Skip to main content
Erschienen in: Endocrine 2/2022

Open Access 26.08.2022 | Mini Review

Radioligand therapy (RLT) as neoadjuvant treatment for inoperable pancreatic neuroendocrine tumors: a literature review

verfasst von: Luca Urso, Alberto Nieri, Ilaria Rambaldi, Angelo Castello, Licia Uccelli, Corrado Cittanti, Stefano Panareo, Irene Gagliardi, Maria Rosaria Ambrosio, Maria Chiara Zatelli, Mirco Bartolomei

Erschienen in: Endocrine | Ausgabe 2/2022

Abstract

In the last 10 years, several literature reports supported radioligand therapy (RLT) in neoadjuvant settings for pancreatic neuroendocrine tumors (PanNETs). Indeed, primary tumor shrinkage has been frequently reported following RLT in unresectable or borderline resectable PanNETs. Moreover, RLT-induced intratumoral modifications facilitate surgery, both on primary tumor and metastasis, having a great impact on progression free survival (PFS), overall survival (OS) and quality of life (QoL). However, prospective controlled investigations are necessary to confirm preliminary data and to define the best RLT scheme and the ideal patient that, in a multidisciplinary approach, should be referred to neoadjuvant RLT.
Hinweise
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Pancreatic neuroendocrine tumors (PanNETs) account for ~10% of all neuroendocrine tumors (NETs). Approximately 70% of these neoplasms are non-functioning and frequently present as relatively indolent and slow-growing masses. Therefore, diagnosis is frequently delayed, occurring when the disease has already spread to lymph nodes, liver, or bone [13].
PanNETs are currently classified according to the 2017 WHO classification. Grading has a strong impact on both clinical behavior and prognosis of patients affected by PanNETs, being a crucial parameter to take into consideration in the global therapeutic strategy [4].
Currently, surgery is potentially the only curative treatment for PanNETs [5]. Conversely, chemotherapy and molecular targeted agents are indicated in patients with advanced, recurrent or metastatic tumors not suitable for surgery (unresectable or borderline resectable), particularly in higher grade forms, while the role of external beam radiotherapy is limited [57].
Well-differentiated PanNETs usually overexpress somatostatin receptors (SSTRs), representing an ideal model for a theranostic approach in Nuclear Medicine. At present, procedures are well established for radiolabelled somatostatin analogs (SSA) with single-photon emission computerized tomography (SPECT) or positron emission tomography (PET) agents, such as Indium-111 (111I) or Gallium-68 (68Ga). Likewise, in the settings of radioligand therapy (RLT), SSA can bind high energy beta emitters, such as Yttrium-90 (90Y) or Lutetium-177 (177Lu) [8, 9].
Since 2017, after the encouraging results from the NETTER-1 phase III clinical trial, RLT has been established as an effective and safe therapy for advanced, metastatic or inoperable gastro-entero pancreatic (GEP) NETs [1012]. RLT is usually proposed as a delayed treatment, when other lines of therapy have already failed [13]. Nevertheless, before NETTER-1 several studies reported a greater efficacy of RLT if offered at a relatively earlier stage of disease, when the tumor burden is lower and patients have a better performance status [14, 15]. Therefore, based on literature evidences, RLT has been proposed as a neoadjuvant treatment, in order to obtain a tumor size decrease or downstaging, allowing a surgical approach and increasing the likelihood of a radical resection [3, 1620].
The present mini-review explores the emerging role of RLT as a potential neoadjuvant treatment for PanNETs.

Methods

We performed a literature search using MEDLINE (PubMed database) on April 30th 2022 to identify articles in English on neoadjuvant RLT. We used Keywords chosen according to Medical Subject Heading (MeSH) terms “neoadjuvant” OR “preoperative” AND “radioligand therapy” OR “Peptide Receptor Radionuclide Therapy”. We included original articles and case reports. Editorials, Letters, and abstracts from conference proceedings were excluded. The full articles of selected studies were examined, and additional searches of their reference lists performed to identify other potentially eligible articles. Moreover, research of active ongoing trials was performed on clinicaltrials.gov.

