You are on page 1of 10

At the Cutting Edge

Neuroendocrinology 2021;111:207–216 Received: March 21, 2020


Accepted: May 13, 2020
DOI: 10.1159/000508633 Published online: May 13, 2020

Bone Metastases in Neuroendocrine


Tumors: Molecular Pathogenesis and
Implications in Clinical Practice
Mauro Cives a Eleonora Pellè a Maria Rinzivillo b Daniela Prosperi c
       

Marco Tucci a Franco Silvestris a Francesco Panzuto b


     

a Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy; b Digestive  

Disease Unit, Sant’ Andrea University Hospital, ENETS Center of Excellence, Rome, Italy; c Nuclear Medicine Unit,
 

Sant’ Andrea University Hospital, ENETS Center of Excellence, Rome, Italy

Keywords ploitation of timely treatments, the management of bone-


Carcinoid tumors · Skeleton-related events · colonizing NETs is today based only on clinical experience
Bisphosphonates · Denosumab · CXCR4 from other osteotropic tumors. Here, we summarize the fun-
damental molecular mechanisms driving bone colonization
and revisit both established and novel treatments for pa-
Abstract tients with bone metastatic NETs. © 2020 S. Karger AG, Basel
Skeletal colonization is often regarded as a rare event in pa-
tients with neuroendocrine tumors (NETs) although both na-
tional registries and retrospective series report an incidence
of bone metastases as high as 20% in subjects with advanced Introduction
disease. While the biological mechanisms leading to bone
metastatic colonization in NETs have been poorly investigat- Neuroendocrine tumors (NETs) are heterogeneous
ed so far, key steps of osteotropic mechanisms, including the neoplasms arising in secretory cells of the diffuse neuro-
epithelial-to-mesenchymal transition, preparation of the endocrine system. They predominate in the gastroentero-
premetastatic niche, migration of circulating tumor cells to- pancreatic (GEP) tract and the bronchopulmonary (BP)
wards the bone marrow as well as the resulting alterations tree and are characterized by a relatively indolent rate of
of the skeletal metabolism, are likely to operate also during growth and the ability to release a variety of peptide hor-
the development of NET bone metastases. The skeleton in- mones and vasoactive substances [1]. The incidence of
volvement by NETs has a detrimental impact on both quality NETs has increased markedly over the last three decades,
of life and patients’ prognosis, leading to pain in the major- probably as a result of trends in imaging and improve-
ity of symptomatic subjects. While it is currently unclear ment in diagnosis, and the projected prevalence of NETs
whether or not the earlier recognition of bone involvement in the US population in 2014 was 171,321 [2]. The clinical
by PET/CT imaging techniques employing 68Ga-DOTA-con- aggressiveness of NETs widely varies in relation to their
jugated peptides might improve outcomes through the ex- primary site as well as the tumor grading. According to

karger@karger.com © 2020 S. Karger AG, Basel Mauro Cives


www.karger.com/nen Department of Internal Medicine and Clinical Oncology
University of Bari Aldo Moro, Piazza Giulio Cesare, 11
IT–70124 Bari (Italy)
mauro.civ @ tiscali.it
the 2019 World Health Organization (WHO) classifica- center study prospectively enrolling 668 patients with
tion, well-differentiated NETs are subdivided as either GEP-NET [6, 16]. Institutional series from tertiary aca-
G1, G2 or G3 tumors, whereas poorly differentiated car- demic centers in the USA reported an incidence of bone
cinomas are always considered G3 and have a dismal metastatic disease in approximately 12% of patients [8, 9],
prognosis [3]. consistent with a recent single-institution German study
NETs are frequently diagnosed at stage IV, with liver assessing 677 patients diagnosed with NET between 2000
and peritoneum being the most common sites of metas- and 2015 [7]. Notably, a 72% relative rise in the frequen-
tasis [2, 4–7]. Skeletal colonization is often regarded as a cy of bone metastasis detection has been reported in this
rare event in patients with NETs, and retrospective series study, where the incidence of skeletal involvement among
reported the incidence of bone metastases as high as 20% patients with the metastatic disease increased from 19 to
in patients with advanced disease [7–10]. However, the 33% in two distinct periods in which both Octreoscan and
clinical advent of PET/CT imaging techniques employing 68
Ga-DOTATOC PET/CT imaging techniques were ex-
68
Ga-DOTA-conjugated peptides has substantially in- clusively used [7]. Furthermore, in a recent study of 162
creased the frequency of NET bone metastasis detection, metastatic lung carcinoids, the bone involvement was ob-
thus allowing their earlier recognition [7]. As result, a served in 42% of cases [17], while a frequency of 38% was
progressive rise in the incidence of bone metastatic dis- reported in a single-institution series enrolling 108 pa-
ease is expected in patients with NETs in the next few tients with metastatic BP-NETs [14]. Overall, the inci-
years. Notably, multiple studies have depicted the bone dence of NET bone metastases appears to be higher in
colonization as a negative prognostic factor in NETs lung carcinoids as compared with GEP-NETs, probably
while skeleton-related events (SREs) including patholog- as result of their different venous drainage. Consistently,
ical fractures, spinal cord compression, need for orthope- rectal NETs appear to have the highest osteotropic poten-
dic surgery or radiotherapy to bone and/or hypercalce- tial in the context of GEP-NETs [4]. Epidemiological in-
mia, are reported to occur in approximately one fifth of consistencies among different retrospective studies may
patients with bone-colonizing NETs [7–14]. Despite their be related to referral patterns, while underreporting can-
clinical relevance, the management of NET bone metas- not be excluded in national registries. Evidence from a
tases is currently guided by limited evidence, and treat- small autopsy series [18], reporting a bone metastasis in-
ment patterns vary substantially among different institu- cidence of 42% in 36 subjects with carcinoid tumors, sup-
tions. ports the idea that the frequency of skeletal involvement
Here, we summarize the fundamental molecular might be clinically underestimated in patients with NETs.
mechanisms driving bone colonization in NETs and crit-
ically discuss the diagnostic and therapeutic opportuni-
ties currently available for NET patients with bone meta- Bone Metastases in NETs: Molecular Pathogenesis
static disease.
Bone metastatic colonization is a complex multistep
process including four distinct phases (Fig. 1). In the first
Epidemiology of Bone Metastases in NETs phase, cancer cells in the primary tumor site acquire the
ability to invade surrounding tissue and spread. In the
The incidence of bone metastases in patients with second phase, primary tumor cells influence the bone
NETs has been investigated only by retrospective series marrow stromal cells via soluble factors, thus preparing
so far. In an analysis of the Swedish Cancer Registry [4], the so-called premetastatic niche. In the third phase, can-
skeletal involvement has been identified in the 16% of cer cells migrate as circulating tumor cells (CTCs) to-
1,842 patients with metastatic NETs diagnosed between wards skeletal premetastatic niches under the pressure of
1987 and 2012, in the presence of a substantial male pre- a specific transcriptional rewiring or as result of cytokine
ponderance. In a study including 14,685 patients with gradients. In the fourth phase, tumor cells colonize the
gastrointestinal NET from the Surveillance, Epidemiol- skeleton and enter a state of dormancy, or aberrantly
ogy and End Results database, bone metastases were re- modulate bone resorption or deposition leading to the
corded in 6% of them with a significantly higher inci- development of lytic or sclerotic metastasis, respectively
dence in males [15]. A similar frequency of skeletal in- [19].
volvement was described in a study of the Spanish Similarly to other cancers, NET cells may acquire in-
National Cancer Registry, as well as in a French multi- vasive potential through the epithelial-to-mesenchymal

