Co-administered antibody improves penetration of antibody-dye conjugate into human cancers with implications for antibody-drug conjugates

Guolan Lu, Naoki Nishio, Nynke S van den Berg, Brock A Martin, Shayan Fakurnejad, Stan van Keulen, Alexander D Colevas, Greg M Thurber, Eben L Rosenthal, Guolan Lu, Naoki Nishio, Nynke S van den Berg, Brock A Martin, Shayan Fakurnejad, Stan van Keulen, Alexander D Colevas, Greg M Thurber, Eben L Rosenthal

Abstract

Poor tissue penetration remains a major challenge for antibody-based therapeutics of solid tumors, but proper dosing can improve the tissue penetration and thus therapeutic efficacy of these biologics. Due to dose-limiting toxicity of the small molecule payload, antibody-drug conjugates (ADCs) are administered at a much lower dose than their parent antibodies, which further reduces tissue penetration. We conducted an early-phase clinical trial (NCT02415881) and previously reported the safety of an antibody-dye conjugate (panitumumab-IRDye800CW) as primary outcome. Here, we report a retrospective exploratory analysis of the trial to evaluate whether co-administration of an unconjugated antibody could improve the intratumoral distribution of the antibody-dye conjugate in patients. By measuring the multiscale distribution of the antibody-dye conjugate, this study demonstrates improved microscopic antibody distribution without increasing uptake (toxicity) in healthy tissue when co-administered with the parent antibody, supporting further clinical investigation of the co-administration dosing strategy to improve the tumor penetration of ADCs.

Conflict of interest statement

E.L.R. acts as consultant for LI-COR Biosciences Inc. and has equipment loans from this company. All other authors declare no competing interests.

