, Y. et al. Sacituzumab tirumotecan in previously treated metastatic triple-negative breast cancer: a randomized phase 3 trial. Nat. Med. 10.1038/s41591-025-03630-w (2025). [DOI] [PubMed]
- 12.Lucas, A. T. et al. Factors affecting the pharmacology of antibody-drug conjugates. Antibodies7, 10 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cilliers, C., Menezes, B., Nessler, I., Linderman, J. & Thurber, G. M. Improved tumor penetration and single-cell targeting of antibody-drug conjugates increases anticancer efficacy and host survival. Cancer Res.78, 758–768 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Heldin, C.-H., Rubin, K., Pietras, K. & Ostman, A. High interstitial fluid pressure—an obstacle in cancer therapy. Nat. Rev. Cancer4, 806–813 (2004). [DOI] [PubMed] [Google Scholar]
- 15.Cilliers, C., Guo, H., Liao, J., Christodolu, N. & Thurber, G. M. Multiscale modeling of antibody-drug conjugates: connecting tissue and cellular distribution to whole animal pharmacokinetics and potential implications for efficacy. AAPS J.18, 1117–1130 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science307, 58–62 (2005). [DOI] [PubMed] [Google Scholar]
- 17.Fujimori, K., Covell, D. G., Fletcher, J. E. & Weinstein, J. N. A modeling analysis of monoclonal antibody percolation through tumors: a binding-site barrier. J. Nucl. Med.31, 1191–1198 (1990). [PubMed] [Google Scholar]
- 18.Ponte, J. F. et al. Antibody co-administration can improve systemic and local distribution of antibody-drug conjugates to increase in vivo efficacy. Mol. Cancer Ther.20, 203–212 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pistilli, B. et al. 340O Efficacy, safety and biomarker analysis of ICARUS-BREAST01: a phase II study of patritumab deruxtecan (HER3-DXd) in patients (pts) with HR+/HER2- advanced breast cancer (ABC). Ann. Oncol.35, S357 (2024). [Google Scholar]
- 20.Corti, C. et al. Future potential targets of antibody-drug conjugates in breast cancer. Breast69, 312–322 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ogitani, Y. et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin. Cancer Res.22, 5097–5108 (2016). [DOI] [PubMed] [Google Scholar]
- 22.Barok, M., Joensuu, H. & Isola, J. Trastuzumab emtansine: mechanisms of action and drug resistance. Breast Cancer Res. BCR16, 209 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tarantino, P. et al. Antibody–drug conjugates: smart chemotherapy delivery across tumor histologies. CA Cancer J. Clin.72, 165–182 (2022). [DOI] [PubMed] [Google Scholar]
- 24.Mosele, F. et al. Trastuzumab deruxtecan in metastatic breast cancer with variable HER2 expression: the phase 2 DAISY trial. Nat. Med.29, 2110–2120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bardia, A. et al. Biomarker analyses in the phase III ASCENT study of sacituzumab govitecan versus chemotherapy in patients with metastatic triple-negative breast cancer☆. Ann. Oncol.32, 1148–1156 (2021). [DOI] [PubMed] [Google Scholar]
- 26.Rugo, H. S. et al. Overall survival with sacituzumab govitecan in hormone receptor-positive and human epidermal growth factor receptor 2-negative metastatic breast cancer (TROPiCS-02): a randomised, open-label, multicentre, phase 3 trial. Lancet402, 1423–1433 (2023). [DOI] [PubMed] [Google Scholar]
- 27.Li, S. et al. Resistance to antibody–drug conjugates: a review. Acta Pharm. Sin. B15, 737–756 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Loganzo, F. et al. Tumor cells chronically treated with a trastuzumab-maytansinoid antibody-drug conjugate develop varied resistance mechanisms but respond to alternate treatments. Mol. Cancer Ther.14, 952–963 (2015). [DOI] [PubMed] [Google Scholar]
- 29.Li, W.-F. et al. OBI-992, a novel TROP2-targeted antibody-drug conjugate, demonstrates antitumor activity in multiple cancer models. Mol. Cancer Ther.24, 163–175 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Coates, J. T. et al. Parallel genomic alterations of antigen and payload targets mediate polyclonal acquired clinical resistance to sacituzumab govitecan in triple-negative breast cancer. Cancer Discov.11, 2436–2445 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yu, H. A. et al. Translational insights and overall survival in the U31402-A-U102 study of patritumab deruxtecan (HER3-DXd) in EGFR-mutated NSCLC. Ann. Oncol.35, 437–447 (2024). [DOI] [PubMed] [Google Scholar]
