Abstract
Antibody–drug conjugates (ADCs) are effective targeted therapeutics but are limited in their ability to incorporate less-potent payloads, varied drug mechanisms of action, different drug release mechanisms and tunable drug-to-antibody ratios. Here we introduce a technology to overcome these limitations called ‘antibody–bottlebrush prodrug conjugates’ (ABCs). An ABC consists of an IgG1 monoclonal antibody covalently conjugated to the terminus of a compact bivalent bottlebrush prodrug that has payloads bound through cleavable linkers and polyethylene glycol branches. This design enables the synthesis of ABCs with tunable average drug-to-antibody ratios up to two orders of magnitude greater than those of traditional ADCs. We demonstrate the functional flexibility and manufacturing efficiency of this technology by synthesizing more than 10 different ABCs targeting either HER2 or MUC1 using drugs with potencies spanning several orders of magnitude as well as imaging agents for ABC visualization and photocatalysts for proximity-based labeling of the ABC interactome. ABCs display high target engagement, high cell uptake and improved efficacy in tumor models compared to conventional HER2-targeted ADCs, suggesting promise for clinical translation.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
All data supporting the findings of this study are available within the article, in its supplementary information and on figshare (https://doi.org/10.6084/m9.figshare.29414048)90 and can be obtained from the corresponding authors upon reasonable request. Source data are provided with this paper.
References
Siegel, R. L., Miller, K. D., Fuchs, H. E. & Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 72, 7–33 (2022).
Wang, Z., Li, H., Gou, L., Li, W. & Wang, Y. Antibody–drug conjugates: recent advances in payloads. Acta Pharm. Sin. B 13, 4025–4059 (2023).
Hobson, A. D. Chapter One - Antibody drug conjugates beyond cytotoxic payloads. Prog. Med. Chem. 62, 1–59 (2023).
Tarantino, P., Ricciuti, B., Pradhan, S. M. & Tolaney, S. M. Optimizing the safety of antibody–drug conjugates for patients with solid tumours. Nat. Rev. Clin. Oncol. 20, 558–576 (2023).
Dumontet, C., Reichert, J. M., Senter, P. D., Lambert, J. M. & Beck, A. Antibody–drug conjugates come of age in oncology. Nat. Rev. Drug Discov. 22, 641–661 (2023).
Chalouni, C. & Doll, S. Fate of antibody-drug conjugates in cancer cells. J. Exp. Clin. Cancer Res. 37, 20 (2018).
Tashima, T. Delivery of drugs into cancer cells using antibody–drug conjugates based on receptor-mediated endocytosis and the enhanced permeability and retention effect. Antibodies 11, 78 (2022).
ADCs’ revival. Nat. Biotechnol. 41, 740 (2023).
Tarantino, P. et al. Antibody–drug conjugates: smart chemotherapy delivery across tumor histologies. CA Cancer J. Clin. 72, 165–182 (2022).
Beck, A., Goetsch, L., Dumontet, C. & Corvaïa, N. Strategies and challenges for the next generation of antibody–drug conjugates. Nat. Rev. Drug Discov. 16, 315–337 (2017).
Drago, J. Z., Modi, S. & Chandarlapaty, S. Unlocking the potential of antibody–drug conjugates for cancer therapy. Nat. Rev. Clin. Oncol. 18, 327–344 (2021).
Gan, H. K., van den Bent, M., Lassman, A. B., Reardon, D. A. & Scott, A. M. Antibody–drug conjugates in glioblastoma therapy: the right drugs to the right cells. Nat. Rev. Clin. Oncol. 14, 695–707 (2017).
Fu, Z., Li, S., Han, S., Shi, C. & Zhang, Y. Antibody drug conjugate: the ‘biological missile’ for targeted cancer therapy. Signal Transduct. Target. Ther. 7, 93 (2022).
Tong, J. T. W., Harris, P. W. R., Brimble, M. A. & Kavianinia, I. An insight into FDA approved antibody-drug conjugates for cancer therapy. Molecules 26, 5847 (2021).
Criscitiello, C., Morganti, S. & Curigliano, G. Antibody–drug conjugates in solid tumors: a look into novel targets. J. Hematol. Oncol. 14, 20 (2021).
Nagayama, A., Ellisen, L. W., Chabner, B. & Bardia, A. Antibody–drug conjugates for the treatment of solid tumors: clinical experience and latest developments. Target. Oncol. 12, 719–739 (2017).
