Antibody–drug conjugates (ADC) using DNA topoisomerase I inhibitor DXd/SN-38 have transformed cancer treatment, yet more effective ADCs are needed for overcoming resistance. We have designed an ADC class using a novel self-immolative T moiety for traceless conjugation and release of exatecan, a more potent topoisomerase I inhibitor with less sensitivity to multidrug resistance (MDR). Characterized by enhanced therapeutic indices, higher stability, and improved intratumoral pharmacodynamic response, antibody–T moiety–exatecan conjugates targeting HER2, HER3, and TROP2 overcome the intrinsic or treatment resistance of equivalent DXd/SN-38 ADCs in low-target-expression, large, and MDR+ tumors. T moiety–exatecan ADCs display durable antitumor activity in patient-derived xenograft and organoid models representative of unmet clinical needs, including EGFR ex19del/T790M/C797S triple-mutation lung cancer and BRAF/KRAS–TP53 double-mutant colon cancer, and show synergy with PARP/ATR inhibitor and anti–PD-1 treatment. High tolerability of the T moiety–exatecan ADC class in nonhuman primates supports its potential to expand the responding patient population and tumor types beyond current ADCs.

Significance:

ADCs combining a novel self-immolative moiety and topoisomerase I inhibitor exatecan as payload show deep and durable response in low-target-expressing and MDR+ tumors resistant to DXd/SN-38 ADCs without increasing toxicity. This new class of ADCs has the potential to benefit an additional patient population beyond current options.

See related commentary by Gupta et al., p. 817.

This article is highlighted in the In This Issue feature, p. 799

Antibody–drug conjugates (ADC) provide targeted tumor-cell delivery of cytotoxic drugs (“payloads”) on antibody carriers. With the approval of more than 10 drugs (7 within the last few years) and more in clinical development, ADCs are undergoing a renaissance (1). This breakthrough is largely driven by a transformative ADC design that uses less potent but safer payload DNA topoisomerase I (TOP1) inhibitor camptothecins (SN38 or DXd) and a higher drug load (DAR 8) to increase therapeutic index, allowing the administration of higher doses of ADC (∼5–10 mg/kg) to improve both drug delivery and tissue penetration without greater toxicity (2). For example, HER2-targeting T-DXd/DS-8201a (Enhertu; Daiichi Sankyo) is composed of highly stable peptide-linker GGFG and payload DXd, which is a derivative of camptothecin exatecan that is 10 to 100 times less potent than microtubule-targeting or DNA-alkylating agents. However, its strong bystander-killing effect overcomes resistance to T-DM1/trastuzumab (Tras), making it the first ADC effective against low HER2 expression tumors (3, 4). IMMU-132 (sacituzumab govitecan/Trodelvy; Gilead Sciences) targeting TROP2 using another TOP1 inhibitor SN-38 as the payload was the first effective ADC for triple-negative breast cancer (5). Additional GGFG–DXd ADCs, including U3-1402 (P–GGFG–DXd) targeting HER3 and datopotamab DXd (Dato-DXd) targeting TROP2, have shown promising therapeutic outcomes in patients, highlighting the unprecedented clinical success of using camptothecin analogues as ADC payload (6, 7).

Despite the breakthrough, DXd and SN-38 ADCs face intrinsic and acquired resistance due to tumor heterogeneity and multidrug resistance (MDR) regulation (8, 9). The typical treatment response rate among patients for payload-sensitive tumor types is only 30% to 50% due to tumor heterogeneity, as low target expression or high intratumoral heterogeneity results in more resistant tumors. DS-8201a generates an initial response of less than 50% and less than 30% in HER2-low-expressing breast and gastric cancers, respectively (10, 11). In non–small cell lung cancer (NSCLC), DS-8201a has improved the response rate of T-DM1 in HER2-overexpressing (IHC 3+ and 2+) patients, but objective response rate (ORR) is still less than 30% (12). DXd ADCs targeting HER3 and TROP2 are similarly less effective in tumors with low target expression in preclinical studies (6, 13). In clinical trials, U3-1402 is effective in patients with HER3-expressing, EGFR-mutated NSCLC, with an ORR of 39%, and patients with higher HER3 expression tend to have better responses (7). TROP2 downregulation due to target mutation after initial responses can cause an acquired resistance for Trodelvy (14). Furthermore, certain tumor types may be more challenging for DXd ADCs, as no meaningful response is observed for the majority of colon cancer cases (HER2 low) in the DS-8201a trial (15). In addition, DXd/SN-38 ADCs may be ineffective in MDR+ tumors due to a higher DXd/SN-38 efflux by the MDR transporter ABCG2 or P-gp (16, 17). Delineation of the inherent resistance mechanism associated with the current TOP1 inhibitor–based ADC class and the design of a limitation-overcoming ADC are clinically imperative.

Exatecan, a camptothecin and the precursor of DXd, has attracted initial attention due to its higher TOP1 inhibition potency (18, 19), lower sensitivity to ABCG2 or P-gp (20), and higher permeability and bystander penetration (21). However, exatecan is too hydrophobic to be conjugated directly to antibodies (22). Commonplace hydrophilic structures such as the β-glucuronidase linker or a peptide-linker modified with polyethylene glycol (PEG) are often used for masking exatecan (23–25). However, β-glucuronidase linkers have not advanced in clinical development, and the significant structural and chemistry changes in dipeptide linker Val-Ala (VA) or Val-Cit (VC) may affect their clinical developability (24–26). As linker chemistry design, payload conjugation strategy, and release mechanism affect physicochemical and pharmacologic performance of an ADC, a holistic approach for ADC design, mechanistic investigation, and a comprehensive comparison of an antibody–exatecan conjugate with the current ADC class are required to achieve successful clinical translation and authentic performance improvement.

To accomplish this goal, here we designed an antibody–exatecan conjugate class with an improved therapeutic index over DXd/SN-38 ADCs by biochemically and pharmacologically profiling exatecan versus DXd/SN-38 as an ADC payload. A novel chemistry (T moiety) compatible with clinically proven ADC linkers was introduced to mask exatecan hydrophobicity for traceless conjugation and exatecan release. T moiety (T800–T1000) translated a superior exatecan payload feature into ADCs, with improved physicochemical and pharmacologic performance demonstrating higher stability, deeper tumor penetration, and more durable antitumor responses than DXd/SN-38 ADCs. T moiety–exatecan ADCs were more effective against large-size tumors due to a higher intratumor pharmacodynamic response and were able to overcome MDR+ (high ABCG2/P-gp expression) tumor resistance to DXd/SN-38 ADCs in vivo, including challenging patient-derived xenograft (PDX) and organoid models. Despite higher potency, T moiety–exatecan ADCs did not incur higher toxicity in nonhuman primates. Initial exploration of combination therapies suggested that exatecan and its ADC had improved synergistic efficacy with DNA damage response (DDR) pathway inhibitors in colon cancer. Our data suggest that T moiety–exatecan ADCs may overcome resistance mechanisms to elicit responses from a larger patient population and a broader variety of tumors compared with prior ADCs.

Exatecan Is a Potent TOP1 Inhibitor Payload with Low Sensitivity to an MDR Transporter–Mediated Efflux

Biochemical and Structural Basis of Exatecan as a Higher Potency TOP1 inhibitor

DXd was derived from exatecan by the chemical modification of NH2 on the F-ring by a 2-hydroxyacetyl (COCH2OH) group (Supplementary Fig. S1A). Exatecan showed an average of 10 to 20 times more in vitro cytotoxic potency than DXd with subnanomolar IC50 (Supplementary Fig. S1B; Supplementary Table S1) in a panel of cancer cell lines.

Cell-based TOP1 inhibition assays and structural modeling provided a mechanism of higher cytotoxic potency of exatecan. The level of DNA-trapped TOP1 (TOP1ccs) of exatecan in comparison with DXd and SN-38 was examined by a modified rapid approach to DNA adduct recovery (RADAR) assay (21). Exatecan was the most potent drug and induced TOP1ccs at a lower concentration than DXd and SN-38 (Fig. 1A; Supplementary Fig. S1C). DNA-trapped TOP1 is typically removed from DNA and degraded. To measure the rate of TOP1 degradation, cells were treated with exatecan or DXd/SN-38 for 2 hours and then allowed to grow without drugs for 30 minutes for the reversal of TOP1ccs and TOP1 degradation. Exatecan induced TOP1 degradation in a dose-dependent manner and was more effective than DXd and SN-38 (Fig. 1B).

Figure 1.

Exatecan potency, MDR sensitivity, and toxicity. A, Detection of DNA-trapped TOP1 by exatecan, DXd, and SN-38. Left, COLO205 cells were treated with the indicated drug concentrations for 30 minutes. TOP1ccs were isolated by RADAR assay. Right, quantitation of TOP1ccs from exposure image (left) in a single experiment. The intensity of TOP1 was analyzed by ImageJ software and normalized to DNA loading. Data were plotted with GraphPad Prism 8. B, TOP1 degradation induced by exatecan. The experimental scheme is shown. Top, COLO205 cells were incubated with the indicated TOP1 inhibitors for 2 hours. After TOP1 reversal for 30 minutes without inhibitors, TOP1 levels were determined by Western blotting. Bottom, quantification of total cellular TOP1 bands. Band intensity was analyzed using the ImageJ software and normalized to Tubulin used as a loading control. C, Modeling of exatecan (yellow) and DXd (light gray) bound to human TOP1–DNA structure (PDB: 1T8I). The structural coordinates of exatecan (PubChem: 151115) and DXd (PubChem: 117888634) were downloaded from the NCBI PubChem Compound (http://www.ncbi.nlm.nih.gov/pccompound) database in 3D SDF file format. Structural modeling by overlapping exatecan and DXd in the TOP1–DNA structure was carried out using Schrödinger and was graphically presented using PyMOL. A red solid dot indicates the primary NH2 (amine) on exatecan that can make two additional hydrogen bonds (shown as dashed lines) with the +1 DNA base oxygen (Base) and the N352 residue of TOP1. D, Efflux ratios for exatecan/DXd/SN-38 in Caco-2 cells with/without inhibitors. Experiments were carried out in Transwell plates for 2 hours at 37°C in the presence or absence of the P-gp inhibitor verapamil, the ABCG2 inhibitor novobiocin, or the dual inhibitor GF120918 (10 μmol/L). Values represent ratios of mean (B to A) to mean (A to B; mean from 3 measurements). E, Representative fluorescence images of intracellular accumulation of exatecan and DXd in NCI-H460 after a 4-hour incubation of cells with drugs. Top, DXd fluorescence was not observed (middle) in the same area with cells observed under brightfield (left). Self-fluorescence (blue) of exatecan was evident (right) in cells (no brightfield cell image control). Bottom, the ABCG2 inhibitor resperpine increased DXd accumulation in cells (middle) but did not affect exatecan accumulation (right). At left is a brightfield of the same area as middle. Scale bar, 20 μm. F, IC50 ratio of DXd/exatecan in high ABCG2 (excluding high P-gp)/P-gp (excluding high ABCG2) and low ABCG2/P-gp mRNA expression cancer cells. Cytotoxicity data are from Supplementary Fig. S1B, and ABCG2/P-gp expression data are from Supplementary Fig. S1D and S1E. ABCG2-high cell lines included ABCG2 mRNA expression >2 and P-gp mRNA <2. P-gp–high cell lines were P-gp mRNA >2 and ABCG mRNA <3. G, ABCG2 and P-gp inhibitors sensitize cancer cells to DXd and SN-38 but not to exatecan. Cytotoxicity of exatecan, DXd, and SN-38 was compared with/without the ABCG2 inhibitor YHO-13177 (ASPC-1 and H2170) or P-gp inhibitor tariquidar (COLO320DM and HCT-15). SKOV3 and COLO205 have low expression of ABCG2/P-gp. Data represent 2 independent measurements. Change of IC50 is quantified. No change of IC50 is labeled by a red dashed line.

Figure 1.

Exatecan potency, MDR sensitivity, and toxicity. A, Detection of DNA-trapped TOP1 by exatecan, DXd, and SN-38. Left, COLO205 cells were treated with the indicated drug concentrations for 30 minutes. TOP1ccs were isolated by RADAR assay. Right, quantitation of TOP1ccs from exposure image (left) in a single experiment. The intensity of TOP1 was analyzed by ImageJ software and normalized to DNA loading. Data were plotted with GraphPad Prism 8. B, TOP1 degradation induced by exatecan. The experimental scheme is shown. Top, COLO205 cells were incubated with the indicated TOP1 inhibitors for 2 hours. After TOP1 reversal for 30 minutes without inhibitors, TOP1 levels were determined by Western blotting. Bottom, quantification of total cellular TOP1 bands. Band intensity was analyzed using the ImageJ software and normalized to Tubulin used as a loading control. C, Modeling of exatecan (yellow) and DXd (light gray) bound to human TOP1–DNA structure (PDB: 1T8I). The structural coordinates of exatecan (PubChem: 151115) and DXd (PubChem: 117888634) were downloaded from the NCBI PubChem Compound (http://www.ncbi.nlm.nih.gov/pccompound) database in 3D SDF file format. Structural modeling by overlapping exatecan and DXd in the TOP1–DNA structure was carried out using Schrödinger and was graphically presented using PyMOL. A red solid dot indicates the primary NH2 (amine) on exatecan that can make two additional hydrogen bonds (shown as dashed lines) with the +1 DNA base oxygen (Base) and the N352 residue of TOP1. D, Efflux ratios for exatecan/DXd/SN-38 in Caco-2 cells with/without inhibitors. Experiments were carried out in Transwell plates for 2 hours at 37°C in the presence or absence of the P-gp inhibitor verapamil, the ABCG2 inhibitor novobiocin, or the dual inhibitor GF120918 (10 μmol/L). Values represent ratios of mean (B to A) to mean (A to B; mean from 3 measurements). E, Representative fluorescence images of intracellular accumulation of exatecan and DXd in NCI-H460 after a 4-hour incubation of cells with drugs. Top, DXd fluorescence was not observed (middle) in the same area with cells observed under brightfield (left). Self-fluorescence (blue) of exatecan was evident (right) in cells (no brightfield cell image control). Bottom, the ABCG2 inhibitor resperpine increased DXd accumulation in cells (middle) but did not affect exatecan accumulation (right). At left is a brightfield of the same area as middle. Scale bar, 20 μm. F, IC50 ratio of DXd/exatecan in high ABCG2 (excluding high P-gp)/P-gp (excluding high ABCG2) and low ABCG2/P-gp mRNA expression cancer cells. Cytotoxicity data are from Supplementary Fig. S1B, and ABCG2/P-gp expression data are from Supplementary Fig. S1D and S1E. ABCG2-high cell lines included ABCG2 mRNA expression >2 and P-gp mRNA <2. P-gp–high cell lines were P-gp mRNA >2 and ABCG mRNA <3. G, ABCG2 and P-gp inhibitors sensitize cancer cells to DXd and SN-38 but not to exatecan. Cytotoxicity of exatecan, DXd, and SN-38 was compared with/without the ABCG2 inhibitor YHO-13177 (ASPC-1 and H2170) or P-gp inhibitor tariquidar (COLO320DM and HCT-15). SKOV3 and COLO205 have low expression of ABCG2/P-gp. Data represent 2 independent measurements. Change of IC50 is quantified. No change of IC50 is labeled by a red dashed line.

