13
Peptide receptor radionuclide therapy (PRRT) for GEP-NETs

https://doi.org/10.1016/j.bpg.2013.01.004Get rights and content

Abstract

Peptide receptor radionuclide therapy (PRRT) with radiolabelled somatostatin analogues plays an increasing role in the treatment of patients with inoperable or metastasised gatroenteropancreatic neuroendocrine tumours (GEP-NETs). 90Y-DOTATOC and 177Lu-DOTATATE are the most used radiopeptides for PRRT with comparable tumour response rates (about 15–35%). The side effects of this therapy are few and mild. However, amino acids should be used for kidney protection, especially during infusion of 90Y-DOTATOC. Options to improve PRRT may include combinations of radioactive labelled somatostatin analogues and the use of radiosensitising drugs combined with PRRT. Other therapeutic applications of PRRT may include intra-arterial administration, neo-adjuvant treatment and additional PRRT cycles in patients with progressive disease, who have benefited from initial therapy. Considering the mild side-effects, PRRT may well become the first-line therapy in patients with metastasised or inoperable GEP-NETs if more widespread use of PRRT can be accomplished.

Introduction

In patients with inoperable metastasised gastroenteropancreatic neuroendocrine tumours (GEP-NETs), therapeutic options are limited. Treatment with somatostatin analogues such as octreotide and lanreotide reduces hormonal overproduction and can relieve symptoms in patients with GEP-NETs [1], [2], [3]. Furthermore, the long acting formula of the somatostatin Octreotide LAR significantly lengthens time to tumour progression in patients with metastatic midgut neuroendocrine tumours (NETs) [4].

The majority of GEP-NETs express somatostatin receptors, mainly somatostatin receptor subtype (sst) 2 and 5 [5]. These can be visualised using radiolabelled somatostatin analogues. The first commercially available diagnostic somatostatin receptor analogues were [111Indium-DTPA0]octreotide (111In-octreotide or Octreoscan) [6] and 99mTechnetium-depreotide (NEOSPECT or NEOTECT). Nowadays newer positron emission tomography (PET) radiopharmaceuticals have been developed, such as [68Ga-DOTA-Tyr3]octreotide [7] and [68Ga-DOTA-Tyr3]octreotate [8].

After the introduction of 111In-octreotide, the therapeutic application of high dose 111In-octreotide showed promising results on symptomatology in patients with neuroendocrine tumours [9]. However, the reported number of objective responses was rather disappointing with a relatively low percentage of patients with tumour shrinkage. In subsequent studies, severe haematological toxicity (myelodysplastic syndrome (MDS) or leukaemia) was observed in a few patients [10], [11].

The use of beta-emitters, such as Yttrium-90 (90Y) and Lutetium-177 (177Lu) was made possible with the introduction of the chelator DOTA (1,4,7,10-tetraazcyclodecane-1,4,7,10-tetraacetic acid). Newer, more efficient, radiolabelled somatostatin analogues were developed with high affinity to the somatostatin receptor, such as [90Y-DOTA0, Tyr3]octreotide (90Y-DOTATOC) and [177Lu-DOTA0, Tyr3]octreotate (177Lu-DOTATATE). The main focus of this review is on this next generation of somatostatin analogues, labelled with 177Lu and 90Y.

Section snippets

Chelator and peptides

The structure of somastostatin analogues that are used for diagnostic and therapeutic purposes consists of three parts: a radionuclide, a chelator and a cyclic octapeptide (Fig. 1).

DTPA (diethylene triamine penta-acetic acid) and DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,20-tetra-acetic acid) are commonly used as chelators for PRRT. The most widely used combinations of peptide-chelators are [DOTA0,Tyr3]octreotide (DOTATOC) and [DOTA0,Tyr3]octreotate (DOTATATE). Other complexes include [DOTA0

Radionuclide characteristics

Over the past two decades, different radionuclides have been used for PRRT. Indium-111 (111In), with its auger electrons caused by gamma-emission, was used in early studies [10], [11]. Besides encouraging results with regard to symptom relief, the reported number of objective responses was rather disappointing. Furthermore, 111In-coupled peptides are not ideal for PRRT due to the small particle range and therefore low tissue penetration.