208 Neuroendocrinology 2021;111:207–216 Cives/Pellè/Rinzivillo/Prosperi/Tucci/


DOI: 10.1159/000508633 Silvestris/Panzuto
Fig. 1. The process of bone metastatic colonization proceeds Once settled in the metastatic niches, cancer cells may either enter
through multiple steps. Under the pressure of a supportive micro- a state of dormancy, as indolent micrometastasis, or proliferate
environment, tumor cells in the primary site undergo epithelial- and aberrantly modulate bone metabolism, leading to the develop-
to-mesenchymal transition as result of a complex transcriptional ment of lytic or sclerotic metastases (phase 4). CAFs, cancer-asso-
rewiring coordinated by transcription factors including snail, slug ciated fibroblasts; CXCR4, C-X-C chemokine receptor type 4;
or twist, among others (phase 1). Concomitantly, malignant cells CXCL12, CXCL10, CXCL16, C-X-C motif chemokine ligands 12,
prepare the so-called premetastatic niche through a variety of sol- 10, 16; RANK, receptor activator of nuclear factor-κB; RANKL,
uble factors, thus priming the nursing ability of bone marrow stro- receptor activator of nuclear factor-κB ligand; BMPs, bone mor-
mal cells to support the tumor (phase 2). After intravasation into phogenetic proteins; TGF-β2, transforming growth factor-β2;
the tumor vasculature, circulating tumor cells migrate towards GAS2, growth arrest-specific 2; TNF-α, tumor necrosis factor-α;
bone marrow niches as result of specific interactions between mar- PTHrP, parathyroid hormone-related protein; IL-11, interleu-
row-derived cytokines such as CXCL12 or RANKL and their cog- kin-11; IGF, insulin-like growth factor; FGF, fibroblast growth fac-
nate receptors expressed on the surface of cancer cells (phase 3). tor; VEGF, vascular endothelial growth factor.

transition (EMT), a transdifferentiation program confer- comes and the overexpression of EMT-related factors in-
ring mesenchymal plasticity to epithelial cells [20]. Mul- cluding snail, slug, twist and vimentin in tumor cells [21–
tiple studies have demonstrated that the EMT drives the 27]. In this context, bone-colonizing NETs have been
progression of both well- and poorly differentiated NETs, shown to harbor specific EMT-related features. In an
and that EMT-undergone tumors have a dismal progno- immunohistochemistry study of 44 samples of GEP- or
sis, as demonstrated by the association between poor out- BP-NETs, osteotropic tumors appeared to overexpress

Bone Metastases in NETs Neuroendocrinology 2021;111:207–216 209


DOI: 10.1159/000508633
C-X-C chemokine receptor type 4 (CXCR4) in the pres- During the process of metastatic colonization, tumor
ence of low levels of TGF-β1 as compared with localized cells are progressively shed into the systemic circulation
neoplasms [11]. In this regard, in vitro evidence suggests as CTCs. Based on available evidence, relatively low num-
that agonist stimulation of CXCR4 by its ligand CXCL12 bers of CTCs can be isolated in patients with either GEP-
is able per se to induce EMT in NET cell lines, enhancing or BP-NETs, and the presence of CTCs has been shown
their migratory and invasive capabilities towards bone to independently predict increased tumor burden and
tissue [28]. Intriguingly, the pharmacological suppres- poor survival outcomes [36–38]. In a study of 254 patients
sion of the CXCL12/CXCR4 axis by ulocuplumab, a with GEP-NETs, the presence of CTCs was significantly
CXCR4 targeting monoclonal antibody, has been prelim- associated with metastatic skeletal colonization, but not
inarily shown to inhibit EMT in pancreatic NET (pan- with lung, peritoneal or lymph node metastases [39]. Giv-
NET) cells, and to downmodulate their metastatic poten- en that cancer cells are able to remain as “niche-engaged”
tial both in vitro and in vivo [29]. dormant cells in the bone marrow for years before switch-
Intratumor hypoxia has been described as a major ing to a proliferative phenotype and eventually causing
driver of EMT in NETs. Gene expression profiling of the overt metastases, the identification of CTCs has been pro-
GOT1 carcinoid cell line revealed a marked upregulation posed as a useful tool for tailoring bone-oriented surveil-
of EMT-related genes under hypoxic conditions [30], and lance programs in patients with NETs.
the activation of EMT has been demonstrated in pan- Several factors are suspected to influence the bone tro-
NETs arising in the context of Von Hippel-Lindau dis- pism of CTCs in NETs. First, circulation patterns dictate
ease, a syndrome caused by germline mutations of VHL, the first capillary bed that CTCs will encounter, and
a critical regulator of the oxygen sensor HIF-1α [31]. therefore it is not surprising that lung carcinoids display
While it is well known that different components of the a higher incidence of skeletal metastases as compared
tumor microenvironment play a crucial role in enhancing with GEP-NETs, which are drained by the mesenteric
cancer invasiveness and metastatic spreading, little is un- stream. Second, specific chemokines including CXCL12,
derstood in this regard in NETs. Preliminary evidence CXCL10, CXCL16 and receptor activator of nuclear
from our group indicates that cancer-associated fibro- factor-κB ligand (RANKL) may attract CTCs towards the
blasts stimulate both EMT and in vitro migration of NET osteogenic niche. In this context, in an analysis of 40 pa-
cells, while tumor-infiltrating platelets, a key regulator of tients with metastatic NETs, the expression of CXCR4 on
EMT, have been recently demonstrated to predict poor the surface of CTCs was apparently associated with an
outcomes and early relapse in patients with resected pan- osteotropic behavior (p = 0.18) [39]. Third, definite pat-
NETs [32]. terns of transcriptional remodeling have been described
Systemic signals produced by the primary tumor can to drive the homing of CTCs towards the skeleton [40].
modulate the microenvironment of distant organs, creat- In this regard, preliminary evidence suggests that acquisi-
ing a pre-metastatic niche prior to the arrival of CTCs. tion of the EMT gene signature primes the skeletal colo-
Extensive research (reviewed in Peinado et al. [33]) has nization in NETs [29].
demonstrated that both inflammatory chemokines, pro- Once they have reached the bone metastatic niche, dis-
angiogenic cytokines, extracellular matrix-remodeling seminated tumor cells may either enter a quiescent state
enzymes as well as extracellular vesicles such as exosomes or evolve in overt metastases [40]. Little is known about
can induce in distant organs the formation of a microen- how NET cells enter the dormant state, what signals sus-
vironment conducive to the survival and outgrowth of tain it, what niches these dormant cells inhabit and, more
disseminated tumor cells, dictating selective tumor organ- importantly, what molecular cues trigger the resumption
otropism [34]. Although it is likely that the mechanisms of aggressive tumor growth eventually leading to overt
driving the osteotropism of NETs are similar to those op- metastasis formation. Bone morphogenetic proteins,
erating in other cancers, no studies have specifically ad- TGF-β2 and growth arrest-specific-6 protein have been
dressed the question of whether and how neuroendocrine reported to enforce the quiescence of disseminated tumor
primary tumors are able to prepare the bone premetastat- cells in a variety of malignancies [40], but their specific
ic niche. In this context, the peripheral trafficking of bone role in NETs is currently unknown. When the bone mi-
marrow-derived stem cells, a key component of the pre- croenvironment becomes suitable for cancer cell prolif-
metastatic niche, appears to be more limited in gastric eration, lytic or sclerotic bone metastases may arise. Os-
NETs as compared with gastric adenocarcinoma, consis- teolytic metastases result from an altered balance between
tent with their lower metastatic potential [35]. bone-resorbing osteoclasts and bone-generating osteo-