Jabbour, E., Paul, S. & Kantarjian, H. The clinical development of antibody–drug conjugates—lessons from leukaemia. Nat. Rev. Clin. Oncol. 18, 418–433 (2021).
Yu, B. & Liu, D. Antibody-drug conjugates in clinical trials for lymphoid malignancies and multiple myeloma. J. Hematol. Oncol. 12, 94 (2019).
Zhao, P. et al. Recent advances of antibody drug conjugates for clinical applications. Acta Pharm. Sin. B 10, 1589–1600 (2020).
Nessler, I., Menezes, B. & Thurber, G. M. Key metrics to expanding the pipeline of successful antibody–drug conjugates. Trends Pharmacol. Sci. 42, 803–812 (2021).
Mckertish, C. M. & Kayser, V. Advances and limitations of antibody drug conjugates for cancer. Biomedicines 9, 872 (2021).
Su, Z. et al. Antibody–drug conjugates: recent advances in linker chemistry. Acta Pharm. Sin. B 11, 3889–3907 (2021).
Abelman, R. O., Wu, B., Spring, L. M., Ellisen, L. W. & Bardia, A. Mechanisms of resistance to antibody–drug conjugates. Cancers 15, 1278 (2023).
Nguyen, T. D., Bordeau, B. M. & Balthasar, J. P. Mechanisms of ADC toxicity and strategies to increase ADC tolerability. Cancers 15, 713 (2023).
Chudasama, V., Maruani, A. & Caddick, S. Recent advances in the construction of antibody–drug conjugates. Nat. Chem. 8, 114–119 (2016).
Lyon, R. P. et al. Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33, 733–735 (2015).
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 6, 100204 (2021).
Goldenberg, D. M. & Sharkey, R. M. Sacituzumab govitecan, a novel, third-generation, antibody-drug conjugate (ADC) for cancer therapy. Expert Opin. Biol. Ther. 20, 871–885 (2020).
Modi, S. et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N. Engl. J. Med. 382, 610–621 (2020).
Rubahamya, B., Dong, S. & Thurber, G. M. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. Sci. Adv. 10, eadk1894 (2024).
Kukkar, D., Kukkar, P., Kumar, V., Hong, J. & Kim, K. H. A. Deep, recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens. Bioelectron. 173, 112787 (2021).
Yurkovetskiy, A. V. et al. Dolaflexin: a novel antibody–drug conjugate platform featuring high drug loading and a controlled bystander effect. Mol. Cancer Ther. 20, 885–895 (2021).
Müllner, M. Molecular polymer bottlebrushes in nanomedicine: therapeutic and diagnostic applications. Chem. Commun. 58, 5683–5716 (2022).
Johnson, J. A. et al. Core-clickable PEG-branch-azide bivalent-bottle-brush polymers by ROMP: grafting-through and clicking-to. J. Am. Chem. Soc. 133, 559–566 (2011).
Golder, M. R. et al. Reduction of liver fibrosis by rationally designed macromolecular telmisartan prodrugs. Nat. Biomed. Eng. 2, 822–830 (2018).
Bhagchandani, S. H. et al. Engineering kinetics of TLR7/8 agonist release from bottlebrush prodrugs enables tumor-focused immune stimulation. Sci. Adv. 9, eadg2239 (2023).
Liao, L. et al. A convergent synthetic platform for single-nanoparticle combination cancer therapy: ratiometric loading and controlled release of cisplatin, doxorubicin, and camptothecin. J. Am. Chem. Soc. 136, 5896–5899 (2014).
Kolb, H. C., Finn, M. G. & Sharpless, K. B. Click chemistry: diverse chemical function from a few good reactions. Angew. Chem. Int. Ed. 40, 2004–2021 (2001).
Sletten, E. M. & Bertozzi, C. R. Bioorthogonal chemistry: fishing for selectivity in a sea of functionality. Angew. Chem. Int. Ed. 48, 6974–6998 (2009).
Agard, N. J., Prescher, J. A. & Bertozzi, C. R. A strain-promoted [3 + 2] azide-alkyne cycloaddition for covalent modification of biomolecules in living systems. J. Am. Chem. Soc. 126, 15046–15047 (2004).
