Skip to main content

Advertisement

Log in

Pre-therapeutic dosimetry of normal organs and tissues of 177Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer

  • Original Article
  • Published:
European Journal of Nuclear Medicine and Molecular Imaging Aims and scope Submit manuscript

Abstract

Purpose

177Lu-617-prostate-specific membrane antigen (PSMA) ligand seems to be a promising tracer for radionuclide therapy of progressive prostate cancer. However, there are no published data regarding the radiation dose given to the normal tissues. The aim of the present study was to estimate the pretreatment radiation doses in patients who will undergo radiometabolic therapy using a tracer amount of 177Lu-labeled PSMA ligand.

Methods

The study included seven patients with progressive prostate cancer with a mean age of 63.9 ± 3.9 years. All patients had prior PSMA positron emission tomography (PET) imaging and had intense tracer uptake at the lesions. The injected 177Lu-PSMA-617 activity ranged from 185 to 210 MBq with a mean of 192.6 ± 11.0 MBq. To evaluate bone marrow absorbed dose 2-cc blood samples were withdrawn in short variable times (3, 15, 30, 60, and 180 min and 24, 48, and 120 h) after injection. Whole-body images were obtained at 4, 24, 48, and 120 h post-injection (p.i.). The geometric mean of anterior and posterior counts was determined through region of interest (ROI) analysis. Attenuation correction was applied using PSMA PET/CT images. The OLINDA/EXM dosimetry program was used for curve fitting, residence time calculation, and absorbed dose calculations.

Results

The calculated radiation-absorbed doses for each organ showed substantial variation. The highest radiation estimated doses were calculated for parotid glands and kidneys. Calculated radiation-absorbed doses per megabecquerel were 1.17 ± 0.31 mGy for parotid glands and 0.88 ± 0.40 mGy for kidneys. The radiation dose given to the bone marrow was significantly lower than those of kidney and parotid glands (p < 0.05). The calculated radiation dose to bone marrow was 0.03 ± 0.01 mGy/MBq.

Conclusion

Our first results suggested that 177Lu-PSMA-617 therapy seems to be a safe method. The dose-limiting organ seems to be the parotid glands rather than kidneys and bone marrow. The lesion radiation doses are within acceptable ranges; however, there is a substantial individual variance so patient dosimetry seems to be mandatory.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Wright GL, Haley C, Beckett ML, Schellhammer PF. Expression of prostate-specific membrane antigen in normal, benign, and malignant prostate tissues. Urol Oncol 1995;1:18–28.

    Article  PubMed  Google Scholar 

  2. Perner S, Hofer MD, Kim R, Shah RB, Li H, Möller P, et al. Prostate-specific membrane antigen expression as a predictor of prostate cancer progression. Hum Pathol 2007;38:696–701.

    Article  CAS  PubMed  Google Scholar 

  3. Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C. Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res 1997;3:81–5.

    CAS  PubMed  Google Scholar 

  4. Eder M, Schäfer M, Bauder-Wüst U, Hull W-E, Wängler C, Mier W, et al. 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging. Bioconjug Chem 2012;23:688–97.

  5. Eder M, Schäfer M, Bauder-Wüst U, Haberkorn U, Eisenhut M, Kopka K. Preclinical evaluation of a bispecific low-molecular heterodimer targeting both PSMA and GRPR for improved PET imaging and therapy of prostate cancer. Prostate 2014;74:659–68.

    Article  CAS  PubMed  Google Scholar 

  6. Afshar-Oromieh A, Malcher A, Eder M, Eisenhut M, Linhart HG, Hadaschik BA, et al. PET imaging with a [68Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour lesions. Eur J Nucl Med Mol Imaging 2013;40:486–95.

  7. Afshar-Oromieh A, Avtzi E, Giesel FL, Holland-Letz T, Linhart HG, Eder M, et al. The diagnostic value of PET/CT imaging with the (68)Ga-labelled PSMA ligand HBED-CC in the diagnosis of recurrent prostate cancer. Eur J Nucl Med Mol Imaging 2015;42:197–209.

