Pretargeted radioimmunotherapy in tumored mice using an in vivo 212Pb/212Bi generator

https://doi.org/10.1016/j.nucmedbio.2005.06.009Get rights and content

Abstract

Objective

Pretargeting is the concept that combines optimal delivery of the antibody and rapid capture and elimination of the radioactivity. In this study, we evaluated the potential of antibody pretargeting to enable the tumor-targeting 212Pb for in vivo generation of 212Bi for α particle radiotherapy.

Methods

The 212Pb/212Bi chelate of DOTA-biotin, as well as their γ-emitting analogues, 203Pb and 205Bi, was prepared and characterized. The radiolabeled compounds were injected in animals for evaluation of tumor targeting and normal tissue uptake and retention. In the pretargeting protocol, injection of 400 μg of NR-LU-10 antibody–streptavidin conjugate was given at t=0 h, then 100 μg of N-acetyl-galatosamine-biotin clearing agent was injected at t=20–24 h; finally, 1 μg of 212Pb/212Bi-DOTA-biotin was injected 6 h later.

Results

Both 203Pb and 205Bi-DOTA-biotin were stable for at least 4 days in the different challenging solutions including PBS, 10 mM DTPA and serum. Contrary to its γ-emitting analogues, radiolabeled 212Pb-DOTA-biotin was not stable. There was greater than 30% of free 212Bi released 4 h after 212Pb-labeled DOTA-biotin. The results of pretargeting protocol of 203Pb and 205Bi-DOTA-biotin showed that the tumor target reached 20% injected dose (ID)/g at 4 h postinjection and remained high for 5 days. The %ID/g in the whole blood and other nontarget organs was low after administration of labeled 203Pb and 205Bi-DOTA-biotin similar to the biodistribution of labeled DOTA-biotin alone. In the animals administered 212Pb-DOTA-biotin, radioactivity in nontarget organs was low except the kidneys. The %ID/g in the kidney for 212Bi was 14.5 at 2 h, higher than 212Pb, but dropped to about 6% ID/g by 4 h. However, tumor uptake for 212Pb and 212Bi was >25% ID/g at 1 h postinjection and remained so through 24 h.

Conclusions

Antibody pretargeting system with Mab-streptavidin, clearing agent and DOTA-biotin provides the potential of 212Bi for solid tumor radiotherapy despite the release of 212Bi after 212Pb decay. Dosimetry calculations resulted in tumor dose at 93 rad/μCi and ratios of tumor to marrow and kidney at 386:1 and 12:1, respectively.

Introduction

The potential use of antibodies that recognize and selectively bind with high affinity to antigens present on tumor cells has been the basis of a great deal of work to deliver radiation to tumors for radioimmunotherapy [1]. A major limitation of this approach has been a mismatch of the properties of radioactivity with the tumor-targeting processes of the large antibody proteins. Antibody proteins are slowly taken up by tumors as well as are slowly disappearing from the circulation. These properties result in irradiation of radiosensitive marrow cells during the extended circulation time as well as decay losses of the radionuclide during the time required for tumor localization.

Antibody pretargeting [2], [3] is a process that addresses the limitations of antibodies in delivery of radiation to tumors. The process [4], which we have investigated, involves an initial step of tumor targeting of an antibody with a high-affinity acceptor such as streptavidin attached. This allows the slow targeting step to be done with a material that poses no radiotoxic effect. The intact immunoglobulin also provides for the maximum fraction of the ID to be localized at the tumor. After time for tumor uptake, about 20 h in mice or 48 h in patients, a clearing agent, synthetic dendrimeric galactose-biotin, is given to remove the remaining circulating antibody–streptavidin. In the final step, the radioactivity is given as a small molecule, radiobiotin, that binds with very high affinity to the pretargeted acceptor, streptavidin (1015/M affinity). The tumor uptake occurs within minutes, while nontargeted radioactivity is rapidly removed by kidneys via the filtration route. The pretargeting system has been extensively studied in animals and in Phase I/II studies [5] in patients using the β-emitting radiotherapy nuclide 90Y.