Results

We found 11 papers including 148 patients as reported in Table 1. Literature evidence is heterogeneous considering sample size, patient characteristics, RLT schemes and outcome assessment. Our analysis and considerations were focused on the following issues: a) tumor response: primary shrinkage, downstaging and downgrading; b) histological and tumor microenvironment modifications; c) influence on tumor burden; d) safety and reproducibility of neoadjuvant RLT.
Table 1
Literature review of neoadjuvant RLT in PanNETs
Study
Year
Patients treated
Radionuclide used
Outcome
Toxicity
Kaemmerer et al. [21]
2009
1
90Y-DOTATOC
(2 cycles)
CR after surgery
Only mild transient hematologic toxicity
Stoeltzing
et al. [34]
2010
1
90Y-DOTATOC
(2 cycles)
CR after 1 year from surgery
Not reported
Ezzidin et al. [22]
2012
1
177Lu-DOTATATE
(3 cycles)
Inoperable PAN-NET became eligible for surgery achieving a complete local remission for 22 months
Minor transient leukopenia (Grade 1)
Barber
et al. [28]
2012
5 (4)
177Lu-DOTATATE
(1–4 cycles)
3 scintigraphic PR; 4 radiological PR and 1SD.
1 patient affected by PanNET became eligible for surgery with a reduction in Ki67 from 10–15% to 5%.
Not reported
Van Vliet
et al. [23]
2015
29
177Lu-DOTATATE
(3–4 cycles)
9/29 patients became amenable for surgery and had an increase in PFS vs. control group (69 vs. 49 months)
No patients showed delayed toxicity
Partelli
et al. [16]
2018
23
177Lu/90Y-DOTATATE
(20/3 pt.)
• No pancreatic fistula development in RLT patients.
• Longer PFS in RLT patients with R0 resection.
Only 1 patient had hematologic toxicity and stopped RLT
Da Silva
et al. [24]
2018
1
177Lu-DOTATATE
Patient became eligible for surgery and was
tumor-free after 3-year follow up
No RLT-induced toxicity reported
Zanata et al. [25]
2021
1
90Y-DOTATOC
Patient became eligible for surgery and was
tumor-free after 108 months follow up. Disease downstaging from stage IV to stage I.
Not reported
Parghane et al. [26]
2021
57 (32)
177Lu-DOTATATE
6/32 PanNETs became resectable after RLT. Primary tumors and liver metastasis decreased in size.
Not reported
Opalinska et al. [27]
2021
9 (6)
177Lu/90Y-DOTATATE
Median primary tumor size decreased by 1.6 cm after RLT, with no differences between Pan-NETs and other GEP-NETs. 4/9 patients became resectable after RLT and in 2 a radical surgery was achieved. Liver metastases became undetectable in 2 patients.
No myelotoxicity or nephrotoxicity found
Minczeles et al. [29]
2022
49
177Lu-DOTATATE
Patients with neoadjuvant RLT followed by surgery had a longer PFS and OS than those who performed only RLT.
A downstaging was found in the 38% of patients with baseline tumor-vessel involvement
47% of patients had transient hematologic toxicity. G3 liver toxicity was found in 1 patient
Treated patients: the number within brackets indicates the number or PanNETS among the cohort of patients treated with RLT.
CR complete response, PanNET pancreatic neuroendocrine tumors, PFS progression free survival, PR partial response, RLT radioligand therapy, SD stable disease.
Moreover, consulting clinicaltrials.gov we found 1 active prospective multicentre trial concerning neoadjuvant RLT for PanNETs.

Tumor response: primary shrinkage, downstaging and downgrading

Overall 148 patients with locally advanced PanNETs undergoing neoadjuvant RLT are described in literature. Seventy-two became eligible for surgery [16, 2129]. Of note, we found 2 cases of extremely large primary tumors who became eligible for surgery following a significant tumor shrinkage and reduction in vascular involvement [24, 25]. Indeed, RLT acts as a cytoreductive agent in well-differentiated NETs, leading to a volumetric reduction of target lesions (Fig. 1). The possibility of performing curative or debulking surgery, in this subset of patients, is associated with an improvement in survival rates and quality of life (QoL), even in advanced or metastatic disease [25]. Moreover, Partelli et al. [16] reported that, among patients with a curative resection (i.e. R0), those treated with neoadjuvant RLT showed a significantly longer progression free survival (PFS) than those who performed surgery up-front. Similarly, Minczeles et al. [29] reported that patients with neoadjuvant RLT followed by surgery had a longer PFS and OS than those who performed RLT only.
The same authors also demonstrated a downstaging in 38% of patients with baseline tumor-vessel involvement.
Grading is a key parameter to assess NETs aggressiveness and, consequently, prognosis [4]. A few papers reported downgrading after RLT [21, 25, 28], mostly from G2 to G1. The reduction in Ki-67 index may be due to the greater radiosensitivity of tumor cells characterized by a higher proliferating cell population [16, 30]. Therefore, we can speculate that neoadjuvant RLT in PanNETs might act by preserving low-grade cell clones compared to the high-grade ones, which are more radiosensitive and selectively killed. The resulting global downgrading obtained could potentially have a significant impact on long surviving patients’ outcomes, both in terms of PFS, OS and QoL [25]. Nevertheless, further studies with larger number of cases are required to validate this hypothesis.

Histological and tumor microenvironment modifications

As reported by Schiavo Lena et al. [31], neoadjuvant RLT can induce intratumoral microenvironment modifications and changes in stroma/cellularity ratio, inducing a progressive scarring process and fibrotic involution. Of note, stromal percentage was higher in patients treated with neoadjuvant RLT in comparison with those who underwent surgery up-front, particularly in patients in which a remarkable reduction in tumor diameter was achieved. Such tissue modifications, together with tumor size decrease can simplify surgery, reducing both operative time and major complications, particularly pancreatic fistula [16]. As a consequence, patients treated with neoadjuvant PRRT could benefit of a faster post-operative recovery, with a concomitant cost reduction for the Healthcare System.

Influence on tumor burden

RLT is a systemic therapy targeting tumor cells expressing SSTRs. In patients with oligometastatic NETs, neoadjuvant RLT could therefore potentially target both primary tumor and metastases. Particularly, liver tumor burden is well known as a main prognostic factor in NETs [12, 30, 32]. Recently, the North American Neuroendocrine Tumor Society suggested several potential advantages of neoadjuvant RLT in reducing liver tumor burden in metastatic PanNETs [33]. In this subset of patients RLT might allow subsequent locoregional hepatic therapies with a potential curative outcome and a consequent increase in PFS, OS and time to recurrence [26, 33]. This recommendation is supported by the positive results obtained with neoadjuvant RLT in reducing liver disease burden, as previously reported [2628, 34]. Moreover, RLT-induced biological modifications can cause liver metastases necrotic involution, avoiding the need of a subsequent therapeutic approach [31]. Theoretically, a similar outcome could be pursued with neoadjuvant chemotherapy, as described by Squires et al. [35], who reported promising results combining neoadjuvant capecitabine and temozolomide in PanNET. Moreover, recent evidence suggests that capecitabine may act as a radiosensitizer in synergy with RLT, increasing the efficacy of this approach in aggressive or radioresistant NETs [36]. Therefore, these results suggest that a combined approach with RLT and chemotherapy in neoadjuvant settings could be effective. Indeed, Barber et al. [28] achieved functional/radiological and symptomatic response over 12–42 month combining neoadjuvant RLT and chemotherapy (5-Fluorouracil). However, further prospective studies, comparing and combining neoadjuvant RLT and neoadjuvant chemotherapy approaches in patients with systemic disease at diagnosis are needed to explore this therapeutic option. A multidisciplinary approach is always preferable for selecting the best treatment and the correct timing.