Close modal

Camptothecins trap TOP1ccs by π–π stacking with base pairs flanking the DNA cleavage site and by three hydrogen bonds with TOP1 residues (R364, D533, and N722; Fig. 1C; ref. 27). Docking simulation suggested that the amino group of exatecan may form two additional hydrogen bonds with the DNA base and the N352 residue of TOP1 (Fig. 1C). This interaction, however, was eliminated with the 2-hydroxyacetyl replacement of the amino group in DXd, possibly contributing to a decreased TOP1cc activity.

Lipophilic ligand efficiency (LLE) measures in vitro potency and lipophilicity. Increasing LLE values has been used to optimize absorption, distribution, metabolism, excretion, and toxicity (ADMET) characteristics (28). Exatecan showed the highest LLE value of 10.08 compared with SN-38 (6.41) and DXd (7.61), reflecting improved “drug-like” properties of exatecan in terms of increased activity and lowered lipophilicity (Supplementary Fig. S1D).

Exatecan Shows Less Sensitivity to MDR Mechanisms than DXd and SN38

The MDR transporter ABCG2- and P-gp–mediated efflux of exatecan, DXd, and SN-38 was determined in Caco-2 cells (Supplementary Fig. S1E). DXd showed an order of magnitude higher efflux ratios than exatecan without or with the ABCG2 inhibitor novobiocin, the P-gp inhibitor verapamil, or the dual inhibitor GF120918 (Fig. 1D). Interestingly, SN-38 efflux ratios were higher than exatecan but lower than DXd. Immunofluorescence (IF) examination of the intracellular accumulation of exatecan and DXd showed that DXd accumulation was lower than exatecan in the ABCG2 high expression cell line NCI-H460 (Fig. 1E; Supplementary Fig. S1F–1J).

Consistent with MDR substrate experiment results, the cytotoxic potency difference between DXd and exatecan showed a correlation with endogenous ABCG2/P-gp expression (mRNA and protein; Supplementary Fig. S2A–S2C). In general, the IC50 ratio of DXd/exatecan was higher in cells with a higher ABCG2 or P-gp expression (Fig. 1F; Supplementary Fig. S2D and S2E). The inhibition of ABCG2 or P-gp improved the cytotoxicity of DXd/SN-38 but had a much smaller effect on exatecan (Fig. 1G). Even with an improved IC50 with inhibitor for DXd/SN-38, exatecan remained more potent (Supplementary Fig. S2F and S2G). Collectively, exatecan demonstrated less sensitivity to the MDR genes than DXd/SN38, potentially conferring higher cytotoxicity in cancer cells along with a more prevalent resistance mechanism.

Exatecan Mesylate Is Well Tolerated in Rats

We conducted a 4-week (days 1, 8, 15, 22, and 29) intermittent intravenous dose toxicity study of exatecan mesylate in rats (6 animals/group) with a 4-week recovery period (3 of the 6 animals). Exatecan mesylate was well tolerated in rats at doses up to 10 mg/kg (calculated as exatecan-free base). At 30 mg/kg, exatecan mesylate resulted in decreased animal body weight, morbidity (decreased food consumption, abnormal clinical signs), and/or mortality (occurred 5 or 6 days after dose). The dose-dependent transient suppression of body weight gain (Supplementary Fig. S3A), decrease of blood cell counts (Supplementary Fig. S3B–S3E), and increase of key serum enzyme levels (Supplementary Fig. S3F and S3G) were observed in 3 mg/kg and 10 mg/kg groups but largely reversible by the end of the 4-week recovery period (Supplementary Table S2), suggesting that exatecan was similarly tolerated to DXd or SN-38 (16).

T Moiety Conjugation Translates Exatecan into a Superior ADC: Physicochemical and Pharmacologic Profile, Stability, and Toxicity

T Moiety Design: Hydrophilic Modulation of the PABC Spacer for Traceless Conjugation

To enable a traceless conjugation and release of exatecan, we used a dipeptide VA linker and a modified self-immolative spacer p-amino benzyl (pAB) called T moiety (Fig. 2A; S1 in Supplementary Fig. S4A). VA was selected because it creates more hydrophilic high-DAR ADCs compared with other peptide linkers VC or Gly–Gly–Phe–Gly (GGFG; S0 in Supplementary Fig. S4A; refs. 29–30). Direct conjugation of HER2-targeting Tras and exatecan using the unmodified peptide linker VA/VC/GGFG caused high aggregation (Supplementary Fig. S4B). Integrating T moiety with a PEG group (T900; S3 in Supplementary Fig. S4A) significantly reduced aggregation to an acceptable level of 2%. However, modification of MC with the same PEG (called M moiety, M900; S2 in Supplementary Fig. S4A) led to a high aggregation of >50% (Supplementary Fig. S4B), highlighting the importance of the judicious selection of modification position. Considering the potential lethal adverse reactions caused by PEG-based biological drugs (31), we decided to use polysarcosine (pSAR) due to its higher solubility and biocompatibility (Supplementary Fig. S4C; ref. 32). Polysarcosine with 10 units (pSAR10) modified pAB (T1000) resulted in homogeneous ADC with a negligible level of aggregation (Supplementary Fig. S4B). Remarkably, an intermediate structure T800 with m-methylaminomethyl modification of pAB for attaching pSAR10 produced homogeneous ADC of high DAR without aggregation. In comparison, the attachment of the same m-methylaminomethyl group to the VA linker (VK-pABC, M800, CLogP = 4.98) caused 10% aggregation with exatecan even when T800 (CLogP = 4.73) and M800 are considered to be chemically equivalent, supporting pAB selection as the optimal modification site. The conjugation yield for T moiety ADCs was >90%. Replacing the VA linker with GGFG in T1000 also yielded homogeneous ADC without aggregation (T1001; Supplementary Fig. S4B).

Figure 2.

T1000–exatecan generates ADCs with a superior physicochemical and pharmacologic profile. A, Antibody–exatecan conjugate by T moiety. Exatecan is attached to a modified (by X) self-immolative p-aminobenzyl carbamate (PABC) spacer (indicated by a red scissor). T moiety modification is shown in the box. The VA linker and maleimidocaproyl (MC) spacer are labeled. An antibody is shown on the left. B,In vitro stability of Tras–T1000–exatecan/MTX-1000 in human and monkey plasma measured by toxin release compared with that of Tras–GGFG–DXd. C, Pharmacokinetics of Tras–GGFG–DXd and MTX-1000 in rats. ADCs were intravenously administered to rats at the dose of 4 mg/kg (n = 5). D, Correlation of exatecan release and target expression. HER2 expression on each cell line was determined by flow cytometry, and exatecan concentration in the culture media at 24 hours after treatment with 100 nmol/L Tras–T1000–exatecan was determined by LC/MS-MS (n = 3). MFI, mean fluorescence intensity. E, Result tabulation of detection of DNA-trapped TOP1 by MTX-1000 and DS-8201a. Data are shown in Supplementary Fig. S9E. Payload data from Fig. 1A are included (dashed curve). F, Result tabulation of TOP1 degradation induced by MTX-1000 and DS-8201a. Data are shown in Supplementary Fig. S9F. G, Patient-derived organoid (PDO) response to MTX-1000 and DS-8201a. Representative images of brightfield (left) and live/dead cells (right) for two individual organoids are shown. Colon cancer PDOs were treated with MTX-1000 or DS-8201a at a concentration of 50 μg/mL for 6 days. Live cells were stained by calcein AM (green) and dead cells by propidium iodide (PI; red). Scale bar, 20 μm. H, Organoid size at days 0 and 6 of ADC treatment. A concentration series of DS-8201a or MTX-1000 was incubated with an organoid for 6 days. Surviving organoid size was measured (see ref. 34). Data shown are means ± SE from 2 independent measurements. I, Quantification of ADC penetration of organoid. Spatial distribution of PI-staining signal of MTX-1000 versus DS-8201a from the organoid surface to core. The analysis was done using a scheme shown in Supplementary Fig. S10F and by ImageJ software. J, Bystander killing effect of MTX-1000, T-DM1, and Tras–GGFG–DXd in coculture conditions in vitro. HCC1954 and MDA-MB-468 cells were cocultured and treated with 10 nmol/L ADCs for 5 days. Cell number and ratio of HER2-positive and HER2-negative cells were determined by a cell counter and a flow cytometer, respectively, after collecting adherent cells. Numbers of HCC1954 and MDA-MB-468 viable cells. Each bar represents the mean and SD (n = 3). K, Myelotoxicity potential of MTX-1000/DS-8201a/Trodelvy and exatecan/DXd/SN-38. Concentration-dependent inhibition of the CellTiter-Glo 3D (CTG-3D) assay with CD34+ human bone marrow hematopoietic stem/progenitor cells. Cells (250 μL/well) were plated in 96 wells for expansion and early differentiation by SFEMII basal medium and myeloid-specific cytokine cocktail. 5-FU and compounds were added on day 1, and cells were transferred into solid-bottomed, 96-well, white plates on day 9. Equal volume CTG-3D was added to detect cell viability and measure the luminescence using a luminometer and calculate the IC50 value for each lineage. Each plot represents mean ± SD (n = 3). Left, data for ADC. Middle, data for payload. Right, IC50 ratio of payload and ADC.

Figure 2.

T1000–exatecan generates ADCs with a superior physicochemical and pharmacologic profile. A, Antibody–exatecan conjugate by T moiety. Exatecan is attached to a modified (by X) self-immolative p-aminobenzyl carbamate (PABC) spacer (indicated by a red scissor). T moiety modification is shown in the box. The VA linker and maleimidocaproyl (MC) spacer are labeled. An antibody is shown on the left. B,In vitro stability of Tras–T1000–exatecan/MTX-1000 in human and monkey plasma measured by toxin release compared with that of Tras–GGFG–DXd. C, Pharmacokinetics of Tras–GGFG–DXd and MTX-1000 in rats. ADCs were intravenously administered to rats at the dose of 4 mg/kg (n = 5). D, Correlation of exatecan release and target expression. HER2 expression on each cell line was determined by flow cytometry, and exatecan concentration in the culture media at 24 hours after treatment with 100 nmol/L Tras–T1000–exatecan was determined by LC/MS-MS (n = 3). MFI, mean fluorescence intensity. E, Result tabulation of detection of DNA-trapped TOP1 by MTX-1000 and DS-8201a. Data are shown in Supplementary Fig. S9E. Payload data from Fig. 1A are included (dashed curve). F, Result tabulation of TOP1 degradation induced by MTX-1000 and DS-8201a. Data are shown in Supplementary Fig. S9F. G, Patient-derived organoid (PDO) response to MTX-1000 and DS-8201a. Representative images of brightfield (left) and live/dead cells (right) for two individual organoids are shown. Colon cancer PDOs were treated with MTX-1000 or DS-8201a at a concentration of 50 μg/mL for 6 days. Live cells were stained by calcein AM (green) and dead cells by propidium iodide (PI; red). Scale bar, 20 μm. H, Organoid size at days 0 and 6 of ADC treatment. A concentration series of DS-8201a or MTX-1000 was incubated with an organoid for 6 days. Surviving organoid size was measured (see ref. 34). Data shown are means ± SE from 2 independent measurements. I, Quantification of ADC penetration of organoid. Spatial distribution of PI-staining signal of MTX-1000 versus DS-8201a from the organoid surface to core. The analysis was done using a scheme shown in Supplementary Fig. S10F and by ImageJ software. J, Bystander killing effect of MTX-1000, T-DM1, and Tras–GGFG–DXd in coculture conditions in vitro. HCC1954 and MDA-MB-468 cells were cocultured and treated with 10 nmol/L ADCs for 5 days. Cell number and ratio of HER2-positive and HER2-negative cells were determined by a cell counter and a flow cytometer, respectively, after collecting adherent cells. Numbers of HCC1954 and MDA-MB-468 viable cells. Each bar represents the mean and SD (n = 3). K, Myelotoxicity potential of MTX-1000/DS-8201a/Trodelvy and exatecan/DXd/SN-38. Concentration-dependent inhibition of the CellTiter-Glo 3D (CTG-3D) assay with CD34+ human bone marrow hematopoietic stem/progenitor cells. Cells (250 μL/well) were plated in 96 wells for expansion and early differentiation by SFEMII basal medium and myeloid-specific cytokine cocktail. 5-FU and compounds were added on day 1, and cells were transferred into solid-bottomed, 96-well, white plates on day 9. Equal volume CTG-3D was added to detect cell viability and measure the luminescence using a luminometer and calculate the IC50 value for each lineage. Each plot represents mean ± SD (n = 3). Left, data for ADC. Middle, data for payload. Right, IC50 ratio of payload and ADC.

Close modal

Tras–T1000–Exatecan (MTX-1000) Shows a Superior Physicochemical Profile

To compare the physicochemical parameters of Tras–T1000–exatecan (MTX-1000) with those of DS-8201a, we used either purchased DS-8201a or internally manufactured Tras–GGFG–DXd with closely matched physicochemical profiles (Supplementary Fig. S5A and S5B), in vivo potency (Supplementary Fig. S5C–S5E), and pharmacokinetics (PK) in mice (Supplementary Fig. S5F). The attachment of exatecan to the HER2 antibody did not affect HER2 antibody binding to target by flow cytometry or ELISA, similar to Tras–GGFG–DXd (Supplementary Fig. S6A and S6B). Tras–exatecan conjugates using T800 or T900 also resulted in more hydrophilic ADCs than Tras–GGFG–DXd (Supplementary Fig. S6C and S6D). Both DAR8 and DAR4 ADCs were readily generated (Supplementary Fig. S6E).