α Particles are 4He ions, approximately 8000 times larger than β-particle electrons, and when emitted, result in high linear energy transfer (LET) of around 100 keV/μm compared to β particles such as 90Y, which have a LET of 0.2 keV/μm [6], [7]. These physical characteristics result in radiation properties that have several advantages compared to β particles for targeted radiotherapy in appropriate clinical situations. The high LET results in a small number of targeted α's required to kill a cell, on the order of 10 to 30 with fewer needed if internalized in cells [8]. Effective cell killing is independent of oxygenation levels, dose rate as achievable with intravenous administration (at or above 5 rad/h) or repair capability. Low penetration, on the order of several cell diameters, provides efficient killing of isolated cells, cell clusters or micrometastases, thus, complementing β emitters that require crossfire field effects and are advantageously used in larger tumors. Finally, there is the potential for lower toxicity in circulation due to the short range. Thus, α particle emitters may be advantageous in clinical applications such as micrometastases that limit the ultimate ability to control or cure cancer [9].

Three α emitters have had extensive study and are candidates for development: 211At with a 7.2-h half-life, 212Bi with a 1.06-h half-life and 213Bi with a 46-min half-life. We have selected the 212Pb/212Bi system [10] because it provides an in vivo generator supply of the 212Bi from the 10.6-h half-life 212Pb. The longer lived 212Pb allows time for tumor targeting of the α emitter 212Bi. Key to its use, however, is the ability to form a stable chelate of 212Pb that controls the in vivo localization through the decay of the 212Bi. Works by Ruegg et al. [11], Junghans et al. [12] and Gansow et al. [13] have demonstrated that the cyclic polyamino acid chelating agent DOTA forms stable chelates with both Pb and Bi.

In theory, the decay of 212Pb should not present a problem in retention of the 212Bi. Conservation principles dictate that the recoil energy of the Bi nucleus is only about 0.5 eV. This is not sufficient to break a chemical bond, which requires approximately 10 eV. However, the γ-ray emitted when 212Pb decays is internally converted over 30% of the time. The resulting cascade of conversion electrons makes the metal–chelate complex unstable. When the 212Pb decays to 212Bi, about two thirds remains as a DOTA chelate, and the other third goes through the decay pathway that results in conversion electrons and “dechelation” of the 212Bi during the process [14]. Loss of 212Bi and its release as free bismuth results in kidney localization and was a major concern for the use of this system.

In this study, we reported the in vitro stability of 212Pb/212Bi-DOTA-biotin and their γ-emitting analogues, 203Pb and 205Bi-DOTA-biotin; biodistribution of 203Pb-DOTA-biotin and 205Bi-DOTA-biotin in animals following the pretargeting protocol; and comparison with that of 212Pb and 212Bi isotopes. We report the dosimetry estimates of targeting 212Pb in mouse xenograft model. The kidney dose by following 212Bi activity after dechelation from 212Pb-DOTA-biotin was also reported.

Section snippets

Radionuclides

203Pb and 205/206Bi (for simplicity, the bismuth radioisotope mixture will hereafter be referred to only as 205Bi) were obtained as chloride forms from Nordion International, Vancouver, BC, and Crocker National laboratory, Davis, CA, respectively. Both radionuclides were received with certificates of radionuclidic purity from the manufacturers and were used directly without further purification. 212Pb/212Bi was obtained by eluting a 224Ra/212Pb generator (Argonne National Laboratory, Argonne,

In vitro stability

The results of in vitro stability of 203Pb and 205Bi-DOTA-biotin by avidin beads assay are compared with 111In-DOTA-biotin and are shown in Table 1. Both Pb-DOTA-biotin and Bi-DOTA-biotin radiochelates were stable for at least 4 days in the different challenging solutions including PBS buffer, 10 mM DTPA and serum. In the case of 212Pb/212Bi, we observed greater than 30% free 212Bi 4 h after we labeled 212Pb-DOTA-biotin by both HPLC and the avidin beads biotin binding assay as shown in Table 2.

Urinary metabolite analysis

Discussion

The quantitative labeling of macrocyclic DOTA-biotin ligand with Pb and Bi isotopes was achieved at elevated temperature with the conditions similar to those used for 111In and 90Y [16]. The in vitro stability study showed that there was very little activity released from the DOTA chelate over 4 days in both 203Pb and 205Bi, which is also very similar to the stability of 111In-DOTA-biotin. Different stability was seen using 212Pb; the 212Pb-DOTA-biotin was stable initially, but there was >30%

Acknowledgment

The funding for this work was provided by the National Cancer Institute SBIR (grant 1R43CA71221-1A1). This work was also performed under the auspices of the U.S. Dept. of Energy, Office of Science, Office of Biological and Environmental Research under contract W-7405-ENG36.

