2020 Volume 10 Issue 5
Article Contents

Jong Hyuk Byun, Anna Park, Il Hyo Jung. RECEPTOR-MEDIATED ENDOCYTOSIS MODELING OF ANTIBODY-DRUG CONJUGATES TO THE RELEASED PAYLOAD WITHIN THE INTRACELLULAR SPACE CONSIDERING TARGET ANTIGEN EXPRESSION LEVELS[J]. Journal of Applied Analysis & Computation, 2020, 10(5): 1848-1868. doi: 10.11948/20190232
Citation: Jong Hyuk Byun, Anna Park, Il Hyo Jung. RECEPTOR-MEDIATED ENDOCYTOSIS MODELING OF ANTIBODY-DRUG CONJUGATES TO THE RELEASED PAYLOAD WITHIN THE INTRACELLULAR SPACE CONSIDERING TARGET ANTIGEN EXPRESSION LEVELS[J]. Journal of Applied Analysis & Computation, 2020, 10(5): 1848-1868. doi: 10.11948/20190232

RECEPTOR-MEDIATED ENDOCYTOSIS MODELING OF ANTIBODY-DRUG CONJUGATES TO THE RELEASED PAYLOAD WITHIN THE INTRACELLULAR SPACE CONSIDERING TARGET ANTIGEN EXPRESSION LEVELS

  • Corresponding author: Email address:ilhjung@pusan.ac.kr(I. H. Jung)
  • Fund Project: This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF- 2020R1C1C1A01004631 and NRF-2019R1A2C2007249)
  • An antibody-drug conjugate (ADC) is one of the effective treatment modalities designed as a targeted therapy for treating tumors. Certain ADCs such as brentuximab vedotin are known to kill negative tumor cells indirectly via membrane permeability and bystander-killing effect and to kill positive tumor cells directly. In this study, we propose a mathematical model to describe the ADC-receptor endocytosis mechanism and to predict payloads over a time profile more accurately, while considering target antigen-positive (Ag+)/negative (Ag–) cells. We discuss how the target-antigen expression levels derived using a ratio of Ag+ to Ag– cells determine the payload release in the intracellular space. The model is aimed at capturing the amount of the payloads based on the target expression levels with the total number of cells fixed. The results indicate that (i) the profile of the total payloads over a time within the intracellular space is less influenced by the target expression levels after a time period, but the slope at the growth phase in which the payload increases is determined by the target expression levels, (ii) the change in the area under the curve of the total intracellularly released payload with a change in the ratio of Ag+ to Ag– cells is more significant due to the initial ADC injection, (iii) the fluctuations in the released payloads within the Ag+ cells increase as the target expression levels decrease, unlike in the case of Ag– cells or extracellular space. In addition, the time $ t_{max} $ that corresponds to the maximum payload concentration $ C_{max} $ is shifted towards the right as the target-antigen levels decrease, and it is strengthened by an increase in the initial free ADCs. The proposed model may reduce the discrepancy between the experiment and the model in the prediction of payloads over time profile.
    MSC: 92B05, 74L15, 92C40
  • 加载中
  • [1] P. J. Aston, G. Derks, B. M. Agoram and P. H. van der Graaf, A mathematical analysis of rebound in a target-mediated drug disposition model: I. without feedback, Journal of Mathematical Biology, 2014, 68(6), 1453-1478. doi: 10.1007/s00285-013-0675-5

    CrossRef Google Scholar

    [2] A. Beck, L. Goetsch, C. Dumontet and N. Corvaïa, Strategies and challenges for the next generation of antibody-drug conjugates, Nature Reviews Drug Discovery, 2017, 16(5), 315-337.

    Google Scholar

    [3] A. Beck, T. Wurch, C. Bailly and N. Corvaia, Strategies and challenges for the next generation of therapeutic antibodies, Nature Reviews. Immunology, 2010, 10(5), 345. doi: 10.1038/nri2747

    CrossRef Google Scholar

    [4] E. R. Boghaert, K. Khandke, L. Sridharan et al., Tumoricidal effect of calicheamicin immuno-conjugates using a passive targeting strategy, International Journal of Oncology, 2006, 28(3), 675-684.