Tras–T moiety–exatecan ADCs (Tras–T800–exatecan/MTX-800 and Tras–T1000–exatecan/MTX-1000, both DAR8) demonstrated similar or better stability than Tras–GGFG–DXd. This was measured by aggregation formed for an extended incubation time at 37°C and under repeat freeze–thaw cycles (Supplementary Fig. S6F). T moiety–exatecan ADCs also showed better thermostability and photostability than DS-8201a (Supplementary Fig. S6G and S6H). Notably, VA and pSAR ADC (T1000) showed better stability and less aggregation than other linker (GGFG) and modification (PEG; T900 or T1001; Supplementary Fig. S6C, S6D, S6F, and S6G), highlighting the impact of linker and conjugation chemistry choice on the physicochemical function of an ADC.

T Moiety for Building ADCs from Additional Target–Antibody Pairs and Payload

T moiety–exatecan-derived ADCs targeting HER3 with patritumab (P; DAR 8), TROP2 using datopotamab (Dato; Dato–T1000–exatecan) or hRS7 antibody from Trodelvy (MTX-132; DAR 4), and CDH6 using DS-6000 antibody (DS6000; see Methods) were more hydrophilic than DXd-based ADCs using the same antibodies, and all ADCs were homogeneous without aggregation [Supplementary Fig. S7A–S7C; hydrophobic interaction chromatography (HIC), size-exclusion chromatography]. T1000 was successfully used to build homogeneous, hydrophilic ADC using Tras and another TOP1 inhibitor belotecan (Supplementary Fig. S7D and S7E). HER2-targeting belotecan ADC showed potent, specific in vitro cytotoxicity in a HER2-high N87 cell line (no activity in HER2-negative MDA-MB-468; Supplementary Fig. S7F). Together, T moiety was proven to be useful for conjugating challenging cytotoxic compounds as ADC payloads.

In Vitro Plasma Stability and In Vivo PK Studies of MTX-1000

The release rate of exatecan from MTX-1000 ranged from 0.4% to 3.6% on day 14 in human, monkey, rat, and mouse plasma by immunocapture followed by reverse-phase LC/MS-MS, compared with 1.4% to 6.6% for Tras–GGFG–DXd (Fig. 2B; Supplementary Fig. S8A), consistent with antibody and ADC stability data (Supplementary Fig. S8B and S8C; Supplementary Table S3). MTX-1000 in particular released more than 10 times less payload in human/monkey plasma than Tras–GGFG–DXd (Fig. 2B; Supplementary Table S3). In vivo, after a single intravenous administration of MTX-1000 at 4 mg/kg in rats, the PK profile of MTX-1000 was similar to that of total antibody (Fig. 2C). MTX-1000 is more stable than Tras–GGFG–DXd, as demonstrated by its longer half-life (T1/2 4.14 ± 1.45 for Tras–GGFG–DXd vs. 7.4 ± 1.5 for MTX-1000) and lower payload release, which resulted in over 50% more AUC for both total antibody and ADC (Supplementary Fig. S8D and S8E; Supplementary Table S4). The higher stability of MTX-1000 was consistent with the more hydrophilic nature of the molecule.

Superior Pharmacologic Profile of MTX-1000: Higher Cellular Stability, Higher TOP1 Inhibition Potency, Better Tumor Penetration, and Stronger Bystander Effect

The internalization rate of MTX-1000 in cancer cell lines was similar to Tras–GGFG–DXd (Supplementary Fig. S9A–S9C; Supplementary Table S5). There was an inverse relationship between HER2 expression level and internalization rate, consistent with a previous report (33). Twenty-four hours after MTX-1000 was internalized into the cells, the quantity of exatecan released in the culture media correlated with the cell-surface HER2 expression level of each cell line (Fig. 2D; Supplementary Table S6). However, DS-8201a showed a higher DXd release in the culture media regardless of HER2 expression due to a high background DXd release unrelated to cell binding and internalization, as shown by a 20- to 40-fold payload release increase in the media (without cells) compared with MTX-1000 at 8 and 24 hours, respectively (Supplementary Fig. S9D and S9E).

MTX-1000 demonstrated a superior pharmacologic profile characterized by higher potency and deeper penetration. MTX-1000 induced higher cytotoxic TOP1ccs and faster TOP1 degradation compared with DS-8201a (Fig. 2E and F; Supplementary Fig. S9F and S9G). The TOP1 inhibition effect of MTX-1000 was confirmed by DNA damage (phosphorylations of Chk1 and histone γH2A.X) and apoptosis (cleaved PARP) in HCC1954 cells in the presence of MTX-1000, exatecan (positive), and Tras (negative; Supplementary Fig. S9H). To gain cellular resolution of ADC potency and payload distribution, tumor penetration of MTX-1000 and DS-8201a was evaluated by cell imaging and HER2-positive patient-derived colon cancer organoids (PDO; refs. 21, 34; Supplementary Fig. S10A and S10B). In a PDO, MTX-1000 caused more cell death to prevent organoid growth than DS-8201a (Fig. 2G and H; Supplementary Fig. S10C–S10E). Notably, the cell death signals (PI staining) of MTX-1000 extended from surface to the core of the organoids compared with mostly peripheral staining for DS-8201a (Fig. 2I; Supplementary Fig. S10F and S10G). Similar results were observed in another colon cancer organoid (Supplementary Fig. S10H).

MTX-1000 showed a higher bystander killing efficacy than Tras–GGFG–DXd in an in vitro coculture system of HER2-negative (MDA-MB-468) and HER2-positive (HCC1954) cell lines (Fig. 2J; Supplementary Fig. S11A–S11D), supported by a 10-fold increase in the cell permeability (A–B) of exatecan compared with DXd/SN-38 (Supplementary Fig. S1E).

MTX-1000 Shows Comparable Myelotoxicity to DS-8201a Despite Higher Payload Myelotoxicity

A CellTiter-Glo 3D assay was performed to evaluate the potential myelotoxicity of ADC and payload. The IC50 of MTX-1000, DS-8201, and Trodelvy was 1.24 μg/mL, 1.47 μg/mL, and 0.15 μg/mL, respectively (Fig. 2K, left). MTX-1000 and DS-8201a showed comparable myelotoxicity despite the 5-fold myelotoxicity potential difference between exatecan and DXd (Fig. 2K, middle and right). MTX-1000's higher stability likely allowed for its decreased myelotoxicity. Trodelvy showed much lower IC50 due to known instability in releasing payload SN-38.

Safety Profiles of MTX-1000 and T1000–exatecan ADCs

MTX-1000 toxicity was evaluated in an exploratory cynomolgus monkey (cross-reactive species) toxicity study (every 3 weeks × 3 at 10 mg/kg and 30 mg/kg). The target organ of toxicity is summarized in Table 1. Primary toxicity findings were found only in lymphatic/hematopoietic organs and digestive systems in monkeys. All findings were minimal or mild, and almost all treatment-related changes showed recovery or a trend of reversibility after a 1-week withdrawal period. Reticulocyte counts showed decreases and rebound beyond the normal range, whereas other hematologic indicator changes were largely within the normal range (Supplementary Fig. S12A–S12G). Serum chemistry also remained normal (Supplementary Fig. S12H and S12I). The manifested digestive system toxicity symptom was limited to only mild diarrhea. Gastrointestinal (GI) toxicity and bone marrow toxicity are typical dose-limiting factors in the clinical use of TOP1 inhibitors, but the effects of MTX-1000 on the intestines were minimal. Based on the toxicology findings, the highest nonseverely toxic dose (HNSTD) for monkeys was considered to be 30 mg/kg, the same as DS-8201a. However, macroscopic toxicity findings in the intestines, bone marrows, and particularly in the lung shown in DS-8201a studies were not observed (Supplementary Fig. S12J). These findings suggest safety profiles of MTX-1000 are similar to or better than those of Tras–GGFG–DXd despite higher antitumor potency. Similarly, TROP2-targeting MTX-132 also displayed mild clinical signs only (skin discolored at forelimb and hindlimb) and its HNSTD was judged to be >30 mg/kg compared with a reported HNSTD of 10 mg/kg for Dato–DXd (7).

Table 1.

Toxicity profiles of MTX-1000 in monkey

SpeciesCynomolgus monkey
Doses 10 and 30 mg/kg 
Regimens Intravenous, every 3 week Days 1, 22, 43 (3 times in total) 
Number of animals 1/sex/group 
Body weight ≤30 mg/kg: normal 
Hematology 10 mg/kg: normal 
 30 mg/kg: decreased RET parameters 
Serum chemistry ≤30 mg/kg: normal 
Gross pathology 10 mg/kg: normal 
 30 mg/kg: small-sized thymuses 
Histopathology 30 mg/kg: lymphocyte moderate reduction 
HNSTD 30 mg/kg 
SpeciesCynomolgus monkey
Doses 10 and 30 mg/kg 
Regimens Intravenous, every 3 week Days 1, 22, 43 (3 times in total) 
Number of animals 1/sex/group 
Body weight ≤30 mg/kg: normal 
Hematology 10 mg/kg: normal 
 30 mg/kg: decreased RET parameters 
Serum chemistry ≤30 mg/kg: normal 
Gross pathology 10 mg/kg: normal 
 30 mg/kg: small-sized thymuses 
Histopathology 30 mg/kg: lymphocyte moderate reduction 
HNSTD 30 mg/kg 

NOTE: Summary of repeated dose toxicity studies in monkeys.

Abbreviations: HNSTD, highest nonseverely toxic dose; RET, reticulocyte.

T Moiety–Exatecan ADCs Show an Enhanced Therapeutic Index and Ability to Overcome Drug Resistance Mechanisms

Quantitative Therapeutic Index Assessment

In vitro, HER-targeting MTX-1000, TROP2-targeting MTX-132, and HER3-targeting P–T1000–exatecan were more cytotoxic than their respective counterpart DXd ADC, especially in low-target-expression cells (Supplementary Fig. S13A–S13C; Supplementary Table S7).

Monkey HNSTD or the mouse PK method was used to assess the therapeutic index of MTX-1000 and MTX-132 against DS-8201a and Trodelvy/Dato–DXd (35). First, minimum effective doses (MED) for inducing tumor regression in vivo were measured by dose–efficacy titration. In HCC1954 with a medium HER2 expression, the MED of MTX-1000 was about one third of DS-8201a (Supplementary Fig. S13D). However, in the HER2-low-expression model COLO205, only 10 mg/kg of MTX-1000 caused tumor regression, whereas 10 mg/kg of DS-8201a was only partially effective (Supplementary Fig. S13E). In the TROP2-high-expression model H2170, the MED of MTX-132 was (single dose) less than 5 mg/kg compared with four doses of 25 mg/kg of Trodelvy (Supplementary Fig. S13F). Dato–GGFG–DXd was less effective even at 10 mg/kg (Supplementary Fig. S13F). In COLO205 with a medium TROP2 expression, 10 mg/kg of MTX-132 produced a tumor regression, but four doses of 25 mg/kg of Trodelvy did not (Supplementary Fig. S13G). Combining HNSTD data from monkey studies, we found that MTX-1000 and MTX-132 improved the therapeutic index two to five times compared with DS-8201a and Trodelvy/Dato–DXd in medium- to high-target-expression models (Supplementary Fig. S13H). The therapeutic index measured by the mouse PK (AUC) of DS-8201a and MTX-1000 yielded a 2-fold improvement (Supplementary Fig. S13H and S13I). In low target expression, tumors resistant to DS-8201a or Trodelvy treatment, MTX-1000, and MTX-132 showed a more significant therapeutic index advantage (Fig. 3A).

Figure 3.

T moiety–exatecan ADCs overcome treatment resistance due to improved therapeutic index and intratumor pharmacodynamic response. A, Therapeutic index comparison based on data from Supplementary Fig. S12D–S12H. MTX-1000/MTX-132 displayed an enhanced therapeutic index compared with DS-8201a/Trodelvy in medium- to high-target-expression tumors due to lowered minimum efficacy dose for tumor regression but similar HNSTD of monkey toxicity study. In low-target-expression tumors (or with high MDR gene expression, see Fig. 4 data) resistant to DS-8201a/Trodelvy, MTX-1000/MTX-132 was still effective. CR, complete response, tumor volume change from baseline % achieve −100%; PR, partial response, tumor volume change from baseline % <0. B, CDH6-targeting DS-6000–T1000–exatecan was effective in a kidney PDX model resistant to DS-6000–GGFG–DXd. Low CDH6 expression is shown as IHC image on untreated mouse tumor tissue and H-score. Scale bar, 20 μm. PDX mice were intravenously administered with indicated ADCs (10 mg/kg) on day 0 (tumor size reached an average of 150–200 mm3). Each value represents the mean and SEM (n = 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, HER3-targeting P–T1000–exatecan caused tumor regression in a lung squamous cell model resistant to P–GGFG–DXd. rMFI, relative mean fluorescence intensity. DF, T moiety–exatecan ADCs overcame DXd/SN-38 ADC treatment resistance. Experimental design and ADC dosing are shown for each model. D, DS-8201a vs. MTX-1000 in COLO025. E, MTX-132 vs. Trodelvy in COLO205. F, P–T1000–exatecan versus P–GGFG–DXd in H2170. G,In vivo tissue distribution of ADC. IHC analysis for γH2AX on COLO205 xenograft tumors treated with control, DS-8201a, and MTX-1000. Each value represents the mean (black bar) and individual data (3 mice, two measurements each). Representative images are shown for days 1 and 3. H and I, Efficacy of MTX-1000 (H) or MTX-132 (I) in large naive tumors. J, Summary of best percentage change of initial tumor volume of MTX-1000 in comparison with DS-8201a. In all in vivo studies, cell line–derived xenograft (CDX) mice were intravenously administered with indicated ADCs (at 10 mg/kg or an indicated dose) on days indicated by an arrow. Each value represents the mean and SEM (n = 5 or otherwise indicated). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. MSS, microsatellite stable.

Figure 3.