- 32.Filho, O. M. et al. Impact of HER2 heterogeneity on treatment response of early-stage HER2-positive breast cancer: phase II neoadjuvant clinical trial of T-DM1 combined with pertuzumab. Cancer Discov.11, 2474–2487 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hurvitz, S. A. et al. Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol.19, 115–126 (2018). [DOI] [PubMed] [Google Scholar]
- 34.Chang, H. L., Schwettmann, B., McArthur, H. L. & Chan, I. S. Antibody-drug conjugates in breast cancer: overcoming resistance and boosting immune response. J. Clin. Invest.133, e172156 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gupta, A. et al. EGFR-directed antibodies promote HER2 ADC internalization and efficacy. Cell Rep. Med.5, 101792 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kalim, M. et al. Intracellular trafficking of new anticancer therapeutics: antibody-drug conjugates. Drug Des. Dev. Ther.11, 2265–2276 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ríos-Luci, C. et al. Resistance to the antibody-drug conjugate T-DM1 is based in a reduction in lysosomal proteolytic activity. Cancer Res.77, 4639–4651 (2017). [DOI] [PubMed] [Google Scholar]
- 38.Sung, M. et al. Caveolae-mediated endocytosis as a novel mechanism of resistance to trastuzumab emtansine (T-DM1). Mol. Cancer Ther.17, 243–253 (2018). [DOI] [PubMed] [Google Scholar]
- 39.Baldassarre, T., Truesdell, P. & Craig, A. W. Endophilin A2 promotes HER2 internalization and sensitivity to trastuzumab-based therapy in HER2-positive breast cancers. Breast Cancer Res. BCR19, 110 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hamblett, K. J. et al. SLC46A3 is required to transport catabolites of noncleavable antibody maytansine conjugates from the lysosome to the cytoplasm. Cancer Res.75, 5329–5340 (2015). [DOI] [PubMed] [Google Scholar]
- 41.Baselga, J. et al. Relationship between tumor biomarkers and efficacy in EMILIA, a phase III study of trastuzumab emtansine in HER2-positive metastatic breast cancer. Clin. Cancer Res.22, 3755–3763 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Breuleux, M. Role of heregulin in human cancer. Cell. Mol. Life Sci. CMLS64, 2358–2377 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Phillips, G. D. L. et al. Dual targeting of HER2-positive cancer with trastuzumab emtansine and pertuzumab: critical role for neuregulin blockade in antitumor response to combination therapy. Clin. Cancer Res.20, 456–468 (2014). [DOI] [PubMed] [Google Scholar]
- 44.Ambrogi, F. et al. Trop-2 is a determinant of breast cancer survival. PLoS ONE9, e96993 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Trerotola, M. et al. Upregulation of Trop-2 quantitatively stimulates human cancer growth. Oncogene32, 222–233 (2013). [DOI] [PubMed] [Google Scholar]
- 46.Garassino, M. C. et al. PL02.11 normalized membrane ratio of TROP2 by quantitative continuous scoring is predictive of clinical outcomes in TROPION-Lung 01. J. Thorac. Oncol.19, S2–S3 (2024). [Google Scholar]
- 47.Kovtun, Y. V. et al. Antibody-maytansinoid conjugates designed to bypass multidrug resistance. Cancer Res.70, 2528–2537 (2010). [DOI] [PubMed] [Google Scholar]
- 48.Cianfriglia, M. The biology of MDR1-P-glycoprotein (MDR1-Pgp) in designing functional antibody drug conjugates (ADCs): the experience of gemtuzumab ozogamicin. Ann. Ist. Super. Sanita49, 150–168 (2013). [DOI] [PubMed] [Google Scholar]
- 49.Takegawa, N. et al. DS-8201a, a new HER2-targeting antibody-drug conjugate incorporating a novel DNA topoisomerase I inhibitor, overcomes HER2-positive gastric cancer T-DM1 resistance. Int. J. Cancer141, 1682–1689 (2017). [DOI] [PubMed] [Google Scholar]
- 50.Pourjamal, N. et al. Comparison of trastuzumab emtansine, trastuzumab deruxtecan, and disitamab vedotin in a multiresistant HER2-positive breast cancer lung metastasis model. Clin. Exp. Metastasis41, 91–102 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sledge, G. et al. Abstract PS13-09: mechanisms of resistance to trastuzumab deruxtecan in breast cancer elucidated by multi-omic molecular profiling. Clin. Cancer Res.31, PS13-09 (2025). [Google Scholar]