  8. Kabasakal L, Demirci E, Ocak M, Akyel R, Nematyazar J, Aygun A, et al. Evaluation of PSMA PET/CT imaging using a 68Ga-HBED-CC ligand in patients with prostate cancer and the value of early pelvic imaging. Nucl Med Commun 2015;36:582–7.

  9. Kratochwil C, Giesel FL, Eder M, Afshar-Oromieh A, Benešová M, Mier W, et al. [(177)Lu]Lutetium-labelled PSMA ligand-induced remission in a patient with metastatic prostate cancer. Eur J Nucl Med Mol Imaging 2015;42:987–8.

  10. Benešová M, Schäfer M, Bauder-Wüst U, Afshar-Oromieh A, Kratochwil C, Mier W, et al. Preclinical evaluation of a tailor-made DOTA-conjugated PSMA inhibitor with optimized linker moiety for imaging and endoradiotherapy of prostate cancer. J Nucl Med 2015;56:914–20.

  11. Siegel JA, Thomas SR, Stubbs JB, Stabin MG, Hays MT, Koral KF, et al. MIRD pamphlet no. 16: techniques for quantitative radiopharmaceutical biodistribution data acquisition and analysis for use in human radiation dose estimates. J Nucl Med 1999;40:37S–61S.

    CAS  PubMed  Google Scholar 

  12. Brown S, Bailey DL, Willowson K, Baldock C. Investigation of the relationship between linear attenuation coefficients and CT Hounsfield units using radionuclides for SPECT. Appl Radiat Isot 2008;66:1206–12.

    Article  CAS  PubMed  Google Scholar 

  13. Wessels BW, Bolch WE, Bouchet LG, Breitz HB, Denardo GL, Meredith RF, et al. Bone marrow dosimetry using blood-based models for radiolabeled antibody therapy: a multiinstitutional comparison. J Nucl Med 2004;45:1725–33.

  14. Thierens HM, Monsieurs MA, Bacher K. Patient dosimetry in radionuclide therapy: the whys and the wherefores. Nucl Med Commun 2005;26:593–9.

    Article  PubMed  Google Scholar 

  15. Glatting G, Bardiès M, Lassmann M. Treatment planning in molecular radiotherapy. Z Med Phys 2013;23:262–9.

  16. Wehrmann C, Senftleben S, Zachert C, Müller D, Baum RP. Results of individual patient dosimetry in peptide receptor radionuclide therapy with 177Lu DOTA-TATE and 177Lu DOTA-NOC. Cancer Biother Radiopharm 2007;22:406–16.

  17. Kwekkeboom D, Bakker W, Kooij P, Konijnenberg M, Srinivasan A, Erion J, et al. [177 Lu-DOTA0,Tyr3]octreotate: comparison with [111In-DTPA0]octreotide in patients. Eur J Nucl Med 2001;28:1319–25.

  18. Forrer F, Krenning EP, Kooij PP, Bernard BF, Konijnenberg M, Bakker WH, et al. Bone marrow dosimetry in peptide receptor radionuclide therapy with [177Lu-DOTA(0),Tyr(3)]octreotate. Eur J Nucl Med Mol Imaging 2009;36:1138–46.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Svensson J, Berg G, Wängberg B, Larsson M, Forssell-Aronsson E, Bernhardt P. Renal function affects absorbed dose to the kidneys and haematological toxicity during 177Lu-DOTATATE treatment. Eur J Nucl Med Mol Imaging 2015;42:947–55..

  20. Sandström M, Garske U, Granberg D, Sundin A, Lundqvist H. Individualized dosimetry in patients undergoing therapy with (177)Lu-DOTA-D-Phe(1)-Tyr(3)-octreotate. Eur J Nucl Med Mol Imaging 2010;37:212–25.