Safety and reproducibility of neoadjuvant RLT

Among available studies, only one reported that a single patient was withdraw from neoadjuvant RLT before completing the treatment, due to severe hematologic toxicity [16]. Mild and transient hematologic toxicity was reported in other studies [21, 23, 24, 27, 29], in line with the most updated clinical and dosimetric evidence [3739].
The available results derive from studies with heterogeneous neoadjuvant RLT schemes. Indeed, different isotopes have been employed: 90Y in 3 studies [21, 25, 34], 177Lu in 6 [2224, 26, 28, 29] and both radioisotopes in 2 [16, 27], always as mono-therapy. It should be noted that, overall, most of the patients (141/148) were treated with [177Lu]-labeled radiopeptides. On the other hand, very different schemes were employed concerning cycles number and injected activity. Therefore, a better standardization of neoadjuvant RLT is desirable and should be investigated in future trials. Although RLT is currently mainly performed with [177Lu]-based radiopharmaceuticals, [90Y]-radiolabeled peptides might still play an important role. Indeed, the greater tissue penetrance of the beta radiations emitted by 90Y, compared to 177Lu, enhances the cross-fire effect that damages even those cellular clones with lower SSTR expression and potentially more undifferentiated. This feature, combined with the higher linear energy transfer (LET), candidates [90Y]-radiopeptides as potentially more effective in the treatment of large and biologically aggressive lesions [8, 40]. On the other hand, [177Lu]-based RLT has proven to be safer than 90Y. 177Lu could be more effective on small lesions, also in relation to its longer half-life, allowing a prolonged dose delivery [41]. Baum et al. [8] reported greater benefits in terms of objective response and survival in a large series of patients affected by NET (including PanNETs) who received combination treatments with [177Lu]-DOTATOC and [90Y]-DOTATOC, but not in neoadjuvant settings. A dual-isotope approach could allow a better risk-benefit ratio in neoadjuvant setting in PanNETs, representing a challenge for planning future multicentre randomized trials in this subset of patients.

Ongoing trials

On the basis of available evidence, a phase II single-arm multicentric trial on neoadjuvant RLT with [177Lu]-DOTATATE, followed by surgery for PanNETs, (NEOLUPANET, EUDRACT 20019-002196-34 – Clinical Trials Identifier NCT04385992) is ongoing [42]. The main purposes of the study are to evaluate safety and efficacy of neoadjuvant RLT followed by surgery in resectable PanNETs at high-risk of recurrence. The secondary endpoints are to investigate the rate of objective radiological response to RLT according to modified RECIST criteria (mRECIST) and patients QoL. The results of this study are not available, yet.
Further ongoing trials proposing neoadjuvant RLT for PanNETS or for NETs of different primary origin are not available at present. Likewise, other trials aiming to compare the efficacy of different RLT schemes in this subset of patients are lacking.

Limits of the study

The present mini-review summarizes the available data in the literature. However, most of the studies on neoadjuvant RLT are single case reports or small sample size studies [16, 2129, 34]. Therefore, the quality of the evidence is quite low. On the other hand, prospective randomized clinical trials are difficult to plan for these patients.

Conclusions

RLT could represent a safe and effective neoadjuvant therapy in unresectable or border line resectable PanNETs. Further prospective controlled investigations are necessary to confirm these preliminary data and to define the ideal therapeutic scheme. In addition, the multidisciplinary approach is preferable to identify patients who could benefit from neoadjuvant RLT to become eligible for second-line surgery.

Acknowledgements

This work has been realized in collaboration with Laboratorio delle Tecnologie Avanzate (LTTA) at the University of Ferrara, Italy. We would like to thank all the Colleagues that participate in the Interdisciplinary group on Neuroendocrine Neoplasms at the Azienda Ospedaliero Univerasitaria di Ferrara.

