We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Pharmacokinetics and toxicity considerations for antibody–drug conjugates: an overview

    Siddhanth Hejmady‡

    Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Rajesh Pradhan‡

    Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Shobha Kumari

    Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India

    ,
    Meghna Pandey

    Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India

    ,
    Sunil K Dubey

    **Author for correspondence: Tel.: +91 823 970 3734;

    E-mail Address: sunilbit2014@gmail.com

    Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India

    Medical Research, R&D Healthcare Division, Emami Ltd, Kolkata 700056, India

    &
    Rajeev Taliyan

    *Author for correspondence: Tel.: +91 637 836 4745;

    E-mail Address: rajeev.taliyan@pilani.bits-pilani.ac.in

    Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India

    Published Online:https://doi.org/10.4155/bio-2023-0104

    Antibody–drug conjugates (ADCs) is one of the fastest-growing drug-delivery systems. It involves a monoclonal antibody conjugated with payload via a ligand that directly targets the expressive protein of diseased cell. Hence, it reduces systemic exposure and provides site-specific delivery along with reduced toxicity. Because of this advantage, researchers have gained interest in this novel system. ADCs have displayed great promise in drug delivery and biomedical applications. However, a lack of understanding exists on their mechanisms of biodistribution, metabolism and side effects. To gain a better understanding of the therapeutics, careful consideration of the pharmacokinetics and toxicity needs to be undertaken. In this review, different pharmacokinetics parameters including distribution, bioanalysis and heterogeneity are discussed for developing novel therapeutics.