Figures

Fig. 1. Overview of the study design.
Fig. 1. Overview of the study design.
Study patients received infusion of antibody–dye conjugate with or without a loading dose of unlabeled antibody. One to five days post infusion, patients underwent surgical tumor resection. Fresh tissue samples obtained from the primary tumor were homogenized to quantify antibody–dye concentration in the tissue. Whole-tumor specimen were then formalin-fixed and cut into 5 mm thick tissue sections for macroscopic imaging. Subsequently, 5 µm histological slides were prepared from each tissue paraffin block made from the 5 mm tissue sections for microscopic imaging. Antibody uptake and distribution were measured from tissue homogenates, macroscopic, and microscopic imaging. Some elements of this figure were created with BioRender.com.
Fig. 2. Co-administration of unlabeled antibody reduced…
Fig. 2. Co-administration of unlabeled antibody reduced muscle uptake while maintained tumor uptake.
a Muscle antibody uptake (%ID kg−1: percent injected dose per kilogram) decreased with increasing total monoclonal antibody (mAb) dose (owing to loading dose (LD)). b, c. Tumor and skin uptake (%ID kg−1) as normalized by muscle uptake (%ID kg−1) were significantly higher in the loading dose group than that in the non-loading dose group, suggesting that tumor and skin were not fully saturated within the total antibody dose range of 0.3 mg kg−1 to 2.6 mg kg−1. d When tumor uptake was normalized by skin uptake, there was no significant differences between groups. The number of independent patient samples available in b: n = 9 (LD), n = 11 (non-LD); cn = 8 (LD), n = 9 (non-LD); dn = 8 (LD), n = 10 (non-LD). p value = 0.010 b, 0.046 c. Mann–Whitney U test (two-tailed) were used for comparison in bd. Graphs plotted mean with standard deviation. (*p < 0.05. **p < 0.01, ns: p > 0.05).
Fig. 3. Antibody–dye distribution demonstrated both macroscopic…
Fig. 3. Antibody–dye distribution demonstrated both macroscopic and microscopic heterogeneity within the solid tumor.
a Antibody–dye uptake (NIR: near-infrared) was higher in the tumor periphery or edge (especially the epithelium) than that in the tumor interior (Scale bar: 2 mm). b, c Antibody penetrated deeper into tumor nests located close to the edge of the bulk tumor than tumor nests located further away from the tumor edge, which was not attributable to EGFR (epidermal growth factor receptor) expression (Scale bar: 50 μm). d EGFR expression level is similar across the entire tumor (Scale bar: 2 mm). e The fluorescence intensity negatively correlated with increasing distance from the edge of the bulk tumor. f The fluorescence intensity plot depicted varying degree of antibody penetration into tumor nests: 1. fully saturated tumor nests at the periphery; 2. partially saturated tumor nests; 3. poorly penetrated tumor nests at the center of the tumor. (The shaded area in e and f means standard deviation). n = 3 tumor nests from the same patient. g Vessel staining (ERG) corresponding to region 1, 2, 3 in f (Scale bar: 50 μm).
Fig. 4. A loading dose of unlabeled…
Fig. 4. A loading dose of unlabeled antibody significantly reduced heterogeneity of antibody distribution in tumors, which can be captured by microscopic imaging but not macroscopic imaging.
a Schematic workflow of reconstructing the whole-tumor fluorescence distribution and measuring antibody delivery by macroscopic imaging. be The tumor uptake as measured by the MFI, and the tumor antibody distribution quantified by IQR,entropy, and uniformity from macroscopic imaging showed no significant difference between the two dosing groups. be Tissue specimens were available in n = 12 patients in the LD group and n = 12 in the non-LD group. f Schematic workflow of measuring antibody delivery in tissue histological sections and tumor microenvironmental factors using microscopic imaging. gj The tumor uptake measured by the MFI from microscopic imaging also showed no difference between the two groups, but the antibody distribution measured by IQR, entropy, and uniformity from microscopic imaging showed significantly higher heterogeneity in the non-LD group (*p < 0.05, ns: p > 0.05). p value = 0.030 h, 0.036 i, 0.025 j. kn Vessel area fraction, EGFR area fraction, αSMA area fraction, and tumor size were not statistically different between the LD and the non-LD group. (Scale bar in a and f: 2 cm; graphs plotted mean with standard deviation). gn Tissue specimens were available in n = 12 patients in LD group and n = 10 in the non-LD group. Mann–Whitney U test (two-tailed) was used in be, gn.
Fig. 5. Antibody–dye has more homogenous spatial…
Fig. 5. Antibody–dye has more homogenous spatial distribution across tumor when co-administered with the unlabeled antibody.
a Each tumor was gridded into small square regions and antibody penetration within each square region was quantified as the ratio of fluorescent positive area to EGFR-positive area. b The ratio of fluorescence to EGFR over the entire tumor area was not significantly different between the LD and non-LD group (Scale bar: 5 mm). ce Comparison of three metrics, including interquartile range (IQR), Pearson’s correlation coefficient between fluorescence and EGFR expression and CLCM correlation, all reflected less heterogeneity in antibody penetration across tumors in patients with loading dose (*p < 0.05; ns: p > 0.05). f, g Co-administration of parent antibody could overcome binding site barrier and improve antibody–dye distribution within the tumors. f As fluorescent antibody extravasates from blood vessels (red tube), it immediately binds EGFR in the tissue (represented by cells in the cube). g By co-administering non-fluorescent antibody, the same amount of fluorescent antibody reaches the tissue, but these antibodies compete for binding sites locally in the tissue. This results in improved distribution of fluorescent antibody at the microscopic scale (Scale bar: 250 μm). Mann–Whitney U test (two-tailed) was used in be. p value = 0.025 c, 0.025 d, 0.036 e. The number of independent patient samples available for be: n = 12 patients in the LD group and n = 10 in the non-LD group. Graphs plotted mean with standard deviation.
Fig. 6. The impact of loading dose…
Fig. 6. The impact of loading dose on total antibody uptake.
a Tumor uptake (%ID kg−1) showed slightly decreasing trend with increasing total antibody dose. b Tumor uptake (%ID kg−1) normalized by vessel area fraction showed a constant trend with increasing total antibody dose, suggested the variations in vessel area between the two groups may have caused the downward trend in tumor uptake before normalization. c–h Examples from a LD patient and non-LD patient demonstrated partial saturation in the tumor periphery with loading dose. (Scale bar in c and f: 2 mm; scale bar in d, e, g, h: 100 μm). Linear regression was performed in a, b with the best-fit line (solid line), the 95% confidence bands (dotted line), goodness of fit (R2), and p value.