Thirumurugan, P., Matosiuk, D. & Jozwiak, K. Click chemistry for drug development and diverse chemical–biology applications. Chem. Rev. 113, 4905–4979 (2013).
Ogba, O. M., Warner, N. C., O’Leary, D. J. & Grubbs, R. H. Recent advances in ruthenium-based olefin metathesis. Chem. Soc. Rev. 47, 4510–4544 (2018).
Fu, L., Zhang, T., Fu, G. & Gutekunst, W. R. Relay conjugation of living metathesis polymers. J. Am. Chem. Soc. 140, 12181–12188 (2018).
Knall, A. C. & Slugovc, C. Inverse electron demand Diels–Alder (iEDDA)-initiated conjugation: a (high) potential click chemistry scheme. Chem. Soc. Rev. 42, 5131–5142 (2013).
Oliveira, B. L., Guo, Z. & Bernardes, G. J. L. Inverse electron demand Diels–Alder reactions in chemical biology. Chem. Soc. Rev. 46, 4895–4950 (2017).
Blackman, M. L., Royzen, M. & Fox, J. M. Tetrazine ligation: fast bioconjugation based on inverse-electron-demand Diels–Alder reactivity. J. Am. Chem. Soc. 130, 13518–13519 (2008).
Oh, D. Y. & Bang, Y. J. HER2-targeted therapies—a role beyond breast cancer. Nat. Rev. Clin. Oncol. 17, 33–48 (2020).
Nakada, T., Sugihara, K., Jikoh, T., Abe, Y. & Agatsuma, T. The latest research and development into the antibody–drug conjugate, [fam-] trastuzumab deruxtecan (DS-8201a), for HER2 cancer therapy. Chem. Pharm. Bull. (Tokyo) 67, 173–185 (2019).
Hou, Y. & Lu, H. Protein PEPylation: a new paradigm of protein–polymer conjugation. Bioconjug. Chem. 30, 1604–1616 (2019).
Chen, P., Yun, W., Sun, T., Lin, J. & Zhang, K. Enabling safer, more potent oligonucleotide therapeutics with bottlebrush polymer conjugates. J. Control. Release 366, 44–51 (2024).
Yu, X. et al. Reducing affinity as a strategy to boost immunomodulatory antibody agonism. Nature 614, 539–547 (2023).
Oostindie, S. C., Lazar, G. A., Schuurman, J. & Parren, P. W. H. I. Avidity in antibody effector functions and biotherapeutic drug design. Nat. Rev. Drug Discov. 21, 715–735 (2022).
Gall, V. A. et al. Trastuzumab increases HER2 uptake and cross-presentation by dendritic cells. Cancer Res. 77, 5374–5383 (2017).
Geri, J. B. et al. Microenvironment mapping via Dexter energy transfer on immune cells. Science 367, 1091–1097 (2020).
Vohidov, F. et al. Design of BET inhibitor bottlebrush prodrugs with superior efficacy and devoid of systemic toxicities. J. Am. Chem. Soc. 143, 4714–4724 (2021).
Zafar, H., Liu, B., Nguyen, H. V. T. & Johnson, J. A. Caspase-3-responsive, fluorogenic bivalent bottlebrush polymers. ACS Macro Lett. 13, 571–576 (2024).
Tsao, L. et al. Effective extracellular payload release and immunomodulatory interactions govern the therapeutic effect of trastuzumab deruxtecan (T-DXd). Nat. Commun. 16, 3167 (2025).
Pyzik, M., Kozicky, L. K., Gandhi, A. K. & Blumberg, R. S. The therapeutic age of the neonatal Fc receptor. Nat. Rev. Immunol. 23, 415–432 (2023).
Akaiwa, M., Dugal-Tessier, J. & Mendelsohn, B. A. Antibody–drug conjugate payloads; study of auristatin derivatives. Chem. Pharm. Bull. (Tokyo) 68, 201–211 (2020).
Junutula, J. R. et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26, 925–932 (2008).
Ejigah, V. et al. Approaches to improve macromolecule and nanoparticle accumulation in the tumor microenvironment by the enhanced permeability and retention effect. Polymers 14, 2601 (2022).
Tolcher, A. W. et al. Randomized phase II study of BR96-doxorubicin conjugate in patients with metastatic breast cancer. J. Clin. Oncol. 17, 478 (1999).