210 Neuroendocrinology 2021;111:207–216 Cives/Pellè/Rinzivillo/Prosperi/Tucci/


DOI: 10.1159/000508633 Silvestris/Panzuto
blasts. Cancer cell-derived parathyroid hormone-related been identified, a correlation with deteriorated perfor-
protein, interleukin-11 and tumor necrosis factor-α stim- mance status has been observed [10]. Of note, a similar
ulate osteoblasts to release RANKL, which promotes os- prevalence of bone lesions and their complications has
teoclast maturation and activation, leading to bone deg- been noted in patients with pancreatic versus gastrointes-
radation. In turn, growth factors physiologically stored in tinal NETs [9, 10].
the bone matrix are released during this process, reignit- Both conventional and functional imaging techniques
ing cancer cell proliferation and skeletal devastation in a are commonly used for the diagnosis of bone metastases in
vicious cycle. In sclerotic metastases, cancer cells produce NET patients. Although in the presence of contrasting evi-
osteoblast-stimulating factors including fibroblast growth dence [7, 8, 11, 13, 46], MRI appears to have a higher sensi-
factor, vascular endothelial growth factor and insulin-like tivity in identifying NET bone lesions as compared with CT,
growth factor, thus driving aberrant bone matrix deposi- and particularly whole-body MRI scans with diffusion-
tion [41]. In patients with NETs, sclerotic bone metasta- weighted imaging sequences seem to have high diagnostic
ses appear to be more prevalent as compared with lytic accuracy in this context (Fig. 2). The ability to detect skel-
ones [11], but the exact molecular mechanisms leading to etal metastases has dramatically increased since the intro-
their development are presently unknown. Similarly to duction of 68Ga-PET/CT, which clearly outperforms both
other bone-devastating malignancies, an imbalance of the bone scintigraphy and somatostatin receptor scintigraphy
RANKL/osteoprotegerin serum levels has been described (OctreoScan). Overall, the diagnostic accuracy of 68Ga-
in patients with NETs with bone metastatic disease as PET/CT in identifying NET bone lesions is comprised be-
compared with those without skeletal colonization, and tween 80 and 100% according to recent studies [43, 47–49].
the measurement of these factors has been proposed as a Notably, 68Ga-PET/CT imaging adds a significant diagnos-
predictor of skeletal colonization in clinical practice [42]. tic yield to conventional radiological procedures and may
change NET management in a substantial proportion of
patients. Compared to contrast-enhanced CT and MRI,
68Ga-PET/CT may provide a 20–25% yield in terms of di-
Bone Metastases in NETs: Clinical Features,
Diagnostic Workup and Prognostic Impact agnostic ability to detect bone metastases [43, 48, 50, 51].
False-positive results with 68Ga-PET/CT may occur in pa-
The diagnosis of bone lesions in patients with NETs is tients with meningiomas, angiomas, lymphomas or inflam-
steadily increasing, mainly due to the improved perfor- matory processes involving the skeleton, whereas false-neg-
mance of radiological and nuclear medicine diagnostic ative findings may be associated with low or absent expres-
techniques [43, 44]. Most frequently, NET bone metasta- sion of somatostatin receptors by tumor cells.
ses appear osteosclerotic and involve with higher fre- The detection of bone metastases is associated with a
quency the axial rather than the appendicular skeleton [7, worsening in patients’ clinical outcome. Significantly low-
8, 11]. While bone involvement is detected incidentally in er progression-free survival (PFS) and overall survival
up to 40% of patients with NETs, clinical complications rates have been reported in patients with skeletal involve-
of NET bone metastases have been observed in approxi- ment compared with those without bone lesions. In a large
mately half of the cases, with no correlation detected be- retrospective analysis from a part of the US National
tween SRE occurrence and gender, radiological appear- Comprehensive Cancer Network database including 691
ance of bone lesions, tumor primary site and WHO grade patients, significantly lower survival rates were observed
[9, 11]. Reportedly, pain is the most frequent symptom in in low-grade NETs arising from both pancreas and small
patients with NET bone metastases and can severely im- bowel [9]. Similar findings have been reported by other
pact the quality of life in a population of long-surviving studies performed in ENETS Centers of Excellence across
patients. In this regard, an analysis of 85 patients with Europe, confirming the negative prognostic impact of
NETs metastatic to the skeleton showed that bone pain bone metastases on patients’ survival [7, 10, 45]. Never-
was reported by 28 and 14% of subjects at the initial diag- theless, in a study enrolling 312 consecutive patients with
nosis and during the follow-up, respectively [7]. On the sporadic, grade 1/2, stage IV, nonfunctioning panNETs,
other hand, fractures have been described as rare events, the presence of bone lesions predicted a shorter PFS, in the
occurring in nearly 10–20% of cases, and even lower rates absence of any impact on overall survival [52]. Overall,
of spinal compression (5%) and hypercalcemia (1–4%) median survival time from bone metastases identification
have been reported [7, 45]. Although no clear factor able ranges from 20 to 60 months [7, 9, 10, 45, 49], a figure that
to predict the development of NET bone metastases has highlights the negative impact of this specific metastatic

