1,131
Views
39
CrossRef citations to date
0
Altmetric
Drug Profile

177Lu-DOTATATE for the treatment of gastroenteropancreatic neuroendocrine tumors

, , &
Pages 1023-1031 | Received 06 Sep 2019, Accepted 22 Oct 2019, Published online: 30 Oct 2019
 

ABSTRACT

Introduction: 177Lutetium-[DOTA°,Tyr3]octreotate (177Lu-DOTATATE) is a type of peptide receptor radionuclide therapy that garnered FDA approval in January 2018 for the treatment of somatostatin receptor-positive gastroenteropancreatic (GEP) neuroendocrine tumor (NET) patients. The therapy approval was based on findings from the randomized international phase III NETTER-1 trial as well as outcome data from a large European registry. The mechanism of the drug stems directly from its structure: a somatostatin analog (octreotate) selectively binding to somatostatin receptor expressing cells and being internalized, along with a chelated beta-emitting isotope 177Lu.

Areas Covered: Herein we describe the pharmacology, clinical efficacy and adverse event data from prospective and retrospective studies with 177Lu-DOTATATE. We discuss the role of 177Lu-DOTATATE within the current treatment landscape for GEP NET patients.

Expert Opinion: 177Lu-DOTATATE represents a unique addition to the treatment armamentarium for GEP NETs because of its potential to elicit tumor cytoreduction, which is rare among other existing treatment options, and prolonged disease control. Where 177Lu-DOTATATE fits into the treatment sequence for GEP NET patients remains an area of active investigation.

Article highlights

  • 177Lu-DOTATATE is a radiolabeled somatostatin agonist (SSA), which is approved for the treatment of somatostatin receptor positive well-differentiated gastroenteropancreatic (GEP) neuroendocrine tumor (NET) patients in Europe, the United States, and Canada.

  • 177Lu-DOTATATE belongs to a class of therapeutic agents called peptide receptor radionuclide therapy (PRRT). In the context of NETs, each agent includes a peptide, which is an SSA or somatostatin receptor antagonist, a chelator which serves as a linking molecule, and a specific radioisotope.

  • Radiolabeled SSAs have been trialed in Europe since 1994. The initial radiolabeled SSAs tested in phase I studies were 111In-DOTATOC (very weakly cytotoxic) and 90Y-DOTATOC. 177Lu-based PRRT was shown to have a more favorable therapeutic index than 90Y, particularly with respect to renal toxicity. Octreotate combined with the chelator DOTA (DOTATATE) showed increased somatostatin receptor 2 avidity and longer intra-tumoral radioactivity residence based on preclinical and dosimetry studies, respectively.

  • The NETTER-1 trial randomized midgut NET patients to four treatments of 177Lu-DOTATATE followed by standard-dose octreotide LAR or high-dose octreotide LAR. It is the only reported randomized phase III study involving any radiolabeled SSA. In the 177Lu-DOTATATE arm, progression-free (PFS) was markedly prolonged compared to the high-dose octreotide arm: hazard ratio [HR] 0.21; p < 0.001.

  • Quality of life (QOL) was analyzed on the NETTER-1 study using the European Organisation for Research and Treatment of Cancer (EORTC) questionnaires. Time to deterioration was significantly prolonged in the 177Lu-DOTATATE arm compared to patients in the high-dose octreotide arm with regards to global health status, physical functioning, diarrhea and fatigue.

  • Other radiolabeled SSAs and somatostatin antagonists which carry theoretical advantages over 177Lu-DOTATATE are now being tested in early safety-focused clinical trials. Two notable agents include the SSA 177Lu-DOTA-EB-TATE and the somatostatin receptor antagonist 177Lu-OPS201. 177Lu-DOTA-EB-TATE has demonstrated increased intra-tumoral radioactivity uptake preclinically and a longer plasma half-life due to its albumin binding moiety, compared to 177Lu-DOTATATE. 177Lu-OPS201 has demonstrated an increased capacity to elicit DNA damage preclinically and increased intra-tumor radioactivity uptake in a small pilot study clinically, compared to 177Lu-DOTATATE.

  • Ongoing studies are exploring whether PRRT is more effective in earlier treatment lines in GEP NET patients, whether additional cytoreductive treatments should be added to PRRT to improve its anti-tumor efficacy and what other patients (such as those with paragangliomas, pheochromocytomas, meningiomas, lung NETs) may benefit clinically from PRRT.

Declaration of interest

S. Das has received honoraria from Targeted Oncology, Lexicon, Medsphere and Clarivate Analytics. J. Strosberg has received honoraria from Novartis, Lexicon and Ipsen. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose

Additional information

Funding

Vanderbilt clinical oncology research career development award: 5K12CA090625-19.

Log in via your institution

Log in to Taylor & Francis Online

PDF download + Online access

  • 48 hours access to article PDF & online version
  • Article PDF can be downloaded
  • Article PDF can be printed
USD 99.00 Add to cart

Issue Purchase

  • 30 days online access to complete issue
  • Article PDFs can be downloaded
  • Article PDFs can be printed
USD 602.00 Add to cart

* Local tax will be added as applicable

Related Research

People also read lists articles that other readers of this article have read.

Recommended articles lists articles that we recommend and is powered by our AI driven recommendation engine.

Cited by lists all citing articles based on Crossref citations.
Articles with the Crossref icon will open in a new tab.