    Article  PubMed  Google Scholar 

  21. Van Essen M, Krenning EP, Kam BLR, de Jong M, Valkema R, Kwekkeboom DJ. Peptide-receptor radionuclide therapy for endocrine tumors. Nat Rev Endocrinol 2009;5:382–93.

  22. Sandström M, Garske-Román U, Granberg D, Johansson S, Widström C, Eriksson B, et al. Individualized dosimetry of kidney and bone marrow in patients undergoing 177Lu-DOTA-octreotate treatment. J Nucl Med 2013;54:33–41.

  23. Lassmann M, Chiesa C, Flux G, Bardiès M, EANM Dosimetry Committee. EANM Dosimetry Committee guidance document: good practice of clinical dosimetry reporting. Eur J Nucl Med Mol Imaging 2011;38:192–200.

  24. Deasy JO, Moiseenko V, Marks L, Chao KSC, Nam J, Eisbruch A. Radiotherapy dose-volume effects on salivary gland function. Int J Radiat Oncol Biol Phys 2010;76:S58–63.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Grundmann O, Mitchell GC, Limesand KH. Sensitivity of salivary glands to radiation: from animal models to therapies. J Dent Res 2009;88:894–903.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Li Y, Taylor JMG, Ten Haken RK, Eisbruch A. The impact of dose on parotid salivary recovery in head and neck cancer patients treated with radiation therapy. Int J Radiat Oncol Biol Phys 2007;67:660–9.

    Article  PubMed Central  PubMed  Google Scholar 

  27. Jeong SY, Kim HW, Lee S-W, Ahn B-C, Lee J. Salivary gland function 5 years after radioactive iodine ablation in patients with differentiated thyroid cancer: direct comparison of pre- and postablation scintigraphies and their relation to xerostomia symptoms. Thyroid 2013;23:609–16.

  28. Grewal RK, Larson SM, Pentlow CE, Pentlow KS, Gonen M, Qualey R, et al. Salivary gland side effects commonly develop several weeks after initial radioactive iodine ablation. J Nucl Med 2009;50:1605–10.

    Article  CAS  PubMed  Google Scholar 

  29. Sreebny LM, Valdini A, Yu A. Xerostomia. Part II: relationship to nonoral symptoms, drugs, and diseases. Oral Surg Oral Med Oral Pathol 1989;68:419–27.

    Article  CAS  PubMed  Google Scholar 

  30. Norrgren K, Svegborn SL, Areberg J, Mattsson S. Accuracy of the quantification of organ activity from planar gamma camera images. Cancer Biother Radiopharm 2003;18:125–31.

    Article  PubMed  Google Scholar 

  31. Garkavij M, Nickel M, Sjögreen-Gleisner K, Ljungberg M, Ohlsson T, Wingårdh K, et al. 177Lu-[DOTA0,Tyr3] octreotate therapy in patients with disseminated neuroendocrine tumors: analysis of dosimetry with impact on future therapeutic strategy. Cancer 2010;116:1084–92.

  32. Kletting P, Müller B, Erentok B, Schmaljohann J, Behrendt FF, Reske SN, et al. Differences in predicted and actually absorbed doses in peptide receptor radionuclide therapy. Med Phys 2012;39:5708–17.

  33. Hindorf C, Glatting G, Chiesa C, Lindén O, Flux G, EANM Dosimetry Committee. EANM Dosimetry Committee guidelines for bone marrow and whole-body dosimetry. Eur J Nucl Med Mol Imaging 2010;37:1238–50.

Download references

Compliance with ethical standards

Funding

This study was not funded

Conflicts of interest

None.

Ethical approval

The study was approved by the local Ethics Committee and was carried out in accordance with the 1964 Declaration of Helsinki.

Informed consent

Informed consent was obtained from all individual participants included in the study

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Levent Kabasakal.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kabasakal, L., AbuQbeitah, M., Aygün, A. et al. Pre-therapeutic dosimetry of normal organs and tissues of 177Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 42, 1976–1983 (2015). https://doi.org/10.1007/s00259-015-3125-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-015-3125-3

Keywords

Navigation