- 52.Gupta, A., Jatwani, K., Gupta, K., Qiu, J. & Dy, G. K. Loss of Rb1 associated with the onset of acquired resistance to trastuzumab deruxtecan in TP53-/HER2-mutated non-small-cell lung cancer: case series. JCO Precis. Oncol.7, e2200476 (2023). [DOI] [PubMed] [Google Scholar]
- 53.Abelman, R. O. et al. TOP1 mutations and cross-resistance to antibody-drug conjugates in patients with metastatic breast cancer. Clin. Cancer Res.10.1158/1078-0432.CCR-24-2771 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Barok, M., Puhka, M., Yazdi, N. & Joensuu, H. Extracellular vesicles as modifiers of antibody-drug conjugate efficacy. J. Extracell. Vesicles10, e12070 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Le Joncour, V. et al. A novel anti-HER2 antibody-drug conjugate XMT-1522 for HER2-positive breast and gastric cancers resistant to trastuzumab emtansine. Mol. Cancer Ther.18, 1721–1730 (2019). [DOI] [PubMed] [Google Scholar]
- 56.Sabbaghi, M. et al. Defective cyclin B1 induction in trastuzumab-emtansine (T-DM1) acquired resistance in HER2-positive breast cancer. Clin. Cancer Res.23, 7006–7019 (2017). [DOI] [PubMed] [Google Scholar]
- 57.Saatci, Ö. et al. Targeting PLK1 overcomes T-DM1 resistance via CDK1-dependent phosphorylation and inactivation of Bcl-2/xL in HER2-positive breast cancer. Oncogene37, 2251–2269 (2018). [DOI] [PubMed] [Google Scholar]
- 58.Wang, L. et al. STAT3 activation confers trastuzumab-emtansine (T-DM1) resistance in HER2-positive breast cancer. Cancer Sci.109, 3305–3315 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Sauveur, J. et al. Esophageal cancer cells resistant to T-DM1 display alterations in cell adhesion and the prostaglandin pathway. Oncotarget9, 21141–21155 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zou, Y. et al. crVDAC3 alleviates ferroptosis by impeding HSPB1 ubiquitination and confers trastuzumab deruxtecan resistance in HER2-low breast cancer. Drug Resist. Update. Rev. Comment. Antimicrob. Anticancer Chemother.77, 101126 (2024). [DOI] [PubMed] [Google Scholar]
- 61.Huppert, L. A. et al. Multicenter retrospective cohort study of the sequential use of the antibody-drug conjugates (ADCs) trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG) in patients with HER2-low metastatic breast cancer (MBC): updated data and subgroup analyses by age, sites of disease, and use of intervening therapies. J. Clin. Oncol.42, 1083–1083 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Poumeaud, F. et al. Abstract PS08-02: efficacy of sacituzumab-govitecan (sg) post trastuzumab-deruxtecan (T-DXd) and vice versa for HER2low advanced or metastatic breast cancer (MBC): a French multicentre retrospective study. Cancer Res.84, PS08-02 (2024). [Google Scholar]
- 63.Abelman, R. O. et al. Abstract PS08-03: sequencing antibody-drug conjugate after antibody-drug conjugate in metastatic breast cancer (A3 study): multi-institution experience and biomarker analysis. Cancer Res.84, PS08-03 (2024). [Google Scholar]
- 64.Mai, N. et al. Real world outcomes of sequential ADC therapy in metastatic breast cancer: patients treated with sacituzumab govitecan and trastuzumab deruxtecan. J. Clin. Oncol.42, 1085–1085 (2024). [Google Scholar]
- 65.Tarantino, P. et al. Outcomes of subsequent treatment regimens after trastuzumab deruxtecan in patients with metastatic breast cancer. JNCI J. Natl. Cancer Inst. 10.1093/jnci/djaf220 (2025). [DOI] [PubMed]
- 66.André, F. et al. Trastuzumab deruxtecan versus treatment of physician’s choice in patients with HER2-positive metastatic breast cancer (DESTINY-Breast02): a randomised, open-label, multicentre, phase 3 trial. Lancet401, 1773–1785 (2023). [DOI] [PubMed] [Google Scholar]
- 67.Fasching, P. A. et al. 322P Human epidermal growth factor receptor 2 (HER2)-directed therapies administered after trastuzumab deruxtecan (T-DXd) remain effective in patients (pts) with metastatic breast cancer (mBC): exploratory analysis from DESTINY-Breast02 and -03. ESMO Open10, 104894 (2025). [Google Scholar]