References (16)

  • J. Schlom

    Monoclonal antibodies in cancer therapy

  • S.F. Roseborough

    Two-step immunological approaches for imaging and therapy

    Q J Nucl Med

    (1996)
  • D.A. Goodwin

    Tumor pretargeting: almost the bottom line

    J Nucl Med

    (1995)
  • P. Abrams et al.

    Phase I clinical evaluation of pretargeted 90Y-radioimmunotherapy

    Proc Am Soc Clin Oncol

    (1995)
  • H.B. Breitz et al.

    Clinical optimization of pretargeted radioimmunotherapy with antibody-streptavidin conjugate and 90Y-DOTA-biotin

    J Nucl Med

    (2000)
  • G. Vaidynathan et al.

    Targeted therapy using alpha emitters

    Phys Med Biol

    (1996)
  • M.R. Zalutsky et al.

    Radioimmunotherapy with α-particle emitting radioimmunoconjugates

    Acta Oncol

    (1996)
  • J.L. Humm et al.

    A model of cell inactivation by α-particle internal emitters

    Radiat Res Phys

    (1993)
There are more references available in the full text version of this article.

Cited by (47)

  • Innovation, Impact, and Strategic Importance of Alpha-Emitting Radionuclides

    2019, Journal of Medical Imaging and Radiation Sciences
    Citation Excerpt :

    Studies show that simple polydentate metal binding does not fully prevent metal complex dissociation after β- decay of the central metal ion. In 2005, these findings were verified by Fu-Min Su et al, who reported that 212Pb–DOTA-biotin was initially stable, but ∼30% of 212Bi activity was released from the DOTA-biotin in 4 h [12]. One of the earliest preclinical studies using this system was reported in 1986 by Kozak et al at NIH [13].

  • <sup>212</sup>Pb-labeled B7-H3-targeting antibody for pancreatic cancer therapy in mouse models

    2018, Nuclear Medicine and Biology
    Citation Excerpt :

    The uptake of 212Pb and 212Bi in the spleens of mice in the current biodistribution studies with 212Pb-376.96 was comparable to that observed in our previous study for this IgG2a isotype RIC [20]. The observed differences in normal tissue retention between 212Pb and its daughter 212Bi are consistent with previous studies that systemically administered 212Pb-labeled compounds [41,42]; ~30% of 212Bi has been shown to be released from the original 212Pb-chelate complex due to internal conversion during decay of 212Pb [43], which would allow distribution of free 212Bi and its daughter radionuclides during circulation of the 212Pb-RICs. Several strategies could be employed to minimize the amount of systemically released 212Bi emitted from 212Pb-conjugates, such as employing small targeting carriers that have rapid pharmacokinetics when i.v. injections are required [41,42], targeting intracavitary (e.g., i.p.) lesions with local administrations when using mAbs as targeting vectors [10,35,36,44], or other approaches to control recoiling daughter radionuclides that emit α-particles [45].

  • Imaging, biodistribution, and toxicology evaluation of <sup>212</sup>Pb-TCMC-trastuzumab in nonhuman primates

    2016, Nuclear Medicine and Biology
    Citation Excerpt :

    Loss of 212Bi from the immunoconjugate is a potential limitation for therapeutic applications, as non-specific distribution of 212Bi could lead to undesirable toxicity. While a growing body of literature has evaluated the safety and efficacy of targeted radiotherapy with 212Pb as an in vivo generator of 212Bi, few reports have quantitatively assessed the relative activity of these radionuclides in vivo [21,22]. Gamma-ray spectroscopy with an HPGe detector provides a method to identify and quantify the activities of individual isotopes when multiple isotopes are present due to the energy resolution (1 keV) of the detector.

View all citing articles on Scopus
1

Current address: Bristol-Myers-Squibb Medical Imaging, North Billerica, MA 01862, USA.

2

Current address: Shin Nippon Biomedical Laboratories, Everett, WA, USA.

3

Current address: 2722 Triton Drive NW, Seattle, WA 98117, USA.

4

Current address: 417 EJ Young Road, Olga, WA 98279, USA.

View full text