T moiety–exatecan ADCs overcome treatment resistance due to improved therapeutic index and intratumor pharmacodynamic response. A, Therapeutic index comparison based on data from Supplementary Fig. S12D–S12H. MTX-1000/MTX-132 displayed an enhanced therapeutic index compared with DS-8201a/Trodelvy in medium- to high-target-expression tumors due to lowered minimum efficacy dose for tumor regression but similar HNSTD of monkey toxicity study. In low-target-expression tumors (or with high MDR gene expression, see Fig. 4 data) resistant to DS-8201a/Trodelvy, MTX-1000/MTX-132 was still effective. CR, complete response, tumor volume change from baseline % achieve −100%; PR, partial response, tumor volume change from baseline % <0. B, CDH6-targeting DS-6000–T1000–exatecan was effective in a kidney PDX model resistant to DS-6000–GGFG–DXd. Low CDH6 expression is shown as IHC image on untreated mouse tumor tissue and H-score. Scale bar, 20 μm. PDX mice were intravenously administered with indicated ADCs (10 mg/kg) on day 0 (tumor size reached an average of 150–200 mm3). Each value represents the mean and SEM (n = 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. C, HER3-targeting P–T1000–exatecan caused tumor regression in a lung squamous cell model resistant to P–GGFG–DXd. rMFI, relative mean fluorescence intensity. DF, T moiety–exatecan ADCs overcame DXd/SN-38 ADC treatment resistance. Experimental design and ADC dosing are shown for each model. D, DS-8201a vs. MTX-1000 in COLO025. E, MTX-132 vs. Trodelvy in COLO205. F, P–T1000–exatecan versus P–GGFG–DXd in H2170. G,In vivo tissue distribution of ADC. IHC analysis for γH2AX on COLO205 xenograft tumors treated with control, DS-8201a, and MTX-1000. Each value represents the mean (black bar) and individual data (3 mice, two measurements each). Representative images are shown for days 1 and 3. H and I, Efficacy of MTX-1000 (H) or MTX-132 (I) in large naive tumors. J, Summary of best percentage change of initial tumor volume of MTX-1000 in comparison with DS-8201a. In all in vivo studies, cell line–derived xenograft (CDX) mice were intravenously administered with indicated ADCs (at 10 mg/kg or an indicated dose) on days indicated by an arrow. Each value represents the mean and SEM (n = 5 or otherwise indicated). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. MSS, microsatellite stable.

Close modal

T moiety–Exatecan ADCs Produce Deeper and More Durable Antitumor Responses in PDX Models

Supported by an improved therapeutic index, T moiety–exatecan ADCs showed superior in vivo antitumor efficacy and a more durable response in clinically relevant PDX models. In a gastric PDX model with high HER expression, MTX-1000 inhibited tumor growth better than Tras–GGFG–DXd, although both ADCs produced only modest inhibition (Supplementary Fig. S14A). In the same gastric PDX model, MTX-132 (DAR 4) showed significantly better tumor inhibition than Dato–GGFG–DXd (DAR 4), with a tumor growth inhibition (TGI) of 65% versus 24% (Supplementary Fig. S14B). In a pancreatic PDX model with moderate HER2 expression, MTX-1000 and MTX-800 initially showed comparable tumor inhibition as Tras–GGFG–DXd but produced more durable tumor inhibition than Tras–GGFG–DXd over 50 days (Supplementary Fig. S14C and S14D). It was noteworthy that in two breast cancer PDX models, Tras–GGFG–DXd and Dato–DXd produced similar levels of tumor inhibition as MTX-1000 and MTX-132 (Supplementary Fig. S14E and S14F), reflecting the clinic observation that breast cancer has been one of the most responsive tumor types for DS-8201a and TROP2-targeting DXd/SN-38 ADCs.

T Moiety–Exatecan ADCs Overcome Intrinsic or Treatment Resistance to DXd ADCs

In low-target-expression tumors, T1000–exatecan ADCs overcame tumor-intrinsic resistance to DXd/SN-38 ADCs. In a CDH6-low-expression (IHC 1+) PDX model of nephroblastoma with hepatic metastasis resistant to DS-6000, T moiety–exatecan ADC using DS-6000 antibody (DS6000–T1000–exatecan, DAR 8) showed strong antitumor activity (Fig. 3B). In H2170 with low HER3 expression, P–GGFG–DXd was ineffective (similar to PBS control) whereas P–T1000–exatecan produced tumor regression (Fig. 3C). COLO205 tumors outgrowth after DS-8201a treatment was tested to see whether T1000–exatecan ADCs could overcome resistance in DXd/SN-38 ADC–treated tumors (Fig. 3D). Those tumors were allowed to grow to 900 mm3 after two doses of DS-8201a. Subsequent administration of one dose of MTX-1000 produced significant tumor regression, whereas one dose of DS-8201a was ineffective (Fig. 3D). MTX-1000 was also effective in growing tumors (at 600 mm3) after DS-8201a treatment in another colon cancer xenograft model, HCT-15 (Supplementary Fig. S15A). Similarly, MTX-132 and P–T1000–exatecan inhibited tumor growth in growing tumors treated with Trodelvy (Fig. 3E; Supplementary Fig. S15B) and Dato–DXd (Supplementary Fig. S15C) or P–GGFG–DXd, respectively (Fig. 3F). In HCT-15, MTX-132 overcame the treatment ineffectiveness of Dato–DXd (Supplementary Fig. S15D). Together, T moiety–exatecan ADCs were effective in DXd/SN-38 ADC treatment–refractory tumors.

T Moiety–Exatecan ADCs Show Superior Intratumor Pharmacodynamic Response and Are Effective against Large Tumors

Following intravenous delivery at the same doses as the efficacy study (Supplementary Fig. S15E), pharmacodynamic response imaging measured by the DNA damaging marker γH2A.X was used to probe the PK profile of MTX-1000 in vivo. Although tumor size difference was insignificant in the first 3 days after ADC administration, the pharmacodynamic marker γH2A.X signal of MTX-1000 was significantly higher than DS-8201a on both days 1 and 3 (Supplementary Fig. S15F and S15G; Fig. 3G).

The improved intratumoral pharmacodynamic response exemplified by MTX-1000 translates into the antitumor effectiveness of T moiety–exatecan ADCs against large-size tumors. In xenograft models of starting tumor size of 500 to 1,000 mm3, MTX-1000 and MTX-132 induced tumor regression and were significantly more effective than DS-8201a and Dato-DXd/Trodelvy, respectively (Fig. 3H and I). Furthermore, MTX-1000 induced tumor regression in the DS-8201a–treated group when the tumor reached 1,300 mm3, whereas another dose of DS-8201a failed to inhibit tumor growth (Fig. 3H). The HER3-targeting ADC P–T1000–exatecan also inhibited tumor growth in tumors 800 mm3 and larger in ASPC-1, demonstrating significantly higher efficacy than P–GGFG–DXd (Supplementary Fig. S15H). The enhanced therapeutic index and improved intratumor pharmacodynamic response of T moiety–exatecan ADCs translate to improved antitumor efficacy in tumors with low target expression, tumors with DXd/SN-38 ADC treatment resistance, or large tumors, as summarized in the efficacy comparison of MTX-1000 versus DS-8201a as an example (Fig. 3J).

T Moiety–Exatecan ADCs overcome MDR to DXd/SN-38 ADCs

MDR transporters such as ABCG2 and P-gp confer chemotherapy resistance, a frequent cause of treatment failure, through active drug efflux. Previously, IMMU-132/Trodelvy was shown to overcome resistance to SN-38 in breast and gastric tumor models due to the combination of the ABCG2 inhibitor YHO-13351 with Trodelvy (17). The effect of MDR dependence of exatecan versus DXd/SN-38 ADCs targeting HER2, HER3, and TROP2 was investigated in representative endogenous ABCG2- and P-gp-high-expression cells (Supplementary Fig. S16A). ABCG2 inhibitor (YHO-13351) and P-gp inhibitor (tariquidar) improved the in vitro cytotoxicity potency of DXd ADCs or Trodelvy significantly but were less effective on IC50 of T1000–exatecan ADCs (Supplementary Fig. S16B–S16D; Fig. 4A), which is largely consistent with payload data (Fig. 1D and G). In vivo, TROP2-targeting Trodelvy or Dato–GGFG–DXd alone was ineffective in HCT-15. When combined with the P-gp inhibitor tariquidar, both showed significant tumor inhibition (Fig. 4B). In comparison, Dato–T1000–exatecan alone was effective. A combination of Dato–T1000–exatecan with tariquidar was slightly more effective (Fig. 4B and C). As control, tariquidar alone or PBS was not effective (Fig. 4C). In HCT-15 and ASPC-1 (high ABCG2), DS-8201a alone was ineffective but led to significant antitumor activity in combination with tariquidar in HCT-15 (Fig. 4D) or the ABCG2 inhibitor YHO-13351 in ASPC-1 (Fig. 4E). MTX-1000 alone or in combination with respective inhibitor was equally effective in both models (Fig. 4DF). Similar results were observed for the HER3-targeting ADC P–GGFG–DXd and counterpart P–T800–exatecan in HCT-15 and ASPC-1 (­Supplementary Fig. S16E; Fig. 4G). The efficacy improvement by YHO-13351 for P–GGFG–DXd in ASPC-1 was short-lived since the tumors started to grow again—even tumor growth in the P–T800–exatecan groups was kept inhibited (Fig. 4G). Finally, in a colon cancer PDX model with low TROP2 and high P-gp expression, tariquidar was able to improve Dato–GGFG–DXd efficacy significantly in large-size (600 mm3) tumor groups (Fig. 4H; Supplementary Fig. S16F). In this model, MTX-132 alone was effective for tumor inhibition (Supplementary Fig. S16F). These data suggest that a combination treatment of DXd/SN-38 ADC with a specific MDR inhibitor or T moiety–exatecan ADC alone was effective for MDR+ tumor resistance.

Figure 4.

Overcoming MDR resistance by T moiety–exatecan ADCs or a combination of MDR inhibitor with DXd/SN-38 ADCs. A,In vitro cytotoxicity improvement by P-gp inhibitor in HCT-15 for HER2/HER3/TROP2-targeting ADCs. IC50 fold change is shown. B and C,In vivo efficacy of TROP2-targeting ADC in HCT-15 with and without the P-gp inhibitor tariquidar. B, Tumor growth and inhibition. n.s., not significant. C, Best percentage change of initial tumor volume from B. Each ADC is labeled by its name and payload used. Tariquidar alone was ineffective in HCT-15. DF, HER2-targeting ADCs in HCT-15 (P-gp) and ASPC-1 (ABCG2) with and without MDR inhibitor. D and E, Tumor growth and inhibition. Tariquidar was used in D, and YHO-13351 was used in E. F, Best percentage change of initial tumor volume from D and E. YHO-13351 alone was ineffective in ASPC-1. G, HER3-targeting ADCs in ASPC-1 with and without the ABCG2 inhibitor YHO-13351. The starting tumor volume was ∼ 900 mm3. Left, tumor growth and inhibition. Blue line is an ADC using T800–exatecan instead of T1000–exatecan. t.i.w. × 2, 3 times a week for 2 weeks. Right, tumor volume comparison at days 68 and 75 for P–GGFG–DXd with and without YHO-13351. n = 3. H, TROP2 targeting Dato–DXd in a high P-gp expression PDX model with and without tariquidar. mRNA of ABCG2 and P-gp is shown. Starting tumor volume was 600 mm3. n = 2. In all in vivo studies, CDX/PDX mice were intravenously administered with indicated ADCs (at 10 mg/kg or at an indicated dose) on days indicated by an arrow. Each value represents the mean and SEM (n = 5 or otherwise indicated). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. For small-molecule inhibitors, the dose of YHO-13351 and tariquidar was 50 mg/kg [p.o., q.d. × 5D× 2 (orally administered every day for 5 days as a cycle and two cycles implemented)] and 15 mg/kg (p.o., q.d. × 5D× 3), respectively.

Figure 4.

Overcoming MDR resistance by T moiety–exatecan ADCs or a combination of MDR inhibitor with DXd/SN-38 ADCs. A,In vitro cytotoxicity improvement by P-gp inhibitor in HCT-15 for HER2/HER3/TROP2-targeting ADCs. IC50 fold change is shown. B and C,In vivo efficacy of TROP2-targeting ADC in HCT-15 with and without the P-gp inhibitor tariquidar. B, Tumor growth and inhibition. n.s., not significant. C, Best percentage change of initial tumor volume from B. Each ADC is labeled by its name and payload used. Tariquidar alone was ineffective in HCT-15. DF, HER2-targeting ADCs in HCT-15 (P-gp) and ASPC-1 (ABCG2) with and without MDR inhibitor. D and E, Tumor growth and inhibition. Tariquidar was used in D, and YHO-13351 was used in E. F, Best percentage change of initial tumor volume from D and E. YHO-13351 alone was ineffective in ASPC-1. G, HER3-targeting ADCs in ASPC-1 with and without the ABCG2 inhibitor YHO-13351. The starting tumor volume was ∼ 900 mm3. Left, tumor growth and inhibition. Blue line is an ADC using T800–exatecan instead of T1000–exatecan. t.i.w. × 2, 3 times a week for 2 weeks. Right, tumor volume comparison at days 68 and 75 for P–GGFG–DXd with and without YHO-13351. n = 3. H, TROP2 targeting Dato–DXd in a high P-gp expression PDX model with and without tariquidar. mRNA of ABCG2 and P-gp is shown. Starting tumor volume was 600 mm3. n = 2. In all in vivo studies, CDX/PDX mice were intravenously administered with indicated ADCs (at 10 mg/kg or at an indicated dose) on days indicated by an arrow. Each value represents the mean and SEM (n = 5 or otherwise indicated). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. For small-molecule inhibitors, the dose of YHO-13351 and tariquidar was 50 mg/kg [p.o., q.d. × 5D× 2 (orally administered every day for 5 days as a cycle and two cycles implemented)] and 15 mg/kg (p.o., q.d. × 5D× 3), respectively.

Close modal

T Moiety–Exatecan ADCs for Colon Cancer

In Vivo Antitumor Efficacy in Challenging Colon Cancer Models

So far, ADCs have failed to demonstrate meaningful clinical efficacy against colon cancer. T moiety–exatecan ADCs targeting HER2, HER3, and TROP2 were evaluated in colon cancer PDX models representing unmet clinical needs (Fig. 5A). In a TP53–BRAF double mutation PDX model with high HER3 expression (IHC 3+), P–T1000–exatecan showed antitumor activity, but the tumors were resistant to P–GGFG–DXd (Fig. 5B). In a KRAS–TP53 double-mutant PDX model with HER2 (low expression 1+) and TROP2 (high expression 3+) positivity, T moiety–exatecan ADCs showed potent antitumor activity (Fig. 5C and D). For HER2, both MTX-1000 and MTX-800 showed significant tumor inhibition with one out of five achieving complete response (CR) in the MTX-1000 group (Fig. 5C). However, tumors were resistant to Tras–GGFG–DXd (Fig. 5D). TROP2-targeting Dato–GGFG–DXd showed comparable tumor inhibition to hRS7–T1000–exatecan (MTX-132) for 40 days before tumors in the Dato–GGFG–DXd treatment group grew again, leading to a significant efficacy gap between these two groups (Fig. 5D). In a TROP2-high-expression tumor with triple mutations of TP53, KRAS, and BRAF and microsatellite stability (MSS), MTX-132 showed more durable response than Dato–DXd (Fig. 5E). Together, T moiety ADCs were found to produce deep and durable antitumor response in colon cancer models resistant to current ADCs.