Bone Metastases in NETs Neuroendocrinology 2021;111:207–216 211


DOI: 10.1159/000508633
Table 1. Key points for approaching neuroendocrine tumor (NET)
patients with bone metastases

Bone metastases are not rare, occurring in up to 20% of stage IV


NETs
They are asymptomatic in approximately 50% of patients
Skeletal fractures and hypercalcemia are rare events
68
Ga-PET/CT is mandatory to correctly stage bone disease
Bone metastases have a negative impact on patients’ clinical
outcomes
Bone-directed symptomatic therapies (surgery, radiotherapy,
bisphosphonates and denosumab) may help preventing skeletal
a complications

Skeletal colonization predicts poor survival also in lung


NETs, irrespective of the tumor grade or the synchronous/
metachronous presentation [14]. Key points to be attend-
ed in daily clinical practice when facing NET patients with
bone metastases are outlined in Table 1.

Treatment of NET Bone Metastases

Despite the availability of guidelines focusing on NET


management published by international scientific societ-
ies, the therapeutic approach to bone metastases in the
clinical practice is not well established yet. Two major
goals need to be taken into account when facing NET pa-
b
tients with advanced disease and bone metastases: (i) sys-
temic therapy of metastatic disease aimed at controlling
Fig. 2. The role of imaging techniques in detecting bone metastases tumor growth; (ii) bone-directed therapy aimed at reduc-
in NET patients. a Multi-field-of-view whole-body MRI shows a ing the risk of skeletal complications while improving the
skeletal metastasis in the right ischiopubic ramus of a patient with patient’s quality of life. With regard to the first point, bone
pancreatic NET (arrow). b A 68Ga-PET/CT scan shows multiple colonization has been described as a predictor of poor PFS
axial and appendicular bone lesions in a patient with small bowel
neuroendocrine tumor (arrowheads).
in patients with advanced, well-differentiated GEP-NETs
treated with first-line somatostatin analogs [54], as well as
in patients with progressive NETs undergoing treatment
with the mammalian target of rapamycin inhibitor evero-
pattern compared with patients with stage IV disease limus [55]. On the other hand, promising results have
without skeletal colonization, where median survival rates been reported in studies investigating the efficacy of pep-
are usually comprised between 80 and 100 months [7, 9, tide receptor radionuclide therapy (PRRT) in NET pa-
53]. Consistently, poorer survival outcomes have been re- tients with bone metastases. To date, PRRT is considered
ported for NET patients diagnosed with synchronous a reference therapy for well-differentiated, progressive,
bone metastases as compared with those presenting meta- somatostatin receptor-expressing, advanced GEP-NETs,
chronous skeletal colonization [11, 13]. Interestingly, thanks to the impressive gain in terms of PFS obtained in
bone metastases seem to have a major impact on the sur- comparison with high doses of octreotide long-acting re-
vival of patients with pancreatic NETs as compared with lease in the NETTER-1 trial [56]. In two single-institution,
those with intestinal NETs [10], confirming that NETs retrospective series from the University of Bonn, PRRT
from different sites should be considered separate entities determined objective responses (complete/major/minor
in terms of tumor behavior and patient’s clinical outcome. responses) in skeletal lesions in up to 50% of NET patients

212 Neuroendocrinology 2021;111:207–216 Cives/Pellè/Rinzivillo/Prosperi/Tucci/


DOI: 10.1159/000508633 Silvestris/Panzuto
Detected only by Systemic anti-NET treatment
functional imaging Active surveillance
(e.g., 68Ga-PET) of the skeletal lesion(s)
Low risk of SRE
Asymptomatic (i.e., rib localization)
(no SREs)
Localized

Detected by High risk of SRE Consider EBRT or


conventional imaging (i.e., femural neck localization) orthopedic surgery

Zoledronate (for 12 weeks)


Denosumab
NET bone Diffuse Calcium/Vitamin D3
metastases Systemic anti-NET
treatment

Consider EBRT or
Localized
orthopedic surgery
Symptomatic
(occurrence of SREs)
Zoledronate (for 4 weeks)
Calcium/Vitamin D3
Diffuse Cytoreductive therapy
(e.g., PRRT,
cytotoxic chemotherapy)

Fig. 3. Treatment tailoring in patients with neuroendocrine tumor (NET) bone metastases: a proposal for a ther-
apeutic algorithm. SRE, skeleton-related event; EBRT, external beam radiotherapy; PRRT, peptide receptor ra-
dionuclide therapy.

with bone metastases [57, 58], although in the presence of survival was 4 months [62]. Main acute toxicities of exter-
a higher risk of myelotoxicity [59]. Nevertheless, lower nal beam radiotherapy include erythema, loss of appetite,
rates of objective responses to PRRT have been docu- fatigue, nausea and vomiting, irrespective of the adoption
mented for NET bone metastases in other studies, and dis- of a single-dose or fractionated treatment schema [63].
crepancies in treatment response between bone and soft- Although their use is recommended by the ENETS guide-
tissue lesions have been described [60]. To the best of our lines [64], the administration of bisphosphonates in pa-
knowledge, no studies have specifically investigated the tients with NETs varies widely across different institutions
impact of sunitinib or chemotherapy on NET bone metas- given an apparent lack of survival benefit associated with
tases. Overall, an accurate assessment of bone disease in this form of bone-directed treatment [7, 8, 65]. Consistent
NET patients at diagnosis and throughout the course of with reports in other malignancies [66], the monthly ad-
the disease might be helpful in categorizing tumor clinical ministration of bisphosphonates did not show any advan-
aggressiveness, thereby supporting physicians in the pro- tage over less intensive treatment schemes in a small group
cess of treatment personalization. of patients with NET [13]. Denosumab has been shown to
Bone-directed therapies include radiotherapy, ortho- be slightly more effective than bisphosphonates in delay-
pedic surgery, bisphosphonates (administered either oral- ing SREs in patients with bone-metastatic cancers [67].
ly or intravenously), and denosumab. Both radiotherapy However, no studies have specifically investigated the ef-
and surgery are often used to prevent or treat SREs in pa- ficacy of the RANKL inhibitor in NET patients. While sev-
tients with NET [11], and bone pain relief has been de- eral toxicities including the osteonecrosis of the jaw, hy-
scribed in up to 90% of patients treated with radiotherapy pocalcemia and atypical fractures are shared by both
[61]. In a study of 45 NET patients receiving palliative ex- bisphosphonates and denosumab, side effects such as
ternal beam radiotherapy, skeletal metastases represented acute phase responses and induction of renal insufficiency
the 75% of target lesions, and the median symptom-free may occur when administering bisphosphonates.

