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

Characterization of the drug-to-antibody ratio distribution for antibody–drug conjugates in plasma/serum

    Keyang Xu

    * Author for correspondence

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.

    ,
    Luna Liu

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Randall Dere

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Elaine Mai

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Rebecca Erickson

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Angela Hendricks

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Kedan Lin

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    ,
    Jagath R Junutula

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    &
    Surinder Kaur

    Genentech, 1 DNA Way, South San Francisco, CA 94080, USA

    Published Online:https://doi.org/10.4155/bio.13.66

    Background: Antibody–drug conjugates (ADCs) are a new class of cancer therapeutics that deliver potent cytotoxins specifically to tumors to minimize systemic toxicity. However, undesirable release of covalently linked drugs in circulation can affect safety and efficacy. The objective of this manuscript was to propose and assess the assays that allow for the characterization of the drug deconjugation in plasma/serum. Results: ADCs of three main drug conjugation platforms, linked via lysine, site-specific engineered cysteine or reduced interchain disulfide cysteine residues, were analyzed using affinity capture for sample enrichment coupled with LC–MS or hydrophobic interaction chromatography–UV for detection. These novel approaches enabled measurement of the relative abundance of individual ADC species with different drug-to-antibody ratios, while maintaining their structural integrity. Conclusion: The characterization data generated by affinity capture LC–MS or hydrophobic interaction chromatography–UV provided critical mechanistic insights into understanding the stability and bioactivity of ADCs in vivo, and also helped the development of appropriate quantitative ELISAs.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Jaracz S, Chen J, Kuznetsova LV, Ojima I. Recent advances in tumor-targeting anticancer drug conjugates. Bioorg. Med. Chem.13(17),5043–5054 (2005).
    • Chari RVJ. Targeted cancer therapy: conferring specificity to cytotoxic drugs. Acc. Chem. Res.41(1),98–107 (2008).
    • Yarden Y, Sliwkowski MX. Untangling the ErbB signaling network. Nat. Rev. Mol. Cell Biol.2(2),127–137 (2001).
    • Carter PJ, Senter PD. Antibody–drug conjugates for cancer therapy. Cancer J.14(3),154–169 (2008).▪▪ Discussion on the advances and challenges of antibody–drug conjugates (ADCs) for cancer therapy.
    • Oflazoglu E, Stone IJ, Gordon K et al. Potent anticarcinoma activity of the humanized anti-CD70 antibody h1F6 conjugated to the tubulin inhibitor auristatin via an uncleavable linker. Clin. Cancer Res.14(19),6171–6180 (2008).
    • Erickson HK, Park PU, Widdison WC et al. Antibody–maytansinoid conjugates are activated in targeted cancer cells by lysosomal degradation and linker-dependent intracellular processing. Cancer Res.66(8),4426–4433 (2006).▪▪ Describes the conjugation platform via lysine side chain amines and its biological activity.
    • Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol. Rev.56(2),185–229 (2004).
    • Boger DL. Design, synthesis, and evaluation of DNA minor groove binding agents: the duocarmycins. Pure Appl. Chem.66(4),837–844 (1994).
    • Tassone P, Gozzini A, Goldmacher V et al.In vitro and in vivo activity of the maytansinoid immunoconjugate huN901-N20-deacetyl-N20-(3-mercapto-1-oxopropyl)-maytansine against CD56+ multiple myeloma cells. Cancer Res.64,4629–4636 (2004).
    • 10  Francisco JA, Cerveny CG, Meyer DL et al. cAC10–vcMMAE an anti-CD30–monomethyl auristatin E conjugate with potent and selective antitumor activity. Blood102(4),1458–1465 (2003).▪▪ Discusses the conjugation platform via cysteine sulfhydryl groups after reduction of the interchain disulfide bonds and its antitumor efficacy.
    • 11  Junutula JR, Raab H, Clark S et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol.26,925–932 (2008).▪▪ Novel conjugation platform via engineered cysteine residues at specific sites in the antibody and its improved bioactivity.
    • 12  Junutula JR, Flagella KM, Graham RA et al. Engineered thio-trastuzumab-DM1 conjugate with an improved therapeutic index to target human epidermal growth factor receptor 2-positive breast cancer. Clin. Cancer Res.16(19),4769–4778 (2010).
