Next Article in Journal
Characteristics and Outcomes in Patients with Ventilator-Associated Pneumonia Who Do or Do Not Develop Acute Respiratory Distress Syndrome. An Observational Study
Next Article in Special Issue
Professional Assessment of the Impact of COVID-19 on Handling NET Patients
Previous Article in Journal
Smoking and Radiolucent Periapical Lesions in Root Filled Teeth: Systematic Review and Meta-Analysis
Previous Article in Special Issue
Essentials of Insulinoma Localization with Selective Arterial Calcium Stimulation and Hepatic Venous Sampling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in the Management of Medullary Thyroid Carcinoma: Focus on Peptide Receptor Radionuclide Therapy

1
Endocrine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
2
NET Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari-Endocrine Unit, AOU Sassari, 07100 Sassari, Italy
3
Endocrine Clinic, Policlinico Sassarese, 07100 Sassari, Italy
4
Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
5
Nuclear Medicine Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
6
Department of Clinical Medicine and Surgery, Endocrinology Unit, University Federico II, 80131 Naples, Italy
*
Authors to whom correspondence should be addressed.
E Grossrubatscher and G Fanciulli are joint first authors.
Membership of NIKE Group is provided in the Acknowledgments.
J. Clin. Med. 2020, 9(11), 3507; https://doi.org/10.3390/jcm9113507
Submission received: 26 September 2020 / Revised: 26 October 2020 / Accepted: 27 October 2020 / Published: 29 October 2020
(This article belongs to the Special Issue Neuroendocrine Tumors: Clinical Challenges)

Abstract

:
Effective treatment options in advanced/progressive/metastatic medullary thyroid carcinoma (MTC) are currently limited. As in other neuroendocrine neoplasms (NENs), peptide receptor radionuclide therapy (PRRT) has been used as a therapeutic option in MTC. To date, however, there are no published reviews dealing with PRRT approaches. We performed an in-depth narrative review on the studies published in this field and collected information on registered clinical trials related to this topic. We identified 19 published studies, collectively involving more than 200 patients with MTC, and four registered clinical trials. Most cases of MTC were treated with PRRT with somatostatin analogues (SSAs) radiolabelled with 90 yttrium (90Y) and 177 lutetium (177Lu). These radiopharmaceuticals show efficacy in the treatment of patients with MTC, with a favourable radiological response (stable disease, partial response or complete response) in more than 60% of cases, coupled with low toxicity. As MTC specifically also expresses cholecystokinin receptors (CCK2Rs), PRRT with this target has also been tried, and some randomised trials are ongoing. Overall, PRRT seems to have an effective role and might be considered in the therapeutic strategy of advanced/progressive/metastatic MTC.

1. Introduction

Medullary thyroid carcinoma (MTC) is a rare neuroendocrine neoplasm (NEN). Surgery, with complete resection of the tumour, is, in most cases, curative [1]. However, in a subgroup of patients, the tumour shows an aggressive behaviour. In this setting (advanced/progressive/metastatic MTC), the management remains challenging [1,2]. Among systemic therapies, tyrosine kinase inhibitors (TKIs) have recently been shown to improve progression-free survival (PFS) in patients with advanced disease [3,4]. Two TKIs, namely vandetanib and cabozantinib, have been approved for the treatment of progressive or symptomatic MTC. Both drugs are effective, resulting in partial response (PR) or stable disease (SD), but tumour escape frequently occurs. On the other hand, both drugs may cause grade III or IV adverse events (classified according to the Common Terminology Criteria for Adverse Events of the National Cancer Institute) [3,4]. Therefore, other systemic options are needed. As in other advanced/progressive/metastatic NENs, radionuclide therapy may represent a therapeutic option, and indeed, it has been applied in MTC since the 1980s. Historically, the first radiopharmaceutical used in therapy was 131Iodine (131I)-metaiodobenzylguanidine (MIBG) [5,6], but its use has been limited in MTC, due to the low percentage of MTC that shows uptake on MIBG scintigraphy [7,8,9].
Somatostatin receptor (SSR) expressions in MTC cells have been reported in vitro and in vivo [10,11,12,13,14,15,16]. This finding provides the basis for peptide receptor radionuclide therapy (PRRT) with radiolabelled somatostatin analogues (SSAs), and indeed, several studies on the therapeutic use of these radiopharmaceuticals in MTC have been performed in the last two decades. Additionally, the cholecystokinin 2 receptor (CCK2R) expressions in MTC [17,18,19,20] offer another promising target for PRRT.
To date, there are no published reviews dealing with the different PRRT approaches in MTC comparing the outcomes in terms of efficacy and safety.