Bone Metastases in NETs Neuroendocrinology 2021;111:207–216 213


DOI: 10.1159/000508633
Several questions remain unanswered regarding the extensively tested in patients with bone-devastating can-
treatment landscape of NET bone metastases. First, when cers, and the calcium-mimetic radium-223 dichloride has
should therapy with antiresorptive agents be initiated? been approved for patients with castration-resistant pros-
Second, should all patients with NET bone metastases re- tate cancer and skeletal metastases [68]. Given that radi-
ceive bone-directed therapies? Third, is there a preferred um-223 is able to form hydroxyapatite complexes in areas
option among bisphosphonates as well as between of high osteoblast activity, and that NET bone metastases
bisphosphonates and denosumab? Fourth, is the month- are prevalently osteosclerotic, future research should de-
ly administration of bisphosphonate inherently superior fine the role of this α-emitter in patients with bone-colo-
to the 3-month schedule, in a population of patients with nizing NETs. Key areas to be prioritized by clinical re-
indolent malignancies and long survival times? Although search in the next few years include the prospective evalu-
in the absence of evidence guiding clinical decisions, a ation of the impact of PRRT on NET bone lesion growth,
possible flowchart outlining the treatment algorithm to the definition of the optimal dosing interval of bisphos-
be potentially followed when facing patients with NET phonates in NET bone metastases, the longitudinal assess-
bone metastases is depicted in Figure 3. ment of quality of life in patients treated or not with bone-
targeting agents, as well as the identification of potential
subgroups of patients who may negligibly benefit of bone-
Conclusions and Future Perspectives directed interventions, thus requiring only observation.
While we envisage that the advent of precision medi-
The skeleton is the third most frequent site of metas- cine will help us identifying NET patients more likely to
tasis in patients with NETs, but the molecular mecha- develop bone metastases and stratifying them according
nisms leading to the development of bone lesions in these to their SRE risk, the development of more sophisticated
malignancies have been poorly investigated so far. In par- diagnostic and therapeutic modalities will progressively
ticular, it is presently unclear whether the dismal out- improve our ability to detect, prevent and treat NET bone
comes observed in patients with bone-colonizing NETs metastases. In the meantime, clinical wisdom is needed
are the result of an inherently aggressive tumor biology, when facing NET patients with skeletal lesions.
rather than the consequence of the late onset or late diag-
nosis of a frequently overlooked condition. The recent
advent of 68Ga-PET/CT has substantially improved our Acknowledgment
ability to detect NET bone metastases, and future studies
The authors thank Dr. Lucrezia Alemanno for editorial assis-
will shed light onto the real frequency of skeletal coloni-
tance.
zation in NET patients, as well as onto the impact of its
earlier recognition on survival outcomes.
Innovative bone-directed treatment approaches are Disclosure Statement
needed to both prevent the occurrence of NET bone me-
tastases and inhibit their growth. In recent years, the evi- The authors declare no affiliation with industries or organiza-
dence that bone-targeting agents interfere with several key tions with a financial interest, direct or indirect, that may affect the
conduct or reporting of the work submitted.
steps throughout the process of skeletal colonization
prompted the initiation of clinical trials investigating the
efficacy of adjuvant bisphosphonates or denosumab in pa-
Funding Sources
tients with osteotropic cancers [19]. While breast cancer
experts currently recommend the use of adjuvant bisphos- This work was supported by a grant from the Italian Associa-
phonates in selected clinical scenarios, the potential util- tion for Cancer Research (AIRC, MFAG23583; Mauro Cives) and
ity of this approach in delaying both skeletal and systemic Apulia Region (Oncogenomic Project, 2015; Franco Silvestris).
recurrences in patients with NETs is presently unknown.
New bone-targeting agents including cathepsin K inhibi-
tors, c-Src antagonists, antisclerostin antibodies, inhibi- Author Contributions
tors of dickkopf-related protein-1 and activin-A blockers
Dr. Cives and Dr. Panzuto have conceived the manuscript. Dr.
are under active clinical scrutiny [19], although studies Cives, Dr. Pellè, Dr. Rinzivillo, Dr. Prosperi and Dr. Panzuto have
specifically focusing on the NET population are presently participated in manuscript writing. Prof. Tucci and Prof. Silvestris
lacking. Bone-targeting radiopharmaceuticals have been have reviewed the manuscript draft.