- 68.Tsao, L.-C. et al. Effective extracellular payload release and immunomodulatory interactions govern the therapeutic effect of trastuzumab deruxtecan (T-DXd). Nat. Commun.16, 3167 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Müller, P. et al. Microtubule-depolymerizing agents used in antibody-drug conjugates induce antitumor immunity by stimulation of dendritic cells. Cancer Immunol. Res.2, 741–755 (2014). [DOI] [PubMed] [Google Scholar]
- 70.Iwata, T. N. et al. A HER2-targeting antibody-drug conjugate, trastuzumab deruxtecan (DS-8201a), enhances antitumor immunity in a mouse model. Mol. Cancer Ther.17, 1494–1503 (2018). [DOI] [PubMed] [Google Scholar]
- 71.Schmid, P. et al. BEGONIA: phase 1b/2 study of durvalumab (D) combinations in locally advanced/metastatic triple-negative breast cancer (TNBC)—initial results from arm 1, d+paclitaxel (P), and arm 6, d+trastuzumab deruxtecan (T-DXd). J. Clin. Oncol.39, 1023–1023 (2021). [Google Scholar]
- 72.Lee, K.-L., Kuo, Y.-C., Ho, Y.-S. & Huang, Y.-H. Triple-negative breast cancer: current understanding and future therapeutic breakthrough targeting cancer stemness. Cancers11, 1334 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Liu, S. & Wicha, M. S. Targeting breast cancer stem cells. J. Clin. Oncol.28, 4006–4012 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Fultang, N., Chakraborty, M. & Peethambaran, B. Regulation of cancer stem cells in triple negative breast cancer. Cancer Drug Resist4, 321–342 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Islam, S. S. et al. Antibody-drug conjugate T-DM1 treatment for HER2+ breast cancer induces ROR1 and confers resistance through activation of Hippo transcriptional coactivator YAP1. EBioMedicine43, 211–224 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Gerber, H.-P., Sapra, P., Loganzo, F. & May, C. Combining antibody-drug conjugates and immune-mediated cancer therapy: what to expect?. Biochem. Pharmacol.102, 1–6 (2016). [DOI] [PubMed] [Google Scholar]
- 77.Müller, P. et al. Trastuzumab emtansine (T-DM1) renders HER2+ breast cancer highly susceptible to CTLA-4/PD-1 blockade. Sci. Transl. Med.7, 315ra188 (2015). [DOI] [PubMed] [Google Scholar]
- 78.Emens, L. A. et al. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised, double-blind trial. Lancet Oncol.21, 1283–1295 (2020). [DOI] [PubMed] [Google Scholar]
- 79.Loi, S. et al. 329TiP KATE3: a phase III study of trastuzumab emtansine (T-DM1) in combination with atezolizumab or placebo in patients with previously treated HER2-positive and PD-L1–positive locally advanced or metastatic breast cancer. Ann. Oncol.32, S509 (2021).
- 80.Schmid, P. et al. 379MO datopotamab deruxtecan (Dato-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic triple-negative breast cancer (a/mTNBC): updated results from BEGONIA, a phase Ib/II study. Am. Oncol.34, S337 (2023). [Google Scholar]
- 81.Schmid, P. et al. Abstract PD11-08: PD11-08 Trastuzumab deruxtecan (T-DXd) + durvalumab (D) as first-line (1L) treatment for unresectable locally advanced/metastatic hormone receptor-negative (HR−), HER2-low breast cancer: updated results from BEGONIA, a phase 1b/2 study (2023).
- 82.Schmid, P. et al. Abstract PO1-19-10: durvalumab + datopotamab deruxtecan in patients with PD-L1 positive advanced/metastatic triple-negative breast cancer: arm 8 of the phase 1b/2, open label, platform BEGONIA study. Cancer Res.84, PO1–19–10 (2024). [Google Scholar]
- 83.Hamilton, E. et al. Trastuzumab deruxtecan with nivolumab in HER2-expressing metastatic breast or urothelial cancer: analysis of the phase Ib DS8201-A-U105 study. Clin. Cancer Res.30, 5548–5558 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Schmid, P. et al. Abstract 181O Interim analysis (IA) of the atezolizumab (atezo) + sacituzumab govitecan (SG) arm in patients (pts) with triple-negative breast cancer (TNBC) in MORPHEUS-pan BC: a phase Ib/II study of multiple treatment (tx) combinations in pts with locally advanced/metastatic BC (LA/mBC). ESMO Open9 (2024).
- 85.Tolaney, S. M. et al. Abstract LBA109: Sacituzumab govitecan (SG) + pembrolizumab (pembro) vs chemotherapy (chemo) + pembro in previously untreated PD-L1-positive advanced triple-negative breast cancer (TNBC): primary results from the randomized phase 3 ASCENT-04/KEYNOTE-D19 study. J. Clin. Oncol. 43 (2025).