2. Aim of the Study

We aimed to perform an in-depth narrative review on the studies published on PRRT and MTC, in order to offer to the physician involved in the management of MTC an updated overview of the PRRT treatments.
Furthermore, we collected information on registered clinical trials (RCTs) related to this topic, in order to give the scientific community a glance about the future of PRRT in MTC.

3. Materials and Methods

3.1. Published Studies

A search on PubMed, Embase and Cochrane was made by using the terms “medullary thyroid carcinoma”, medullary thyroid cancer”, “radionuclide”, lutetium”, “yttrium”, “indium”, “peptide receptor radionuclide therapy”, “radiolabelled somatostatin analogues”, “gastrin-like peptides” and “cholecystokinin-like peptides”. Additional studies were identified by reviewing the references of all selected articles. Only studies in the English language were included. We excluded from the analysis: (1) single-case reports, (2) abstracts from congresses and proceedings from workshops and (3) letters to the editor. The search was last updated 16 October 2020.

3.2. RCTs

By using the same terms used in the section describing published studies, we performed a search on the Registries Clinical Trials Gov and EudraCT. The search was last updated 16 October 2020.

4. Results

4.1. Published Studies

We found 18 PRRT studies targeting SSRs involving patients with MTC: 14 with SSA labelled with the radionuclide(s) 90 yttrium (90Y) and/or 177 lutetium (177Lu) and four with SSA labelled with 111 indium (111In). Additionally, we found one study targeting cholecystokinin receptors (CCK2Rs), employing a gastrin analogue labelled with 90Y.
PRRT with 90Y and 177Lu: Fourteen studies (publication years: 1999–2020) were found [21,22,23,24,25,26,27,28,29,30,31,32,33,34]. The cumulative number of patients with MTC treated by PRRT with radiolabelled SSA was 186. The results are summarised in Table 1.
Disease data. The disease status at baseline was represented by progressive or inoperable or advanced diseases in 142/149 patients (95.3%) in 10 studies [22,27,30,32,34].
Treatment schedule. SSA labelled with 177Lu was employed in 72 patients (38.7%) [29,30,32,33,34]. SSA labelled with 90Y was employed in 78 patients (41.9%) [21,22,23,24,25,26,27]. SSA labelled with 90Y or 177Lu were employed in 36 patients (19.4%) [28,31]. In one study, 177Lu-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-octreotate (DOTATATE) was used with concomitant low-dose oral capecitabine therapy as a potential radiosensitiser [34].
Radiographic response. The response was evaluated with heterogeneous criteria. Evaluation data were available for 117 patients (62.9%) in 11 studies: 44 patients showed progressive disease (PD) (37.6%), 64 patients showed SD (54.7%), six patients showed PR (5.1%), and three patients showed complete response (CR) (2.6%) [21,22,23,24,25,26,29,30,32,33,34].
Biochemical response. The calcitonin measurement was available for 101 patients (54.3%) in five studies [25,26,27,33,34]. In the four studies employing calcitonin plasma concentrations, evaluation data were available in 70 patients (37.6%): after PRRT, six patients showed values < 15 pg/mL (8.6%), 20 patients showed a decrease ≥ 50% in basal values (28.6%), and the remaining 44 patients showed lower reduction or increase in basal values (62.8%) [25,26,33,34]. Another study reported a post-PRRT prolongation of calcitonin doubling time ≥ 100% in 18/31 patients (58.1%) [27].
Symptomatic response and Quality of life (QoL). Data regarding the subjective response were nearly absent. The evaluation of the symptomatic response was available for 53 patients (28.5%) in two studies: the first one reported “no improvement” in all 10 patients (100%) [32]; the second one reported PD (new symptoms or symptoms increase > 30%) in 21 patients (49%), SD (symptoms reduction < 30%) in two patients (4%), PR (symptoms reduction 30–75%) in 12 patients (28%), and CR (symptoms reduction 100%) in eight patients (19%) [33]. Only one study specifically evaluated the QoL through the Short Form-36 Health Survey (SF-36) questionnaire on a small case series of six patients; all the responders (patients who had at least some tumour shrinkage) had an improvement in QoL six to 12 months after therapy [29].