Compliance with ethical standards

Conflict of interest

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat M. Falconi, B. Eriksson, G. Kaltsas, D.K. Bartsch, J. Capdevila, M. Caplin, B. Kos-Kudla, D. Kwekkeboom, G. Rindi, G. KlÖppel, N. Reed, R. Kianmanesh, R.T. Jensen, ENETS consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology 153–171 (2016). https://doi.org/10.1159/000443171 M. Falconi, B. Eriksson, G. Kaltsas, D.K. Bartsch, J. Capdevila, M. Caplin, B. Kos-Kudla, D. Kwekkeboom, G. Rindi, G. KlÖppel, N. Reed, R. Kianmanesh, R.T. Jensen, ENETS consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology 153–171 (2016). https://​doi.​org/​10.​1159/​000443171
8.
Zurück zum Zitat R.P. Baum, H.R. Kulkarni, A. Singh, D. Kaemmerer, D. Mueller, V. Prasad, M. Hommann, F.C. Robiller, K. Niepsch, H. Franz, A. Jochems, P. Lambin, D. Hörsch, Results and adverse events of personalized peptide receptor radionuclide therapy with 90 Yttrium and 177 Lutetium in 1048 patients with neuroendocrine neoplasms. Oncotarget 9, 16932–16950 (2018) R.P. Baum, H.R. Kulkarni, A. Singh, D. Kaemmerer, D. Mueller, V. Prasad, M. Hommann, F.C. Robiller, K. Niepsch, H. Franz, A. Jochems, P. Lambin, D. Hörsch, Results and adverse events of personalized peptide receptor radionuclide therapy with 90 Yttrium and 177 Lutetium in 1048 patients with neuroendocrine neoplasms. Oncotarget 9, 16932–16950 (2018)
10.
Zurück zum Zitat J. Strosberg, G. El-Haddad, E. Wolin, A. Hendifar, J. Yao, B. Chasen, E. Mittra, P.L. Kunz, M.H. Kulke, H. Jacene, D. Bushnell, T.M. O’Dorisio, R.P. Baum, H.R. Kulkarni, M. Caplin, R. Lebtahi, T. Hobday, E. Delpassand, E. Van Cutsem, A. Benson, R. Srirajaskanthan, M. Pavel, J. Mora, J. Berlin, E. Grande, N. Reed, E. Seregni, K. Öberg, M. Lopera Sierra, P. Santoro, T. Thevenet, J.L. Erion, P. Ruszniewski, D. Kwekkeboom, E. Krenning, Phase 3 trial of 177 lu-dotatate for midgut neuroendocrine tumors. N. Engl. J. Med. 376, 125–135 (2017). https://doi.org/10.1056/nejmoa1607427CrossRefPubMedPubMedCentral J. Strosberg, G. El-Haddad, E. Wolin, A. Hendifar, J. Yao, B. Chasen, E. Mittra, P.L. Kunz, M.H. Kulke, H. Jacene, D. Bushnell, T.M. O’Dorisio, R.P. Baum, H.R. Kulkarni, M. Caplin, R. Lebtahi, T. Hobday, E. Delpassand, E. Van Cutsem, A. Benson, R. Srirajaskanthan, M. Pavel, J. Mora, J. Berlin, E. Grande, N. Reed, E. Seregni, K. Öberg, M. Lopera Sierra, P. Santoro, T. Thevenet, J.L. Erion, P. Ruszniewski, D. Kwekkeboom, E. Krenning, Phase 3 trial of 177 lu-dotatate for midgut neuroendocrine tumors. N. Engl. J. Med. 376, 125–135 (2017). https://​doi.​org/​10.​1056/​nejmoa1607427CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat T. Brabander, W.A. Van Der Zwan, J.J.M. Teunissen, B.L.R. Kam, R.A. Feelders, W.W. De Herder, C.H.J. Van Eijck, G.J.H. Franssen, E.P. Krenning, D.J. Kwekkeboom, Long-term efficacy, survival, and safety of [177Lu-DOTA0,Tyr3]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin. Cancer Res. 23, 4617–4624 (2017). https://doi.org/10.1158/1078-0432.CCR-16-2743 T. Brabander, W.A. Van Der Zwan, J.J.M. Teunissen, B.L.R. Kam, R.A. Feelders, W.W. De Herder, C.H.J. Van Eijck, G.J.H. Franssen, E.P. Krenning, D.J. Kwekkeboom, Long-term efficacy, survival, and safety of [177Lu-DOTA0,Tyr3]octreotate in patients with gastroenteropancreatic and bronchial neuroendocrine tumors. Clin. Cancer Res. 23, 4617–4624 (2017). https://​doi.​org/​10.​1158/​1078-0432.​CCR-16-2743
13.
14.
Zurück zum Zitat G. Paganelli, M. Sansovini, S. Nicolini, I. Grassi, T. Ibrahim, E. Amadori, V. Di Iorio, M. Monti, E. Scarpi, A. Bongiovanni, M. Altini, L. Urso, C. Cittanti, F. Matteucci, S. Severi, 177Lu-PRRT in advanced gastrointestinal neuroendocrine tumors: 10-year follow-up of the IRST phase II prospective study, Eur. J. Nucl. Med. Mol. Imaging (2020). https://doi.org/10.1007/s00259-020-04873-0 G. Paganelli, M. Sansovini, S. Nicolini, I. Grassi, T. Ibrahim, E. Amadori, V. Di Iorio, M. Monti, E. Scarpi, A. Bongiovanni, M. Altini, L. Urso, C. Cittanti, F. Matteucci, S. Severi, 177Lu-PRRT in advanced gastrointestinal neuroendocrine tumors: 10-year follow-up of the IRST phase II prospective study, Eur. J. Nucl. Med. Mol. Imaging (2020). https://​doi.​org/​10.​1007/​s00259-020-04873-0
15.