Hendrikx, J. J. M. A. et al. Fixed dosing of monoclonal antibodies in oncology. Oncologist 22, 1212 (2017).
Li, Y. et al. Targeted immunotherapy for HER2-low breast cancer with 17p loss. Sci. Transl. Med. 13, eabc6894 (2021).
Burslem, G. M. & Crews, C. M. Proteolysis-targeting chimeras as therapeutics and tools for biological discovery. Cell 181, 102–114 (2020).
Dale, B. et al. Advancing targeted protein degradation for cancer therapy. Nat. Rev. Cancer 21, 638–654 (2021).
Wu, T. et al. Targeted protein degradation as a powerful research tool in basic biology and drug target discovery. Nat. Struct. Mol. Biol. 27, 605–614 (2020).
Li, X. & Song, Y. Proteolysis-targeting chimera (PROTAC) for targeted protein degradation and cancer therapy. J. Hematol. Oncol. 13, 50 (2020).
Raina, K. et al. PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc. Natl Acad. Sci. USA 113, 7124–7129 (2016).
Noblejas-López, M. et al. Activity of BET-proteolysis targeting chimeric (PROTAC) compounds in triple negative breast cancer. J. Exp. Clin. Cancer Res. 38, 383 (2019).
Sun, X. et al. A chemical approach for global protein knockdown from mice to non-human primates. Cell Discov. 5, 10 (2019).
Noblejas-López, M. et al. MZ1 co-operates with trastuzumab in HER2 positive breast cancer. J. Exp. Clin. Cancer Res. 40, 106 (2021).
Dragovich, P. S. Degrader-antibody conjugates. Chem. Soc. Rev. 51, 3886–3897 (2022).
Chen, W. et al. MUC1: structure, function, and clinic application in epithelial cancers. Int. J. Mol. Sci. 22, 6567 (2021).
Cheever, M. A. et al. The prioritization of cancer antigens: a National Cancer Institute pilot project for the acceleration of translational research. Clin. Cancer Res. 15, 5323–5337 (2009).
Liu, B. et al. An organometallic swap strategy for bottlebrush polymer–protein conjugate synthesis. Chem. Commun. 60, 4238–4241 (2024).
Hamblett, K. J. et al. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin. Cancer Res. 10, 7063–7070 (2004).
Bai, S. et al. Cylindrical polymer brushes-anisotropic unimolecular micelle drug delivery system for enhancing the effectiveness of chemotherapy. Bioact. Mater. 6, 2894–2904 (2021).
Rabanel, J. M. et al. Deep tissue penetration of bottle-brush polymers via cell capture evasion and fast diffusion. ACS Nano 16, 21583–21599 (2022).
Strasser, P. et al. Degradable bottlebrush polypeptides and the impact of their architecture on cell uptake, pharmacokinetics, and biodistribution in vivo. Small 19, 2300767 (2023).
Qi, Y. et al. A brush-polymer/exendin-4 conjugate reduces blood glucose levels for up to five days and eliminates poly(ethylene glycol) antigenicity. Nat. Biomed. Eng. 1, 0002 (2016).
Gil Alvaradejo, G. et al. Polyoxazoline-based bottlebrush and brush-arm star polymers via ROMP: syntheses and applications as organic radical contrast agents. ACS Macro Lett. 8, 473–478 (2019).
Shieh, P., Nguyen, H. V. T. & Johnson, J. A. Tailored silyl ether monomers enable backbone-degradable polynorbornene-based linear, bottlebrush, and star copolymers through ROMP. Nat. Chem. 11, 1124–1132 (2019).
Trowbridge, A. D. et al. Small molecule photocatalysis enables drug target identification via energy transfer. Proc. Natl Acad. Sci. USA 119, e2208077119 (2022).
Oakley, J. V. et al. Radius measurement via super-resolution microscopy enables the development of a variable radii proximity labeling platform. Proc. Natl Acad. Sci. USA 119, e2203027119 (2022).
Pan, C., Knutson, S. D., Huth, S. W. & MacMillan, D. W. C. µMap proximity labeling in living cells reveals stress granule disassembly mechanism. Nat. Chem. Biol. 21, 490–500 (2025).
Demichev, V., Messner, C. B., Vernardis, S. I., Lilley, K. S. & Ralser, M. DIA-NN: neural networks and interference correction enable deep proteome coverage in high throughput. Nat. Methods 17, 41–44 (2020).