Outcome. Data about time to progression (TTP) and/or PFS and/or overall survival (OS) were reported in five studies [22,26,31,32,33]. A longer survival was reported in patients with disease control (PR + SD) in the study by Salavati et al. [31] and with biochemical response in the study by Iten et al. [27]. Finally, in the study by Parghane et al. [33], a longer OS was observed in patients with longer calcitonin doubling time.
Toxicity. Adverse reactions included two cases of treatment discontinuation (“kidney toxicity”) among 154 patients (1.3%) [21,22,23,24,25,26,27,28,29,32,33,34], one case of grade III-IV nephrotoxicity among 133 patients (0.8%) [21,22,23,24,26,27,28,29,32,33,34], and nine cases of grade III-IV haemotoxicity among 139 patients (6.5%) [21,24,25,26,27,28,29,32,33,34]. Moreover, one case of haemoptysis was reported, presumably due to the progression of pulmonary metastases [32]. No other grade III-IV treatment-related adverse events were reported in 143 patients [21,22,24,25,26,27,29,32,33,34].
PRRT with 111In: Four studies (publication years: 2000–2004) were found [35,36,37,38]. The cumulative number of patients with MTC treated by 111In-SSA was 10. The results are summarised in Table 2.
Disease data. In all cases, the disease status at baseline was represented by progressive or inoperable disease [35,36,37,38].
Radiographic response. Data about the radiologic response, evaluated with heterogeneous criteria, were available for eight patients (80.0%) in three studies: overall, four patients showed PD (40.0%), two patients showed SD (25.0%), and two patients showed CR (25.0%) [36,37,38].
Biochemical response. This parameter was evaluated with heterogeneous criteria. The evaluation of calcitonin was available for seven patients (70.0%) in three studies: after PRRT, one patient showed “normalisation” (14.2%), three patients showed “SD” (42.9%), and three patients showed “PD” (42.9%) [35,36,38].
Symptomatic response and QoL. The evaluation of the symptomatic response was available for five patients (50.0%) in the study of Valkema et al. [36]: only one patient (20.0%) showed symptomatic improvement, but the evaluation criteria were not specified. Only the study of Pasieka et al. specifically evaluated the QoL [38]: the single patient studied showed no improvement.
Outcome. Data about PFS and OS were not available in the studies.
Toxicity. Information about the adverse reactions included no cases of treatment discontinuation among four patients [35,37,38], no cases of grade III-IV nephrotoxicity among 10 patients [35,36,37,38], no cases of grade III-IV haemotoxicity among four patients [35,37,38], and no other grade III-IV treatment-related adverse events [35,36,37,38].
PRRT targeting CCK2Rs: Only one study, including eight patients, was found (publication year: 2002) [17]. Results are summarised in Table 3.
Disease data. In all cases, the disease status at baseline was represented by progressive and advanced disease.
Radiographic response. Since neither imaging procedures nor response criteria were specified, these data were not included in Table 3. However, patient responses were defined by the authors as follows: two PD (25.0%), four SD (50.0%), and two PR (25.0%).
Biochemical response, symptomatic response, QoL, and outcome. No data were available.
Toxicity. Information about adverse reactions included no cases of treatment discontinuation, no cases of grade III-IV nephrotoxicity, and three cases of grade III-IV haemotoxicity (37.5%).

4.2. RCTs

We identified four RCTs on ClinicalTrials.gov, two on SSRs, and two on CCK2Rs. Details of the RCTs are reported in Table 4.

