Treatment of HER2-positive breast carcinomatous meningitis with intrathecal administration of α-particle-emitting 211At-labeled trastuzumab

https://doi.org/10.1016/j.nucmedbio.2009.04.003Get rights and content

Abstract

Introduction

Carcinomatous meningitis (CM) is a devastating disease characterized by the dissemination of malignant tumor cells into the subarachnoid space along the brain and spine. Systemic treatment with monoclonal antibody (mAb) trastuzumab can be effective against HER2-positive systemic breast carcinoma but, like other therapies, is ineffective against CM. The goal of this study was to evaluate the therapeutic effect of α-particle emitting 211At-labeled trastuzumab following intrathecal administration in a rat model of breast carcinoma CM.

Methods

Athymic rats were injected intrathecally with MCF-7/HER2-18 breast carcinoma cells through a surgically implanted indwelling intrathecal catheter. In Experiment 1, animals received 33 or 66 μCi 211At-labeled trastuzumab, cold trastuzumab or saline. In Experiment 2, animals were inoculated with a lower tumor burden and received 46 or 92 μCi 211At-labeled trastuzumab or saline. In Experiment 3, animals received 28 μCi 211At-labeled trastuzumab, 30 μCi 211At-labeled TPS3.2 control mAb or saline. Histopathological analysis of the neuroaxis was performed at the end of the study.

Results

In Experiment 1, median survival increased from 21 days for the saline and cold trastuzumab groups to 45 and 48 days for 33 and 66 μCi 211At-labeled trastuzumab, respectively. In Experiment 2, median survival increased from 23 days for saline controls to 68 and 92 days for 46 and 92 μCi 211At-labeled trastuzumab, respectively. In Experiment 3, median survival increased from 20 days to 29 and 36 days for animals treated with 211At-labeled TPS3.2 and 211At-labeled trastuzumab, respectively. Long-term survivors were observed exclusively in the 211At-trastuzumab-treated groups.

Conclusion

Intrathecal 211At-labeled trastuzumab shows promise as a treatment for patients with HER2-positive breast CM.

Introduction

Carcinomatous meningitis (CM) is a devastating disease characterized by the dissemination of malignant tumor cells within the leptomeningeal space and metastatic spread throughout the cerebrospinal fluid (CSF) compartment along the brain and spine. The disease occurs in approximately 4–15% of patients with solid tumors and is most frequently diagnosed in patients with breast carcinoma (27–50%), lung carcinoma (22–36%), lymphoma or leukemia (5–15%) as well as tumors of the central nervous system (CNS), mostly notably, medulloblastoma (30%) [1], [2]. Developing better treatments for CM accompanying breast cancer is particularly important not only because of its relative frequency but also due to the fact that its incidence appears to be increasing in part because systemic disease has become more effectively managed [3]. The prognosis for patients with CM is grim with a median survival of only 2–6 months. This is due in large part to the fact that external beam radiotherapy and chemotherapy are generally self-limited in this setting by their cytotoxicity to normal tissues, particularly those of the CNS. An attractive approach to increasing the specificity of CM treatment is targeted radiotherapy, in which a vector such as a monoclonal antibody (mAb) is utilized to selectively deliver a radionuclide to cancer cells.

With regard to the choice of vehicle for this purpose, the epidermal growth factor receptor tyrosine kinase known as both ErbB2 and HER2/neu is a potentially valuable target for cancer therapeutics. The p185 transmembrane protein HER2 oncogene product is overexpressed in about 25% of breast carcinomas and other malignancies but only at low levels on normal tissues [4], [5], [6]. Trastuzumab (Herceptin, Genentech, South San Francisco, CA, USA) is a humanized mAb that specifically binds to a cysteine-rich motif within the extracellular domain of this p185 protein [7]. Systemically administered Trastuzumab is broadly utilized, primarily in combination with chemotherapy, for the treatment of patients with HER2-positive breast carcinoma with responses observed in about half of the time [8], [9].