- 86.Abelman, R. O. et al. A phase 2 study of response-guided neoadjuvant sacituzumab govitecan and pembrolizumab (SG/P) in patients with early-stage triple-negative breast cancer: results from the NeoSTAR trial. J. Clin. Oncol.43, 511–511 (2025). [Google Scholar]
- 87.Shatsky, R. A. et al. Datopotamab–deruxtecan plus durvalumab in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial. Nat. Med.30, 3737–3747 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Smith, L. M., Willmore, E., Austin, C. A. & Curtin, N. J. The novel poly(ADP-Ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks. Clin. Cancer Res.11, 8449–8457 (2005). [DOI] [PubMed] [Google Scholar]
- 89.Cardillo, T. M. et al. Synthetic lethality exploitation by an anti-trop-2-SN-38 antibody-drug conjugate, IMMU-132, plus PARP inhibitors in BRCA1/2-wild-type triple-negative breast cancer. Clin. Cancer Res.23, 3405–3415 (2017). [DOI] [PubMed] [Google Scholar]
- 90.Bardia, A. et al. Antibody–drug conjugate sacituzumab govitecan enables a sequential TOP1/PARP inhibitor therapy strategy in patients with breast cancer. Clin. Cancer Res.30, 2917–2924 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Yap, T. A. et al. Phase Ib SEASTAR study: combining rucaparib and sacituzumab govitecan in patients with cancer with or without mutations in homologous recombination repair genes. JCO Precis. Oncol.6, e2100456 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.La Monica, S. et al. Trastuzumab emtansine delays and overcomes resistance to the third-generation EGFR-TKI osimertinib in NSCLC EGFR mutated cell lines. J. Exp. Clin. Cancer Res. CR36, 174 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Haikala, H. M. et al. EGFR inhibition enhances the cellular uptake and antitumor-activity of the HER3 antibody-drug conjugate HER3-DXd. Cancer Res.82, 130–141 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Abraham, J. et al. Safety and efficacy of T-DM1 plus neratinib in patients with metastatic HER2-positive breast cancer: NSABP Foundation Trial FB-10. J. Clin. Onco.37, 2601–2609 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kulukian, A. et al. Abstract PS10-08: tucatinib potentiates the activity of the antibody-drug conjugate T-DM1 in preclinical models of HER2-positive breast cancer. Cancer Res.81, PS10-08 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Murthy, R. K. et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N. Engl. J. Med.382, 597–609 (2020). [DOI] [PubMed] [Google Scholar]
- 97.Scaltriti, M. et al. Lapatinib, a HER2 tyrosine kinase inhibitor, induces stabilization and accumulation of HER2 and potentiates trastuzumab-dependent cell cytotoxicity. Oncogene28, 803–814 (2009). [DOI] [PubMed] [Google Scholar]
- 98.Patel, T. A. et al. A randomized, controlled phase II trial of neoadjuvant ado-trastuzumab emtansine, lapatinib, and nab-paclitaxel versus trastuzumab, pertuzumab, and paclitaxel in HER2-positive breast cancer (TEAL study). Breast Cancer Res. BCR21, 100 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wang B. et al. Abstract e13020: Safety and preliminary efficacy of GQ1001, a next generation HER2-targeting ADC, combined with pyrotinib in pretreated patients with HER2-positive metastatic breast cancer: a phase 1b clinical trial. J. Clin. Oncol.