5. Discussion

The largest part of cases of MTC reported in our review were treated with PRRT with SSAs radiolabelled with 90Y and 177Lu. The radiographic response showed a significant disease control rate (DCR = CR + PR + SD) in 62.4% of cases (CR in 2.6%, PR in 5.1%, and SD in 54.7%), most of which were in progression at the baseline. PD was observed in 37.6% of cases [21,22,23,24,25,26,29,30,32,33,34]. In two [22,30,32] of the three studies [22,30,32] in which the percentage of PD was higher, the population under study was of older age. This is in-line with the evidence that age is an important prognostic determinant in MTC [39].
The use of a concomitant radiosensitising agent, like low-dose oral capecitabine, could be even more beneficial, as suggested by a study from Satapathy et al. [40] in which the DCR was 86%. These results are in-line with similar observations made in gastroenteropancreatic NENs and malignant phaeocromocytomas about the combination of PRRT and chemotherapy [41,42,43].
In responsive patients, PRRT seems to stabilise the disease. However, the effects of the treatment on PFS and OS rates have yet to be determined. Indeed, only five studies evaluated these outcome endpoints. In particular, one study demonstrated longer survival in patients with a radiological disease control [31], and two studies [27,33] demonstrated longer survival in patients with a biochemical response, evaluated through calcitonin assessment, which, in MTC, can indicate tumour recurrence or progression [44] and indicate a treatment response with good sensitivity [45,46].
The TKI vandetanib and cabozantinib have been shown to improve PFS in patients with advanced MTC [3,4] and have been approved for the treatment of progressive or symptomatic MTC. However, both are associated in many patients with grade III-IV adverse events [3,4], which reduce the QoL of patients. Data from our review showed that a treatment discontinuation for toxicity was observed in only 1.3% of cases, grade III-IV haemotoxicity in 6.5% of cases, grade III-IV nephrotoxicity in 0.8%, and other grade III-IV side effects in no cases. The apparently higher rates of haemotoxicity compared with nephrotoxicity were derived from the high percentage of haemotoxicity found in a study from Bodei et al. [47], in which a transient lymphocytopenia was demonstrated in 88% of cases. These data could be explained by the fact that the series included patients who had received bone radiotherapy with the consequence of a potential reduction of bone marrow reserve [47]. On the contrary, the low nephrotoxicity that emerged from the data of the present review may be linked to the duration of follow-up. Indeed, it is unknown whether late renal damage may become evident four or five years after treatment administration [47].
PRRT has been used mainly in the treatment of gastroenteropancreatic NENs expressing SSRs, with promising results. Following the publication of the Neuroendocrine Tumors Therapy (NETTER-1) study [48], PRRT with 177Lu-oxodotreotide was approved by the Food and Drug Administration, European Medicines Agency, and National Institute for Health and Care Excellence in SSR-positive, well-differentiated (G1-G2) gastroenteropancreatic NENs. As for MTC, the guidelines of the European Society for Medical Oncology [49] and American Thyroid Association [50] agree that current experiences about PRRT with radiolabelled SSA in MTC are limited, and no randomised clinical trials have been carried out to compare the efficacies of PRRT versus TKIs. Consequently, at this time, PRRT should only be considered in selected patients in the context of clinical trials. However, its potential benefit when TKIs are contraindicated is recognised.
In order to establish the role of PRRT in the therapeutic algorithm of advanced/progressive/metastatic MTC, more solid data from randomised controlled studies with survival endpoints are needed. Nevertheless, the rarity and the long natural history of the disease have hindered the research in this field.
As with other NENs, 111In-diethylenetriaminepentacetic acid (DTPA)-octreotide was the first therapeutic radiopharmaceutical used as a PRRT in MTC. The contrasting results and the limited number of patients with MTC treated do not allow a judgment on this radiopharmaceutical, which was soon abandoned, due to the low rate of objective tumour responses and the significant haematologic toxicity in favour of SSAs labelled with the more efficient 90Y and 177Lu radionuclides. Moreover, the only RCT we found, namely NCT00002947, was terminated early (reason not stated).
The limitations of this review study about data on PRRT targeting SSRs in MTC are as follows: first, most analysed case series are small, and prospective studies are few. Second, most of the data—including treatment schedule, follow-up, response evaluation, and treatments before PRRT—are highly heterogeneous and sometimes incomplete. Finally, the outcome data are available only in a few studies, due to the small sample sizes and limited lengths of follow-up, which underpowered the evaluations of PFS and OS.
New data with the primary outcome of the tumour response are expected in 2022 (RCT NCT04106843).
As previously mentioned, a PRRT could also target CCK2Rs, which are overexpressed in more than 90% of MTCs [17,18,19,20]. In the past two decades, a variety of CCK2-related peptides radiolabelled with 90Y/177Lu have been synthesised and categorised based on the sequence of their parent peptide (CCK or gastrin). To date, most studies about PRRT targeting CCK2Rs were preclinical trials and investigated specific basic criteria of candidate radioligands such as nanomolar receptor affinity, rapid and efficient accumulation in the tumour, low uptake and rapid wash-out in/from normal tissues, and in vivo stability. Only a few pilot clinical studies have been carried out in small cohorts of patients. An important landmark study in this area was the study conducted in 2002 by Behr and Behe [17], which investigated 45 patients with metastatic MTC with 111In-DTPA-minigastrin scintigraphy (23 with known and 32 with occult diseases). The radiotracer physiological uptake was related to CCK2R-specific binding and to the route of excretion. As for the binding, the normal uptake mainly regarded the stomach and, to a lower extent, gallbladder and breasts. Regarding the route of excretion, the kidneys were identified as the only excretory organs. All tumour manifestations known by conventional imaging were visualised, and at least one lesion was detected in 29/32 patients with occult disease. Furthermore, eight patients with advanced metastatic disease were injected in a dose-escalation study with potentially therapeutic activities of a 90Y-labelled minigastrin derivative at four–six-weekly intervals (Table 3, Results: section PRRT targeting CCK2Rs); haematologic and renal were identified as dose-limiting toxicities.
In general, although very promising in a theoretical context, the clinical translation of PRRT targeting CCK2Rs has been prevented by two major problems: high kidney uptake and/or fast in vivo degradation of the different radiopeptides developed. Radiopharmaceutical research has tried to overcome these limitations by introducing various modifications in the peptide sequence of gastrin or CCK-related peptides, with the aim of reducing kidney retention and improving metabolic stability. A co-injection of plasma expanders has been proposed to reduce kidney uptake and retention, consequently minimising radiation-induced nephrotoxicity [19,20,51]. In order to reduce in vivo radiopeptide degradation, a co-injection of neutral endopeptidase inhibitors is also under investigation, since endopeptidases are the major enzymes implicated in the catabolism of gastrin/CCK-based peptides, and they are widely distributed in the body [52,53]. Research in this field is actively ongoing, as evidenced by the two clinical trials pertaining to PRRT in CCK2Rs mentioned above (Table 4); this lesser-known but promising therapy is expected to establish a new and efficient strategy for the treatment of MTC, which is still a challenging entity.