Demichev, V. et al. dia-PASEF data analysis using FragPipe and DIA-NN for deep proteomics of low sample amounts. Nat. Commun. 13, 3944 (2022).
Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).
Liu, B. et al. Data for ‘antibody-bottlebrush prodrug conjugates for targeted cancer therapy’. figshare. https://doi.org/10.6084/m9.figshare.29414048 (2025).
Acknowledgements
We thank the National Institutes of Health (2R01CA220468-06A1) and the Yosemite–American Cancer Society Award (YACS-24-1343208-01-YACS) for support. B.L. thanks the Ludwig Center of the Koch Institute for Integrative Cancer Research at MIT for a postdoctoral fellowship and the Koch Institute Frontier Research Program for support. P.M. would like to acknowledge support from the National Science Foundation Graduate Research Fellowship Program (NSF-GRFP, grant no. 2141064). Z.H.B., P.A.R. and D.W.C.M. are thankful for the financial support provided by the National Institutes of Health, National Institute of General Medical Sciences (R35GM134897-04); the Princeton Catalysis Initiative; Princeton University; and kind gifts from Merck, Janssen, Bristol Myers Squibb, Genentech, Genmab and Pfizer. N.J. is supported by the US Department of Defense (DOD award W81XWH-22-1-1122). We are grateful for the use of open-access tools, including DIA-NN, Perseus and BioGRID.
Author information
Authors and Affiliations
Contributions
B.L. and J.A.J. conceived the project idea. B.L. designed and performed the polymerization and bioconjugation. B.L. and H.V.-T.N. synthesized and characterized the macromonomers. B.L. performed the in vitro experiments. B.L., H.V.-T.N. and Y.J. performed the in vivo experiments. A.X.W., V.L., Z.S., Y.D., P.L.M., Y.W., W.W., S.B., S.H., P.S., S.L.K. and M.B. assisted with the experiments. Z.H.B. and P.A.R. performed µMap optimization and proteomic analysis, under the guidance of D.W.C.M. N.J., M.D. and R.M.E. provided the site-specific thiol-mAb. J.A.J. supervised the research. B.L. and J.A.J. analyzed the data and wrote the manuscript, and all authors discussed and commented on it.
Corresponding author
Ethics declarations
Competing interests
J.A.J., H.V.-T.N. and Y.J. are shareholders of Window Therapeutics. D.W.C.M. declares an ownership interest in Dexterity Pharma, which has commercialized materials used in this work. The remaining authors declare no competing interests.
Peer review
Peer review information
Nature Biotechnology thanks Charles Dumontet and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 ABC synthesis using different stoichiometries of BP-Tz and Ab-TCO.
a. Non-reducing SDS-PAGE analysis of model IgG1-based ABCs as a function of synthesis stoichiometry. BAR = average brush–antibody ratio. b. Non-reducing SDS-PAGE analysis of Trastuzumab-based ABCs as a function of synthesis stoichiometry. c. Flow cytometry histograms showing Cy5.5-HER2 uptake into HER2 + BT-474 cells as a function of synthesis stoichiometry (that is, the constructs are present as a mixture of BAR values). The x-axis represents the Cy5.5 fluorescence intensity. Since the number of Cy5.5 dyes per ABC increases with BAR, histograms are normalized to the total Cy5.5 loading for each construct to enable comparison (normalized to 40 µg/mL Cy5.5-BP, 1 h). d. Flow cytometry histograms for the same constructs shown in panel c normalized by antibody dose (normalized to 10 µg/mL mAb, 1 h). The x-axis represents the Cy5.5 fluorescence intensity. e. and f. Flow cytometry histograms for BT-474 cell uptake of isolated Cy5.5-HER2 ABCs with different BAR values (25 µg/mL, 1 h) (that is, ABCs with each BAR were separated from the synthesis mixture). Results are presented as mean ± SEM (n = 3 biological replicates). Statistical analysis was done using a 2-tailed t-test. For these statistical tests, **** denotes P < 0.001.