    • 13  Wu AM, Senter PD. Arming antibodies: prospects and challenges for immunoconjugates. Nat. Biotechnol.23,1137–1146 (2005).
    • 14  Lambert JM. Drug-conjugated monoclonal antibodies for the treatment of cancer. Curr. Opin. Pharmacol.5(5),543–549 (2005).
    • 15  Alley SC, Okeley NM, Senter PD. Antibody–drug conjugates: targeted drug delivery for cancer. Curr. Opin. Chem. Biol.14(4),529–537 (2010).
    • 16  Younes A, Bartlett NL, Leonard JP et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N. Engl. J. Med.363,1812–1821 (2010).
    • 17  Sanderson RJ, Hering MA, James SF et al.In vivo drug-linker stability of an anti-CD30 dipeptide-linked auristatin immunoconjugate. Clin. Cancer Res.11,843–852 (2005).
    • 18  Ducry L, Stump B. Antibody–drug conjugates: linking cytotoxic payloads to monoclonal antibodies. Bioconjug. Chem.21(1),5–13 (2010).
    • 19  Xie H, Audette C, Hoffee M, Lambert JM, Blättler WA. Pharmacokinetics and biodistribution of the antitumor immunoconjugate, cantuzumab mertansine (huC242-DM1), and its two components in mice. J. Pharmacol. Exp. Ther.308(3),1073–1082 (2004).
    • 20  Doronina SO, Bovee TD, Meyer DW et al. Novel peptide linkers for highly potent antibody–auristatin conjugate. Bioconjug. Chem.19(10),1960–1963 (2008).
    • 21  Doronina SO, Mendelsohn BA, Bovee TD et al. Enhanced activity of monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and toxicity. Bioconjug. Chem.17(1),114–124 (2006).
    • 22  Stephan JP, Chan P, Lee C et al. Anti-CD22-MCC–DM1 and MC–MMAF conjugates: impact of assay format on pharmacokinetic parameters determination. Bioconjug. Chem.19(8),1673–1683 (2008).
    • 23  Xu K, Liu L, Saad OM et al. Characterization of intact antibody–drug conjugates from plasma/serum in vivo by affinity capture capillary liquid chromatography–mass spectrometry. Anal. Biochem.412(1),56–66 (2011).▪ Novel approach to effectively measure changes in the drug-to-antibody ratio distribution of ADCs in biological fluids while preserving their structural integrity.
    • 24  Shen BQ, Xu K, Liu L et al. Conjugation site modulates the in vivo stability and therapeutic activity of antibody–drug conjugates. Nat. Biotechnol.30(2),184–189 (2012).
    • 25  Valliere-Douglass JF, McFee WA, Salas-Solano O. Native intact mass determination of antibodies conjugated with monomethyl Auristatin E and F at interchain cysteine residues. Anal. Chem.84(6),2843–2849 (2012).▪ Novel method to determine the intact mass of noncovalently associated ADCs via interchain cysteine residues.
    • 26  Kaur S, Xu K, Saad O, Dere R, Carrasco-Triguero M. A perspective on bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics. Bioanalysis5(2),201–226 (2013).▪ Provides detailed discussions on the complexity of ADCs and the strategies to address challenges in ADC bioanalysis with highlights of data from nonclinical and clinical case studies.
    • 27  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).
    • 28  Hamblett KJ, Senter PD, Chace DF et al. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin. Cancer Res.10(20),7063–7070 (2004).
    • 29  Alley SC, Benjamin DR, Jeffrey SC et al. Contribution of linker stability to the activities of anticancer immunoconjugates. Bioconjug. Chem.19(3),759–765 (2008).
    • 30  Chari RV, Martell BA, Gross JL et al. Immunoconjugates containing novel maytansinoids: promising anticancer drugs. Cancer Res.52(1),127–131 (1992).
    • 31  Lazar AC, Wang L, Blättler WA, Amphlett G, Lambert JM, Zhang W. Analysis of the composition of immunoconjugates using size-exclusion chromatography coupled to mass spectrometry. Rapid Commun. Mass Spectrom.19(13),1806–1814 (2005).
    • 32  Wang L, Amphlett G, Blättler WA, Lambert JM, Zhang W. Structural characterization of the maytansinoid–monoclonal antibody immunoconjugate, huN901–DM1, by mass spectrometry. Protein Sci.14(9),2436–2446 (2005).▪ Determination of the composition of lysine linked ADCs.
    • 33  Wakankar AA, Feeney MB, Rivera J et al. Physicochemical stability of the antibody–drug conjugate trastuzumab–DM1: changes due to modification and conjugation processes. Bioconjug. Chem.21(9),1588–1595 (2010).
    • 34  Sun MM, Beam KS, Cerveny CG et al. Reduction-alkylation strategies for the modification of specific monoclonal antibody disulfides. Bioconjug. Chem.16(5),1282–1290 (2005).