Your browser doesn't support javascript.
loading
Targeting of the Cholecystokinin-2 Receptor with the Minigastrin Analog 177Lu-DOTA-PP-F11N: Does the Use of Protease Inhibitors Further Improve In Vivo Distribution?
Sauter, Alexander W; Mansi, Rosalba; Hassiepen, Ulrich; Muller, Lionel; Panigada, Tania; Wiehr, Stefan; Wild, Anna-Maria; Geistlich, Susanne; Béhé, Martin; Rottenburger, Christof; Wild, Damian; Fani, Melpomeni.
Afiliação
  • Sauter AW; Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland.
  • Mansi R; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany.
  • Hassiepen U; Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland.
  • Muller L; Novartis Pharma AG, Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland; and.
  • Panigada T; Novartis Pharma AG, Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland; and.
  • Wiehr S; Novartis Pharma AG, Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland; and.
  • Wild AM; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany.
  • Geistlich S; Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tuebingen, Germany.
  • Béhé M; Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, Villigen, Switzerland.
  • Rottenburger C; Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, Villigen, Switzerland.
  • Wild D; Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland.
  • Fani M; Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland.
J Nucl Med ; 60(3): 393-399, 2019 03.
Article em En | MEDLINE | ID: mdl-30002107
ABSTRACT
Patients with metastatic medullary thyroid cancer (MTC) have limited systemic treatment options. The use of radiolabeled gastrin analogs targeting the cholecystokinin-2 receptor (CCK2R) is an attractive approach. However, their therapeutic efficacy is presumably decreased by their enzymatic degradation in vivo. We aimed to investigate whether the chemically stabilized analog 177Lu-DOTA-PP-F11N (177Lu-DOTA-(dGlu)6-Ala-Tyr-Gly-Trp-Nle-Asp-Phe-NH2) performs better than reference analogs with varying in vivo stability, namely 177Lu-DOTA-MG11 (177Lu-DOTA-dGlu-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2) and 177Lu-DOTA-PP-F11 (177Lu-DOTA-(dGlu)6-Ala-Tyr-Gly-Trp-Met-Asp-Phe-NH2), and whether the use of protease inhibitors further improves CCKR2 targeting. First human data on 177Lu-DOTA-PP-F11N are also reported.

Methods:

In vitro stability of all analogs was assessed against a panel of extra- and intracellular endoproteases, whereas their in vitro evaluation was performed using the human MTC MZ-CRC-1 and the transfected A431-CCK2R(+) cell lines. Biodistribution without and with the protease inhibitors phosphoramidon and thiorphan was assessed 4 h after injection in MZ-CRC-1 and A431-CCK2R(+) dual xenografts. Autoradiography of 177Lu-DOTA-PP-F11N (without and with phosphoramidon) and NanoSPECT/CT were performed. SPECT/CT images of 177Lu-DOTA-PP-F11N in a metastatic MTC patient were also acquired.

Results:

natLu-DOTA-PP-F11N is less of a substrate for neprilysins than the other analogs, whereas intracellular cysteine proteases, such as cathepsin-L, might be involved in the degradation of gastrin analogs. The uptake of all radiotracers was higher in MZ-CRC-1 tumors than in A431-CCK2R(+), apparently because of the higher number of binding sites on MZ-CRC-1 cells. 177Lu-DOTA-PP-F11N had the same biodistribution as 177Lu-DOTA-PP-F11; however, uptake in the MZ-CRC-1 tumors was almost double (20.7 ± 1.71 vs. 11.2 ± 2.94 %IA [percentage injected activity]/g, P = 0.0002). Coadministration of phosphoramidon or thiorphan increases 177Lu-DOTA-MG11 uptake significantly in the CCK2R(+) tumors and stomach. Less profound was the effect on 177Lu-DOTA-PP-F11, whereas no influence or even reduction was observed for 177Lu-DOTA-PP-F11N (20.7 ± 1.71 vs. 15.6 ± 3.80 [with phosphoramidon] %IA/g, P < 0.05 in MZ-CRC-1 tumors). The first clinical data show high 177Lu-DOTA-PP-F11N accumulation in tumors, stomach, kidneys, and colon.

Conclusion:

The performance of 177Lu-DOTA-PP-F11N without protease inhibitors is as good as the performance of 177Lu-DOTA-MG11 in the presence of inhibitors. The human application of single compounds without unessential additives is preferable. Preliminary clinical data spotlight the stomach as a potential dose-limiting organ besides the kidneys.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Inibidores de Proteases / Radioisótopos / Gastrinas / Receptor de Colecistocinina B / Compostos Heterocíclicos com 1 Anel / Lutécio Limite: Animals / Female / Humans Idioma: En Revista: J Nucl Med Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Suíça

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Inibidores de Proteases / Radioisótopos / Gastrinas / Receptor de Colecistocinina B / Compostos Heterocíclicos com 1 Anel / Lutécio Limite: Animals / Female / Humans Idioma: En Revista: J Nucl Med Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Suíça