References

    1. Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat. Rev. Cancer. 2006;6:583–592. doi: 10.1038/nrc1893.
    1. Lu G, et al. Predicting therapeutic antibody delivery into human head and neck cancers. Clin. Cancer Res. 2020;26:2582–2594. doi: 10.1158/1078-0432.CCR-19-3717.
    1. Martin JD, Cabral H, Stylianopoulos T, Jain RK. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat. Rev. Clin. Oncol. 2020;17:251–266. doi: 10.1038/s41571-019-0308-z.
    1. Khongorzul P, Ling CJ, Khan FU, Ihsan AU, Zhang J. Antibody–drug conjugates: a comprehensive review. Mol. Cancer Res. 2020;18:3–19. doi: 10.1158/1541-7786.MCR-19-0582.
    1. Bartelink IH, et al. Tumor drug penetration measurements could be the neglected piece of the personalized cancer treatment puzzle. Clin. Pharmacol. Ther. 2019;106:148–163. doi: 10.1002/cpt.1211.
    1. Rhoden JJ, Wittrup KD. Dose dependence of intratumoral perivascular distribution of monoclonal antibodies. J. Pharm. Sci. 2012;101:860–867. doi: 10.1002/jps.22801.
    1. Cilliers C, Menezes B, Nessler I, Linderman J, Thurber GM. Improved tumor penetration and single-cell targeting of antibody–drug conjugates increases anticancer efficacy and host survival. Cancer Res. 2018;78:758–768. doi: 10.1158/0008-5472.CAN-17-1638.
    1. Nessler, I. et al. Increased tumor penetration of single-domain antibody drug conjugates improves in vivo efficacy in prostate cancer models. Cancer Res.80, 1268–1278 (2020).
    1. Thurber GM, Schmidt MM, Wittrup KD. Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance. Adv. Drug Deliv. Rev. 2008;60:1421–1434. doi: 10.1016/j.addr.2008.04.012.
    1. Castaigne S, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet (Lond., Engl.) 2012;379:1508–1516. doi: 10.1016/S0140-6736(12)60485-1.
    1. Ribrag V, et al. A dose-escalation study of SAR3419, an anti-CD19 antibody maytansinoid conjugate, administered by intravenous infusion once weekly in patients with relapsed/refractory B-cell non-Hodgkin lymphoma. Clin. Cancer Res. 2014;20:213–220. doi: 10.1158/1078-0432.CCR-13-0580.
    1. Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates. mAbs. 2016;8:659–671. doi: 10.1080/19420862.2016.1156829.
    1. Cilliers C, Guo H, Liao J, Christodolu N, Thurber GM. Multiscale modeling of antibody-drug conjugates: connecting tissue and cellular distribution to whole animal pharmacokinetics and potential implications for efficacy. AAPS J. 2016;18:1117–1130. doi: 10.1208/s12248-016-9940-z.
    1. Singh AP, et al. Antibody coadministration as a strategy to overcome binding-site barrier for ADCs: a auantitative investigation. AAPS J. 2020;22:28. doi: 10.1208/s12248-019-0387-x.
    1. Gao RW, et al. Safety of panitumumab-IRDye800CW and cetuximab-IRDye800CW for fluorescence-guided surgical navigation in head and neck cancers. Theranostics. 2018;8:2488–2495. doi: 10.7150/thno.24487.
    1. Zinn KR, et al. IND-directed safety and biodistribution study of intravenously injected cetuximab-IRDye800 in cynomolgus macaques. Mol. Imaging Biol. 2015;17:49–57. doi: 10.1007/s11307-014-0773-9.
    1. Hamblett KJ, et al. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin. Cancer Res. 2004;10:7063–7070. doi: 10.1158/1078-0432.CCR-04-0789.
    1. Stylianopoulos T, Munn LL, Jain RK. Reengineering the physical microenvironment of tumors to improve drug delivery and efficacy: from mathematical modeling to bench to bedside. Trends Cancer. 2018;4:292–319. doi: 10.1016/j.trecan.2018.02.005.
    1. Fujimori K, Covell DG, Fletcher JE, Weinstein JN. A modeling analysis of monoclonal antibody percolation through tumors: a binding-site barrier. J. Nucl. Med. 1990;31:1191–1198.
    1. Thurber GM, Weissleder R. Quantitating antibody uptake in vivo: conditional dependence on antigen expression levels. Mol. Imaging Biol. 2011;13:623–632. doi: 10.1007/s11307-010-0397-7.
    1. Thurber, G. M. In Targeted radionuclide therapy (ed. Speer TW) 168–181 (Lippincott Williams and Wilkins, 2011).
