Abstract
Antibody-drug conjugates (ADC) offer a targeted cancer treatment approach by delivering cytotoxic payloads directly to tumor cells. However, resistance mechanisms, poor tumor penetration, and off-target toxicity often limit clinical efficacy. Vartumab targets oncofetal chondroitin sulfate (ofCS), a pan-cancer target present on tumor cells and in the malignant stroma, with low expression in normal tissues. As part of transitioning Vartumab to clinical evaluation, two linker-payloads known to mediate bystander effects, valine-citrulline (vc)—monomethyl auristatin E (MMAE) and glycine-glycine-phenylalanine-glycine (ggfg)—Deruxtecan (DXd), were investigated for design of Vartumab ADCs. We show that the ADCs maintain specificity to ofCS proteoglycans, cancer cells, and tissue biopsies, exhibiting specific binding to a wide range of malignant and metastatic tissues. Biodistribution assessment of Vartumab ADCs in mice shows strong and specific tumor uptake, with minimal accumulation in other organs. Both ADCs induced bystander killing of antigen-negative cells in the presence of antigen-positive cells and displayed potent anti-tumor activities in a cell-derived xenograft melanoma model. Furthermore, we show that Vartumab conjugates with bystander-capable linker-payloads exhibit greater in vivo potency compared to those with payloads lacking significant bystander effect. Finally, toxicity assessment in rats indicates that the ADC-MMAE is well-tolerated upon repeated doses, with similar dose-limiting toxicities as reported for clinically approved MMAE-conjugated ADCs. Our data support further clinical development of Vartumab-based ADCs.
Similar content being viewed by others
Data availability
All data relating to the findings in the article are contained in the manuscript and supporting information.
References
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2024;74:229–63.
GarcÃa-Fernández C, Saz A, Fornaguera C, Borrós S. Cancer immunotherapies revisited: state of the art of conventional treatments and next-generation nanomedicines. Cancer Gene Ther. 2021;28:935–46.
Zhou W, Xu Z, Liu S, Lou X, Liu P, Xie H, et al. Landscape of clinical drug development of ADCs used for the pharmacotherapy of cancers: an overview of clinical trial registry data from 2002 to 2022. BMC Cancer. 2024;24:898.
Nguyen TD, Bordeau BM, Balthasar JP. Mechanisms of ADC toxicity and strategies to increase ADC tolerability. Cancers. Cancers. 2023;15:713
Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R. et al. Mechanisms of resistance to antibody-drug conjugates. Int J Mol Sci. 2023;24:1278
Fuso Nerini I, Morosi L, Zucchetti M, Ballerini A, Giavazzi R, D’Incalci M. Intratumor heterogeneity and its impact on drug distribution and sensitivity. Clin Pharmacol Ther. 2014;96:224–38.
Heist RS, Sands J, Bardia A, Shimizu T, Lisberg A, Krop I, et al. Clinical management, monitoring, and prophylaxis of adverse events of special interest associated with datopotamab deruxtecan. Cancer Treat Rev. 2024;125:102720.
Stepan LP, Trueblood ES, Hale K, Babcook J, Borges L, Sutherland CL. Expression of Trop2 cell surface glycoprotein in normal and tumor tissues: potential implications as a cancer therapeutic target. J Histochem Cytochem. 2011;59:701–10.
Bardia A, Mayer IA, Vahdat LT, Tolaney SM, Isakoff SJ, Diamond JR, et al. Sacituzumab Govitecan-hziy in Refractory Metastatic Triple-Negative Breast Cancer. N Engl J Med. 2019;380:741–51.
Baston-Bust DM, Gotte M, Janni W, Krussel JS, Hess AP. Syndecan-1 knock-down in decidualized human endometrial stromal cells leads to significant changes in cytokine and angiogenic factor expression patterns. Reprod Biol Endocrinol. 2010;8:133.
Matsumura Y. Cancer stromal targeting (CAST) therapy. Adv Drug Deliv Rev. 2012;64:710–9.
Modi S, Saura C, Yamashita T, Park YH, Kim S-B, Tamura K, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382:610–21.
Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–46.
Giugliano F, Corti C, Tarantino P, Michelini F, Curigliano G. Bystander effect of antibody-drug conjugates: fact or fiction? Curr Oncol Rep. 2022;24:809–17.
Wang Z, Li H, Gou L, Li W, Wang Y. Antibody-drug conjugates: Recent advances in payloads. Acta Pharmaceutica Sin B. 2023;13:4025–59.
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer. 2022;21:208.
Yasunaga M, Manabe S, Tarin D, Matsumura Y. Tailored immunoconjugate therapy depending on a quantity of tumor stroma. Cancer Sci. 2013;104:231–7.