214 Neuroendocrinology 2021;111:207–216 Cives/Pellè/Rinzivillo/Prosperi/Tucci/


DOI: 10.1159/000508633 Silvestris/Panzuto
References
  1 Cives M, Strosberg JR. Gastroenteropancre- 13 Alexandraki KI, Pizanias M, Uri I, Thomas D, 25 Ikezono Y, Koga H, Akiba J, Abe M, Yoshida
atic neuroendocrine tumors. CA Cancer J Page T, Kolomodi D, et al. The prognosis and T, Wada F, et al. Pancreatic Neuroendocrine
Clin. 2018 Nov;68(6):471–87. management of neuroendocrine neoplasms- Tumors and EMT Behavior Are Driven by the
  2 Dasari A, Shen C, Halperin D, Zhao B, Zhou related metastatic bone disease: lessons from CSC Marker DCLK1. Mol Cancer Res. 2017
S, Xu Y, et al. Trends in the Incidence, Preva- clinical practice. Endocrine. 2019 Jun; 64(3): Jun;15(6):744–52.
lence, and Survival Outcomes in Patients 690–701. 26 Kim JY, Lee SH, An S, Kim SJ, Sung YN, Song
With Neuroendocrine Tumors in the United 14 Peri M, Botteri E, Pisa E, De Marinis F, Un- KB, et al. Carbonic anhydrase 9 expression in
States. JAMA Oncol. 2017 Oct; 3(10): 1335– garo A, Spada F, et al. A single-institution ret- well-differentiated pancreatic neuroendo-
42. rospective analysis of metachronous and syn- crine neoplasms might be associated with ag-
  3 Klimstra DS, Kloppel G, La Rosa S, Rindi G. chronous metastatic bronchial neuroendo- gressive behavior and poor survival. Vir-
Classification of neuroendocrine neoplasms crine tumors. J Thorac Dis. 2018 Jul; 10(7): chows Arch. 2018 May;472(5):739–48.
of the digestive system. WHO classification of 3928–39. 27 Yonemori K, Kurahara H, Maemura K, Mat-
tumors of the digestive system. Volume 1. 5th 15 Zheng Z, Chen C, Jiang L, Zhou X, Dai X, aki Y, Sakoda M, Iino S, et al. Impact of Snail
ed. Lyon, France: IARC Press; 2019. Song Y, et al. Incidence and risk factors of gas- and E-cadherin expression in pancreatic neu-
 4 Riihimäki M, Hemminki A, Sundquist K, trointestinal neuroendocrine neoplasm me- roendocrine tumors. Oncol Lett. 2017 Aug;
Sundquist J, Hemminki K. The epidemiology tastasis in liver, lung, bone, and brain: A pop- 14(2):1697–702.
of metastases in neuroendocrine tumors. Int ulation-based study. Cancer Med. 2019 Dec; 28 Cives M, Quaresmini D, Rizzo FM, Felici C,
J Cancer. 2016 Dec;139(12):2679–86. 8(17):7288–98. D’Oronzo S, Simone V, et al. Osteotropism of
  5 Pape UF, Berndt U, Müller-Nordhorn J, Böh- 16 Lombard-Bohas C, Mitry E, O’Toole D, Lou- neuroendocrine tumors: role of the CXCL12/
mig M, Roll S, Koch M, et al. Prognostic fac- vet C, Pillon D, Cadiot G, et al.; FFCD-AN- CXCR4 pathway in promoting EMT in vitro.
tors of long-term outcome in gastroentero- GH-GERCOR. Thirteen-month registration Oncotarget. 2017 Apr;8(14):22534–49.
pancreatic neuroendocrine tumours. Endocr of patients with gastroenteropancreatic endo- 29 Pellé E, Cives M, Quaresmini D, Lovero D,
Relat Cancer. 2008 Dec;15(4):1083–97. crine tumours in France. Neuroendocrinolo- Felici C, Cafforio P, et al. CXCR4 inhibition
 6 Nuñez-Valdovinos B, Carmona-Bayonas A, gy. 2009;89(2):217–22. by ulocuplumab prevents EMT of pNET cells
Jimenez-Fonseca P, Capdevila J, Castaño- 17 Robelin P, Hadoux J, Forestier J, Planchard D, in vitro. Ann Oncol. 2017;28 Suppl 5:142–57.
Pascual Á, Benavent M, et al. Neuroendocrine Hervieu V, Berdelou A, et al. Characteriza- 30 Arvidsson Y, Bergström A, Arvidsson L, Kris-
Tumor Heterogeneity Adds Uncertainty to tion, Prognosis, and Treatment of Patients tiansson E, Ahlman H, Nilsson O. Hypoxia
the World Health Organization 2010 Classifi- With Metastatic Lung Carcinoid Tumors. J stimulates CXCR4 signalling in ileal carci-
cation: Real-World Data from the Spanish Thorac Oncol. 2019 Jun;14(6):993–1002. noids. Endocr Relat Cancer. 2010 Jun; 17(2):
Tumor Registry (R-GETNE). Oncologist. 18 Ross EM, Roberts WC. The carcinoid syn- 303–16.
2018 Apr;23(4):422–32. drome: comparison of 21 necropsy subjects 31 Speisky D, Duces A, Bièche I, Rebours V,