    1. Lambert JM. Antibody–drug conjugates (ADCs): magic bullets at last! Mol. Pharmaceutics. 2015;12:1701–1702. doi: 10.1021/acs.molpharmaceut.5b00302.
    1. Hoffmann RM, et al. Antibody structure and engineering considerations for the design and function of antibody drug conjugates (ADCs) Oncoimmunology. 2017;7:e1395127–e1395127. doi: 10.1080/2162402X.2017.1395127.
    1. Nessler I, Khera E, Thurber GM. Quantitative pharmacology in antibody-drug conjugate development: armed antibodies or targeted small molecules? Oncoscience. 2018;5:161–163. doi: 10.18632/oncoscience.435.
    1. Wittrup KD. Antitumor antibodies can drive therapeutic T Cell responses. Trends Cancer. 2017;3:615–620. doi: 10.1016/j.trecan.2017.07.001.
    1. Rosenberg JE, et al. Study EV-103: preliminary durability results of enfortumab vedotin plus pembrolizumab for locally advanced or metastatic urothelial carcinoma. J. Clin. Oncol. 2020;38:441–441. doi: 10.1200/JCO.2020.38.6_suppl.441.
    1. Bardia A, et al. Sacituzumab govitecan-hziy in refractory metastatic triple-negative breast cancer. N. Engl. J. Med. 2019;380:741–751. doi: 10.1056/NEJMoa1814213.
    1. Modi S, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N. Engl. J. Med. 2020;382:610–621. doi: 10.1056/NEJMoa1914510.
    1. Moore K, et al. 992O - FORWARD I (GOG 3011): a phase III study of mirvetuximab soravtansine, a folate receptor alpha (FRa)-targeting antibody-drug conjugate (ADC), versus chemotherapy in patients (pts) with platinum-resistant ovarian cancer (PROC) Ann. Oncol. 2019;30:v403. doi: 10.1093/annonc/mdz250.
    1. Gao RW, et al. Determination of tumor margins with surgical specimen mapping using near-infrared fluorescence. Cancer Res. 2018;78:5144–5154. doi: 10.1158/0008-5472.CAN-18-0878.
    1. Nishio N, et al. Optimal dosing strategy for fluorescence gcuided surgery with panitumumab-IRDye800CW in head and neck cancer. Mol. Imaging Biol. 2020;22:156–164. doi: 10.1007/s11307-019-01358-x.
    1. Thurber GM, Dane Wittrup K. A mechanistic compartmental model for total antibody uptake in tumors. J. Theor. Biol. 2012;314:57–68. doi: 10.1016/j.jtbi.2012.08.034.
    1. Liu JF, et al. Phase I study of safety and pharmacokinetics of the anti-MUC16 antibody-drug conjugate DMUC5754A in patients with platinum-resistant ovarian cancer or unresectable pancreatic cancer. Ann. Oncol. 2016;27:2124–2130. doi: 10.1093/annonc/mdw401.
    1. Moore K, et al. Abstract CT036: targeting MUC16 with the THIOMAB TM-drug conjugate DMUC4064A in patients with platinum-resistant ovarian cancer: a phase I expansion study. Cancer Res. 2018;78:CT036–CT036.
    1. Baker JHE, et al. Direct visualization of heterogeneous extravascular distribution of trastuzumab in human epidermal growth factor receptor type 2 overexpressing xenografts. Clin. Cancer Res. 2008;14:2171–2179. doi: 10.1158/1078-0432.CCR-07-4465.
    1. Baker JHE, et al. Heterogeneous distribution of trastuzumab in HER2-positive xenografts and metastases: role of the tumor microenvironment. Clin. Exp. Metastasis. 2018;35:691–705. doi: 10.1007/s10585-018-9929-3.
    1. Gough A, et al. Biologically relevant heterogeneity: metrics and practical insights. SLAS Discov. 2017;22:213–237.
    1. Aerts HJWL, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat. Commun. 2014;5:4006. doi: 10.1038/ncomms5006.
    1. Gu, Y. An Introduction to the Theory of Statistics. (Charles Griffin and Company 1911).
    1. Shannon, C. E., Weaver, W. The Mathematical Theory of Communication. (University of Illinois Press, 1949).
    1. Gonzalez, R. C., Woods, R. E. & Eddins, S. L. Digital Image Processing Using MATLAB. (Prentice-Hall, 2013).
    1. Boswell, C. A. et al. Biodistribution and efficacy of an anti-TENB2 antibody-drug conjugate in a patient-derived model of prostate cancer. Oncotarget10, 6234–6244 (2019).
    1. Boswell CA, et al. Differential effects of predosing on tumor and tissue uptake of an 111In-labeled anti-TENB2 antibody-drug conjugate. J. Nucl. Med. 2012;53:1454–1461. doi: 10.2967/jnumed.112.103168.