Zurück zum Zitat D. Campana, G. Capurso, S. Partelli, F. Nori, F. Panzuto, D. Tamburrino, G. Cacciari, G. Delle Fave, M. Falconi, P. Tomassetti, Radiolabelled somatostatin analogue treatment in gastroenteropancreatic neuroendocrine tumours: Factors associated with response and suggestions for therapeutic sequence. Eur. J. Nucl. Med. Mol. Imaging 40, 1197–1205 (2013). https://doi.org/10.1007/s00259-013-2402-2CrossRefPubMed D. Campana, G. Capurso, S. Partelli, F. Nori, F. Panzuto, D. Tamburrino, G. Cacciari, G. Delle Fave, M. Falconi, P. Tomassetti, Radiolabelled somatostatin analogue treatment in gastroenteropancreatic neuroendocrine tumours: Factors associated with response and suggestions for therapeutic sequence. Eur. J. Nucl. Med. Mol. Imaging 40, 1197–1205 (2013). https://​doi.​org/​10.​1007/​s00259-013-2402-2CrossRefPubMed
16.
Zurück zum Zitat S. Partelli, E. Bertani, M. Bartolomei, C. Perali, F. Muffatti, C.M. Grana, M. Schiavo Lena, C. Doglioni, S. Crippa, N. Fazio, G. Zamboni, M. Falconi, Peptide receptor radionuclide therapy as neoadjuvant therapy for resectable or potentially resectable pancreatic neuroendocrine neoplasms. Surgery 163, 761–767 (2018). https://doi.org/10.1016/j.surg.2017.11.007CrossRefPubMed S. Partelli, E. Bertani, M. Bartolomei, C. Perali, F. Muffatti, C.M. Grana, M. Schiavo Lena, C. Doglioni, S. Crippa, N. Fazio, G. Zamboni, M. Falconi, Peptide receptor radionuclide therapy as neoadjuvant therapy for resectable or potentially resectable pancreatic neuroendocrine neoplasms. Surgery 163, 761–767 (2018). https://​doi.​org/​10.​1016/​j.​surg.​2017.​11.​007CrossRefPubMed
18.
Zurück zum Zitat C. Ricci, S. Pusceddu, N. Prinzi, S. Tafuto, T. Ibrahim, A. Filice, M.P. Brizzi, F. Panzuto, S. Baldari, C.M. Grana, D. Campana, M.V. Davì, D. Giuffrida, M.C. Zatelli, S. Partelli, P. Razzore, R. Marconcini, S. Massironi, F. Gelsomino, A. Faggiano, E. Giannetta, E. Bajetta, F. Grimaldi, M. Cives, F. Cirillo, V. Perfetti, F. Corti, C. Ricci, L. Giacomelli, L. Porcu, M. Di Maio, E. Seregni, M. Maccauro, S. Lastoria, A. Bongiovanni, A. Versari, I. Persano, M. Rinzivillo, S.A. Pignata, P.A. Rocca, G. Lamberti, S. Cingarlini, I. Puliafito, M.R. Ambrosio, I. Zanata, A. Bracigliano, S. Severi, F. Spada, V. Andreasi, R. Modica, F. Scalorbi, M. Milione, G. Sabella, J. Coppa, R. Casadei, M. Di Bartolomeo, M. Falconi, F. De Braud, Association of upfront peptide receptor radionuclide therapy with progression-free survival among patients with enteropancreatic neuroendocrine tumors. JAMA Netw. Open 5, e220290–e220290 (2022). https://doi.org/10.1001/JAMANETWORKOPEN.2022.0290CrossRefPubMedPubMedCentral C. Ricci, S. Pusceddu, N. Prinzi, S. Tafuto, T. Ibrahim, A. Filice, M.P. Brizzi, F. Panzuto, S. Baldari, C.M. Grana, D. Campana, M.V. Davì, D. Giuffrida, M.C. Zatelli, S. Partelli, P. Razzore, R. Marconcini, S. Massironi, F. Gelsomino, A. Faggiano, E. Giannetta, E. Bajetta, F. Grimaldi, M. Cives, F. Cirillo, V. Perfetti, F. Corti, C. Ricci, L. Giacomelli, L. Porcu, M. Di Maio, E. Seregni, M. Maccauro, S. Lastoria, A. Bongiovanni, A. Versari, I. Persano, M. Rinzivillo, S.A. Pignata, P.A. Rocca, G. Lamberti, S. Cingarlini, I. Puliafito, M.R. Ambrosio, I. Zanata, A. Bracigliano, S. Severi, F. Spada, V. Andreasi, R. Modica, F. Scalorbi, M. Milione, G. Sabella, J. Coppa, R. Casadei, M. Di Bartolomeo, M. Falconi, F. De Braud, Association of upfront peptide receptor radionuclide therapy with progression-free survival among patients with enteropancreatic neuroendocrine tumors. JAMA Netw. Open 5, e220290–e220290 (2022). https://​doi.​org/​10.​1001/​JAMANETWORKOPEN.​2022.​0290CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat E. Giannetta, F. Sesti, R. Modica, E.M. Grossrubatscher, V. Guarnotta, A. Ragni, I. Zanata, A. Colao, A. Faggiano, Case report: unmasking hypercalcemia in patients with neuroendocrine neoplasms. Experience from six Italian referral centers. Front. Endocrinol. (Lausanne). 12, 1 (2021). https://doi.org/10.3389/FENDO.2021.665698CrossRef E. Giannetta, F. Sesti, R. Modica, E.M. Grossrubatscher, V. Guarnotta, A. Ragni, I. Zanata, A. Colao, A. Faggiano, Case report: unmasking hypercalcemia in patients with neuroendocrine neoplasms. Experience from six Italian referral centers. Front. Endocrinol. (Lausanne). 12, 1 (2021). https://​doi.​org/​10.​3389/​FENDO.​2021.​665698CrossRef
20.
Zurück zum Zitat G. Puliani, V. Di Vito, T. Feola, F. Sesti, R. Centello, C. Pandozzi, M.G. Tarsitano, M. Verrico, A. Lenzi, A.M. Isidori, E. Giannetta, A. Faggiano, NETest: a systematic review focusing on the prognostic and predictive role. Neuroendocrinology 112, 523–536 (2022). https://doi.org/10.1159/000518873CrossRefPubMed G. Puliani, V. Di Vito, T. Feola, F. Sesti, R. Centello, C. Pandozzi, M.G. Tarsitano, M. Verrico, A. Lenzi, A.M. Isidori, E. Giannetta, A. Faggiano, NETest: a systematic review focusing on the prognostic and predictive role. Neuroendocrinology 112, 523–536 (2022). https://​doi.​org/​10.​1159/​000518873CrossRefPubMed