- 100.Qu, F., Li, W. & Yi, Y. 397P Efficacy and safety of the recombinant humanized anti-HER2 monoclonal antibody-MMAE conjugate RC48-ADC in patients with HER2-positive or HER2-low expressing, locally advanced or metastatic breast cancer: a single-arm phase II study. Ann. Oncol.34, S348 (2023). [Google Scholar]
- 101.Masuda, N. et al. 67TiP HER2CLIMB-02: a randomized, double-blind, phase III study of tucatinib or placebo with T-DM1 for unresectable locally-advanced or metastatic HER2+ breast cancer. Ann. Oncol.31, S1267–S1268 (2020). [Google Scholar]
- 102.Hurvitz, S. et al. Abstract GS01-10: HER2CLIMB-02: randomized, double-blind phase 3 trial of tucatinib and trastuzumab emtansine for previously treated HER2-positive metastatic breast cancer. Cancer Res.84, GS01–GS10 (2024). [Google Scholar]
- 103.Ponte, J. F. et al. Mirvetuximab soravtansine (IMGN853), a Folate receptor alpha-targeting antibody-drug conjugate, potentiates the activity of standard of care therapeutics in ovarian cancer models. Neoplasia18, 775–784 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Quanz, M. et al. Anetumab ravtansine inhibits tumor growth and shows additive effect in combination with targeted agents and chemotherapy in mesothelin-expressing human ovarian cancer models. Oncotarget9, 34103–34121 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gilbert, L. et al. Safety and efficacy of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol. Oncol.170, 241–247 (2023). [DOI] [PubMed] [Google Scholar]
- 106.Bordeau, B. M., Yang, Y. & Balthasar, J. P. Transient competitive inhibition bypasses the binding site barrier to improve tumor penetration of trastuzumab and enhance T-DM1 efficacy. Cancer Res.81, 4145–4154 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Miller, K. D. et al. Phase IIa trial of trastuzumab emtansine with pertuzumab for patients with human epidermal growth factor receptor 2-positive, locally advanced, or metastatic breast cancer. J. Clin. Oncol.32, 1437–1444 (2014). [DOI] [PubMed] [Google Scholar]
- 108.Perez, E. A. et al. Trastuzumab emtansine with or without pertuzumab versus trastuzumab with taxane for human epidermal growth factor receptor 2-positive advanced breast cancer: final results from MARIANNE. Cancer125, 3974–3984 (2019). [DOI] [PubMed] [Google Scholar]
- 109.Hurvitz, S. A. et al. Neoadjuvant trastuzumab emtansine and pertuzumab in human epidermal growth factor receptor 2-positive breast cancer: three-year outcomes from the phase III KRISTINE Study. J. Clin. Oncol.37, 2206–2216 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tolaney, S. M. et al. Trastuzumab deruxtecan (T-DXd) + pertuzumab (P) vs taxane + trastuzumab + pertuzumab (THP) for first-line (1L) treatment of patients (pts) with human epidermal growth factor receptor 2-positive (HER2+) advanced/metastatic breast cancer (a/mBC): interim results from DESTINY-Breast09. J. Clin. Oncol.43, LBA1008–LBA1008 (2025). [Google Scholar]
- 111.Tolaney, S. M. et al. 328TiP Phase III study of trastuzumab deruxtecan (T-DXd) with or without pertuzumab vs a taxane, trastuzumab and pertuzumab in first-line (1L), human epidermal growth factor receptor 2-positive (HER2+) metastatic breast cancer (mBC): DESTINY-Breast09. Ann. Oncol.32, S507–S508 (2021). [Google Scholar]
- 112.Fukuyo, Y., Hunt, C. R. & Horikoshi, N. Geldanamycin and its anti-cancer activities. Cancer Lett.290, 24–35 (2010). [DOI] [PubMed] [Google Scholar]
- 113.McCombs, J. R., Chang, H. P., Shah, D. K. & Owen, S. C. Antibody-drug conjugate and free geldanamycin combination therapy enhances anti-cancer efficacy. Int. J. Pharm.610, 121272 (2021). [DOI] [PubMed] [Google Scholar]
- 114.McGregor, B. A. et al. The double antibody drug conjugate (DAD) phase I trial: sacituzumab govitecan plus enfortumab vedotin for metastatic urothelial carcinoma. Ann. Oncol.35, 91–97 (2024). [DOI] [PubMed] [Google Scholar]
- 115.Conilh, L., Sadilkova, L., Viricel, W. & Dumontet, C. Payload diversification: a key step in the development of antibody-drug conjugates. J. Hematol. Oncol.16, 3 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Takahashi, M. et al. Eribulin penetrates brain tumor tissue and prolongs survival of mice harboring intracerebral glioblastoma xenografts. Cancer Sci.110, 2247–2257 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wang, Y. et al. Preclinical studies of BB-1701, a HER2-targeting eribulin-containing ADC with potent bystander effect and ICD activity. Antib. Ther.7, 221–232 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ma, F. et al. Updated safety and antitumor activity of BB-1701 from study 101 in the multiple dose level cohort of patients with locally advanced/metastatic HER2-low expressing breast cancer. J. Clin. Oncol.42, 1110–1110 (2024).38215351 [Google Scholar]