Figure 5.

T moiety–exatecan ADCs overcome resistance and synergize with PARP/ATR inhibitors in colon cancer cells. A, Information on colon cancer PDX models. Shown is the mutation status of TP53/KRAS/BRAF and micosatellite instability (MSI) status. Target expression level is indicated based on IHC score. WT, wild-type. BE,In vivo efficacy of T moiety–exatecan (blue) and GGFG–DXd (red) ADCs in colon cancer xenograft (CDX) and PDX models. Target and cell line/PDX tumor type are labeled. Target expression in each model is shown as flow cytometry (relative MFI) and/or IHC image on untreated mouse tumor tissue and H-score. Scale bars, 20 μm. CDX/PDX mice were intravenously administered with indicated ADCs (10 mg/kg unless otherwise labeled) on day 0 (tumor size reached an average of 150–200 mm3) and subsequent dates indicated by red arrows. Each value represents the mean and SEM (n = 4 or 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. n.s., not significant; T800–e, T800–exatecan; T1000–e, T1000–exatecan. ADCs used for each model: HER3-targeting P–T1000–exatecan and P–GGFG–DXd (B); HER2-targeting MTX-1000, MTX-800, and Tras–GGFG–DXd (C); and TROP2-targeting Dato–GGFG–DXd and MTX-132 using hRS7 (IMMU-132 antibody; D and E). E, Spider plot of individual tumor volume of Dato–GGFG–DXd and MTX-132 for PDX 360837. F and G, CI of MTX-1000 and Tras–GGFG–DXd with Tala (F) or M4344 (G). Calculated from cell viability data of Supplementary Fig. S17G. MTX-1000 showed synergy with Tala at a wider concentration range than Tras–GGFG–DXd and at lower concentrations with M4344 than Tras–GGFG–DXd. H and I,In vivo efficacy of MTX-1000 (blue) or Tras–GGFG–DXd (red) in combination with Tala (H) or M4344 (I) in a COLO205 xenograft model. Mice were intravenously administered with indicated ADCs (10 mg/kg for Tras–GGFG–DXd and 3 mg/kg for MTX-1000 based on data from Fig. 3A) on days 0 (tumor size reached about 200 mm3) and 7 (red arrows). Tala was given daily and M4344 every week orally (not labeled on the curves). Each value represents the mean and SEM (n = 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 5.

T moiety–exatecan ADCs overcome resistance and synergize with PARP/ATR inhibitors in colon cancer cells. A, Information on colon cancer PDX models. Shown is the mutation status of TP53/KRAS/BRAF and micosatellite instability (MSI) status. Target expression level is indicated based on IHC score. WT, wild-type. BE,In vivo efficacy of T moiety–exatecan (blue) and GGFG–DXd (red) ADCs in colon cancer xenograft (CDX) and PDX models. Target and cell line/PDX tumor type are labeled. Target expression in each model is shown as flow cytometry (relative MFI) and/or IHC image on untreated mouse tumor tissue and H-score. Scale bars, 20 μm. CDX/PDX mice were intravenously administered with indicated ADCs (10 mg/kg unless otherwise labeled) on day 0 (tumor size reached an average of 150–200 mm3) and subsequent dates indicated by red arrows. Each value represents the mean and SEM (n = 4 or 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. n.s., not significant; T800–e, T800–exatecan; T1000–e, T1000–exatecan. ADCs used for each model: HER3-targeting P–T1000–exatecan and P–GGFG–DXd (B); HER2-targeting MTX-1000, MTX-800, and Tras–GGFG–DXd (C); and TROP2-targeting Dato–GGFG–DXd and MTX-132 using hRS7 (IMMU-132 antibody; D and E). E, Spider plot of individual tumor volume of Dato–GGFG–DXd and MTX-132 for PDX 360837. F and G, CI of MTX-1000 and Tras–GGFG–DXd with Tala (F) or M4344 (G). Calculated from cell viability data of Supplementary Fig. S17G. MTX-1000 showed synergy with Tala at a wider concentration range than Tras–GGFG–DXd and at lower concentrations with M4344 than Tras–GGFG–DXd. H and I,In vivo efficacy of MTX-1000 (blue) or Tras–GGFG–DXd (red) in combination with Tala (H) or M4344 (I) in a COLO205 xenograft model. Mice were intravenously administered with indicated ADCs (10 mg/kg for Tras–GGFG–DXd and 3 mg/kg for MTX-1000 based on data from Fig. 3A) on days 0 (tumor size reached about 200 mm3) and 7 (red arrows). Tala was given daily and M4344 every week orally (not labeled on the curves). Each value represents the mean and SEM (n = 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Exatecan/MTX-1000 and PARP/ATR Inhibitor Synergize in Colon Cancer Cells

Antitumor agents targeting DDR pathway mediators, such as PARP and ATM/ATR/WEE1, have shown synergistic effects with ADCs (36, 37). A synergistic mechanism of combined TOP1 and CHEK1 inhibition was demonstrated in colon cancer (38). To explore the synthetic lethality of DXd or exatecan-based ADCs with DDR inhibitors, we first tested in vitro cytotoxicity of exatecan and DXd in combination with the PARP inhibitor talazoparib (Tala; Supplementary Fig. S17A) and a novel ATR inhibitor, M4344 (Supplementary Fig. S17B), at respective nontoxic doses (IC20) for each inhibitor in COLO205 (39). Both DXd and exatecan show synergy with Tala and M4344, with an improvement of IC50 30- to 200-fold for Tala and 7- to 20-fold for M4344 (tabulated in Supplementary Fig. S17C and S17D). Tala improved the IC50 of exatecan and DXd by roughly 200-fold, whereas M4344 did by only 10-fold (Supplementary Fig. S17E and S17F). Combining exatecan with Tala or M4344 was much more potent (10–60 times) than DXd plus Tala or M4344 (Supplementary Fig. S17G). Combination index (CI) score calculation suggested that exatecan synergized with both Tala and M4344 at a lower concentration than DXd and Tala synergized with both exatecan and DXd better than M4344 (Supplementary Fig. S17H and S17I). Cytotoxicity of MTX-1000 or Tras–GGFG–DXd was then tested with Tala or M4344. In COLO205, all molecules showed some activity in vitro (Supplementary Fig. S17J), and CI calculation showed that MTX-1000 synergized better with both Tala and M4344 compared with Tras–GGFG–DXd (Fig. 5F and G). In vivo, a combined use of MTX-1000 (3 mg/kg) with Tala produced significantly better tumor inhibition than MTX-1000 (effective), Tala (ineffective), or 10 mg/kg of Tras–GGFG–DXd with Tala (Fig. 5H). Even at 10 mg/kg, Tras–GGFG–DXd did not show synergy with Tala. Tras–GGFG–DXd combined with Tala inhibited tumor growth in a similar way to just Tras–GGFG–DXd. Similarly, MTX-1000 at 3 mg/kg was synergistic with M4344, and Tras–GGFG–DXd at 10 mg/kg was not (Fig. 5I). These data demonstrated that exatecan/MTX-1000 can synergize with DDR pathway mediators to achieve improved antitumor efficacy, outperforming DXd/Tras–GGFG–DXd.

T Moiety–Exatecan ADCs Targeting Lung Cancer Resistance

We evaluated T moiety–exatecan ADCs targeting TROP2, HER2, and HER3 in lung cancer PDX models representative of unmet medical needs (Fig. 6A). Lung squamous cell cancer is a more challenging type of lung cancer to treat due to a lack of actionable mutations. MTX-132 caused significant tumor regression (CR in one model) in two models, whereas Dato–DXd was ineffective in one model and less effective in another (Fig. 6B and C). In a PDX model derived from an osimertinib (AZD9291; third-generation tyrosine kinase inhibitor)-resistant patient with EGFR del19/T790M/C797S (triple mutation) that lacked approved therapeutic options, combination therapy of the ALK inhibitor brigatinib with the anti-EGFR antibody cetuximab was able to induce tumor inhibition, which is consistent with a previous study (ref. 40; Supplementary Fig. S18A–S18D). Despite very low HER2 and HER3 expression in this model, MTX-1000 and P–T1000–exatecan produced significant tumor inhibition, with 3 of 5 and 1 of 5 mice, respectively, achieving CR (Fig. 6D and E). P–GGFG–DXd was ineffective in tumor inhibition, whereas Tras–GGFG–DXd initially produced three of five CR, but two tumors in this group started to regrow rapidly after 50 days compared with slight regrowth of two tumors in the MTX-1000 treatment group, leading to a significant difference between MTX-1000 and Tras–GGFG–DXd (Fig. 6D). In comparison, both MTX-1000 and P–T1000–exatecan significantly outperformed brigatinib combined with cetuximab to inhibit tumor growth in this model (Fig. 6F). Finally, in a PDX model with EGFR ex19del and MET amplification, P–T1000–exatecan produced CR in all 5 mice, whereas P–GGFG–DXd showed significant tumor inhibition in 3 mice, but no growth inhibition for the other two tumors, showing a significant difference between the efficacy of two ADCs (Fig. 6G and H; Supplementary Fig. S18E). This tumor was resistant to both erlotinib and osimertinib (Supplementary Fig. S18F).

Figure 6.

T moiety–exatecan ADCs targeting lung cancer resistance. A, Information on lung cancer PDX models. Shown is the mutation status of EGFR and amplification. B–H,In vivo efficacy of ADCs [T1000–exatecan (blue) and DXd (red)] and other drugs in xenograft (CDX) and PDX models. Target and PDX tumor type are labeled. Target expression in each model is shown as an IHC image on untreated mouse tumor tissue and H-score. Scale bars, 20 μm. PDX mice were intravenously administered with indicated ADCs (10 mg/kg unless otherwise labeled) or other drugs (dose indicated) on day 0 (tumor size reached an average of 150–200 mm3) and subsequent dates indicated by red arrows. Each value represents the mean and SEM (n = 4 or 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. ADCs/drugs used for each model are as follows: B and C,In vitro efficacy of TROP2-targeting ADCs in lung squamous cell cancer PDX. D, MTX-1000 and Tras–GGFG–DXd, including spider plot of individual tumor size. E, P–T1000–exatecan and P–GGFG–DXd. F, Comparison of MTX-1000, P–T1000–exatecan (–e), and tyrosine kinase inhibitor/cetuximab tumor volume at day 30. G and H, P–T1000–exatecan and P–GGFG–DXd. Spider plot of individual tumor volume is shown in G, and a comparison is shown in H.

Figure 6.

T moiety–exatecan ADCs targeting lung cancer resistance. A, Information on lung cancer PDX models. Shown is the mutation status of EGFR and amplification. B–H,In vivo efficacy of ADCs [T1000–exatecan (blue) and DXd (red)] and other drugs in xenograft (CDX) and PDX models. Target and PDX tumor type are labeled. Target expression in each model is shown as an IHC image on untreated mouse tumor tissue and H-score. Scale bars, 20 μm. PDX mice were intravenously administered with indicated ADCs (10 mg/kg unless otherwise labeled) or other drugs (dose indicated) on day 0 (tumor size reached an average of 150–200 mm3) and subsequent dates indicated by red arrows. Each value represents the mean and SEM (n = 4 or 5). Unpaired two-sided t test. *, P < 0.05; **, P < 0.01; ***, P < 0.001. ADCs/drugs used for each model are as follows: B and C,In vitro efficacy of TROP2-targeting ADCs in lung squamous cell cancer PDX. D, MTX-1000 and Tras–GGFG–DXd, including spider plot of individual tumor size. E, P–T1000–exatecan and P–GGFG–DXd. F, Comparison of MTX-1000, P–T1000–exatecan (–e), and tyrosine kinase inhibitor/cetuximab tumor volume at day 30. G and H, P–T1000–exatecan and P–GGFG–DXd. Spider plot of individual tumor volume is shown in G, and a comparison is shown in H.

Close modal

Exatecan and MTX-1000 Induce Immunogenic Cell Death In Vitro and Enhanced Antitumor Effect of Anti–PD-1 in Combination with MTX-1000

Both microtubule-disrupting payloads and DXd-derived ADCs induce immunogenic cell death (ICD) and promote an antitumor response (41). We sought to determine whether MTX-1000 has an immunomodulatory effect. We first assayed exatecan's capability for inducing ICD through induction of endoplasmic reticulum stress and autophagy as well as involving calreticulin (CRT) cell-surface exposure. In the exatecan-sensitive cell line COLO205, treatment with exatecan and MTX-1000 led to a significant externalization and plasma membrane exposure of CRT (Supplementary Fig. S19A and S19B), suggesting that exatecan and released exatecan from MTX-1000 through target-dependent internalization of MTX-1000 induced strong ICD. The increases in relative mean fluorescence intensity (rMFI) and percentage of positive cells of surface CRT for exatecan/MTX-1000 were significantly higher than that of DXd/T–GGFG–DXd's (Supplementary Fig. S19A and S19B), suggesting that exatecan/MTX-1000 may have better immunogenic properties. The enhanced CRT was observed despite 30% to 70% decreases in cell number after 48 hours (Supplementary Fig. S19C), compared with controls.

To translate the in vitro exatecan/MTX-1000–induced ICD phenotype into enhanced antitumor activity in vivo, we generated a syngeneic mouse model using CT-26 with transfected human HER2 (CT26-hHER2). CT26-hHER2 had medium (rMFI of 30–50) hHER2 expression determined by flow cytometry (Supplementary Fig. S19D). The combination of MTX-1000 and Tras–GGFG–DXd with an anti–PD-1 antibody was examined. At day 12, all experimental groups showed significant tumor inhibition compared with the vehicle control (Supplementary Fig. S19E). Similar to other models, monotherapy of MTX-1000 (10 mg/kg, once a week, twice) showed better tumor inhibition than Tras–GGFG–DXd (**, P = 0.09), with a TGI of 61% compared with the latter's 38% (Supplementary Fig. S19F). Interestingly, monotherapy of MTX-1000 was similarly effective as a combination therapy of Tras–GGFG–DXd with PD-1 (not significant), with a TGI of 61% and 62%. Combination efficacy of MTX-1000 with PD-1 (TGI = 78%) was significantly higher than PD-1 alone (**, P = 0.002; TGI = 45%) than to MTX-1000 alone (*, P = 0.0179; TGI = 61%), suggesting that MTX-1000 might play a more prominent role in the improved efficacy of combination therapy (P = 0.002). The combined use of MTX-1000 and anti–PD-1 antibody produced an antitumor effect that was superior to both monotherapies (Supplementary Fig. S19F, right). No body weight abnormalities were observed in mice (Supplementary Fig. S19G). The survival rates for vehicle, Tras–GGFG–DXd, anti–PD-1 antibody, MTX-1000, Tras–GGFG–DXd + PD-1, and MTX-1000 + PD-1 at day 21 were 0/8, 1/8, 2/8, 3/8, 4/8, and 7/8, respectively. TGI of MTX-1000 combined with PD-1 was significantly better (*, P = 0.0195) than that of Tras–GGFG–DXd combined with PD-1 (Supplementary Fig. S19F tabulation).