22.
Zurück zum Zitat S. Ezziddin, H. Lauschke, M. Schaefers, C. Meyer, M. Van Essen, H.J. Biersack, D.J. Kwekkeboom, H. Ahmadzadehfar, Neoadjuvant downsizing by internal radiation: a case for preoperative peptide receptor radionuclide therapy in patients with pancreatic neuroendocrine tumors. Clin. Nucl. Med. 37, 102–104 (2012). https://doi.org/10.1097/RLU.0B013E318238F111CrossRefPubMed S. Ezziddin, H. Lauschke, M. Schaefers, C. Meyer, M. Van Essen, H.J. Biersack, D.J. Kwekkeboom, H. Ahmadzadehfar, Neoadjuvant downsizing by internal radiation: a case for preoperative peptide receptor radionuclide therapy in patients with pancreatic neuroendocrine tumors. Clin. Nucl. Med. 37, 102–104 (2012). https://​doi.​org/​10.​1097/​RLU.​0B013E318238F111​CrossRefPubMed
23.
Zurück zum Zitat E.I. van Vliet, C.H. van Eijck, R.R. de Krijger, E.J.N. van Dijkum, J.J. Teunissen, B.L. Kam, W.Wde. Herder, R.A. Feelders, B.A. Bonsing, T. Brabander, E.P. Krenning, D.J. Kwekkeboom, Neoadjuvant treatment of nonfunctioning pancreatic neuroendocrine tumors with [177Lu-DOTA0,Tyr3] octreotate. J. Nucl. Med. 56, 1647–1653 (2015). https://doi.org/10.2967/JNUMED.115.158899 E.I. van Vliet, C.H. van Eijck, R.R. de Krijger, E.J.N. van Dijkum, J.J. Teunissen, B.L. Kam, W.Wde. Herder, R.A. Feelders, B.A. Bonsing, T. Brabander, E.P. Krenning, D.J. Kwekkeboom, Neoadjuvant treatment of nonfunctioning pancreatic neuroendocrine tumors with [177Lu-DOTA0,Tyr3] octreotate. J. Nucl. Med. 56, 1647–1653 (2015). https://​doi.​org/​10.​2967/​JNUMED.​115.​158899
24.
Zurück zum Zitat T.N. da Silva, M.L.F. van Velthuysen, C.H.J. van Eijck, J.J. Teunissen, J. Hofland, W.W. de Herder, Successful neoadjuvant peptide receptor radionuclide therapy for an inoperable pancreatic neuroendocrine tumour. Endocrinol. Diabetes Metab. Case Rep. 2018 (2018). https://doi.org/10.1530/edm-18-0015 T.N. da Silva, M.L.F. van Velthuysen, C.H.J. van Eijck, J.J. Teunissen, J. Hofland, W.W. de Herder, Successful neoadjuvant peptide receptor radionuclide therapy for an inoperable pancreatic neuroendocrine tumour. Endocrinol. Diabetes Metab. Case Rep. 2018 (2018). https://​doi.​org/​10.​1530/​edm-18-0015
29.
Zurück zum Zitat N.S. Minczeles, C.H.J. van Eijck, M.J. van Gils, M.L.F. van Velthuysen, E.J.M. Nieveen van Dijkum, R.A. Feelders, W.W. de Herder, T. Brabander, J. Hofland. Induction therapy with 177Lu-DOTATATE procures long-term survival in locally advanced or oligometastatic pancreatic neuroendocrine neoplasm patients. Eur. J. Nucl. Med. Mol. Imaging 1–12 (2022). https://doi.org/10.1007/S00259-022-05734-8/TABLES/5 N.S. Minczeles, C.H.J. van Eijck, M.J. van Gils, M.L.F. van Velthuysen, E.J.M. Nieveen van Dijkum, R.A. Feelders, W.W. de Herder, T. Brabander, J. Hofland. Induction therapy with 177Lu-DOTATATE procures long-term survival in locally advanced or oligometastatic pancreatic neuroendocrine neoplasm patients. Eur. J. Nucl. Med. Mol. Imaging 1–12 (2022). https://​doi.​org/​10.​1007/​S00259-022-05734-8/​TABLES/​5
30.
Zurück zum Zitat J. Strosberg, P.L. Kunz, A. Hendifar, J. Yao, D. Bushnell, M.H. Kulke, R.P. Baum, M. Caplin, P. Ruszniewski, E. Delpassand, T. Hobday, C. Verslype, A. Benson, R. Srirajaskanthan, M. Pavel, J. Mora, J. Berlin, E. Grande, N. Reed, E. Seregni, G. Paganelli, S. Severi, M. Morse, D.C. Metz, C. Ansquer, F. Courbon, A. Al-Nahhas, E. Baudin, F. Giammarile, D. Taïeb, E. Mittra, E. Wolin, T.M. O’Dorisio, R. Lebtahi, C.M. Deroose, C.M. Grana, L. Bodei, K. Öberg, B.D. Polack, B. He, M.F. Mariani, G. Gericke, P. Santoro, J.L. Erion, L. Ravasi, E. Krenning, Impact of liver tumour burden, alkaline phosphatase elevation, and target lesion size on treatment outcomes with 177Lu-Dotatate: an analysis of the NETTER-1 study. Eur. J. Nucl. Med. Mol. Imaging 47, 2372–2382 (2020). https://doi.org/10.1007/s00259-020-04709-xCrossRefPubMedPubMedCentral J. Strosberg, P.L. Kunz, A. Hendifar, J. Yao, D. Bushnell, M.H. Kulke, R.P. Baum, M. Caplin, P. Ruszniewski, E. Delpassand, T. Hobday, C. Verslype, A. Benson, R. Srirajaskanthan, M. Pavel, J. Mora, J. Berlin, E. Grande, N. Reed, E. Seregni, G. Paganelli, S. Severi, M. Morse, D.C. Metz, C. Ansquer, F. Courbon, A. Al-Nahhas, E. Baudin, F. Giammarile, D. Taïeb, E. Mittra, E. Wolin, T.M. O’Dorisio, R. Lebtahi, C.M. Deroose, C.M. Grana, L. Bodei, K. Öberg, B.D. Polack, B. He, M.F. Mariani, G. Gericke, P. Santoro, J.L. Erion, L. Ravasi, E. Krenning, Impact of liver tumour burden, alkaline phosphatase elevation, and target lesion size on treatment outcomes with 177Lu-Dotatate: an analysis of the NETTER-1 study. Eur. J. Nucl. Med. Mol. Imaging 47, 2372–2382 (2020). https://​doi.​org/​10.​1007/​s00259-020-04709-xCrossRefPubMedPubMedCentral