For unknown reasons, HER2-positive breast carcinoma patients have a relatively high incidence of CM [3]. The impermeability of the blood-brain barrier (BBB) hinders the delivery of systemically administered macromolecules to lesions located within the CNS, such as brain metastasis or CM, in a therapeutically significant manner: following intravenous administration, the CSF concentration of trastuzumab remains 300-fold lower than its systemic concentration [10]. Compartmental administration (intratumoral, intrathecal) bypasses the BBB, thereby allowing for significantly higher doses available for binding to HER2-positive tumor cells. This has lead to a number of case reports investigating the therapeutic effectiveness of high-dose intrathecal trastuzumab, with two of five patients treated surviving for more than 6 months [11], [12], [13], [14], [15].

We hypothesize that the efficacy of intrathecal trastuzumab could be enhanced by combining the mAb with a radionuclide possessing emission characteristics that are well matched to the geometrical features of CM. Because leptomeningeal spread of malignancies present as free-floating cancer cells in the CSF and sheet-like deposits on compartmental walls, radionuclides emitting short range radiation are recommended to minimize radiation dose to the spinal cord [16]. Alpha particles such as those emitted by 211At have a range in tissue of only a few cell diameters and, thus, might be ideally suited to this purpose. In addition, the 7.2-h half-life of this radiohalogen reduces the risk of systemic toxicity after CSF protein resorption into the general circulation. Finally, as a consequence of the high linear energy transfer nature of α-particles, the cytotoxicity of 211At-labeled compounds is considerably higher than those labeled with β-emitters such as 131I and 90Y in routine use for clinical radioimmunotherapy [17].

In the present study, we describe a rat model for HER2-positive breast carcinoma CM. This model was utilized to evaluate the therapeutic potential of 211At-labeled trastuzumab and our results indicate that significant survival prolongation could be obtained after intrathecal administration of this targeted radiotherapeutic.

Section snippets

Antibodies

Trastuzumab was obtained from the Duke University Medical Center hospital pharmacy and was dialyzed overnight into 100-mM pH 8.5 borate buffer prior to labeling. The chimeric human/murine TPS3.2 mAb, produced as described previously [18], was treated in similar fashion and served as a control. Fluorescence-activated cell sorting analysis of this anti-dansyl IgG2 mAb confirmed its lack of reactivity to the HER2-expressing cell line used in these studies (data not shown).

Labeling mAbs with 211At

Astatine-211 was produced

Breast carcinomatous meningitis model

The effect of altering the number and volume of inoculated cells was evaluated in preliminary experiments where it was found that CM could be established reproducibly with volumes of 80–100 μl and 0.5–1.25×107 MCF-7/HER2-18 cells. As observed previously, in order to use this cell line to establish intracerebral xenografts in this rat strain [22], consistent tumor growth required use of a continuous-release 17β-estradiol pellet. At these cell doses, animals developed progressive neurological

Discussion

Better treatments for malignancies that metastasize to the subarachnoid space are critically needed because current therapies are largely ineffective [1], [2]. Although radiation has been utilized in patients with CM, external beam therapy can rarely achieve a meaningful therapeutic effect because of dose-limiting toxicity to the normal neuraxis [3]. Targeted radiotherapy is attractive in this setting because of the potential for achieving high levels of tumor cell irradiation while sparing

Acknowledgments

This work was supported by grants CA42324 and NS20023 from the National Institutes of Health and Grant DOE-FG02-08ER64697 from the Department of Energy. Dr. Abraham Boskovitz was supported by the SICPA Foundation (Prilly, Switzerland), the Swiss Cancer League and the 450th Anniversary of the University of Lausanne Foundation (Lausanne, Switzerland). We thank Dr. Xiao-Guang Zhao for his assistance in preparing the 211At-labeled mAbs.

References (37)

  • PradosM.D.

    Primary neoplasms of the central nervous system in adults

  • ChamberlainM.C.

    Neoplastic meningitis

    J Clin Oncol

    (2005)
  • PressM.F. et al.