Cortes J, Kim SB, Chung WP, Im SA, Park YH, Hegg R, et al. Trastuzumab deruxtecan versus trastuzumab emtansine for breast cancer. N Engl J Med. 2022;386:1143–54.
Tsao LC, Wang JS, Ma X, Sodhi S, Ragusa JV, Liu B, et al. Effective extracellular payload release and immunomodulatory interactions govern the therapeutic effect of trastuzumab deruxtecan (T-DXd). Nat Commun. 2025;16:3167.
Salanti A, Clausen TM, Agerbæk MO, Al Nakouzi N, Dahlbäck M, Oo HZ. et al. Targeting human cancer by a glycosaminoglycan-binding malaria protein. Cancer Cell. 2015;28:500–14.
Dancey J, Eisenhauer EA. Current perspectives on camptothecins in cancer treatment. Br J Cancer. 1996;74:327–38.
Diamantis N, Banerji U. Antibody-drug conjugated-an emerging class of cancer treatment. Br J Cancer. 2016;114:362–7.
Theocharis AD, Tsara ME, Papageorgacopoulou N, Karavias DD, Theocharis DA. Pancreatic carcinoma is characterized by elevated content of hyaluronan and chondroitin sulfate with altered disaccharide composition. Biochim Biophys Acta. 2000;1502:201–6.
Kalathas D, Theocharis DA, Bounias D, Kyriakopoulou D, Papageorgakopoulou N, Stavropoulos MS, et al. Chondroitin synthases I, II, III and chondroitin sulfate glucuronyltransferase expression in colorectal cancer. Mol Med Rep. 2011;4:363–8.
Xu G, Guimond MJ, Chakraborty C, Lala PK. Control of proliferation, migration, and invasiveness of human extravillous trophoblast by decorin, a decidual product. Biol Reprod. 2002;67:681–9.
Salanti A, Dahlb„ck M, Turner L, Nielsen MA, Barfod L, Magistrado P. Evidence for the involvement of VAR2CSA in pregnancy-associated malaria. J Exp Med. 2004;200:1197–203.
Agerbaek MO, Bang-Christensen SR, Yang MH, Clausen TM, Pereira MA, Sharma S, et al. The VAR2CSA malaria protein efficiently retrieves circulating tumor cells in an EpCAM-independent manner. Nat Commun. 2018;9:3279.
Spliid CB, Toledo AG, Sanderson P, Mao Y, Gatto F, Gustavsson T. et al. The specificity of the malarial VAR2CSA protein for chondroitin sulfate depends on 4-O-sulfation and ligand accessibility. J Biol Chem. 2021;297:101391
Vidal-Calvo EE, Martin-Salazar A, Choudhary S, Dagil R, Raghavan SSR, Duvnjak L, et al. Tumor-agnostic cancer therapy using antibodies targeting oncofetal chondroitin sulfate. Nat Commun. 2024;15:7553.
Bax DA, Little SE, Gaspar N, Perryman L, Marshall L, Viana-Pereira M, et al. Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development. PLoS ONE. 2009;4:e5209.
Yao B, Gao X, Dan M, Yuan C, Hu X, Sun Z, et al. Selection of payloads for antibody-drug conjugates targeting ubiquitously expressed tumor-associated antigens: a case study. AAPS J. 2022;24:70.
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. 2019;67:173–85.
Caculitan NG, Dela Cruz Chuh J, Ma Y, Zhang D, Kozak KR, Liu Y, et al. Cathepsin B is dispensable for cellular processing of cathepsin B-cleavable antibody-drug conjugates. Cancer Res. 2017;77:7027–37.
Li Z, Zhang J, Wu Y, Wang F, Cai T. Integrated strategy for biotransformation of antibody-drug conjugates and multidimensional interpretation via high-resolution mass spectrometry. Drug Metab Dispos. 2025;53:100081.
Balamkundu S, Liu CF. Lysosomal-cleavable peptide linkers in antibody-drug conjugates. Biomedicines. 2023;11:3080
Szijj PA, Bahou C, Chudasama V. Minireview: addressing the retro-Michael instability of maleimide bioconjugates. Drug Discov Today Technol. 2018;30:27–34.
Buecheler JW, Winzer M, Tonillo J, Weber C, Gieseler H. Impact of payload hydrophobicity on the stability of antibody-drug conjugates. Mol Pharm. 2018;15:2656–64.
Yerroum M, Braconnier F, Chariot P. Influence of handling procedures on rat plasma creatine kinase activity. Muscle Nerve. 1999;22:1119–21.