  7 Scharf M, Petry V, Daniel H, Rinke A, Gress with carcinoid heart disease to 15 necropsy Hammel P, Sauvanet A, et al.; GTE Group,
TM. Bone Metastases in Patients with Neuro- subjects without carcinoid heart disease. Am Groupe d’Etude des Tumeurs Endocrines.
endocrine Neoplasm: Frequency and Clinical, J Med. 1985 Sep;79(3):339–54. Molecular profiling of pancreatic neuroendo-
Therapeutic, and Prognostic Relevance. Neu- 19 D'Oronzo S, Coleman R, Brown J, Silvestris F. crine tumors in sporadic and Von Hippel-
roendocrinology. 2018;106(1):30–7. Metastatic bone disease: pathogenesis and Lindau patients. Clin Cancer Res. 2012 May;
  8 Kavecansky J, Wei L, Caronia L, Ramirez MT, therapeutic options: update on bone metasta- 18(10):2838–49.
Bloomston M, Shah MH. Bone metastases in sis management. J Bone Oncol. 2018; 15: 004- 32 Xu SS, Xu HX, Wang WQ, Li S, Li H, Li TJ, et
well-to-moderately differentiated neuroen- 4. al. Tumor-infiltrating platelets predict post-
docrine tumors: a single institutional review 20 Lambert AW, Pattabiraman DR, Weinberg operative recurrence and survival in resect-
from the Ohio State University Medical Cen- RA. Emerging biological principles of metas- able pancreatic neuroendocrine tumor.
ter. Pancreas. 2015 Mar;44(2):198–203. tasis. Cell. 2017 Feb;168(4):670–91. World J Gastroenterol. 2019 Nov; 25(41):
  9 Van Loon K, Zhang L, Keiser J, Carrasco C, 21 Fendrich V, Waldmann J, Esni F, Ramaswa- 6248–57.
Glass K, Ramirez MT, et al. Bone metastases my A, Mullendore M, Buchholz M, et al. Snail 33 Peinado H, Zhang H, Matei IR, Costa-Silva B,
and skeletal-related events from neuroendo- and Sonic Hedgehog activation in neuroen- Hoshino A, Rodrigues G, et al. Pre-metastatic
crine tumors. Endocr Connect. 2015 Mar; docrine tumors of the ileum. Endocr Relat niches: organ-specific homes for metastases.
4(1):9–17. Cancer. 2007 Sep;14(3):865–74. Nat Rev Cancer. 2017 May;17(5):302–17.
10 Panzuto F, Puscedddu S, Faggiano A, Rinzi­ 22 Fendrich V, Maschuw K, Waldmann J, Buch- 34 Tucci M, Mannavola F, Passarelli A, Stucci LS,
villo M, Brighi N, Prinzi N, et al.; Itanet (Ital- holz M, Rehm J, Gress TM, et al. Epithelial- Cives M, Silvestris F. Exosomes in melanoma:
ian Association for Neuroendocrine Tu- mesenchymal transition is a critical step in a role in tumor progression, metastasis and
mours). Prognostic impact of tumour burden tumorgenesis of pancreatic neuroendocrine impaired immune system activity. Oncotar-
in stage IV neuroendocrine neoplasia: A com- tumors. Cancers (Basel). 2012 Mar;4(1):281– get. 2018 Apr;9(29):20826–37.
parison between pancreatic and gastrointesti- 94. 35 Błogowski W, Zuba-Surma E, Sałata D, Bud-
nal localizations. Pancreatology. 2019 Dec; 23 Galván JA, Astudillo A, Vallina A, Fonseca PJ, kowska M, Dołęgowska B, Starzyńska T. Pe-
19(8):1067–73. Gómez-Izquierdo L, García-Carbonero R, et ripheral trafficking of bone-marrow-derived
11 Cives M, Rizzo F, Simone V, Bisceglia F, Stuc- al. Epithelial-mesenchymal transition mark- stem cells in patients with different types of
ci S, Seeber A, et al. Reviewing the Osteotro- ers in the differential diagnosis of gastroen- gastric neoplasms. OncoImmunology. 2015
pism in Neuroendocrine Tumors: The Role of teropancreatic neuroendocrine tumors. Am J Oct;5(4):e1099798.
Epithelial-Mesenchymal Transition. Neuro- Clin Pathol. 2013 Jul;140(1):61–72. 36 Khan MS, Tsigani T, Rashid M, Rabouhans
endocrinology. 2016;103(3-4):321–34. 24 Galván JA, Astudillo A, Vallina A, Crespo G, JS, Yu D, Luong TV, et al. Circulating tumor
12 Strosberg J, Gardner N, Kvols L. Survival and Folgueras MV, González MV. Prognostic and cells and EpCAM expression in neuroendo-
prognostic factor analysis of 146 metastatic diagnostic value of epithelial to mesenchymal crine tumors. Clin Cancer Res. 2011 Jan;
neuroendocrine tumors of the mid-gut. Neu- transition markers in pulmonary neuroendo- 17(2):337–45.
roendocrinology. 2009;89(4):471–6. crine tumors. BMC Cancer. 2014 Nov; 14(1):
855.