- 119.Yao, H. et al. Safety, efficacy, and pharmacokinetics of SHR-A1811, a human epidermal growth factor receptor 2-directed antibody-drug conjugate, in human epidermal growth factor receptor 2-expressing or mutated advanced solid tumors: a global phase I trial. J. Clin. Oncol.42, 3453–3465 (2024). [DOI] [PubMed] [Google Scholar]
- 120.Yao, H. et al. Abstract CT175: safety, tolerability, pharmacokinetics, and antitumor activity of SHR-A1811 in HER2-expressing/mutated advanced solid tumors: a global phase 1, multi-center, first-in-human study. Cancer Res.83, CT175 (2023). [Google Scholar]
- 121.Yamazaki, C. M. et al. Antibody-drug conjugates with dual payloads for combating breast tumor heterogeneity and drug resistance. Nat. Commun.12, 3528 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Dong, W., Wang, W. & Cao, C. The evolution of antibody-drug conjugates: toward accurate DAR and multi-specificity. Chem Med Chem19, e202400109 (2024). [DOI] [PubMed] [Google Scholar]
- 123.Nilchan N. et al. Dual-mechanistic antibody-drug conjugate via site-specific selenocysteine/cysteine conjugation. Antibody Ther.2, 71–78 (2019). [DOI] [PMC free article] [PubMed]
- 124.Kumar, A. et al. Synthesis of a heterotrifunctional linker for the site-specific preparation of antibody-drug conjugates with two distinct warheads. Bioorg. Med. Chem. Lett.28, 3617–3621 (2018). [DOI] [PubMed] [Google Scholar]
- 125.Li, J., Chen, X., Lu, A. & Liang, C. Targeted protein degradation in cancers: orthodox PROTACs and beyond. Innovation4, 100413 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tecalco-Cruz, A. C., Zepeda-Cervantes, J., Ramírez-Jarquín, J. O. & Rojas-Ochoa, A. Proteolysis-targeting chimeras and their implications in breast cancer. Explor. Target. Anti-Tumor Ther.2, 496–510 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ocaña, A. & Pandiella, A. Proteolysis targeting chimeras (PROTACs) in cancer therapy. J. Exp. Clin. Cancer Res. CR39, 189 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hong, K. B. & An, H. Degrader–antibody conjugates: emerging new modality. J. Med. Chem.66, 140–148 (2023). [DOI] [PubMed] [Google Scholar]
- 129.Maneiro, M. A. et al. Antibody-PROTAC conjugates enable HER2-dependent targeted protein degradation of BRD4. ACS Chem. Biol.15, 1306–1312 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Poudel, Y. B., Thakore, R. R. & Chekler, E. P. The new frontier: merging molecular glue degrader and antibody–drug conjugate modalities to overcome strategic challenges. J. Med. Chem.67, 15996–16001 (2024). [DOI] [PubMed] [Google Scholar]
- 131.Zhang, D., Lin, P. & Lin, J. Molecular glues targeting GSPT1 in cancers: a potent therapy. Bioorg. Chem.143, 107000 (2024). [DOI] [PubMed] [Google Scholar]
- 132.Palacino, J. et al. Abstract 3933: ORM-5029: a first-in-class targeted protein degradation therapy using antibody neodegrader conjugate (AnDC) for HER2-expressing breast cancer. Cancer Res.82, 3933 (2022). [Google Scholar]
- 133.Hurvitz S. A. et al. Abstract TPS1114: A phase 1, first-in-human, open label, escalation and expansion study of ORM-5029, a highly potent GSPT1 degrader targeting HER2, in patients with HER2-expressing advanced solid tumors. J. Clin. Oncol. (2023).
- 134.Fu, C. et al. When will the immune-stimulating antibody conjugates (ISACs) be transferred from bench to bedside?. Pharmacol. Res.203, 107160 (2024). [DOI] [PubMed] [Google Scholar]
- 135.Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol.15, 760–770 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wu, Y.-T. et al. Tumor-targeted delivery of a STING agonist improvescancer immunotherapy. Proc. Natl. Acad. Sci. USA119, e2214278119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Bukhalid, R. A. et al. XMT-2056, a HER2-directed STING agonist antibody-drug conjugate, induces innate anti-tumor immune responses by acting on cancer cells and tumor-resident immune cells. Clin. Cancer Res. 10.1158/1078-0432.CCR-24-2449 (2025). [DOI] [PMC free article] [PubMed]
- 138.Metz, H. et al. SBT6050, a HER2-directed TLR8 therapeutic, as a systemically administered, tumor-targeted human myeloid cell agonist. J. Clin. Oncol.38, 3110–3110 (2020). [Google Scholar]
- 139.Klempner, S. et al. 393 A phase 1/2 study of SBT6050 combined with trastuzumab deruxtecan (T-DXd) or trastuzumab and tucatinib with or without capecitabine in patients with HER2-expressing or HER2-amplified cancers. J. Immunother. Cancer9, A426–A426 (2021).