6. Conclusions

On the basis of the reported efficacy and low toxicity, PRRT could represent a therapeutic option in the treatment of advanced/progressive/metastatic MTC. As in other NENs, most of the experience was obtained with PRRT targeting SSRs, but CCK2Rs might offer in the future an alternative PRRT option.

Author Contributions

E.G., G.F., L.P., F.S., C.D., and F.d.C. were responsible for the design, the methodology, the writing—original draft preparation, and the reviewing and editing. A.C. and A.F. were responsible for the supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Italian Ministry of Education, University and Research (MIUR): PRIN 2017Z3N3YC, and by the University of Sassari, Italy: Specialist International Clinical Training (SICT) Project (G.F.).

Acknowledgments

This review is part of the “Neuroendocrine Tumors Innovation Knowledge and Education” project led by Annamaria Colao and Antongiulio Faggiano, which aims at increasing the knowledge on NET. We would like to acknowledge all the Collaborators of the “NIKE” project: Manuela Albertelli—Genova; Barbara Altieri—Wurzburg; Luigi Barrea—Napoli; Filomena Bottiglieri—Napoli; Severo Campione—Napoli; Sergio Di Molfetta—Bari; Alessandra Dicitore—Milano; Tiziana Feola—Roma; Diego Ferone—Genova; Francesco Ferraù—Messina; Marco Gallo—Torino; Elisa Giannetta—Roma; Federica Grillo—Genova; Elia Guadagno—Napoli; Valentina Guarnotta—Palermo; Andrea M. Isidori—Roma; Andrea Lania—Milano; Andrea Lenzi—Roma; Fabio Lo Calzo—Avellino; Pasquale Malandrino—Catania; Erika Messina—Messina; Roberta Modica—Napoli; Giovanna Muscogiuri—Napoli; Genoveffa Pizza—Avellino; Riccardo Pofi—Roma; Giulia Puliani—Roma; Carmen Rainone—Napoli; Paola Razzore—Torino; Laura Rizza—Roma; Manila Rubino—Milano; Rosa Maria Ruggieri—Messina; Emilia Sbardella—Roma; Mary Anna Venneri—Roma; Giovanni Vitale—Milano; Maria Chiara Zatelli—Ferrara.

