Is Peptide Receptor Radionuclide Therapy on the Horizon for Unresectable or Metastatic Gastroenteropancreatic Neuroendocrine Tumours?
EDITORIAL
Is Peptide Receptor Radionuclide Therapy on the Horizon for Unresectable or Metastatic Gastroenteropancreatic Neuroendocrine Tumours?
VHF Lee
Department of Clinical Oncology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
Correspondence: Prof VHF Lee, Department of Clinical Oncology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. Email: vhflee@hku.hk
Contributor: The author solely contributed to the editorial, approved the final version for publication, and takes responsibility for its accuracy and integrity.
Conflicts of Interest: The author has disclosed no conflicts of interest.
With their insidious onset and indolent clinical
behaviours,[1] unresectable or metastatic neuroendocrine
tumours (NETs) have long been regarded as one of the
most intractable malignancies. The digestive system is
the most common site of involvement. In particular, the
gastroenteropancreatic (GEP) location of NETs (GEP-NETs)
as the most common well-differentiated NET has
been of great interest to clinicians. Well-differentiated
NETs are classified into grade 1 (Ki-67 index <3%),
grade 2 (Ki-67 index 3-20%), and grade 3 (Ki-67 index
>20%).[2] The abundant expression of somatostatin
receptors (SSRTs), especially subtype 2 (SSRT2) on
the NET cell surface makes SSRT-directed therapy a
promising treatment option.[3] [4]
Traditionally, somatostatin analogues, including
octreotide, lanreotide, and pasireotide, alone or in
combination with targeted therapy, have been the
most commonly used treatments for unresectable or
metastatic NETs, showing durable and effective tumour
control with favourable safety profiles.[5] [6] [7] Most recently,
peptide receptor radionuclide therapy (PRRT) with
either lutetium-177 (177Lu) or yttrium-90 has established
itself as a safe and effective treatment for unresectable
or metastatic GEP-NETs.[5] The NETTER-1 study, which
compared 177Lu-dotatate plus standard-dose long-acting
octreotide to high-dose long-acting octreotide alone as
second-line therapy in patients with advanced midgut
NETs demonstrated a significantly higher objective response to 177Lu-dotatate and a better progression-free
survival (PFS), although the secondary endpoint of
overall survival (OS) was not met.[8] [9] In view of such
encouraging result using 177Lu-dotatate PRRT as second-line
treatment, the NETTER-2 study was subsequently
conducted to evaluate the efficacy and safety of first-line
177Lu-dotatate PRRT plus long-acting repeatable
(LAR) octreotide versus high-dose LAR octreotide
alone in patients with higher grade 2 (Ki-67 indices
≥10% and ≤20%) and grade 3 (Ki-67 indices >20% and
≤55%) NETs.[10] The results again demonstrated a better
objective response (43% vs. 9%) and longer PFS (22.8
months vs. 8.5 months; hazard ratio 0.28, p < 0.0001)
but not a lengthened OS, when compared to LAR
indium-111 octreotide (111In-octreotide) alone.[10] The
absence of a significant OS benefit is often observed in
cross-over studies, where patients in the control group
may later receive investigational treatment. It should
be noted that almost all published studies have mainly
recruited Caucasians. The efficacy and safety of PRRT
has been less well assessed in the Chinese population.
Wong et al[11] reported the outcomes of PRRT in
their retrospective study of 21 Chinese patients with
metastatic NETs treated in a single institution in
Hong Kong, including one patient who had grade 3
NET. Most of them (85.7%) had received at least one
prior line of systemic therapy, including somatostatin
analogues, targeted therapy, and/or chemotherapy, while the remaining three patients (14.3%) received PRRT
as their first-line systemic treatment. All patients had
undergone 111In-octreotide scintigraphy or gallium-68
dotatate (68Ga-dotatate) positron emission tomography
with computed tomography (PET/CT) scanning prior
to treatment, which confirmed that the amount of SSRT
uptake by tumour cells was equal to or greater than that
of normal tissues. Both 177Lu and yttrium-90 were used
in two patients. The recruited patients underwent an
average of four PRRT sessions, ranging from one to six
sessions.