31.
Zurück zum Zitat M. Schiavo Lena, S. Partelli, P. Castelli, V. Andreasi, C.E. Smart, E. Pisa, M. Bartolomei, E. Bertani, G. Zamboni, M. Falconi, C. Doglioni, Histopathological and immunophenotypic changes of pancreatic neuroendocrine tumors after neoadjuvant peptide receptor radionuclide therapy (PRRT). Endocr. Pathol. 31, 119–131 (2020). https://doi.org/10.1007/s12022-020-09623-4 M. Schiavo Lena, S. Partelli, P. Castelli, V. Andreasi, C.E. Smart, E. Pisa, M. Bartolomei, E. Bertani, G. Zamboni, M. Falconi, C. Doglioni, Histopathological and immunophenotypic changes of pancreatic neuroendocrine tumors after neoadjuvant peptide receptor radionuclide therapy (PRRT). Endocr. Pathol. 31, 119–131 (2020). https://​doi.​org/​10.​1007/​s12022-020-09623-4
32.
Zurück zum Zitat F. Panzuto, S. Puscedddu, A. Faggiano, M. Rinzivillo, N. Brighi, N. Prinzi, F. Riccardi, E. Iannicelli, I. Maggio, D. Femia, S. Tafuto, L. Manuzzi, A. Di Sarno, B. Annibale, F. de Braud, D. Campana, Prognostic impact of tumour burden in stage IV neuroendocrine neoplasia: a comparison between pancreatic and gastrointestinal localizations. Pancreatology 19, 1067–1073 (2019). https://doi.org/10.1016/j.pan.2019.09.015CrossRefPubMed F. Panzuto, S. Puscedddu, A. Faggiano, M. Rinzivillo, N. Brighi, N. Prinzi, F. Riccardi, E. Iannicelli, I. Maggio, D. Femia, S. Tafuto, L. Manuzzi, A. Di Sarno, B. Annibale, F. de Braud, D. Campana, Prognostic impact of tumour burden in stage IV neuroendocrine neoplasia: a comparison between pancreatic and gastrointestinal localizations. Pancreatology 19, 1067–1073 (2019). https://​doi.​org/​10.​1016/​j.​pan.​2019.​09.​015CrossRefPubMed
33.
Zurück zum Zitat J.R. Howe, N.B. Merchant, C. Conrad, X.M. Keutgen, J. Hallet, J.A. Drebin, R.M. Minter, T.C. Lairmore, J.F. Tseng, H.J. Zeh, S.K. Libutti, G. Singh, J.E. Lee, T.A. Hope, M.K. Kim, Y. Menda, T.R. Halfdanarson, J.A. Chan, R.F. Pommier, The North American neuroendocrine tumor society consensus paper on the surgical management of pancreatic neuroendocrine tumors. Pancreas 49, 1–33 (2020). https://doi.org/10.1097/MPA.0000000000001454CrossRefPubMedPubMedCentral J.R. Howe, N.B. Merchant, C. Conrad, X.M. Keutgen, J. Hallet, J.A. Drebin, R.M. Minter, T.C. Lairmore, J.F. Tseng, H.J. Zeh, S.K. Libutti, G. Singh, J.E. Lee, T.A. Hope, M.K. Kim, Y. Menda, T.R. Halfdanarson, J.A. Chan, R.F. Pommier, The North American neuroendocrine tumor society consensus paper on the surgical management of pancreatic neuroendocrine tumors. Pancreas 49, 1–33 (2020). https://​doi.​org/​10.​1097/​MPA.​0000000000001454​CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat O. Stoeltzing, E. Huber, M. Loss, V. Gross, C. Eilles, J. Mueller-Brand, H.J. Schlitt, Staged surgery with neoadjuvant 90Y-DOTATOC therapy for down-sizing synchronous bilobular hepatic metastases from a neuroendocrine pancreatic tumor. Langenbeck’s Arch. Surg. 395, 185–192 (2010). https://doi.org/10.1007/s00423-009-0520-xCrossRef O. Stoeltzing, E. Huber, M. Loss, V. Gross, C. Eilles, J. Mueller-Brand, H.J. Schlitt, Staged surgery with neoadjuvant 90Y-DOTATOC therapy for down-sizing synchronous bilobular hepatic metastases from a neuroendocrine pancreatic tumor. Langenbeck’s Arch. Surg. 395, 185–192 (2010). https://​doi.​org/​10.​1007/​s00423-009-0520-xCrossRef
35.
Zurück zum Zitat M.H. Squires, P.J. Worth, B. Konda, M.H. Shah, M.E. Dillhoff, S. Abdel-Misih, J.A. Norton, B.C. Visser, M. Dua, T.M. Pawlik, C.R. Schmidt, G. Poultsides, J.M. Cloyd, Neoadjuvant capecitabine/temozolomide for locally advanced or metastatic pancreatic neuroendocrine tumors. Pancreas 355–360 (2020). https://doi.org/10.1097/MPA.0000000000001500 M.H. Squires, P.J. Worth, B. Konda, M.H. Shah, M.E. Dillhoff, S. Abdel-Misih, J.A. Norton, B.C. Visser, M. Dua, T.M. Pawlik, C.R. Schmidt, G. Poultsides, J.M. Cloyd, Neoadjuvant capecitabine/temozolomide for locally advanced or metastatic pancreatic neuroendocrine tumors. Pancreas 355–360 (2020). https://​doi.​org/​10.​1097/​MPA.​0000000000001500​
36.