    References

    • 1. Khongorzul P, Ling CJ, Khan FU et al. Antibody–drug conjugates: a comprehensive review. Mol. Cancer Res. 18(1), 3–19 (2020).
    • 2. Hedrich WD, Fandy TE, Ashour HM et al. Antibody–drug conjugates: pharmacokinetic/pharmacodynamic modeling, preclinical characterization, clinical studies, and lessons learned. Clin. Pharmacokinet. 57(6), 687–703 (2018).
    • 3. Nejadmoghaddam M-R, Minai-Tehrani A, Ghahremanzadeh R et al. Antibody–drug conjugates: possibilities and challenges. Avicenna J. Med. Biotechnol. 11(1), 3–23 (2019).
    • 4. Kamath AV, Iyer S. Preclinical pharmacokinetic considerations for the development of antibody drug conjugates. Pharm. Res. 32(11), 3470–3479 (2015).
    • 5. Lucas A, Price L, Schorzman A et al. Factors affecting the pharmacology of antibody–drug conjugates. Antibodies. 7(1), 10 (2018).
    • 6. Beaumont M, Tomazela D, Hodges D et al. Antibody–drug conjugates: integrated bioanalytical and biodisposition assessments in lead optimization and selection. AAPS Open. 4(1), 1–17 (2018).
    • 7. Kraynov E, Kamath AV, Walles M et al. Current approaches for absorption, distribution, metabolism, and excretion characterization of antibody–drug conjugates: an industry white paper. Drug Metab. Dispos. 44(5), 617–623 (2016).
    • 8. Matsuda Y, Seki T, Yamada K et al. Chemical site-specific conjugation platform to improve the pharmacokinetics and therapeutic index of antibody–drug conjugates. Mol. Pharm. 18(11), 4058–4066 (2021).
    • 9. Matsuda Y, Kliman M, Mendelsohn BA. Application of native ion exchange mass spectrometry to intact and subunit analysis of site-specific antibody–drug conjugates produced by AJICAP first generation technology. J. Am. Soc. Mass Spectrom. 31(8), 1706–1712 (2020).
    • 10. Junutula JR, Raab H, Clark S et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26(8), 925–932 (2008).
    • 11. Mahalingaiah PK, Ciurlionis R, Durbin KR et al. Potential mechanisms of target-independent uptake and toxicity of antibody-drug conjugates. Pharmacol. Ther. 200, 110–125 (2019).
    • 12. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody–drug conjugates. MAbs. 8(4), 659–671 (2016).
    • 13. Mou S, Huang Y, Rosenbaum A. ADME considerations and bioanalytical strategies for pharmacokinetic assessments of antibody–drug conjugates. Antibodies. 7(4), 41 (2018).
    • 14. Engvall E, Perlmann P. Enzyme-linked immunosorbent assay (ELISA) quantitative assay of immunoglobulin G. Immunochemistry. 8(9), 871–874 (1971).
    • 15. Kaur S, Xu K, Saad OM et al. Bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics. Bioanalysis 5(2), 201–226 (2013).
    • 16. Stephan JP, Kozak KR, Wong WLT. Challenges in developing bioanalytical assays for characterization of antibody–drug conjugates. Bioanalysis 3(6), 677–700 (2011).
    • 17. Li H, Ortiz R, Tran L et al. General LC–MS/MS method approach to quantify therapeutic monoclonal antibodies using a common whole antibody internal standard with application to preclinical studies. Anal. Chem. 84(3), 1267–1273 (2012).
    • 18. Liu A, Kozhich A, Passmore D et al. Quantitative bioanalysis of antibody-conjugated payload in monkey plasma using a hybrid immuno-capture LC–MS/MS approach: Assay development, validation, and a case study. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1002, 54–62 (2015).
    • 19. Shen B-Q, Bumbaca D, Saad O et al. Catabolic fate and pharmacokinetic characterization of trastuzumab emtansine (T-DM1): an emphasis on preclinical and clinical catabolism. Curr. Drug Metab. 13(7), 901–910 (2012).
    • 20. Tang Y, Tang F, Yang Y et al. Real-time analysis on drug-antibody ratio of antibody–drug conjugates for synthesis, process optimization, and quality control. Sci. Rep. 7(1), 2–11 (2017).
    • 21. Singh AP, Shah DK. Measurement and mathematical characterization of cell-level pharmacokinetics of antibody–drug conjugates: a case study with trastuzumab-vc-MMAE. Drug Metab. Dispos. 45(11), 1120–1132 (2017).
    • 22. Cahuzac H, Devel L. Analytical methods for the detection and quantification of adcs in biological matrices. Pharmaceuticals 13(12), 1–16 (2020).
    • 23. Taneja N, Gota V, Gurjar M, Singh KK. Development and validation of high-performance liquid chromatographic method for quantification of irinotecan and its active metabolite SN-38 in colon tumor bearing NOD/SCID mice plasma samples: application to pharmacokinetic study. Acta Chromatogr. 31(3), 166–172 (2019).
    • 24. Leal M, Wentland JA, Han X et al. Preclinical development of an anti-5T4 antibody–drug conjugate: pharmacokinetics in mice, rats, and NHP and tumor/tissue distribution in mice. Bioconjug. Chem. 26(11), 2223–2232 (2015).
    • 25. He J, Yu SF, Yee S et al. Characterization of in vivo biotransformations for trastuzumab emtansine by high-resolution accurate-mass mass spectrometry. MAbs. 10(7), 960–967 (2018).
    • 26. Lewis Phillips GD, Li G, Dugger DL et al. Targeting HER2-positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 68(22), 9280–9290 (2008).
    • 27. Dowell JA, Korth-Bradley J, Liu H et al. Pharmacokinetics of gemtuzumab ozogamicin, an antibody-targeted chemotherapy agent for the treatment of patients with acute myeloid leukemia in first relapse. J. Clin. Pharmacol. 41(11), 1206–1214 (2001).
    • 28. Scottish Medicines Consortium. Polatuzumab vedotin 140 mg powder for concentrate for solution for infusion (Polivy) (2020). www.scottishmedicines.org.uk/media/5360/polatuzumab-vedotin-polivy-final-august-2020-amended-180820-for-website.pdf
    • 29. Committee for Medicinal Products for Human Use. CHMP assessment report: Enhertu (2020). www.ema.europa.eu/contact
    • 30. EMA. Assessment report: Adcetris. CHMP Assess. Rep. 44, (July), 1–105 (2012).
    • 31. Malik P, Phipps C, Edginton A, Blay J. Pharmacokinetic considerations for antibody–drug conjugates against cancer. Pharm. Res. 34(12), 2579–2595 (2017).