The study by Wong et al[11] reported an objective response
rate of 47.6% was observed, in addition to 23.8% stable
disease. The median PFS and OS were 22.3 months
and 45.2 months, respectively, after a median follow-up
duration of 19.0 months. Multivariable analysis
revealed that bone metastasis and a high liver tumour
burden of more than 50% were significant negative
prognostic factors in OS. Lymphopenia, followed
by anaemia, neutropenia, thrombocytopenia, and
hepatotoxicity were the most common adverse events
after PRRT. Grade 3/4 toxicities with lymphopoenia
and hepatotoxicity were reported in 42.9% and 4.8%
of patients, respectively. Of interest, two patients
received retreatment with PRRT after their initial
courses of PRRT, with tolerable and manageable
grade 3 lymphopenia noted in one patient. No patient
developed myelodysplastic syndrome, which had been
seen in one patient who had this possibly PRRT-related
toxicity approximately 14 months after the first dose of
PRRT in the NETTER-2 study.[10]
Despite the relatively small number of patients
and retrospective nature of this study, Wong et al[11]
demonstrated for the first time that PRRT is a safe
and effective treatment for metastatic GEP-NETs
in a Chinese population, echoing the results of the
NETTER-1[8] [9] and NETTER-2 trials.[10] Patient selection
and eligibility screening based on the uptake of 111In-octreotide
in octreotide scintigraphy or 68Ga-dotatate in
68Ga-dotatate PET/CT scans may merit discussion. The
Krenning score, a semi-quantitative tool, is commonly
used to assess SSRT uptake based on octreotide
scintigraphy and is defined as follows[12]: Grade 1 as
uptake less than normal liver background activity; grade
2 as uptake equal to normal liver background activity;
grade 3 as uptake greater than normal liver background
activity; and grade 4 as uptake greater than spleen or
kidney background activity. In the NETTER-1 study,[9]
pretreatment screening was performed using octreotide scintigraphy and the Krenning score, with patients
eligible if their score was grade 2, 3, or 4. The NETTER-2
study,[10] used either 111In-octreotide scintigraphy with the
Krenning score or 68Ga-dotatate PET/CT with a modified
Krenning score (an adaptation of the original Krenning
score applied to 68Ga-dotatate PET/CT) for eligibility.
However, it is still unclear whether the modified
Krenning scores are equivalent between these imaging
modalities. A post-hoc head-to-head comparison study
of 68Ga-dotatate PET/CT and 111In-octreotide scan-based Krenning scores in 150 patients from a phase 2
prospective study (NCT01967537) revealed that the
Krenning score was significantly higher with PET/CT
than with two-dimensional scintigraphy or 111In-octreotide
scintigraphy.[13] In patients with a Krenning
score of 3 or above on PET/CT, the detection rates
of two-dimensional scintigraphy and 111In-octreotide
scintigraphy were significantly lower for lesions smaller
than 2 cm compared to lesions of 2 cm or larger: 15%
and 24% versus 78% and 89%, respectively (p < 0.001).
On the other hand, for lesions greater than 5 cm, the
Krenning scores between PET/CT scan and octreotide
scintigraphy were comparable. Lesion size did not
affect PET/CT-based Krenning scores. In other words,
octreotide scintigraphy may miss smaller lesions (<2
cm) that would otherwise be detected on 68Ga-dotatate
PET/CT. Prospective studies are warranted to standardise
the use of a single imaging modality for eligibility
screening.
Of concern, a recent notification was announced by the
drug sponsor of the NETTER-2 study that the application
for using 177Lu-PRRT as first-line systemic therapy to the
European Medicines Agency was withdrawn.[14] The lack
of OS prolongation because of immature OS data and
potentially unfavourable risks including myelodysplastic
syndrome, radiation-associated second malignancies,
and haematological and renal toxicities in a treatment-naïve
population were the key concerns.[10] For now,
the current European Medicines Agency approval for
177Lu-dotatate is confined to advanced, progressive,
grade 1 and grade 2 GEP-NETs.[15] Patients with grade 3
NETs are still denied access to PRRT.
In summary, PRRT is a novel and promising treatment
modality for grade 1 and 2 unresectable or metastatic
GEP-NETs after failure of prior systemic therapy. More
prospective and mature data for grade 3 NETs and OS
are awaited to confirm whether PRRT can also work
favourably in this histological subgroup and as first-line
therapy for treatment-naïve patients.