Zurück zum Zitat S. Nicolini, L. Bodei, A. Bongiovanni, M. Sansovini, I. Grassi, T. Ibrahim, M. Monti, P. Caroli, A. Sarnelli, D. Diano, V. Di Iorio, C.M. Grana, C. Cittanti, F. Pieri, S. Severi, G. Paganelli, Combined use of 177Lu-DOTATATE and metronomic capecitabine (Lu-X) in FDG-positive gastro-entero-pancreatic neuroendocrine tumors. Eur. J. Nucl. Med. Mol. Imaging (2021). https://doi.org/10.1007/s00259-021-05236-z/Published S. Nicolini, L. Bodei, A. Bongiovanni, M. Sansovini, I. Grassi, T. Ibrahim, M. Monti, P. Caroli, A. Sarnelli, D. Diano, V. Di Iorio, C.M. Grana, C. Cittanti, F. Pieri, S. Severi, G. Paganelli, Combined use of 177Lu-DOTATATE and metronomic capecitabine (Lu-X) in FDG-positive gastro-entero-pancreatic neuroendocrine tumors. Eur. J. Nucl. Med. Mol. Imaging (2021). https://​doi.​org/​10.​1007/​s00259-021-05236-z/​Published
37.
Zurück zum Zitat J.R. Strosberg, M.E. Caplin, P.L. Kunz, P.B. Ruszniewski, L. Bodei, A. Hendifar, E. Mittra, E.M. Wolin, J.C. Yao, M.E. Pavel, E. Grande, E. Van Cutsem, E. Seregni, H. Duarte, G. Gericke, A. Bartalotta, M.F. Mariani, A. Demange, S. Mutevelic, E.P. Krenning, 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. 22, 1752–1763 (2021). https://doi.org/10.1016/S1470-2045(21)00572-6CrossRefPubMed J.R. Strosberg, M.E. Caplin, P.L. Kunz, P.B. Ruszniewski, L. Bodei, A. Hendifar, E. Mittra, E.M. Wolin, J.C. Yao, M.E. Pavel, E. Grande, E. Van Cutsem, E. Seregni, H. Duarte, G. Gericke, A. Bartalotta, M.F. Mariani, A. Demange, S. Mutevelic, E.P. Krenning, 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. 22, 1752–1763 (2021). https://​doi.​org/​10.​1016/​S1470-2045(21)00572-6CrossRefPubMed
38.
Zurück zum Zitat K. Sjögreen Gleisner, N. Chouin, P.M. Gabina, F. Cicone, S. Gnesin, C. Stokke, M. Konijnenberg, M. Cremonesi, F.A. Verburg, P. Bernhardt, U. Eberlein, J. Gear, EANM dosimetry committee recommendations for dosimetry of 177Lu-labelled somatostatin-receptor- and PSMA-targeting ligands. Eur. J. Nucl. Med. Mol. Imaging (2022). https://doi.org/10.1007/s00259-022-05727-7 K. Sjögreen Gleisner, N. Chouin, P.M. Gabina, F. Cicone, S. Gnesin, C. Stokke, M. Konijnenberg, M. Cremonesi, F.A. Verburg, P. Bernhardt, U. Eberlein, J. Gear, EANM dosimetry committee recommendations for dosimetry of 177Lu-labelled somatostatin-receptor- and PSMA-targeting ligands. Eur. J. Nucl. Med. Mol. Imaging (2022). https://​doi.​org/​10.​1007/​s00259-022-05727-7
39.
Zurück zum Zitat U. Garske-Román, M. Sandström, K. Fröss Baron, L. Lundin, P. Hellman, S. Welin, S. Johansson, T. Khan, H. Lundqvist, B. Eriksson, A. Sundin, D. Granberg, Prospective observational study of 177Lu-DOTA-octreotate therapy in 200 patients with advanced metastasized neuroendocrine tumours (NETs): feasibility and impact of a dosimetry-guided study protocol on outcome and toxicity. Eur. J. Nucl. Med. Mol. Imaging 45, 970–988 (2018). https://doi.org/10.1007/s00259-018-3945-zCrossRefPubMedPubMedCentral U. Garske-Román, M. Sandström, K. Fröss Baron, L. Lundin, P. Hellman, S. Welin, S. Johansson, T. Khan, H. Lundqvist, B. Eriksson, A. Sundin, D. Granberg, Prospective observational study of 177Lu-DOTA-octreotate therapy in 200 patients with advanced metastasized neuroendocrine tumours (NETs): feasibility and impact of a dosimetry-guided study protocol on outcome and toxicity. Eur. J. Nucl. Med. Mol. Imaging 45, 970–988 (2018). https://​doi.​org/​10.​1007/​s00259-018-3945-zCrossRefPubMedPubMedCentral
Metadaten
Titel
Radioligand therapy (RLT) as neoadjuvant treatment for inoperable pancreatic neuroendocrine tumors: a literature review
verfasst von
Luca Urso
Alberto Nieri
Ilaria Rambaldi
Angelo Castello
Licia Uccelli
Corrado Cittanti
Stefano Panareo
Irene Gagliardi
Maria Rosaria Ambrosio
Maria Chiara Zatelli
Mirco Bartolomei
Publikationsdatum
26.08.2022
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 2/2022
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-022-03170-0

Weitere Artikel der Ausgabe 2/2022

Endocrine 2/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Klimaschutz beginnt bei der Wahl des Inhalators

14.05.2024 Klimawandel Podcast

Auch kleine Entscheidungen im Alltag einer Praxis können einen großen Beitrag zum Klimaschutz leisten. Die neue Leitlinie zur "klimabewussten Verordnung von Inhalativa" geht mit gutem Beispiel voran, denn der Wechsel vom klimaschädlichen Dosieraerosol zum Pulverinhalator spart viele Tonnen CO2. Leitlinienautor PD Dr. Guido Schmiemann erklärt, warum nicht nur die Umwelt, sondern auch Patientinnen und Patienten davon profitieren.

Zeitschrift für Allgemeinmedizin, DEGAM

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.