Extended Data Fig. 2 HER2-targeted ABCs synthesized via site-specific cysteine conjugation.
a. Non-reducing SDS-PAGE analysis of site-specifically conjugated Cy5.5-HER ABCs. Two independent experiments were performed with similar results. b. Flow cytometry histograms showing enhanced BT474 cell uptake for site-specific Cy5.5-HER2 ABC compared to BPD alone. The x-axis represents the Cy5.5 fluorescence intensity. c. and d. Quantification of cell uptake based on mean fluorescence intensity from flow cytometry (n = 3 biological replicates). e. and f. Flow cytometry histograms (The x-axis represents the Cy5.5 fluorescence intensity) and quantification, respectively, for cell uptake studies comparing site-specific Cy5.5-HER2 to stochastically functionalized lysine-based Cy5.5-HER2 ABC with BAR of 1 (BT474 cells, 20 µg/mL, 60 min incubation). Both ABCs have the same 5% Cy5.5 concentration. ABCK is the lysine-conjugated Cy5.5-HER2 ABC prepared from commercial Trastuzumab. We note that this ABC was stored at 4 °C for ~1.5 years prior to this study, demonstrating excellent long-term storage stability. T-ABCK is a stochastic Lys conjugate prepared using the engineered Trastuzumab designed for cysteine conjugation; this construct is designed to rule out differences in cell uptake between Lys-conjugated commercial Trastuzumab and the engineered antibody. Finally, T-ABCC is the site-specific cysteine conjugate Cy5.5-HER2. Results are presented as mean ± SEM (n = 3 biological replicates). Statistical analysis was done using a 2-tailed t-test. For these statistical tests, NS denotes non-significant; **, P < 0.01.
Extended Data Fig. 3 HER2+BT474 cell uptake and toxicity experiments comparing C5.5-labeled constructs.
a. Cell uptake studies comparing Cy5.5-HER2 to non-targeted controls Cy5.5-IgG1 and Cy5.5-BP, each containing 1% Cy5.5 loading (40 µg/mL, 60 min incubation, BAR = 3 for ABCs). The x-axis represents the Cy5.5 fluorescence intensity. b. Cell uptake for similar constructs with 5% Cy5.5 loadings (40 µg/mL, 60 min incubation, BAR = 3 for ABCs). The x-axis represents the Cy5.5 fluorescence intensity. c. Cytotoxicity of PTX-HER2 ABC compared to non-targeted PTX-BP (BPD only), a mixture of PTX-BPD and Trastuzumab (aHER2), and Trastuzumab (aHER2) alone for 24 h incubation. Results are presented as mean ± SEM (n = 3 biological replicates). The ABC PTX-HER2 displays improved cytotoxicity compared to controls. d. Confocal fluorescence microscopy images showed greater cell engagement and uptake for Cy5.5-HER2 compared to non-targeted controls (50 μg/mL, 6 h incubation, 1% Cy5.5 labeled ABC). Two independent experiments were performed with similar results.
Extended Data Fig. 4 In vitro targeting abilities of ABCs across cell lines with different HER2 expression.
a) Flow cytometry histograms demonstrating that Cy5.5-HER2 ABC uptake is dependent on HER2 expression. The x-axes represents the Cy5.5 fluorescence intensity. Cell uptake was studied using cell lines with varied HER2 expression: MCF-10A (HER2–); SKOV-3 (HER2 medium); SKBR-3 (HER2 high). Cells were treated with Cy5.5-HER2 ABC or non-targeting Cy5.5-BP polymer (1% Cy5.5 labeling) under the conditions listed at the bottom of the figure (varied times and concentrations). (b-d) Cytotoxicity results for HER2-targeted ABCs comprising different payloads in cell lines with varied HER2 expression. Results are presented as mean ± SEM (n = 3 biological replicates). b. MTT assay results for PTX-HER2 compared to non-targeted PTX-BP in HER2 high, medium, and negative (from left to right), respectively, cell lines following 24 h and 72 h incubation. c. HER2 + SKBR3 cell viability results for ABCs with different payloads (from left to right: MMAE, SN-38, and DOX). d. HER2– MCF-10A cell viability results for ABCs with different payloads (from left to right: MMAE, SN-38, and DOX).