90Y has high tissue penetration (12 mm) due to

90Y-labelled somatostatin analogues

Early studies with 90Y labelled somatostatin analogues were done in Basel by Otte et al [16], [17]. In their pilot study, 29 patients with mostly neuroendocrine tumours were treated with 90Y-DOTATOC in a mean cumulative administered activity of 6120 ± 1347 MBq/m2 given in four or more single injections. Sixteen patients were diagnosed with inoperable or metastasised GEP-NETs. Disease stabilisation (SD) was reported in 14 (88%) GEP-NET patients and one (6%) patient had a partial remission (PR).

177Lu-labelled somatostatin analogues

De Jong et al [26] compared different radiolabelled somatostatin analogues in a preclinical setting. 111In-octreotide, 90Y-DOTATOC and 177Lu-DOTATATE were injected in rats bearing Somatostatin receptor subtype 2 (sst2) tumours. 177Lu-DOTATATE demonstrated the highest tumour uptake together with excellent tumour-to-kidney ratios.

Forrer et al [27] injected 222 MBq 111In-DOTATOC and 111In-DOTATATE within two weeks in five patients with metastatic neuroendocrine tumours. They examined whether one

Side effects

The kidneys are the dose-limiting organs for PRRT, when 90Y labelled somatostatin analogues are used. Valkema et al [34] compared the decline in creatinine clearance after treatment with 90Y-DOTATOC or 177Lu-DOTATATE. A median decline in creatinine of 7.3% per year was observed in 28 patients treated with 90Y-DOTATOC with a median follow-up of 2.9 years [35]. The median decline in creatinine was 3.8% per year in 37 patients treated with 177Lu-DOTATATE (cumulative administered activity of

Retreatment

Van Essen et al [39] reported successful retreatment with 177Lu-DOTATATE in 33 NET patients. All patients had benefit from prior therapy with 18.5–29.6 GBq of 177Lu-DOTATATE and later again experienced progressive disease, documented by CT or somatostatin receptor scintigraphy (SRS). Two additional cycles of 7.4 GBq 177Lu-DOTATATE were given. Tumour response was evaluated using SWOG criteria. Two (6%) patients had PR, six (18%) patients responded with MR, SD was reported in ten (30%) patients

Dosimetry

In the first PRRT biodistribution and therapy studies, it was found that the kidneys and bone marrow are the critical organs for PRRT. The current role of dosimetry is that of a guide with respect to safety, especially to prevent kidney damage.

In a recent 177Lu-DOTATATE dose escalating study of Bodei et al [33], evaluation of pharmakinetics, activity biodistribution and absorbed dose to normal organs and tumours was performed. Fifty-one patients with metastatic sst2-positive tumours (mainly

Combination of compounds

A preclinical study by de Jong et al [15] compared the antitumoral effects of the combination of 177Lu- and 90Y-labelled somatostatin analogues with either 90Y- or 177Lu-analogue alone in animals bearing tumours of various sizes. The combination of both compounds was best for tumour control of both large and small tumours and resulted in the longest survival of the animals. The tumour in this rat model was rapidly growing and this may led to necrosis in parts of the tumour, therefore causing a

Summary

Peptide receptor radionuclide therapy (PRRT) with radiolabelled somatostatin analogues plays an increasing role in the treatment of patients with inoperable or metastasised gatroenteropancreatic neuroendocrine tumours (GEP-NETs). 90Y-DOTATOC and 177Lu-DOTATATE are the most used radiopeptides for PRRT with comparable tumour response rates (about 15–35%). The side effects of this therapy are few and mild. However, amino acids should be used for kidney protection, especially during infusion of 90

Conflict of interest

None.

References (62)

  • J.G. Jurcic et al.