    1. Lee, H. et al. 64Cu-MM-302 Positron emission tomography quantifies variability of enhanced permeability and retention of nanoparticles in relation to treatment response in patients with metastatic breast cancer. Clin. Cancer Res.23, 4190–4202 (2017).
    1. Lindenberg L, et al. Dosimetry and first human experience with (89)Zr-panitumumab. Am. J. Nucl. Med. Mol. Imaging. 2017;7:195–203.
    1. Lamberts LE, et al. Antibody positron emission tomography imaging in anticancer drug development. J. Clin. Oncol. Oncol. 2015;33:1491–1504. doi: 10.1200/JCO.2014.57.8278.
    1. Solon EG. Autoradiography techniques and quantification of drug distribution. Cell Tissue Res. 2015;360:87–107. doi: 10.1007/s00441-014-2093-4.
    1. Stumpf WE. Whole-body and microscopic autoradiography to determine tissue distribution of biopharmaceuticals - target discoveries with receptor micro-autoradiography engendered new concepts and therapies for vitamin D. Adv. Drug Deliv. Rev. 2013;65:1086–1097. doi: 10.1016/j.addr.2012.11.008.
    1. Scott AM, et al. A phase I trial of humanized monoclonal antibody A33 in patients with colorectal carcinoma: biodistribution, pharmacokinetics, and quantitative tumor uptake. Clin. Cancer Res. 2005;11:4810–4817. doi: 10.1158/1078-0432.CCR-04-2329.
    1. Ilovich O, et al. Dual-isotope cryoimaging quantitative autoradiography: investigating antibody-drug conjugate distribution and payload delivery through imaging. J. Nucl. Med. 2018;59:1461–1466. doi: 10.2967/jnumed.118.207753.
    1. Heskamp S, et al. Noninvasive Imaging of Tumor PD-L1 Expression Using Radiolabeled Anti-PD-L1 Antibodies. Cancer Res. 2015;75:2928–2936. doi: 10.1158/0008-5472.CAN-14-3477.
    1. Bensch, F. et al. 89 Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat. Med.24, 1852–1858 (2018).
    1. Koller M, et al. Implementation and benchmarking of a novel analytical framework to clinically evaluate tumor-specific fluorescent tracers. Nat. Commun. 2018;9:3739. doi: 10.1038/s41467-018-05727-y.
    1. van Keulen S, et al. Rapid, non-invasive fluorescence margin assessment: optical specimen mapping in oral squamous cell carcinoma. Oral. Oncol. 2019;88:58–65. doi: 10.1016/j.oraloncology.2018.11.012.
    1. van Keulen S, et al. The sentinel margin: intraoperative ex vivo specimen mapping using relative fluorescence intensity. Clin. Cancer Res. 2019;25:4656–4662. doi: 10.1158/1078-0432.CCR-19-0319.
    1. de Jongh, S. J. et al. Back-table fluorescence-guided imaging for circumferential resection margin evaluation in locally advanced rectal cancer patients using bevacizumab-800CW. J. Nucl. Med.61, 655–661 (2019).
    1. Junutula JR, et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 2008;26:925–932. doi: 10.1038/nbt.1480.
    1. Conner KP, et al. Evaluation of near infrared fluorescent labeling of monoclonal antibodies as a tool for tissue distribution. Drug Metab. Dispos. 2014;42:1906–1913. doi: 10.1124/dmd.114.060319.
    1. Cilliers C, Nessler I, Christodolu N, Thurber GM. Tracking antibody distribution with near-infrared fluorescent dyes: impact of dye structure and degree of labeling on plasma clearance. Mol. Pharmaceutics. 2017;14:1623–1633. doi: 10.1021/acs.molpharmaceut.6b01091.
    1. Haber MA, et al. ERG is a novel and reliable marker for endothelial cells in central nervous system tumors. Clin. Neuropathol. 2015;34:117–127. doi: 10.5414/NP300817.
    1. Otsu N. A threshold selection method from gray-level histograms. IEEE Trans. Syst., Man, Cybern. 1979;9:62–66. doi: 10.1109/TSMC.1979.4310076.
    1. Thurber GM, Wittrup KD. Quantitative spatiotemporal analysis of antibody fragment diffusion and endocytic consumption in tumor spheroids. Cancer Res. 2008;68:3334–3341. doi: 10.1158/0008-5472.CAN-07-3018.
    1. Haralick RM, Shanmugam K, Dinstein I. Textural features for image classification. IEEE Trans. Syst., Man, Cybern. 1973;SMC-3:610–621. doi: 10.1109/TSMC.1973.4309314.

Source: PubMed

3
Sottoscrivi