Bone Metastases in NETs Neuroendocrinology 2021;111:207–216 215


DOI: 10.1159/000508633
37 Khan MS, Kirkwood A, Tsigani T, Garcia- 48 Albanus DR, Apitzsch J, Erdem Z, Erdem O, 58 Ezziddin S, Sabet A, Heinemann F, Yong-
Hernandez J, Hartley JA, Caplin ME, et al. Verburg FA, Behrendt FF, et al. Clinical value Hing CJ, Ahmadzadehfar H, Guhlke S, et al.
Circulating tumor cells as prognostic markers of ⁶⁸Ga-DOTATATE-PET/CT compared to Response and long-term control of bone me-
in neuroendocrine tumors. J Clin Oncol. 2013 stand-alone contrast enhanced CT for the de- tastases after peptide receptor radionuclide
Jan;31(3):365–72. tection of extra-hepatic metastases in patients therapy with (177)Lu-octreotate. J Nucl Med.
38 Hsieh JC, Chen GY, Jhou DD, Chou WC, Yeh with neuroendocrine tumours (NET). Eur J 2011 Aug;52(8):1197–203.
CN, Hwang TL, et al. The Prognostic Value of Radiol. 2015 Oct;84(10):1866–72. 59 Cives M, Strosberg J. Radionuclide Therapy
Circulating Tumor Cells in Asian Neuroen- 49 Ambrosini V, Nanni C, Zompatori M, Cam- for Neuroendocrine Tumors. Curr Oncol
docrine Tumors. Sci Rep. 2019 Dec; 9(1): pana D, Tomassetti P, Castellucci P, et al. (68) Rep. 2017 Feb;19(2):9.
19917. Ga-DOTA-NOC PET/CT in comparison 60 van Vliet EI, Hermans JJ, de Ridder MA,
39 Rizzo FM, Vesely C, Childs A, Marafioti T, with CT for the detection of bone metastasis Teunissen JJ, Kam BL, de Krijger RR, et al.
Khan MS, Mandair D, et al. Circulating tu- in patients with neuroendocrine tumours. Tumor response assessment to treatment
mour cells and their association with bone Eur J Nucl Med Mol Imaging. 2010 Apr;37(4): with [177Lu-DOTA0,Tyr3]octreotate in pa-
metastases in patients with neuroendocrine 722–7. tients with gastroenteropancreatic and bron-
tumours. Br J Cancer. 2019 Feb; 120(3): 294– 50 Crown A, Rocha FG, Raghu P, Lin B, Funk G, chial neuroendocrine tumors: differential re-
300. Alseidi A, et al. Impact of initial imaging with sponse of bone versus soft-tissue lesions. J
40 Massagué J, Obenauf AC. Metastatic coloni- gallium-68 dotatate PET/CT on diagnosis and Nucl Med. 2012 Sep;53(9):1359–66.
zation by circulating tumour cells. Nature. management of patients with neuroendocrine 61 Guan M, He I, Luu M, David J, Gong J, Pla-
2016 Jan;529(7586):298–306. tumors. J Surg Oncol. 2020 Mar; 121(3): 480– cencio-Hickok VR, et al. Palliative Radiation
41 Weilbaecher KN, Guise TA, McCauley LK. 5. Therapy for Bone Metastases in Neuroendo-
Cancer to bone: a fatal attraction. Nat Rev 51 Skoura E, Michopoulou S, Mohmaduvesh M, crine Neoplasms. Adv Radiat Oncol. 2019
Cancer. 2011 Jun;11(6):411–25. Panagiotidis E, Al Harbi M, Toumpanakis C, Apr;4(3):513–9.
42 Milone F, Pivonello C, Cariati F, Sarnataro M, et al. The impact of 68Ga-DOTATATE PET/ 62 Yalchin M, Tharmalingam H, Owen C, Walk-
Ramundo V, Marotta V, et al. Assessment and CT imaging on management of patients with er M, Caplin M, Stewart G, et al. The role of
clinical implications of RANK/RANKL/OPG neuroendocrine tumors: experience from a external beam radiotherapy in the manage-
pathway as markers of bone tumor progres- national referral center in the United King- ment of advanced neuroendocrine tumors.
sion in patients with NET harboring bone dom. J Nucl Med. 2016 Jan;57(1):34–40. Presented at the 12th Annual ENETS Confer-
metastases. Biomarkers. 2013 Mar;18(2):121– 52 Jiménez-Fonseca P, Krug S, Tamagno G, Fi- ence, Barcelona, Spain. Neuroendocrinology.
5. erro Maya F, Monléon Getino A, Rodriguez 2015;102:136.
43 Merola E, Pavel ME, Panzuto F, Capurso G, Casado CI, et al. Identifying Prognostic Fac- 63 Lutz S, Balboni T, Jones J, Lo S, Petit J, Rich
Cicchese N, Rinke A, et al. Functional Imag- tors for Well-Differentiated Metastatic Pan- SE, et al. Palliative radiation therapy for bone
ing in the Follow-Up of Enteropancreatic creatic Neuroendocrine Tumours: A Retro- metastases: Update of an ASTRO Evidence-
Neuroendocrine Tumors: Clinical Usefulness spective International Multicentre Cohort Based Guideline. Pract Radiat Oncol. 2017
and Indications. J Clin Endocrinol Metab. Study. Neuroendocrinology. 2018; 107(4): Jan - Feb;7(1):4–12.
2017 May;102(5):1486–94. 315–23. 64 Kos-Kudła B, O’Toole D, Falconi M, Gross D,
44 Sadowski SM, Neychev V, Millo C, Shih J, Ni- 53 Panzuto F, Merola E, Rinzivillo M, Partelli S, Klöppel G, Sundin A, et al.; Palma de Mal-
lubol N, Herscovitch P, et al. Prospective Campana D, Iannicelli E, et al. Advanced di- lorca Consensus Conference Participants.
study of 68Ga-DOTATATE positron emis- gestive neuroendocrine tumors: metastatic ENETS consensus guidelines for the manage-
sion tomography/computed tomography for pattern is an independent factor affecting ment of bone and lung metastases from neu-
detecting gastro-entero-pancreatic neuroen- clinical outcome. Pancreas. 2014 Mar; 43(2): roendocrine tumors. Neuroendocrinology.
docrine tumors and unknown primary sites. J 212–8. 2010;91(4):341–50.
Clin Oncol. 2016 Feb;34(6):588–96. 54 Carmona-Bayonas A, Jiménez-Fonseca P, La- 65 Altieri B, Di Dato C, Martini C, Sciammarella
45 Lim KHJ, Raja H, D'Arienzo P, Barriuso J, marca Á, Barriuso J, Castaño Á, Benavent M, C, Di Sarno A, Colao A, et al.; NIKE Group.
McNamara MG, Hubner RA, et al. Identifica- et al. Prediction of Progression-Free Survival Bone Metastases in Neuroendocrine Neo-
tion of areas for improvement in the manage- in Patients With Advanced, Well-Differenti- plasms: From Pathogenesis to Clinical Man-
ment of bone metastases in patients with neu- ated, Neuroendocrine Tumors Being Treated agement. Cancers (Basel). 2019 Sep;
roendocrine neoplasms. Neuroendocrinolo- With a Somatostatin Analog: the GETNE- 11(9):E1332.
gy. 2020;110:688–96. TRASGU Study. J Clin Oncol. 2019 Oct; 66 Himelstein AL, Foster JC, Khatcheressian JL,
46 Shi W, Johnston CF, Buchanan KD, Ferguson 37(28):2571–80. Roberts JD, Seisler DK, Novotny PJ, et al. Ef-
WR, Laird JD, Crothers JG, et al. Localization 55 Benslama N, Bollard J, Vercherat C, Massoma fect of Longer-Interval vs Standard Dosing of
of neuroendocrine tumours with [111In] DT- P, Roche C, Hervieu V, et al. Prediction of re- Zoledronic Acid on Skeletal Events in Pa-
PA-octreotide scintigraphy (Octreoscan): a sponse to everolimus in neuroendocrine tu- tients With Bone Metastases: A Randomized
comparative study with CT and MR imaging. mors: evaluation of clinical, biological and Clinical Trial. JAMA. 2017 Jan;317(1):48–58.
QJM. 1998 Apr;91(4):295–301. histological factors. Invest New Drugs. 2016 67 Lipton A, Fizazi K, Stopeck AT, Henry DH,
47 Gauthé M, Testart Dardel N, Ruiz Santiago F, Oct;34(5):654–62. Brown JE, Yardley DA, et al. Superiority of
Ohnona J, Nataf V, Montravers F, et al. Ver- 56 Strosberg J, El-Haddad G, Wolin E, Hendifar denosumab to zoledronic acid for prevention
tebral metastases from neuroendocrine tu- A, Yao J, Chasen B, et al; NETTER-1 Trial In- of skeletal-related events: a combined analysis
mours: how to avoid false positives on 68Ga- vestigators. Phase 3 trial of 177Lu-Dotatate of 3 pivotal, randomised, phase 3 trials. Eur J
DOTA-TOC PET using CT pattern analysis? for midgut neuroendocrine tumors. N Engl J Cancer. 2012 Nov;48(16):3082–92.
Eur Radiol. 2018 Sep;28(9):3943–52. Med. 2017 Jan;376(2):125–35. 68 Parker C, Nilsson S, Heinrich D, Helle SI,
57 Sabet A, Khalaf F, Haslerud T, Al-Zreiqat A, O’Sullivan JM, Fosså SD, et al; ALSYMPCA
Sabet A, Simon B, et al. Bone metastases in Investigators. Alpha emitter radium-223 and
GEP-NET: response and long-term outcome survival in metastatic prostate cancer. N Engl
after PRRT from a follow-up analysis. Am J J Med. 2013 Jul;369(3):213–23.
Nucl Med Mol Imaging. 2013 Sep; 3(5): 437–
45.

216 Neuroendocrinology 2021;111:207–216 Cives/Pellè/Rinzivillo/Prosperi/Tucci/


DOI: 10.1159/000508633 Silvestris/Panzuto

You might also like