Nadkarni DV, Lee J, Jiang Q, Patel V, Sriskanda V, Dutta K, et al. Impact of drug conjugation and loading on target antigen binding and cytotoxicity in cysteine antibody-drug conjugates. Mol Pharm. 2021;18:889–97.
Li F, Emmerton KK, Jonas M, Zhang X, Miyamoto JB, Setter JR, et al. Intracellular released payload influences potency and bystander-killing effects of antibody-drug conjugates in preclinical models. Cancer Res. 2016;76:2710–9.
Okamoto H, Oitate M, Hagihara K, Shiozawa H, Furuta Y, Ogitani Y, et al. Pharmacokinetics of trastuzumab deruxtecan (T-DXd), a novel anti-HER2 antibody-drug conjugate, in HER2-positive tumour-bearing mice. Xenobiotica. 2020;50:1242–50.
Bocci M, Zana A, Principi L, Lucaroni L, Prati L, Gilardoni E, et al. In vivo activation of FAP-cleavable small molecule-drug conjugates for the targeted delivery of camptothecins and tubulin poisons to the tumor microenvironment. J Control Release. 2024;367:779–90.
Tsumura R, Anzai T, Koga Y, Takashima H, Matsumura Y, Yasunaga M. Anti-tissue factor antibody conjugated with monomethyl auristatin E or deruxtecan in pancreatic cancer models. Cancer Sci. 2024;115:3986–96.
Zhang P, Tao C, Shimura T, Huang AC, Kong N, Dai Y, et al. ICAM1 antibody drug conjugates exert potent antitumor activity in papillary and anaplastic thyroid carcinoma. iScience. 2023;26:107272.
Zhu B, Wang X, Shimura T, Huang AC, Kong N, Dai Y, et al. Development of potent antibody drug conjugates against ICAM1(+) cancer cells in preclinical models of cholangiocarcinoma. NPJ Precis Oncol. 2023;7:93.
Colombo R, Rich JR. The therapeutic window of antibody drug conjugates: a dogma in need of revision. Cancer Cell. 2022;40:1255–63.
Colombo R, Tarantino P, Rich JR, LoRusso PM, de Vries EGE. The journey of antibody-drug conjugates: lessons learned from 40 years of development. Cancer Discov. 2024;14:2089–108.
Acknowledgements
The authors would like to acknowledge Chumpol Hueyploo, Sabrina Damiri, Julie Zeberg Salwin, Akiko Shirashi, Benjamin Jacobsen, Muhammad Tauseef Mukhtar, Raminta Jonauskaite, Yasmin Ibrahim, and Mikkel Snog Vestergård for excellent technical support. At the University of Copenhagen, Faculty of Health and Medical Sciences, we would like to thank the Core Facility for Flow Cytometry and Single Cell Analysis for assistance in flow cytometry experiments, the Histolab for preparing tissues blocks and slides for us, and the Core Facility for Integrated BioImaging for helping with image acquisition… We would also like to thank the Department of Cellular and Molecular Medicine for access to their MALDI-TOF instrument.
Funding
EEVC is supported by the Novo Nordisk Foundation (NNF) BRIDGE Translational Excellence Program grant (NNF23SA0087869). JM is supported by the NNF Project grant (NNF23OC0086106). MD is supported by the Terry Fox Research Institute New Frontier Program Project Grant (1109). ASa is supported by the NNF Tandem grant (NNF21OC0068192); NNF Distinguished Innovator grant (NNF22OC0076055).
Author information
Authors and Affiliations
Contributions
ASk, EVC, SC, RD, TG and ASa conceived and designed the study; ASk, EVC and ASa wrote the original manuscript draft; ASk and EVC prepared the main figures and supplementary figure files; HZO prepared Fig. 5g; ASk, EVC, SC, JM, RD, AMS, HZO and LD, performed the experiments; MD, TG and ASa supervised; ASk, EVC, SC, JM, RD, AMS, HZO, LD, TGT, MD, TG and ASa contributed to discussing the data, reviewing the manuscript and approved the final manuscript.
Corresponding author
Ethics declarations
Competing interests
ASa, MD, and TGT are shareholders of VAR2 Pharmaceuticals. The antibodies are subject to a patenting process owned by VAR2 Pharmaceuticals. The remaining authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Edited by Professor Boris Zhivotovsky
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Skafte, A., Vidal-Calvo, E.E., Choudhary, S. et al. Preclinical profiling of antibody drug conjugates targeting oncofetal chondroitin sulfate. Cell Death Dis (2026). https://doi.org/10.1038/s41419-026-08420-x
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41419-026-08420-x