REFERENCES
1. Abboud Y, Shah A, Sutariya R, Shah VP, Al-Khazraji A, Gaglio PJ, et al. Gastroenteropancreatic neuroendocrine tumor incidence by sex and age in the US. JAMA Oncol. 2025;11:345-9. Crossref
2. Rindi G, Mete O, Uccella S, Basturk O, La Rosa S, Brosens LA,
et al. Overview of the 2022 WHO classification of neuroendocrine
neoplasms. Endocr Pathol. 2022;33:115-54. Crossref
3. Mizutani G, Nakanishi Y, Watanabe N, Honma T, Obana Y, Seki T,
et al. Expression of somatostatin receptor (SSTR) subtypes (SSTR-1, 2A, 3, 4 and 5) in neuroendocrine tumors using real-time RT-PCR
method and immunohistochemistry. Acta Histochem Cytochem.
2012;45:167-76. Crossref
4. Han G, Hwang E, Lin F, Clift R, Kim D, Guest M, et al. RYZ101
(Ac-225 DOTATATE) opportunity beyond gastroenteropancreatic
neuroendocrine tumors: preclinical efficacy in small-cell lung
cancer. Mol Cancer Ther. 2023;22:1434-43. Crossref
5. Rinke A, Wittenberg M, Schade-Brittinger C, Aminossadati B, Ronicke E, Gress TM, 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 (PROMID): results of long-term
survival. Neuroendocrinology. 2017;104:26-32. Crossref
6. Caplin ME, Pavel M, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371:224-33. Crossref
7. Kulke MH, Ruszniewski P, Van Cutsem E, Lombard-Bohas C,
Valle JW, De Herder WW, et al. A randomized, open-label, phase
2 study of everolimus in combination with pasireotide LAR or
everolimus alone in advanced, well-differentiated, progressive
pancreatic neuroendocrine tumors: COOPERATE-2 trial. Ann
Oncol. 2019;30:1846. Crossref
8. Strosberg J, El-Haddad G, Wolin E, Hendifar A, Yao J, Chasen
B, et al. Phase 3 trial of 177Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376:125-35. Crossref
9. Strosberg JR, Caplin ME, Kunz PL, Ruszniewski PB, Bodei
L, Hendifar A, et al. 177Lu-dotatate plus long-acting octreotide
versus high-dose long-acting octreotide in patients with midgut
neuroendocrine tumours (NETTER-1): final overall survival and
long-term safety results from an open-label, randomised, controlled,
phase 3 trial. Lancet Oncol. 2021;22:1752-63. Crossref
10. Singh S, Halperin D, Myrehaug S, Herrmann K, Pavel M, Kunz
PL, et al. [177Lu]Lu-DOTA-TATE plus long-acting octreotide
versus high dose long-acting octreotide for the treatment of
newly diagnosed, advanced grade 2-3, well-differentiated,
gastroenteropancreatic neuroendocrine tumours (NETTER-2): an
open-label, randomised, phase 3 study. Lancet. 2024;403:2807-17. Crossref
11. Wong WH, Lam HC, Au Yong TK. Outcomes of peptide receptor radionuclide therapy in metastatic neuroendocrine tumours. Hong Kong J Radiol. 2025;28:e163-71. Crossref
12. Kwekkeboom DJ, Krenning EP. Somatostatin receptor imaging.
Semin Nucl Med. 2002;32:84-91. Crossref
13. Hope TA, Calais J, Zhang L, Dieckmann W, Millo C. 111In-pentetreotide
scintigraphy versus 68Ga-dotatate PET: impact
on Krenning scores and effect of tumor burden. J Nucl Med.
2019;60:1266-9. Crossref
14. European Medicines Agency. Withdrawal of application to
change the marketing authorisation for Lutathera (lutetium (177Lu) oxodotreotide). 23 May 2025. Available from: https://www.ema.europa.eu/en/documents/medicine-qa/questions-answers-withdrawal-application-change-marketing-authorisation-lutathera-lutetium-177lu-oxodotreotide-ii-52_en.pdf. Accessed 25 Jul 2025.
15. European Medicines Agency. Lutathera, INN-lutetium (177Lu)
oxodotreotide. Annex I. Summary of product characteristics.
Available from: https://www.ema.europa.eu/en/documents/product-information/lutathera-epar-product-information_en.pdf.
Accessed 25 Jul 2025.