    • 32. Cao M, De Mel N, Jiao Y et al. Site-specific antibody–drug conjugate heterogeneity characterization and heterogeneity root cause analysis. MAbs 11(6), 1064–1076 (2019).
    • 33. Behrens CR, Ha EH, Chinn LL et al. Antibody–drug conjugates (ADCs) derived from interchain cysteine cross-linking demonstrate improved homogeneity and other pharmacological properties over conventional heterogeneous ADCs. Mol. Pharm. 12(11), 3986–3998 (2015).
    • 34. Mager DE, Jusko WJ. General pharmacokinetic model for drugs exhibiting target-mediated drug disposition. J. Pharmacokinet. Pharmacodyn. 28(6), 507–532 (2001).
    • 35. Ritchie M, Tchistiakova L, Scott N. Implications of receptor-mediated endocytosis and intracellular trafficking dynamics in the development of antibody drug conjugates. MAbs 5(1), 13–21 (2013).
    • 36. Han TH, Zhao B. Absorption, distribution, metabolism, and excretion considerations for the development of antibody–drug conjugates. Drug Metab. Dispos. 42(11), 1914–1920 (2014).
    • 37. Grimm HP. Gaining insights into the consequences of target-mediated drug disposition of monoclonal antibodies using quasi-steady-state approximations. J. Pharmacokinet. Pharmacodyn. 36(5), 407–420 (2009).
    • 38. Acchione M, Kwon H, Jochheim CM, Atkins WM. Impact of linker and conjugation chemistry on antigen binding, Fc receptor binding and thermal stability of model antibody-drug conjugates. MAbs. 4(3), 362–372 (2012).
    • 39. Brachet G, Respaud R, Arnoult C et al. Increment in drug loading on an antibody–drug conjugate increases its binding to the human neonatal Fc receptor in vitro. Mol. Pharm. 13(4), 1405–1412 (2016).
    • 40. Hamblett KJ, Le T, Rock BM et al. Altering antibody–drug conjugate binding to the neonatal Fc receptor impacts efficacy and tolerability. Mol. Pharm. 13(7), 2387–2396 (2016).
    • 41. Berger C, Madshus IH, Stang E. Cetuximab in combination with anti-human IgG antibodies efficiently down-regulates the EGF receptor by macropinocytosis. Exp. Cell Res. 318(20), 2578–2591 (2012).
    • 42. Ingle GS, Chan P, Elliott JM et al. High CD21 expression inhibits internalization of anti-CD19 antibodies and cytotoxicity of an anti-CD19-drug conjugate. Br. J. Haematol. 140(1), 46–58 (2008).
    • 43. Williams S-P, Ogasawara A, Tinianow JN et al. ImmunoPET helps predicting the efficacy of antibody–drug conjugates targeting TENB2 and STEAP1.Oncotarget. 7(18), 25103–25112 (2016).
    • 44. Eaton JS, Miller PE, Mannis MJ, Murphy CJ. Ocular adverse events associated with antibody–drug conjugates in human clinical trials. J. Ocul. Pharmacol. Ther. 31(10), 589–604 (2015).
    • 45. Al-Tweigeri T, Nabholtz JM, Mackey JR. Ocular toxicity and cancer chemotherapy. Cancer 78(7), 1359–1373 (1996).
    • 46. Matulonis UA, Birrer MJ, O'Malley DM et al. Evaluation of prophylactic corticosteroid eye drop use in the management of corneal abnormalities induced by the antibody–drug conjugate mirvetuximab soravtansine. Clin. Cancer Res. 25(6), 1727–1736 (2019).
    • 47. Thon JN, Devine MT, Begonja AJ et al. High-content live-cell imaging assay used to establish mechanism of trastuzumab emtansine (T-DM1)-mediated inhibition of platelet production. Blood. 120(10), 1975–1984 (2012).
    • 48. Zhao H, Gulesserian S, Malinao MC et al. A potential mechanism for ADC-induced neutropenia: Role of neutrophils in their own demise. Mol. Cancer Ther. 16(9), 1866–1876 (2017).
    • 49. Uppal H, Doudement E, Mahapatra K et al. Potential mechanisms for thrombocytopenia development with trastuzumab emtansine (T-DM1). Clin. Cancer Res. 21(1), 123–133 (2015).
    • 50. de Goeij BECG, Lambert JM. New developments for antibody–drug conjugate-based therapeutic approaches. Curr. Opin. Immunol. 40, 14–23 (2016).
    • 51. Hock MB, Thudium KE, Carrasco-Triguero M, Schwabe NF. Immunogenicity of antibody drug conjugates: bioanalytical methods and monitoring strategy for a novel therapeutic modality. AAPS J. 17(1), 35–43 (2015).
    • 52. Pratt KP. Anti-drug antibodies: Emerging approaches to predict, reduce or reverse biotherapeutic immunogenicity. Antibodies. 7(2),1–18 (2018).
    • 53. Chalouni C, Doll S. Fate of antibody–drug conjugates in cancer cells. J. Exp. Clin. Cancer Res. 37(1), 1–12 (2018).
    • 54. Zhang S, Zhou D, Zheng C et al. Preclinical evaluation of a novel antibody–drug conjugate targeting DR5 for lymphoblastic leukemia therapy. Mol. Ther. Oncolytics 21(908), 329–339 (2021).
    • 55. Meyer DW, Bou LB, Shum S et al. An in vitro assay using cultured Kupffer cells can predict the impact of drug conjugation on in vivo antibody pharmacokinetics. Mol. Pharm. 17(3), 802–809 (2020).
    • 56. Sievers EL, Larson RA, Stadtmauer EA et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. J. Clin. Oncol. 19(13), 3244–3254 (2001).
    • 57. Palanca-Wessels MCA, Czuczman M, Salles G et al. Safety and activity of the anti-CD79B antibody–drug conjugate polatuzumab vedotin in relapsed or refractory B-cell non-Hodgkin lymphoma and chronic lymphocytic leukaemia: a phase 1 study. Lancet Oncol. 16(6), 704–715 (2015).
    • 58. Younes A, Bartlett NL, Leonard JP et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N. Engl. J. Med. 363(19), 1812–1821 (2010).
    • 59. European Society for Medical Oncology. FDA approves ado-trastuzumab emtansine for early breast cancer (2019). https://www.esmo.org/oncology-news/fda-approves-ado-trastuzumab-emtansine-for-early-breast-cancer
    • 60. Deonarain MP, Yahioglu G, Stamati I, Marklew J. Emerging formats for next-generation antibody drug conjugates. Expert Opin. Drug Discov. 10(5), 463–481 (2015).
    • 61. Mokhtari RB, Homayouni TS, Baluch N et al. Combination therapy in combating cancer. Oncotarget 8(23), 38022–38043 (2017).
    • 62. Boshuizen J, Koopman LA, Krijgsman O et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody–drug conjugate and BRAF/MEK inhibitors. Nat. Med. 24(2), 203–212 (2018).