    Expression of the HER-2/neu proto-oncogene in normal human adult and fetal tissues

    Oncogene

    (1990)
  • BerchuckA. et al.

    Overexpression of HER-2/neu is associated with poor survival in advanced epithelial ovarian cancer

    Cancer Res

    (1990)
  • NataliP.G. et al.

    Expression of the p185 encoded by HER2 oncogene in normal and transformed human tissues

    Int J Cancer

    (1990)
  • SliwkowskiM.X. et al.

    Nonclinical studies addressing the mechanism of action of trastuzumab (Herceptin)

    Semin Oncol

    (1999)
  • BaselgaJ.

    Clinical trials of Herceptin (trastuzumab)

    Eur J Cancer

    (2001)
  • PestalozziB.C. et al.

    Trastuzumab in CSF

    J Clin Oncol

    (2000)
  • Cited by (37)

    • Recent progress of astatine-211 in endoradiotherapy: Great advances from fundamental properties to targeted radiopharmaceuticals

      2022, Chinese Chemical Letters
      Citation Excerpt :

      Akabani et al. [204] pointed out that the relative biological effect of 211At-trastuzumab was twice higher than that of external irradiation therapy, and it was able to inhibit the proliferation of HER2-positive tumor cells. Furthermore, the therapeutic effect of 211At-trastuzumab on carcinomatous meningitis was verified in murine models [205]. It had been proved that the median survival of tumor-bearing mice increased from 21 days in saline group to 45 and 48 days after intracranial administration of 1.22 and 2.44 MBq of 211At-trastuzumab, respectively.

    • Radiopharmaceutical Chemistry and Drug Development—What's Changed?

      2021, Seminars in Radiation Oncology
      Citation Excerpt :

      An advantage of 211At is the ability to label a broad range of molecules for uses as therapeutic radiopharmaceuticals, such as 211At-labeled macromolecules.12 A preclinical study of [211At]trastuzumab targeted to HER2-expressing leptomeningeal breast cancer shows promising results.20 Similarly, 211At-labeled monoclonal antibodies can be targeted to leukemia (CD-45), lymphoma (CD-20), and multiple myeloma (CD38), with active clinical trials in leukemia and promising pre-clinical studies in lymphoma and multiple myeloma.21-24

    • Phase I feasibility study for intrathecal administration of trastuzumab in patients with HER2 positive breast carcinomatous meningitis

      2018, European Journal of Cancer
      Citation Excerpt :

      Indeed, cerebrospinal fluid (CSF) levels of trastuzumab in patients given i.v. infusions were 49–420 times lower than matched serum levels [4]. Trastuzumab IT administration in rat models of CNS metastatic HER2-BC was safe without toxicity events and was superior to systemic delivery or to isotype-matched control antibody in terms of survival [5–7]. In a recent review of the literature pooling 17 case reports of IT administration of trastuzumab for MC in HER2-BC, Zagouri et al. reported no serious adverse events in 88.2% of cases and found significant clinical improvement in 68.8% of cases [8].

    • Astatine-211 labeled anti-HER2 5F7 single domain antibody fragment conjugates: radiolabeling and preliminary evaluation

      2018, Nuclear Medicine and Biology
      Citation Excerpt :

      The relative biological effectiveness of 211At-labeled trastuzumab was about 10 times higher than that of conventional external beam therapy, with significant reduction in survival achieved with only a few 211At atoms bound per cell. A subsequent study was performed in a HER2-positive breast carcinomatous meningitis model to evaluate the therapeutic efficacy of a single intrathecal injection of 211At-labeled trastuzumab [15]. Significant prolongation in median survival with some long-term survivors was observed; however, even with direct injection into the intrathecal compartment, histopathological analyses revealed that regions of the neuroaxis had escaped treatment in some animals.

    View all citing articles on Scopus

    This work was supported by Grants CA42324 and NS20023 from the National Institutes of Health and Grant DOE-FG02-08ER64697 from the Department of Energy.

    View full text