    Google Scholar

    [5] C. A. Boswell, D. B. Yadav, E. E. Mundo et al., Biodistribution and efficacy of an anti-tenb2 antibody-drug conjugate in a patient-derived model of prostate cancer, Oncotarget, 2019, 10(58), 6234. doi: 10.18632/oncotarget.27263

    CrossRef Google Scholar

    [6] J. H. Byun and I. H. Jung, Modeling to capture bystander-killing effect by released payload in target positive tumor cells, BMC Cancer, 2019, 19(1), 194. doi: 10.1186/s12885-019-5336-7

    CrossRef Google Scholar

    [7] R. V. Chari, Targeted cancer therapy: conferring specificity to cytotoxic drugs, Accounts of Chemical Research, 2007, 41(1), 98-107.

    Google Scholar

    [8] R. Deng, C. Zhou, D. Li et al., Preclinical and translational pharmacokinetics of a novel thiomabTM antibody-antibiotic conjugate against staphylococcus aureus, in mAbs, Taylor & Francis, 2019, 1-13.

    Google Scholar

    [9] H. P. Gerber, F. E. Koehn and R. T. Abraham, The antibody-drug conjugate: an enabling modality for natural product-based cancer therapeutics, Natural Product Reports, 2013, 30(5), 625-639. doi: 10.1039/c3np20113a

    CrossRef Google Scholar

    [10] L. Gibiansky and E. Gibiansky, Target-mediated drug disposition model and its approximations for antibody-drug conjugates, Journal of Pharmacokinetics and Pharmacodynamics, 2014, 41(1), 35-47.

    Google Scholar

    [11] C. P. Graff and K. D. Wittrup, Theoretical analysis of antibody targeting of tumor spheroids: importance of dosage for penetration, and affinity for retention, Cancer Research, 2003, 63(6), 1288-1296.

    Google Scholar

    [12] K. J. Hamblett, P. D. Senter, D. F. Chace et al., Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate, Clinical Cancer Research, 2004, 10(20), 7063-7070. doi: 10.1158/1078-0432.CCR-04-0789

    CrossRef Google Scholar

    [13] N. Jain, S. W. Smith, S. Ghone and B. Tomczuk, Current adc linker chemistry, Pharmaceutical Research, 2015, 32(11), 3526-3540. doi: 10.1007/s11095-015-1657-7

    CrossRef Google Scholar

    [14] M. C. Janin-Bussat, M. Dillenbourg, N. Corvaia et al., Characterization of antibody drug conjugate positional isomers at cysteine residues by peptide mapping lc-ms analysis, Journal of Chromatography B, 2015, 981, 9-13.

    Google Scholar

    [15] J. Jin, G. Park, J. B. Park et al., An anti-egfr $\times$ cotinine bispecific antibody complexed with cotinine-conjugated duocarmycin inhibits growth of egfr-positive cancer cells with kras mutations, Experimental & Molecular Medicine, 2018, 50(5), 67.

    $\times$ cotinine bispecific antibody complexed with cotinine-conjugated duocarmycin inhibits growth of egfr-positive cancer cells with kras mutations" target="_blank">Google Scholar

    [16] E. Khera, C. Cilliers, S. Bhatnagar and G. M. Thurber, Computational transport analysis of antibody-drug conjugate bystander effects and payload tumoral distribution: implications for therapy, Molecular Systems Design & Engineering, 2018, 3(1), 73-88.

    Google Scholar

    [17] Y. V. Kovtun, C. A. Audette, Y. Ye et al., Antibody-drug conjugates designed to eradicate tumors with homogeneous and heterogeneous expression of the target antigen, Cancer Research, 2006, 66(6), 3214-3221. doi: 10.1158/0008-5472.CAN-05-3973

    CrossRef Google Scholar

    [18] A. Krol, J. Maresca, M. W. Dewhirst and F. Yuan, Available volume fraction of macromolecules in the extravascular space of a fibrosarcoma: implications for drug delivery, Cancer Research, 1999, 59(16), 4136-4141.

    Google Scholar

    [19] F. Li, K. K. Emmerton, M. Jonas et al., Intracellular Released Payload Influences Potency and Bystander-Killing Effects of Antibody-Drug Conjugates in Preclinical Models, Cancer Research, 2016, 76(9), 2710-2719. doi: 10.1158/0008-5472.CAN-15-1795

    CrossRef Google Scholar

    [20] Q. Li, A. Barrett, B. Vijayakrishnan et al., Improved inhibition of tumor growth by diabody-drug conjugates via half-life extension, Bioconjugate Chemistry, 2019, 30(4), 1232-1243. doi: 10.1021/acs.bioconjchem.9b00170

    CrossRef Google Scholar

    [21] D. E. Mager and W. J. Jusko, General pharmacokinetic model for drugs exhibiting target-mediated drug disposition, Journal of Pharmacokinetics and Pharmacodynamics, 2001, 28(6), 507-532. doi: 10.1023/A:1014414520282