    Targeted alpha particle immunotherapy for myeloid leukemia

    Blood

    (2002)
  • S. Nilsson et al.

    Bone-targeted radium-223 in symptomatic, hormone-refractory prostate cancer: a randomised, multicentre, placebo-controlled phase II study

    Lancet Oncol

    (2007)
  • T.K. Nayak et al.

    Somatostatin-receptor-targeted alpha-emitting 213Bi is therapeutically more effective than beta(-)-emitting 177Lu in human pancreatic adenocarcinoma cells

    Nucl Med Biol

    (2007)
  • R. Arnold et al.

    Gastroenteropancreatic endocrine tumours: effect of Sandostatin on tumour growth. The German Sandostatin Study Group

    Digestion

    (1993)
  • E.T. Janson et al.

    Long-term management of the carcinoid syndrome. Treatment with octreotide alone and in combination with alpha-interferon

    Acta Oncol

    (1993)
  • A. Rinke et al.

    Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group

    J Clin Oncol

    (2009)
  • J.C. Reubi et al.

    Somatostatin receptor sst1-sst5 expression in normal and neoplastic human tissues using receptor autoradiography with subtype-selective ligands

    Eur J Nucl Med

    (2001)
  • H.R. Balon et al.

    Procedure guideline for somatostatin receptor scintigraphy with (111)In-pentetreotide

    J Nucl Med

    (2001)
  • M. Gabriel et al.

    68Ga-DOTA-Tyr3-octreotide PET in neuroendocrine tumors: comparison with somatostatin receptor scintigraphy and CT

    J Nucl Med

    (2007)
  • I. Kayani et al.

    Functional imaging of neuroendocrine tumors with combined PET/CT using 68Ga-DOTATATE (DOTA-DPhe1, Tyr3-octreotate) and 18F-FDG

    Cancer

    (2008)
  • E.P. Krenning et al.

    Radiotherapy with a radiolabeled somatostatin analogue, [111In-DTPA-D-Phe1]-octreotide. A case history

    Ann N Y Acad Sci

    (1994)
  • J.C. Reubi et al.

    Affinity profiles for human somatostatin receptor subtypes SST1-SST5 of somatostatin radiotracers selected for scintigraphic and radiotherapeutic use

    Eur J Nucl Med

    (2000)
  • S. Walrand et al.

    Experimental facts supporting a red marrow uptake due to radiometal transchelation in 90Y-DOTATOC therapy and relationship to the decrease of platelet counts

    Eur J Nucl Med Mol Imaging

    (2011)
  • J.A. O'Donoghue et al.

    Relationships between tumor size and curability for uniformly targeted therapy with beta-emitting radionuclides

    J Nucl Med

    (1995)
  • M. de Jong et al.

    Combination radionuclide therapy using 177Lu- and 90Y-labeled somatostatin analogs

    J Nucl Med

    (2005)
  • A. Otte et al.

    DOTATOC: a powerful new tool for receptor-mediated radionuclide therapy

    Eur J Nucl Med

    (1997)
  • A. Otte et al.

    Yttrium-90 DOTATOC: first clinical results

    Eur J Nucl Med

    (1999)
  • D.L. Bushnell et al.

    90Y-edotreotide for metastatic carcinoid refractory to octreotide

    J Clin Oncol

    (2010)
  • A.K. Pfeifer et al.

    Peptide receptor radionuclide therapy with Y-DOTATOC and (177)Lu-DOTATOC in advanced neuroendocrine tumors: results from a Danish cohort treated in Switzerland

    Neuroendocrinology

    (2011)
  • A. Imhof et al.

    Response, survival, and long-term toxicity after therapy with the radiolabeled somatostatin analogue [90Y-DOTA]-TOC in metastasized neuroendocrine cancers

    J Clin Oncol

    (2011)
  • C. Waldherr et al.

    Tumor response and clinical benefit in neuroendocrine tumors after 7.4 GBq (90)Y-DOTATOC

    J Nucl Med

    (2002)
  • Cited by (0)

    View full text