Camptothecin class TOP1 inhibitors have become more prominent as ADC payloads thanks to the clinical success of DS-8201a and Trodelvy (30, 42–44). The characterization of novel camptothecins largely focuses on the structure–activity relationship, and the scope of preclinical ADC studies is limited to routine efficacy and toxicity examination (23, 30, 42, 43). However, the development of an optimal ADC necessitates a more holistic approach in selecting linker chemistry and the incorporation position of a hydrophilic polymer moiety (often required) within the ADC architecture (30, 45–47). In order to improve DXd/SN-38 ADC performance, an ADC with high clinical translatability will need to be developed, which will warrant a thorough mechanistic investigation of the new TOP1 inhibitor–based ADC design.

Here we present our effort to develop a class of exatecan ADC with such clinical potential. Exatecan was selected as the payload due to its higher potency, known clinical and toxicity profile in humans, and lower MDR-mediated efflux and higher cell permeability compared with DXd/SN-38 (48). Mechanistic differences in the biochemical, structural, and pharmacologic profiles of exatecan and DXd/SN-38 were investigated. Exatecan was shown to have a 10-fold higher potency and 1/10th efflux ratio in MDR+ cells, two traits that correspond with higher efficacy and the ability to overcome resistance toward low-target-expression and MDR+ tumors. Higher cell permeability and stronger bystander penetration of exatecan may further enhance its ADC function (21). On the other hand, a higher potency could imply more severe side effects. Therefore, exatecan's linker chemistry and the conjugation strategy for exatecan must incorporate its superior pharmacologic feature while maintaining or improving upon safety in the final ADC. To achieve these objectives, we focused on the physicochemical evaluation of distinctive ADC designs to compare and select a novel chemistry architecture, T moiety. The modification of pAB adjacent to exatecan not only led to a highly homogeneous and hydrophilic ADC through efficient masking of payload hydrophobicity, but also created a simple, ready process to test established linker structure without additional chemistry. The selection of a VA linker in combination with pAB modifications by pSAR (T1000; or a minimal m-methylaminomethyl group, T800) resulted in more hydrophilic, less aggregated, and more stable antibody–exatecan conjugates than alternative linkers (GGFG or VC), modification site (linker/MC), or modification chemistry (PEG), which was consistent with previous observations and principles (30, 45–47). T moiety chemistry was validated by the successful translation of higher potency and less MDR sensitivity of exatecan into corresponding ADCs. Performance improvement of T moiety–exatecan ADC was mediated by exatecan through stronger TOP1 inhibition, lower MDR-mediated efflux of released payload, and a deeper and broader intratumoral pharmacodynamic response. The mechanistic understanding of T moiety–exatecan ADC may provide a broader implication for designing new TOP1 ADCs. The consistent high-quality antitumor response of T moiety–exatecan ADCs in relevant PDXs and organoids with a high patient response predictability bodes well for clinical translation (34, 49).

Low-target-expression and MDR+ tumors represent important resistance mechanisms for ADC (8, 9). We demonstrated here that DXd ADC was susceptible to similar resistance. However, T moiety–exatecan ADCs showed less resistance and often displayed potent and durable antitumor activity in low-target-expression and MDR+ tumors, compared with a combination treatment of ABCG2/p-pg inhibitor and DXd/SN-38 ADCs that was shown to be effective but less persistent. Although MDR inhibitors, including ABCG2 inhibitor (YHO-13351) and P-gp inhibitor (tariquidar), are commonly used in ADC studies (17), the mechanism of the observed sensitivity difference among various TOP1 inhibitors remains to be clarified in the future due to the complexity of MDR biology (50). Due to potential selectivity issues of MDR inhibitors, there remains the possibility of other substrates of the inhibitors contributing to the overall cytotoxicity differences. Nevertheless, enhanced therapeutic indices of T moiety–exatecan ADCs targeting classic tumor antigens such as HER2/TROP2/HER3 may provide a new opportunity to treat target-positive patient populations beyond the reach of current ADCs. For example, T moiety–exatecan ADCs generated durable and deep responses in resistant or treatment-refractory colon cancer models. In colon cancer cells, MTX-1000 showed highly synergistic antitumor efficacy with PARP/ATR inhibitors, suggesting that a combination therapy strategy of exatecan-based ADCs with DDR pathway inhibitors could augment antitumor potency (30, 38). Similarly, T moiety–exatecan ADCs showed improved efficacy against lung cancer intrinsic resistance mechanisms. Although the degree of improvements varied among tumor models, likely due to a combination of factors such as target antigen expression level, tumor cell sensitivity toward payload, degree of bystander effect, and tumor microenvironment, T moiety ADCs showed greater efficacy gain in more challenging models. One possible explanation was that the combined mechanistic improvements of T moiety–exatecan ADCs resulted in an elevated cumulated payload inside the tumor cells beyond the functional inhibition threshold, thus yielding more qualitative differences in these models. More preclinical data and eventually clinical data will help establish the most responsive tumor types for each class of ADC.

T moiety–exatecan ADCs maintained a higher potency without increasing toxicity with HNSTDs in nonhuman primate studies at a level that was similar to or higher than that of other TOP1 inhibitor ADCs. Notably, side effects from the administration of T moiety–exatecan ADC were often more prominently observed in the GI tract (diarrhea) and the hematologic system (reticulocyte reduction), though symptoms in both were fully reversible. The hematologic and digestive organ toxicities for T moiety–exatecan ADCs mirrored those of exatecan as a single agent in human trials (48). However, T moiety–exatecan ADC achieved more favorable safety signals in the GI tract and minimal myelotoxicity than exatecan, possibly reflecting the ADC design's superior physicochemical features. Improved stability, longer ADC half-life, lower free payload release, and the reduced myelotoxicity potential of ADC in comparison with free payload may all contribute to the favorable toxicity profile of T moiety–exatecan ADC. Interstitial lung disease occurred in just over 10% of DXd-based ADC–treated patients during clinical trials. Although we did not observe any structural damage or signs of inflammation in the lungs of animals dosed with T moiety–exatecan ADCs, it remains uncertain whether prolonged treatment will yield additional pulmonary toxicity. Nevertheless, it is conceivable that T ­moiety–exatecan ADCs could have different target-independent tissue distributions than DXd-based ADCs, resulting in the manifestation of diverse symptoms.

For tumors less responsive or resistant to TOP1 inhibitors, payload classes with different mechanisms may be necessary. T moiety is a modular structure compatible with diverse linker chemistry and is amenable to building ADCs with dual payloads. Taken together, T moiety–exatecan ADCs have the potential to address patient needs unfulfilled by current ADCs, whereas the versatility and scalability of T moiety can facilitate the introduction of payloads of completely different MOAs to meet the continuous challenges of drug resistance.

Synthesis of T Moiety

Synthesis of T moiety and all other chemical compounds is described in the Supplementary Note.

Antibodies and ADCs, and Compounds

The anti-HER2 antibody (Tras) was purchased from Geneway Biotechnology Co., Ltd. The anti-CDH6 antibody (hG019), anti-TROP2 antibodies (Dato and sacituzumab), and anti-HER3 antibody (patritumab) were all generated in the CHO or 293F expression system and purified by Mabselect Sure Resin (GE Healthcare) in gravity column. The sequences of these antibodies were obtained from the corresponding patents and literature. T-DM1 was purchased from F. Hoffmann-La Roche (Basel, Switzerland). Control IgG1 was purchased from Beijing Solarbio Science and Technology Co., Ltd. Anti-mouse PD-1 antibody is produced by ChemPartner (batch#: 201630002). ADCs were synthesized as described above, and their DAR values were determined by reverse-phase chromatography. The drug distribution was analyzed by HIC and further characterized by reduced reverse-phase LC/MS (Supplementary Note). T-DXd/DS-8201a was made in-house with Tras (Batch#: FX03DS1901, Geneway Biotech) using linker–payload directly purchased from MedChemExpress: MC-GGFG–DXd (HY-114233, MedChemExpress). Talazoparib (HY-108413), exatecan (HY-13631), DXd (HY-13631E), and M4344 (HY-136270) were all purchased from MedChemExpress.

ADC Preparation

A solution of naked mAb (10 mg/mL in PBS (7.0) + 2 mmol/L EDTA) was treated with 7 molar equivalents of tris(2-carboxyethyl)phosphine (TCEP) for 2 hours at 37°C. Then 14 molar equivalents of the linker drug (from a 10 mmol/L DMA stock solution) were added to the fully reduced antibody while keeping residual DMA concentration below 10% (v/v). The mixture was incubated at room temperature for an hour followed by purification over a Zeba spin desalting column (Thermo Fisher Scientific) to remove excess reagents. In this step, the resulting ADC was also buffer exchanged into formulation buffer (20 mmol/L histidine, 150 mmol/L NaCl, pH 5.5). Moreover, the excess linker drug could be removed thoroughly by treating the product with activated charcoal (30 mg of charcoal to 1 mL ADC solution) followed by vortexing for 2 hours at room temperature. The charcoal was then removed via sterile filtration (0.2 μm PES filters), and the final ADC was stored at −80°C.

ADC Concentration, DAR, and Aggregation Measurement

The concentration of mAb and the conjugated drug in the ADC was calculated by measuring UV absorbance of an aqueous solution of the ADC at two wavelengths of 280 nm and 370 nm, as the total absorbance at any given wavelength is equal to the sum of the absorbance of all light-absorbing chemical species that are present in a system (additivity of absorbance). The equations were as follows:

The values of εA,280, εA,370, εD,280, and εD,370 were estimated based on calculation of the amino acid sequence of the antibody or obtained by UV measurement of the compound according to the Lambert–Beer law. Simultaneous equations (1) and (2) can be solved by substitution of the above values, and then CA and CD were determined. Dividing CD by CA, the average number of conjugated drug molecules per antibody was determined.

Aggregation was assessed by size exclusion chromatography, which was performed on an Agilent 1260 Infinity II HPLC system with UV detection at 280 nm. The column was a TSKgel G3000SWXL column (Tosoh Bioscience). Samples were injected at a 50 μg load. The mobile phase was 200 mmol/L sodium phosphate and 150 mmol/L sodium chloride (pH 7.0). Also, 15% (v/v) isopropanol was added to the mobile phase to minimize secondary hydrophobic interactions with the stationary phase and prevent bacterial growth. The flow rate was 0.75 mL/minute, and the column temperature was set at room temperature.

ADCs with different numbers of drugs per antibody were separated using a butyl HIC column (TSK-gel Butyl-NPR 4.6 × 35 mm 2.5 μm, Tosoh Bioscience) at room temperature. HIC was also performed on an Agilent 1260 Infinity II HPLC system with UV detection at 280 nm. Mobile phase A was 1.5 mol/L (NH4)2SO4, 50 mmol/L K2HPO4, pH 7.0, and mobile phase B was 21.3 mmol/L KH2PO4, 28.6 mmol/L K2HPO4, 25% (v/v) isopropanol, pH 7.0. The gradient program was as follows: B %: 0% to 25% (0–1 minutes, 0.8 mL/minute), 25% (1–3 minutes, 0.6 mL/minute), 25% to 80% (3–13 minutes 0.6 mL/minute), 80% (13–17 minutes, 0.6 mL/minute), 80% to 0% (17–17.10 minutes, 0.5 mL/minute), and 0% (17.10–25 minutes, 0.7 mL/minute).

Flow Cytometry

For cell-surface proteins, IF and FACS assays were performed under nonpermeable conditions without detergent in the buffers. Antibody binding signal was detected using Alexa Fluor 488 and 594 goat anti-mouse IgG secondary antibodies (Jackson ImmunoResearch, no. 115-545-003 and no. 115-585-003). Briefly, cells attached on coverslips (IF assays) or suspended in 1× PBS (FACS assays) were first fixed in 4% paraformaldehyde (PFA) for 10 minutes. PFA was then removed, and cells were rinsed 3 times with 1× PBS. Cells were blocked overnight at 4°C in blocking buffer (1× PBS containing 10% normal goat serum; 0.1% Triton was added for intracellular proteins). After removing the blocking buffer, cells were incubated with primary antibody (dilution in the blocking buffer at 1:100 to 1,000) for 3 hours at room temperature. Cells were rinsed 6 times in 1× PBS before being incubated with fluorescence-labeled secondary antibody (diluted in blocking buffer at 1:500 dilution ratio with 1:10,000 Hoechst 33258; Sigma, no. 94403) for 1 hour. Last, cells were rinsed 3 times with 1× PBS. IF images were recorded with a Nikon confocal system A1Si. The 3D reconstruction of the IF results was performed in ImageJ (NCBI, NIH free software). The FACS data were collected using a BD Accuri C6 Plus system. A control sample without a primary antibody and another sample with an isotype control antibody were used. The LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit (Thermo Fisher Scientific) was used to exclude dead cells.

Cell Lines and PDX Models

COLO205 (RRID:CVCL_0218), HCT-15 (RRID:CVCL_0292), and DLD-1 (RRID: CVCL_0248) cell lines were purchased from the ATCC in 2020. HT-29 (RRID:CVCL_0320), SK-MES-1 (RRID:CVCL_0630), NCI-H292 (RRID:CVCL_0455), MDA-MB-468 (RRID:CVCL_0419), SK-OV-3 (RRID:CVCL_0532), LoVo (RRID:CVCL_0399), NCI-H460 (RRID:CVCL_0459), BxPC-3 (RRID:CVCL_0186), and PC9 (RRID:CVCL_B260) cell lines were purchased from a stem cell bank, Chinese Academy of Sciences (Shanghai, China), in 2018. Cells were maintained in RPMI 1640 medium (COLO205, NCI-H292, HCT-15, DLD-1, NCI-H460, BxPC-3, and PC9) or DMEM (SK-MES-1 and MDA-MB-468) or F-12K medium (LoVo) or McCoy's 5A medium (HT-29 and SK-OV-3) supplemented with 10% heat-inactivated FBS and cultured at 37°C in a 5% CO2 atmosphere. When cells were grown to ∼80% confluence, they were dissociated from culture plates by treatment with 1× PBS and 1 mmol/L EDTA for 10 to 20 minutes. Trypsin was not used to avoid damage on cell-surface proteins. rMFI was calculated by the ratio of MFI of antitarget Ab-FITC/MFI of isotype control-FITC.