Extended Data Fig. 5 Imaging of cell uptake and payload release in BT-474 cells.
a. Confocal fluorescence images of BT-474 cells incubated with Cy5.5-HER2 (20 μg/mL) for different times. b. Confocal fluorescence images of BT-474 cells incubated with “theranostic” ABC SN38-Cy5.5-HER2 (50 μg/mL) for different times. White arrows point to cell nuclei where SN38 has localized following release from the ABC. In panels a and b, 4 h + 24 h and 4 h + 72 h mean that the cells were incubated with Cy5.5-HER2 or SN38-Cy5.5-HER2, respectively, for 4 h. Then, the cells were washed with PBS buffer three times and further incubated for an additional 24 h or 72 h before imaging. Two independent experiments were performed with similar results.
Extended Data Fig. 6 Proposed ABC cell uptake and drug release mechanism.
First, ABCs bind to the cell surface through antibody-antigen interactions (upper left). Then, bound ABCs enter the cells through receptor-mediated endocytosis. Inside the endosome or lysosome, covalently attached payloads are released and subsequently diffuse to regions of the cell (for example, the nucleus or cytosol) to perform their MoA. Graphic created with BioRender.com.
Extended Data Fig. 7 Time-dependent biodistribution (BD) studies.
a. Ex vivo images of organs from mice (n = 3) at different time points following administration of Cy5.5-HER2 and non-targeted controls Cy5.5-IgG1 and Cy5.5-BP. b. Quantification of time-dependent BD for targeted and non-targeted constructs as quantified by fluorescence imaging. The bottom bar graphs correspond to the tumor fluorescence signals for different treatment groups, showing substantially greater tumor accumulation for HER2-targeting Cy5.5-HER2. Results are presented as mean ± SEM (n = 3 biological replicates). Statistical analysis was done using a 2-tailed t-test. For these statistical tests, NS denotes non-significant; *, P < 0.05; **, P < 0.01.
Extended Data Fig. 8 In vitro and in vivo evaluation of the anticancer efficiency of ABCs with different payloads on BT-474 cells or xenograft tumor model.
a. Cell viability studies for ABCs with different payloads compared to their nontargeting BPs (top and middle row, from left to right: MMAE, SN-38, and DOX; top row: 2 days incubation; bottom row: 5 days incubation) and different ADCs (bottom row, from left to right: T-DM1 and T-DXd, 2 d or 5 d incubation). Each data point represents the mean of three independent replicates (n = 3 biological replicates). b. BT-474 tumor volumes at day 40 for mice given MMAE-based ABCs and non-targeted controls (n = 4 mice/group). c. Ex vivo images of the tumors at day 40 for mice given MMAE-based constructs. d. Enlarged Fig. 3d for tumor volume measurement with dose schedule illustrated via green arrows (n = 6 mice/group). e. SDS-PAGE analysis of ABCs with DOX as the payload. f. BT-474 tumor volumes at day 40 for mice given DOX-based ABCs and non-targeted controls (n = 6 mice/group). g. Ex vivo images of the tumors at day 40 for mice given DOX-based constructs. Results are presented as mean ± SEM. Statistical analysis was done using a 2-tailed t-test. For these statistical tests, NS denotes non-significant; *, P < 0.05; **, P < 0.01.
Extended Data Fig. 9 Images of mice organs and xenografts examined after ABC treatments in the BT-474 xenograft tumor model.
Tissue sections were formalin-fixed paraffin-embedded (FFPE), stained with hematoxylin and eosin (H&E), and evaluated by a board-certified veterinary pathologist. a. No sign of toxicity was observed in the heart, lung, liver, spleen, and kidney, supporting a good safety profile. b. Xenografts with different magnifications. Tumor cells were only visible in the PBS, MMAE-BP, and MMAE-IgG1 groups. Tumor-bearing mice were treated with MMAE-based constructs (5 mg/kg mAb; 1.7 mg/kg MMAE per dose). Mice were dosed once a week for a total of 4 doses as illustrated via green arrows in panel a of Fig. 3. Scale bar = 300 μm. For each representative histology image, tissue sections from 4 mice were analyzed independently, with 3 slices examined per mouse, yielding similar results across all samples.
Supplementary information
Supplementary Information
Supplementary Discussion and Supplementary Figs. 1–45; Synthesis and Bioconjugation; Spectral Data; Supplementary References
Supplementary Data
Uncropped gels for Supplementary Figures
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Liu, B., Nguyen, H.VT., Jiang, Y. et al. Antibody–bottlebrush prodrug conjugates for targeted cancer therapy. Nat Biotechnol (2025). https://doi.org/10.1038/s41587-025-02772-z
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41587-025-02772-z