- 140.Dugal-Tessier, J., Thirumalairajan, S. & Jain, N. Antibody-oligonucleotide conjugates: a twist to antibody-drug conjugates. J. Clin. Med.10, 838 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yao, Y. et al. Targeted delivery of PLK1-siRNA by ScFv suppresses Her2+ breast cancer growth and metastasis. Sci. Transl. Med.4, 130ra48–130ra48 (2012). [DOI] [PubMed] [Google Scholar]
- 142.Deonarain, M. P. et al. Small-format drug conjugates: a viable alternative to ADCs for solid tumours?. Antibodies7, 16 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gu, Y., Wang, Z. & Wang, Y. Bispecific antibody drug conjugates: making 1+1>2. Acta Pharm. Sin. B14, 1965–1986 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kast, F. et al. Engineering an anti-HER2 biparatopic antibody with a multimodal mechanism of action. Nat. Commun.12, 3790 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Li, J. Y. et al. A biparatopic HER2-targeting antibody-drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-targeted therapy. Cancer Cell29, 117–129 (2016). [DOI] [PubMed] [Google Scholar]
- 146.Andreev, J. et al. Bispecific antibodies and antibody-drug conjugates (ADCs) bridging HER2 and prolactin receptor improve efficacy of HER2 ADCs. Mol. Cancer Ther.16, 681–693 (2017). [DOI] [PubMed] [Google Scholar]
- 147.Hassani, D. et al. A novel tumor inhibitory hybridoma monoclonal antibody with dual specificity for HER3 and HER2. Curr. Res. Transl. Med.69, 103277 (2021). [DOI] [PubMed] [Google Scholar]
- 148.Ma, Y. et al. BL-B01D1, a first-in-class EGFR–HER3 bispecific antibody–drug conjugate, in patients with locally advanced or metastatic solid tumours: a first-in-human, open-label, multicentre, phase 1 study. Lancet Oncol.25, 901–911 (2024). [DOI] [PubMed] [Google Scholar]
- 149.Zhang, J. et al. Abstract 302MO: Phase I study of iza-bren (BL-B01D1), an EGFR x HER3 bispecific antibody-drug conjugate (ADC), in patients with locally advanced or metastatic breast cancer (BC). ESMO Open10 (2025).
- 150.Xu, R.-H. et al. Safety and efficacy of TQB2102, a novel bispecific anti-HER2 antibody–drug conjugate, in patients with advanced solid tumors: preliminary data from the first-in-human phase 1 trial. J. Clin. Oncol.43, 3003–3003 (2025). [Google Scholar]
- 151.Yao H. et al. Abstract e13138: First-in-human study of JSKN016, a bispecific anti-TROP2/HER3 antibody drug conjugate (ADC): antitumor activity in patients (pts) with metastatic triple-negative breast cancer (mTNBC) and safety results. J. Clin. Oncol. (2025).
- 152.Rautio, J., Meanwell, N. A., Di, L. & Hageman, M. J. The expanding role of prodrugs in contemporary drug design and development. Nat. Rev. Drug Discov.17, 559–587 (2018). [DOI] [PubMed] [Google Scholar]
- 153.Fu, Z., Li, S., Han, S., Shi, C. & Zhang, Y. Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct. Target. Ther.7, 1–25 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Bhagyalalitha, M. et al. Advances in HER2-targeted therapies: from monoclonal antibodies to dual inhibitors developments in cancer treatment. Bioorg. Chem.151, 107695 (2024). [DOI] [PubMed] [Google Scholar]
- 155.Zhou, C. et al. A phase Ia study of a novel anti-HER2 antibody–drug conjugate GQ1001 in patients with previously treated HER2 positive advanced solid tumors. J. Transl. Med.23, 37 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Lu, J. M. et al. Targeting HER2-positive metastatic breast cancer with ARX788, a novel anti-HER2 antibody-drug conjugate in patients whose disease is resistant or refractory to T-DM1, and/or T-DXd, and/or tucatinib-containing regimens. J. Clin. Oncol.40, TPS1112–TPS1112 (2022). [Google Scholar]
- 157.Hu, X. et al. ACE-Breast-02: a randomized phase III trial of ARX788 versus lapatinib plus capecitabine for HER2-positive advanced breast cancer. Signal Transduct. Target. Ther.10, 56 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hamilton, E. et al. Abstract 298MO: Clinical activity of emiltatug ledadotin (Emi-Le), a B7-H4-directed ADC, in patients with TNBC who received at least one prior topoisomerase-1 inhibitor (Topo-1) ADC. ESMO Open10 (2025).
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analyzed during the current study.