The mouse colon cancer cell line CT26 (RRID:CVCL_7254) was purchased from the ATCC in 2020. An empty vector (pQCXIN; Clontech) or human HER2 gene (sequence based on NM_004448.3) was retrovirally introduced into CT26 cells (CT26 and CT26-hHER2, respectively). The cells were cultured in RPMI 1640 medium supplemented with 10% heat-inactivated FBS and 250 μg/mL geneticin (Thermo Fisher Scientific) at 37°C in a 5% CO2 atmosphere. Human HER2 expression was confirmed by flow cytometry.

All PDX models (362014, 361795, 360837, 200717, 200662, 200615, and 360961) were obtained from Lidebiotech Co., Ltd. in 2021.

All cell lines were authenticated by short tandem repeat analysis according to suppliers’ information. All cells were tested negative for Mycoplasma contamination using the Mycoplasma PCR Detection Kit (MP004, GeneCopoeia) prior to conducting cell-based assays. All cells and PDX models were used for the experiments within 5 to 15 passages and 2 to 5 passages, respectively, after collection.

Cytotoxic Assay

Cells were seeded to a 96-well plate at an appropriate density depending on the cell line (typically 1,000–3,000 cells per well in 100 μL of appropriate culture media). After overnight incubation, a concentration series of small-molecule or ADC drugs and controls was added. Cell viability was evaluated after 5 or 6 days. MTT (5 mg/mL, 20 μL, Sigma-Aldrich) was added into the wells, and incubation was continued for 2 to 4 hours at 37°C. Culture media were then removed, and well content was homogeneously dissolved with 0.1 N HCl/isopropanol. Absorbance values were measured on a Thermo Scientific Multiskan EX microplate reader using a wavelength of 570 nm (with a reference wavelength of 690 nm). The IC50 values compared with untreated cells or treated with other controls were determined using inhibition dose–response curve fitting (GraphPad Prism 9). Target expression on each cell line was measured by flow cytometry as described above.

Cell Line and PDX Studies

All tumor-bearing models were established in female BALB/c nude mice that were purchased from the Shanghai Family Planning Research Institute. All in vivo studies were performed in accordance with the local guidelines of the Institutional Animal Care and Use Committee of Multitude Therapeutics and with the approval of the committee. Briefly, 4- to 6-week-old mice were housed together in sterilized cages and maintained under required pathogen-free conditions. Each cell suspension or tumor fragment was inoculated subcutaneously into female nude mice. When the tumor had grown to an appropriate volume, the tumor-bearing mice were randomized into treatment and control groups based on the tumor volumes, and dosing was started. Antibodies and ADCs were administered intravenously to the mice. Small-molecule drugs were administered orally. Tumor volume defined as 1/2 × length × width2 was measured twice (3–4 days) a week. TGI (%) was calculated as follows: TGI (%) = [1 – (mean of treatment group tumor volume on evaluation day)/(mean of control group tumor volume on evaluation day)] × 100. The mice were euthanized with CO2 gas when they reached endpoints (tumor volume exceeding 3,000 mm3, >10% reduction of body weight, or clinical signs indicating that mice should be euthanized for ethical reasons).

Syngeneic Model

Female BALB/c mice, weighing approximately 18 to 20 g, were purchased from Shanghai Lingchang Biotech Co., Ltd. and were maintained with 5 mice per cage in an individually ventilated cage in a specific pathogen–free animal facility at ChemPartner with autoclaved cages. After 9 days of acclimation, animals were subcutaneously inoculated with CT26-hHER2 cells (3 × 105 cells/0.1 mL/mouse) on the right flank for tumor development. Ten days after inoculation, 36 mice with tumor sizes ranging from 100 to 200 mm3 (average tumor size 178 mm3) were enrolled into the efficacy study and randomized into six therapy groups, and the treatments were initiated on the randomization day (defined as D0). MTX-1000, DS-8201a (10 mg/kg), and anti–PD-1 antibody (5 mg/kg) were administered intravenously at a volume of 10 mL/kg to mice. As a control, ABS buffer (10 mmol/L acetate buffer, 5% sorbitol, pH 5.5) was administered at the same volume as the drugs. MTX-1000 and DS-8201a were administered on days 0 and 7. Anti–PD-1 antibody was administered on days 0, 3, 7, and 10.

For flow-cytometric analysis of T cells, dendritic cells, and tumor cells, when the average volume of tumors reached approximately 250 to 400 mm3 (8 days after tumor inoculation), the mice were treated with vehicle or DS-8201a (10 mg/kg, once, intravenously; day 0). The mice were euthanized with CO2 asphyxiation on day 8, and tumors were cut into small pieces and dissociated with the Tumor Dissociation Kit (Miltenyi Biotec) by the gentleMACS Octo Dissociator with Heaters (Miltenyi Biotec). The resultant single cells were blocked with Mouse BD Fc Block reagent (Becton Dickinson) and stained with antibodies against mouse CD3, CD4, CD8, CD11c, CD45, CD86, granzyme B, MHC class I, MHC class II, and PD-L1 and human HER2.

Tissue Multiplex IHC

Sections from formalin-fixed, paraffin-embedded tumor tissue blocks were stained using Opal multiplex according to the manufacturer's protocol (PerkinElmer) for DAPI, CD45, CD4, and CD8. Slide scanning was performed on the Vectra 3.0 instrument. All the images were analyzed with the HALOTM Image Analysis platform. The whole section was analyzed, and the big necrosis area and stroma area were excluded. Single-positive cells were counted separately. Images were spectrally unmixed, evaluated for staining intensities and morphology, tissue segmented based on tissue markers, cell segmented based on nuclear and membrane markers, and phenotypically scored.

PK of MTX-1000/DS-8201a in Rat/Mouse

Concentrations of ADC and the total antibody in plasma were determined with a validated ligand-binding assay; the lower limit of quantitation was 0.02 μg/mL. Briefly, immunoplates were coated with 1 μg/mL Human HER2 Protein, His Tag (Acrobiosystems Inc., Beijing) in coating buffer and kept overnight at 4°C. After washing, the plates were blocked, and each serially diluted sample was added to the wells. After incubation for 1 to 2 hours at 37°C, the plates were washed and incubated with HRP-conjugated anti-human IgG Fc secondary antibody (Abcam Inc.) for total antibody measurement or a biotin-labeled anti-exatecan/DXd antibody (Abmart) for ADC measurement. After reaction at 37°C for 1 to 2 hours, TMB solution (KPL Inc.) was added directly or after incubation with Streptavidin Protein, HRP (Thermo Scientific) for 40 to 60 minutes at 37°C. A450 in each well was measured with a microplate reader. Concentrations of payload exatecan/DXd in plasma were determined with a validated LC/MS-MS method; the lower limit of quantitation was 0.05 ng/mL. MTX-1000/DS-8201a was intravenously administered at 4.0 mg/kg to rats. Plasma concentrations of ADC, total antibody, and DXd were measured up to 21 days after dose.

In Vitro Stability of MTX-1000/DS-8201a in Plasma

The release rate of exatecan/DXd from MTX-1000/DS-8201a at the concentration of 10 mg/mL at 37°C up to 21 days was evaluated in mouse, rat, monkey, and human plasma.

ELISA

For a binding assay, immune plates were coated with 2.5 mg/mL His-tagged HER2-ECD protein (Sino Biological) in coating buffer and kept overnight at 4°C. After washing, the plates were blocked, and each serially diluted substance was added to the wells. After incubation for 1.5 hours at 37°C, the plates were washed and incubated with HRP-conjugated anti-human IgG secondary antibody for 1 hour at 37°C. After washing, TMB solution was added and A450 in each well was measured with a microplate reader. For the detection of phosphorylated Akt (pAkt), SK-BR-3 cells were preincubated in a 96-well plate for 4 days and then incubated with each substance for 24 hours. After incubation, the cells were lysed and intercellular pAkt and total Akt were detected using a PathScan Phospho-Akt1 (Ser473) Sandwich ELISA Kit and PathScan Total-Akt1 Sandwich ELISA Kit (Cell Signaling Technology) according to the manufacturer's instructions. The relative pAkt of each sample well was calculated by dividing treated normalized pAkt values by untreated normalized pAkt values.

Immunoblotting

KPL-4 cells were treated with each substance. After 24, 48, or 72 hours, the cells were harvested and lysed with M-PER lysis buffer containing Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific). The samples were loaded and separated by SDS-PAGE and blotted onto polyvinylidene difluoride membranes. The membranes were blocked and probed overnight with anti–phospho-Chk1 (Ser345; 133D3) rabbit mAb, anti-Chk1 (2G1D5) mouse mAb, anti–cleaved PARP (Asp214) antibody, anti–β-actin (8H10D10) mouse mAb (Cell Signaling Technology), anti–phospho-Histone H2A.X (Ser139) antibody (Millipore), and anti–Histone H2A.X antibody (Abcam) at 4°C. Then, the membranes were washed and incubated with fluorescence-labeled secondary antibodies for 10 minutes using SNAP intradermally (Millipore). The fluorescence signal was detected using an Odyssey imaging system (LI-COR, Inc.).

Detection of Cellular TOP1ccs

DNA-trapped TOP1 was determined by a modified RADAR assay following a previous study (19). After treatment with TOP1 inhibitors, COLO205 cells (1–3 × 106 cells/sample) were washed with 1× PBS and lysed with 600 μL of DNAzol (Invitrogen) followed by precipitation with 300 μL of 200-proof ethanol by centrifugation at 14,000 rpm. The nucleic acids were collected, washed with 75% ethanol, resuspended in 200 μL of TE buffer, and then heated at 65°C for 15 minutes followed by shearing with sonication (40% output for 10-second pulse and 10-second rest repeated 4 times). The samples were centrifuged at 15,000 rpm for 5 minutes at 4°C, and the supernatant was collected. The sample (1 μL) was saved for the spectrophotometric measurement of absorbance at 260 nm to quantitate DNA content (NanoDrop). Two milligrams of each sample were subjected to slot blot for immunoblotting with anti-TOP1 antibody (no. 556597, BD Biosciences) or anti-dsDNA antibody (no. 3519, Abcam) as a loading control. The intensity of TOP1 and DNA was quantified by densitometric analysis using ImageJ.

Toxicity Studies in Rats and Monkeys

MTX-1000 was intravenously administered intermittently at 3-week intervals over a 6-week period to cynomolgus monkeys (Table 1). Clinical signs, body weight, food consumption, and clinical pathology were monitored throughout the study. A necropsy was conducted on the day after the last administration. The reversibility of the toxic changes was assessed in a subsequent 6-week recovery period in cynomolgus monkeys.

Organoids Preparation for Drug Tests

Human colon cancer organoids were prepared as previously described (34). Organoids were harvested and seeded in a 96-well cell culture plate (Corning, 3799) when organoids grew to 50 μm in diameter. The sandwich method was used for drug tests. Before seeding, 50 μL 50% Matrigel (Corning, 356231, Matrigel mixed with PBS 1:1) was dropped on the bottom of the culture plate as the bottom layer. Then, 10 μL 10% Matrigel (Matrigel mixed with PBS 1:9) containing 50 ± 20 organoids was dropped on the bottom layer as the middle layer. Organoid culture medium (200 μL) was added in each well as the upper layer. Drug tests would start after 1 day of culture.

Colon Cancer Organoids Drug Test Assays

For drug tests, calcein AM was used for living cell staining at day 0. RCO culture medium was removed and replaced by a 200 μL drug-containing culture medium. The drug-containing culture medium was refreshed again after 3 days. Then, calcein AM/PI was used for living/dead cell staining at day 6. Z-stack imaging technique was used for the photographing of organoids at days 0 and 6 (Nikon, Ti2-E). The size of alive organoids was measured using NIS-Elements AR software (Nikon).

Statistical Analysis

All statistical analyses except for toxicity studies were performed using SAS System Release 9.1.3 and 9.2 (SAS Institute Inc.). Statistical analysis in toxicity studies was performed using MUSCOT (YukmsCo., Ltd.). All IC50 and ED50 values were determined by a Sigmoid Emax model, and dose dependency was evaluated by a Spearman rank correlation coefficient hypothesis test.

Statistics and Data

Tests to determine significance are detailed in the relevant figure legends. Briefly, t tests or one-way ANOVAs were used to determine the statistical significance of treatment conditions and two-way ANOVA to discriminate between the contributions of both dose and transformation.

Data Availability

Correspondence and requests for data underlying the findings or materials should be addressed to Xun Meng ([email protected]).

S.-H. Liu reports other support from Multitude Therapeutics outside the submitted work. No disclosures were reported by the other authors.

W. Weng: Resources, investigation, visualization, methodology. T. Meng: Conceptualization, resources, formal analysis, investigation, writing–review and editing. Q. Zhao: Resources, investigation, visualization, methodology. Y. Shen: Investigation, visualization. G. Fu: Investigation, visualization. J. Shi: Investigation, visualization. Y. Zhang: Validation, investigation, visualization. Z. Wang: Investigation, visualization. M. Wang: Resources, validation, investigation, visualization. R. Pan: Investigation, visualization. L. Ma: Investigation, visualization. C. Chen: Investigation, visualization. L. Wang: Investigation, visualization. B. Zhou: Investigation, visualization. H. Zhang: Resources, investigation, visualization. J. Pu: Investigation, visualization. J. Zhang: Investigation, visualization. Y.P. Hu: Investigation, visualization. G. Hua: Conceptualization, resources, project administration, writing–review and editing. Y. Qian: Investigation, visualization. S.-H. Liu: Validation, investigation, visualization, writing–original draft, writing–review and editing. W. Hu: Validation, investigation, visualization, writing–original draft, writing–review and editing. X. Meng: Conceptualization, resources, formal analysis, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing.

This study was funded by Multitude Therapeutics, Abmart Inc., HySlink, Mabcare, D1 Medical Technology Company, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery (2019B030301005), and Fudan University Shanghai Cancer Center. We thank Y. Tang of Multitude Therapeutics for initiating this project and for providing encouragement and support throughout the project. We thank X.X. Meng for reading and editing the manuscript. Finally, we thank Professor Hongbin Ji at the State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China, for encouragement and helpful suggestions.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Cancer Discovery Online (http://cancerdiscovery.aacrjournals.org/).