    CrossRef Google Scholar

    [22] D. E. Mager and W. Krzyzanski, Quasi-equilibrium pharmacokinetic model for drugs exhibiting target-mediated drug disposition, Pharmaceutical Research, 2005, 22(10), 1589-1596. doi: 10.1007/s11095-005-6650-0

    CrossRef Google Scholar

    [23] R. K. Mittapalli, S. Stodtmann, A. Friedel et al., An integrated population pharmacokinetic model versus individual models of depatuxizumab mafodotin, an anti-egfr antibody drug conjugate, in patients with solid tumors likely to overexpress egfr, The Journal of Clinical Pharmacology, 2019.

    Google Scholar

    [24] A. Mullard, Maturing antibody-drug conjugate pipeline hits 30, 2013.

    Google Scholar

    [25] N. M. Okeley, J. B. Miyamoto, X. Zhang et al., Intracellular activation of sgn-35, a potent anti-cd30 antibody-drug conjugate, Clinical Cancer Research, 2010, 16(3), 888-897. doi: 10.1158/1078-0432.CCR-09-2069

    CrossRef Google Scholar

    [26] H. L. Perez, P. M. Cardarelli, S. Deshpande et al., Antibody-drug conjugates: current status and future directions, Drug Discovery Today, 2014, 19(7), 869-881. doi: 10.1016/j.drudis.2013.11.004

    CrossRef Google Scholar

    [27] G. D. L. Phillips, G. Li, D. L. Dugger et al., Targeting her2-positive breast cancer with trastuzumab-dm1, an antibody-cytotoxic drug conjugate, Cancer Research, 2008, 68(22), 9280-9290. doi: 10.1158/0008-5472.CAN-08-1776

    CrossRef Google Scholar

    [28] L. Schwartz and I. de mathématique (Strasbourg), Théorie des distributions, 2, Hermann Paris, 1957.

    Google Scholar

    [29] A. P. Singh, L. Guo, A. Verma et al., A cell-level systems pk-pd model to characterize in vivo efficacy of adcs, Pharmaceutics, 2019, 11(2), 98. doi: 10.3390/pharmaceutics11020098

    CrossRef Google Scholar

    [30] A. P. Singh and D. K. Shah, A "dual" cell-level systems pk-pd model to characterize the bystander effect of adc, Journal of Pharmaceutical Sciences, 2019, 108(7), 2465-2475. doi: 10.1016/j.xphs.2019.01.034

    CrossRef Google Scholar

    [31] A. P. Singh, S. Sharma and D. K. Shah, Quantitative characterization of in vitro bystander effect of antibody-drug conjugates, Journal of Pharmacokinetics and Pharmacodynamics, 2016, 43(6), 567-582. doi: 10.1007/s10928-016-9495-8

    CrossRef Google Scholar

    [32] A. H. Staudacher and M. P. Brown, Antibody drug conjugates and bystander killing: is antigen-dependent internalisation required?, British Journal of Cancer, 2017, 117(12), 1736. doi: 10.1038/bjc.2017.367

    CrossRef Google Scholar

    [33] P. A. Trail, Antibody drug conjugates as cancer therapeutics, Antibodies, 2013, 2(1), 113-129.

    Google Scholar

    [34] C. Vasalou, G. Helmlinger and B. Gomes, A mechanistic tumor penetration model to guide antibody drug conjugate design, Plos One, 2015, 10(3), e0118977. doi: 10.1371/journal.pone.0118977

    CrossRef Google Scholar

    [35] J. Wang, W. Shen and J. L. Zaro, Antibody-Drug Conjugates: The 21st Century Magic Bullets for Cancer, 17, Springer, 2015.

    Google Scholar

    [36] Wikipedia contributors, Brentuximab vedotin - Wikipedia, the free encyclopedia, 2018.

    Google Scholar

    [37] H. Yao, L. Feng, S. R. Suthe et al., Therapeutic efficacy, pharmacokinetic profiles, and toxicological activities of humanized antibody-drug conjugate zt/g4-mmae targeting ron receptor tyrosine kinase for cancer therapy, Journal for Immunotherapy of Cancer, 2019, 7(1), 75. doi: 10.1186/s40425-019-0525-0

    CrossRef Google Scholar

Figures(9)  /  Tables(1)

Article Metrics

Article views(3828) PDF downloads(354) Cited by(0)

Access History

Other Articles By Authors

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint