Abstract

BackgroundWe applied a non-linear immunokinetic model to quantitatively compare absolute antibody uptake and turnover in subcutaneous LNCaP human prostate cancer (PCa) xenografts of two radiolabeled forms of the humanized anti-prostate-specific membrane antigen (PSMA) monoclonal antibody J591 (124I-J591 and 89Zr-J591). Using the model, we examined the impact of dose on the tumor and plasma positron emission tomography (PET)-derived time-activity curves. We also sought to predict the optimal targeting index (ratio of integrated-tumor-to-integrated-plasma activity concentrations) for radioimmunotherapy.MethodsThe equilibrium rates of antibody internalization and turnover in the tumors were derived from PET images up to 96 h post-injection using compartmental modeling with a non-linear transfer rate. In addition, we serially imaged groups of LNCaP tumor-bearing mice injected with 89Zr-J591 antibody doses ranging from antigen subsaturating to saturating to examine the suitability of using a non-linear approach and derived the time-integrated concentration (in μM∙hours) of administered tracer in tumor as a function of the administered dose of antibody.ResultsThe comparison of 124I-J591 and 89Zr-J591 yielded similar model-derived values of the total antigen concentration and internalization rate. The association equilibrium constant (ka) was twofold higher for 124I, but there was a ~tenfold greater tumoral efflux rate of 124I from tumor compared to that of 89Zr. Plots of surface-bound and internalized radiotracers indicate similar behavior up to 24 h p.i. for both 124I-J591 and 89Zr-J591, with the effect of differential clearance rates becoming apparent after about 35 h p.i. Estimates of J591/PSMA complex turnover were 3.9–90.5 × 1012 (for doses from 60 to 240 μg) molecules per hour per gram of tumor (20 % of receptors internalized per hour).ConclusionsUsing quantitative compartmental model methods, surface binding and internalization rates were shown to be similar for both 124I-J591 and 89Zr-J591 forms, as expected. The large difference in clearance rates of the radioactivity from the tumor is likely due to differential trapping of residualizing zirconium versus non-residualizing iodine. Our non-linear model was found to be superior to a conventional linear model. This finding and the calculated activity persistence time in tumor have important implications for radioimmunotherapy and other antibody-based therapies in patients.

Highlights

  • We applied a non-linear immunokinetic model to quantitatively compare absolute antibody uptake and turnover in subcutaneous LNCaP human prostate cancer (PCa) xenografts of two radiolabeled forms of the humanized anti-prostate-specific membrane antigen (PSMA) monoclonal antibody J591 (124I-J591 and 89Zr-J591)

  • A variety of monoclonal antibodies specific for intracellular and extracellular epitopes of PSMA have been prepared [3,4,5,6], with two variants in particular demonstrating high affinity and specific and efficient targeting in vivo: the murine mAb 7E11, which binds an intracellular domain of PSMA, and the humanized mAb J591, which binds to an extracellular domain of PSMA. 7E11 has been investigated clinically as a SPECT imaging agent for recurrent and metastatic PCa [7] (111In-7E11, ProstaScintTM) as well as therapeutically as the Y-90 conjugate [8]

  • The thyroid is clearly visible in the 124I-J591 images, indicating that significant de-iodination occurred in vivo

Read more

Summary

Introduction

We applied a non-linear immunokinetic model to quantitatively compare absolute antibody uptake and turnover in subcutaneous LNCaP human prostate cancer (PCa) xenografts of two radiolabeled forms of the humanized anti-prostate-specific membrane antigen (PSMA) monoclonal antibody J591 (124I-J591 and 89Zr-J591). We examined the impact of dose on the tumor and plasma positron emission tomography (PET)-derived time-activity curves. While PSMA is expressed in normal human prostate cells and certain other normal tissues, it is highly upregulated in PCa, making it a promising diagnostic and therapeutic target [2]. Like 7E11, J591 has been clinically investigated in a variety of radiolabeled forms for both positron emission tomography (PET) imaging (e.g., in the form of 89Zr-J591) [9] as well as therapy (e.g., as 177Lu-J591) [10]. The surface binding and subsequent internalization are the fundamental rationale for using J591 as a carrier for residualizing radiometals and cytotoxic drug conjugates [11,12,13] that rely on such internalization to maximize their therapeutic effectiveness

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call