1.
Joubert
N
,
Beck
A
,
Dumontet
C
,
Denevault-Sabourin
C
.
Antibody–drug conjugates: the last decade
.
Pharmaceuticals
2020
;
13
:
245
.
2.
Nessler
I
,
Menezes
B
,
Thurber
GM
.
Key metrics to expanding the pipeline of successful antibody–drug conjugates
.
Trends Pharmacol Sci
2021
;
42
:
803
12
.
3.
Ogitani
Y
,
Aida
T
,
Hagihara
K
,
Yamaguchi
J
,
Ishii
C
,
Harada
N
, 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
2016
;
22
:
5097
108
.
4.
Modi
S
,
Saura
C
,
Yamashita
T
,
Park
YH
,
Kim
SB
,
Tamura
K
, et al
.
Trastuzumab deruxtecan in previously treated HER2-positive breast cancer
.
N Engl J Med
2020
;
382
:
610
21
.
5.
Adams
E
,
Wildiers
H
,
Neven
P
,
Punie
K
.
Sacituzumab govitecan and trastuzumab deruxtecan: two new antibody-drug conjugates in the breast cancer treatment landscape
.
ESMO Open
2021
;
6
:
100204
.
6.
Okajima
D
,
Yasuda
S
,
Maejima
T
,
Karibe
T
,
Sakurai
K
,
Aida
T
, et al
.
Datopotamab deruxtecan, a novel TROP2-directed antibody-drug conjugate, demonstrates potent antitumor activity by efficient drug delivery to tumor cells
.
Mol Cancer Ther
2021
;
20
:
2329
40
.
7.
Janne
PA
,
Baik
C
,
Su
WC
,
Johnson
ML
,
Hayashi
H
,
Nishio
M
, et al
.
Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer
.
Cancer Discov
2022
;
12
:
74
89
.
8.
Garcia-Alonso
S
,
Ocana
A
,
Pandiella
A
.
Resistance to antibody-drug conjugates
.
Cancer Res
2018
;
78
:
2159
65
.
9.
Collins
DM
,
Bossenmaier
B
,
Kollmorgen
G
,
Niederfellner
G
.
Acquired resistance to antibody-drug conjugates
.
Cancers (Basel)
2019
;
11
:394.
10.
Modi
S
,
Park
H
,
Murthy
RK
,
Iwata
H
,
Tamura
K
,
Tsurutani
J
, et al
.
Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer: results from a phase Ib study
.
J Clin Oncol
2020
;
38
:
1887
96
.
11.
Shitara
K
,
Bang
Y-J
,
Iwasa
S
,
Sugimoto
N
,
Ryu
M-h
,
Sakai
D
, et al
.
Trastuzumab deruxtecan (T-DXd; DS-8201) in patients with HER2-positive advanced gastric or gastroesophageal junction (GEJ) adenocarcinoma: a randomized, phase II, multicenter, open-label study (DESTINY-Gastric01)
.
J Clin Oncol
38
:
2020
(suppl; abstr 4513)
.
12.
Nakagawa
K
,
Nagasaka
M
,
Felip
E
,
Pacheco
J
,
Baik
C
,
Goto
Y
, et al
.
Trastuzumab deruxtecan in HER2-overexpressing metastatic non-small cell lung cancer: interim results of DESTINY-Lung01
.
J Thorac Oncol
2021
;
16
:
S109
S10
. Abstract nr OA04.05.
13.
Hashimoto
Y
,
Koyama
K
,
Kamai
Y
,
Hirotani
K
,
Ogitani
Y
,
Zembutsu
A
, et al
.
A novel HER3-targeting antibody-drug conjugate, U3-1402, exhibits potent therapeutic efficacy through the delivery of cytotoxic payload by efficient internalization
.
Clin Cancer Res
2019
;
25
:
7151
61
.
14.
Coates
JT
,
Sun
S
,
Leshchiner
I
,
Thimmiah
N
,
Martin
EE
,
McLoughlin
D
, 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
2021
;
11
:
2436
45
.
15.
Siena
S
,
Bartolomeo
MD
,
Raghav
KPS
,
Masuishi
T
,
Loupakis
F
,
Kawakami
H
, et al
.
A phase II, multicenter, open-label study of trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressing metastatic colorectal cancer (mCRC): DESTINY-CRC01
.
J Clin Oncol
38
:
2020
(suppl; abstr 4000)
.
16.
Nagai
Y
,
Oitate
M
,
Shiozawa
H
,
Ando
O
.
Comprehensive preclinical pharmacokinetic evaluations of trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody–drug conjugate, in cynomolgus monkeys
.
Xenobiotica
2019
;
49
:
1086
96
.
17.
Chang
CH
,
Wang
Y
,
Zalath
M
,
Liu
D
,
Cardillo
TM
,
Goldenberg
DM
.
Combining ABCG2 inhibitors with IMMU-132, an anti-trop-2 antibody conjugate of SN-38, overcomes resistance to SN-38 in breast and gastric cancers
.
Mol Cancer Ther
2016
;
15
:
1910
9
.
18.
Joto
N
,
Ishii
M
,
Minami
M
,
Kuga
H
,
Mitsui
I
,
Tohgo
A
.
DX-8951f, a water-soluble camptothecin analog, exhibits potent antitumor activity against a human lung cancer cell line and its SN-38-resistant variant
.
Int J Cancer
1997
;
72
:
680
6
.
19.
Jo
U
,
Murai
Y
,
Agama
KK
,
Sun
Y
,
Saha
LK
,
Yang
X
, et al
.
TOP1-DNA trapping by exatecan and combination therapy with ATR inhibitor
.
Mol Cancer Ther
2022
;
21
:
1090
102
.
20.
van Hattum
AH
,
Hoogsteen
IJ
,
Schluper
HM
,
Maliepaard
M
,
Scheffer
GL
,
Scheper
RJ
, et al
.
Induction of breast cancer resistance protein by the camptothecin derivative DX-8951f is associated with minor reduction of antitumour activity
.
Br J Cancer
2002
;
87
:
665
72
.
21.
Khera
E
,
Dong
S
,
Huang
H
,
de Bever
L
,
van Delft
FL
,
Thurber
GM
.
Cellular-resolution imaging of bystander payload tissue penetration from antibody-drug conjugates
.
Mol Cancer Ther
2022
;
21
:
310
21
.
22.
Ogitani
Y
,
Abe
Y
,
Iguchi
T
,
Yamaguchi
J
,
Terauchi
T
,
Kitamura
M
, et al
.
Wide application of a novel topoisomerase I inhibitor-based drug conjugation technology
.
Bioorg Med Chem Lett
2016
;
26
:
5069
72
.
23.
Conilh
L
,
Fournet
G
,
Fourmaux
E
,
Murcia
A
,
Matera
EL
,
Joseph
B
, et al
.
Exatecan antibody drug conjugates based on a hydrophilic polysarcosine drug-linker platform
.
Pharmaceuticals (Basel)
2021
;
14
:
247
63
.
24.
Burke
PJ
,
Senter
PD
,
Meyer
DW
,
Miyamoto
JB
,
Anderson
M
,
Toki
BE
, et al
.
Design, synthesis, and biological evaluation of antibody-drug conjugates comprised of potent camptothecin analogues
.
Bioconjug Chem
2009
;
20
:
1242
50
.
25.
Bryden
F
,
Martin
C
,
Letast
S
,
Lles
E
,
Vieitez-Villemin
I
,
Rousseau
A
, et al
.
Impact of cathepsin B-sensitive triggers and hydrophilic linkers on in vitro efficacy of novel site-specific antibody-drug conjugates
.
Org Biomol Chem
2018
;
16
:
1882
9
.
26.
Bargh
JD
,
Isidro-Llobet
A
,
Parker
JS
,
Spring
DR
.
Cleavable linkers in antibody-drug conjugates
.
Chem Soc Rev
2019
;
48
:
4361
74
.
27.
Pommier
Y
,
Cushman
M
.
The indenoisoquinoline noncamptothecin topoisomerase I inhibitors: update and perspectives
.
Mol Cancer Ther
2009
;
8
:
1008
14
.
28.
Hopkins
AL
,
Keseru
GM
,
Leeson
PD
,
Rees
DC
,
Reynolds
CH
.
The role of ligand efficiency metrics in drug discovery
.
Nat Rev Drug Discov
2014
;
13
:
105
21
.
29.
Jeffrey
SC
,
Nguyen
MT
,
Andreyka
JB
,
Meyer
DL
,
Doronina
SO
,
Senter
PD
.
Dipeptide-based highly potent doxorubicin antibody conjugates
.
Bioorg Med Chem Lett
2006
;
16
:
358
62
.
30.
Kinneer
K
,
Wortmann
P
,
Cooper
ZA
,
Dickinson
NJ
,
Masterson
L
,
Cailleau
T
, et al
.
Design and preclinical evaluation of a novel B7-H4–directed antibody–drug conjugate, AZD8205, alone and in combination with the PARP1-selective inhibitor AZD5305
.
Clin Cancer Res
2023
;
29
:
1086
101
.
31.
Hoang Thi
TT
,
Pilkington
EH
,
Nguyen
DH
,
Lee
JS
,
Park
KD
,
Truong
NP
.
The importance of Poly(ethylene glycol) alternatives for overcoming PEG immunogenicity in drug delivery and bioconjugation
.
Polymers (Basel)
2020
;
12
:
298
318
.
32.
Singhai
M
,
Bhattacharya
S
.
Polysarcosine: the best alternative of poly (Ethylene Glycol)
.
Curr Appl Polym Sci
2021
;
4
:
93
8
.
33.
Sharma
S
,
Li
Z
,
Bussing
D
,
Shah
DK
.
Evaluation of quantitative relationship between target expression and antibody-drug conjugate exposure inside cancer cells
.
Drug Metab Dispos
2020
;
48
:
368
77
.
34.
Yao
Y
,
Xu
X
,
Yang
L
,
Zhu
J
,
Wan
J
,
Shen
L
, et al
.
Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer
.
Cell Stem Cell
2020
;
26
:
17
26
.
35.
Muller
PY
,
Milton
MN
.
The determination and interpretation of the therapeutic index in drug development
.
Nat Rev Drug Discov
2012
;
11
:
751
61
.
36.
Cardillo
TM
,
Sharkey
RM
,
Rossi
DL
,
Arrojo
R
,
Mostafa
AA
,
Goldenberg
DM
.
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
2017
;
23
:
3405
15
.
37.
Xing
L
,
Lin
L
,
Yu
T
,
Li
Y
,
Cho
SF
,
Liu
J
, et al
.
A novel BCMA PBD-ADC with ATM/ATR/WEE1 inhibitors or bortezomib induce synergistic lethality in multiple myeloma
.
Leukemia
2020
;
34
:
2150
62
.
38.
Jaaks
P
,
Coker
EA
,
Vis
DJ
,
Edwards
O
,
Carpenter
EF
,
Leto
SM
, et al
.
Effective drug combinations in breast, colon and pancreatic cancer cells
.
Nature
2022
;
603
:
166
73
.
39.
Jo
U
,
Senatorov
IS
,
Zimmermann
A
,
Saha
LK
,
Murai
Y
,
Kim
SH
, et al
.
Novel and highly potent ATR inhibitor M4344 kills cancer cells with replication stress, and enhances the chemotherapeutic activity of widely used DNA damaging agents
.
Mol Cancer Ther
2021
;
20
:
1431
41
.
40.
Uchibori
K
,
Inase
N
,
Araki
M
,
Kamada
M
,
Sato
S
,
Okuno
Y
, et al
.
Brigatinib combined with anti-EGFR antibody overcomes osimertinib resistance in EGFR-mutated non-small-cell lung cancer
.
Nat Commun
2017
;
8
:
14768
.
41.
Kepp
O
,
Senovilla
L
,
Vitale
I
,
Vacchelli
E
,
Adjemian
S
,
Agostinis
P
, et al
.
Consensus guidelines for the detection of immunogenic cell death
.
Oncoimmunology
2014
;
3
:
e955691
.
42.
Lyski
RD
,
Bou
LB
,
Lau
UY
,
Meyer
DW
,
Cochran
JH
,
Okeley
NM
, et al
.
Development of novel antibody-camptothecin conjugates
.
Mol Cancer Ther
2021
;
20
:
329
39
.
43.
Li
W
,
Veale
KH
,
Qiu
QF
,
Sinkevicius
KW
,
Maloney
EK
,
Costoplus
JA
, et al
.
Synthesis and evaluation of camptothecin antibody-drug conjugates
.
ACS Med Chem Lett
2019
;
10
:
1386
92
.
44.
Sugimori
M
,
Ejima
A
,
Ohsuki
S
,
Uoto
K
,
Mitsui
I
,
Kawato
Y
, et al
.
Synthesis and antitumor activity of ring A- and F-modified hexacyclic camptothecin analogues
.
J Med Chem
1998
;
41
:
2308
18
.
45.
Tiberghien
AC
,
Levy
JN
,
Masterson
LA
,
Patel
NV
,
Adams
LR
,
Corbett
S
, et al
.
Design and synthesis of tesirine, a clinical antibody-drug conjugate pyrrolobenzodiazepine dimer payload
.
ACS Med Chem Lett
2016
;
7
:
983
7
.
46.
Lyon
RP
,
Bovee
TD
,
Doronina
SO
,
Burke
PJ
,
Hunter
JH
,
Neff-LaFord
HD
, et al
.
Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index
.
Nat Biotechnol
2015
;
33
:
733
5
.
47.
Burke
PJ
,
Hamilton
JZ
,
Jeffrey
SC
,
Hunter
JH
,
Doronina
SO
,
Okeley
NM
, et al
.
Optimization of a PEGylated glucuronide-monomethylauristatin E linker for antibody–drug conjugates
.
Mol Cancer Ther
2017
;
16
:
116
23
.
48.
Rowinsky
EK
.
Preclinical and clinical development of exatecan (DX-8951f): a hexacyclic camptothecin analog
.
In:
Adams
VR
,
Burke
TG
,
editors
.
Camptothecins in cancer therapy. Totowa
(NJ)
:
Humana Press
;
2018
. p
317
41
.
49.
Bleijs
M
,
van de Wetering
M
,
Clevers
H
,
Drost
J
.
Xenograft and organoid model systems in cancer research
.
EMBO J
2019
;
38
:
e101654
.
50.
Kukal
S
,
Guin
D
,
Rawat
C
,
Bora
S
,
Mishra
MK
,
Sharma
P
, et al
.
Multidrug efflux transporter ABCG2: expression and regulation
.
Cell Mol Life Sci
2021
;
78
:
6887
939
.

Supplementary data