Conflicts of Interest

All authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

CCK: cholecystokinin; CCK2R: cholecystokinin 2 receptor; CT: computerized tomography; CR: complete response; DCR: disease control rate; DOTA: 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid; DOTATATE: DOTA-octreotate; DTPA-octreotide: diethylenetriaminepentacetic acid-octreotide; 68Ga: 68 gallium; 111In: 111 indium; 131I: 131 iodine; 177Lu: 177 lutetium; MIBG: metaiodobenzylguanidine; MTC: medullary thyroid carcinoma; MEN: multiple endocrine neoplasia; NEN: neuroendocrine neoplasm; OS: overall survival; PD: progressive disease; PET: positron emission tomography; PFS: progression-free survival; PR: partial response; PP-F11N: DOTA-(D-Glu)6-Ala-Tyr-Gly-Trp-Nle-Asp-PheNH2; PRRT: peptide receptor radionuclide therapy; QoL: quality of life; SD: stable disease; SS: somatostatin; SSA: somatostatin analogue; SSR: somatostatin receptor; RCT: registered clinical trials; TKI: tyrosine kinase inhibitor; TTP: time-to-progression; and 90Y: 90 Yttrium.

References

  1. Ceolin, L.; Duval, M.; Benini, A.F.; Ferreira, C.V.; Maia, A.L. Medullary thyroid carcinoma beyond surgery: Advances, challenges, and perspectives. Endocr. Relat. Cancer 2019, 26, R499–R518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Viola, D.; Elisei, R. Management of Medullary Thyroid Cancer. Endocrinol. Metab. Clin. N. Am. 2019, 48, 285–301. [Google Scholar] [CrossRef] [PubMed]
  3. Wells, S.A., Jr.; Robinson, B.G.; Gagel, R.F.; Dralle, H.; Fagin, J.A.; Santoro, M.; Baudin, E.; Elisei, R.; Jarzab, B.; Vasselli, J.R.; et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: A randomized, double-blind phase III trial. J. Clin. Oncol. 2012, 30, 134–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Elisei, R.; Schlumberger, M.J.; Muller, S.P.; Schoffski, P.; Brose, M.S.; Shah, M.H.; Licitra, L.; Jarzab, B.; Medvedev, V.; Kreissl, M.C.; et al. Cabozantinib in progressive medullary thyroid cancer. J. Clin. Oncol. 2013, 31, 3639–3646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Clarke, S.E.; Lazarus, C.R.; Edwards, S.; Murby, B.; Clarke, D.G.; Roden, T.M.; Fogelman, I.; Maisey, M.N. Scintigraphy and treatment of medullary carcinoma of the thyroid with iodine-131 metaiodobenzylguanidine. J. Nucl. Med. 1987, 28, 1820–1825. [Google Scholar]
  6. Voute, P.A.; Hoefnagel, C.A.; de Kraker, J.; Evans, A.E.; Hayes, A.; Green, A. Radionuclide therapy of neural crest tumors. Med. Pediatr. Oncol. 1987, 15, 192–195. [Google Scholar] [CrossRef]
  7. Baulieu, J.L.; Guilloteau, D.; Delisle, M.J.; Perdrisot, R.; Gardet, P.; Delepine, N.; Baulieu, F.; Dupont, J.L.; Talbot, J.N.; Coutris, G.; et al. Radioiodinated meta-iodobenzylguanidine uptake in medullary thyroid cancer. A French cooperative study. Cancer 1987, 60, 2189–2194. [Google Scholar] [CrossRef]
  8. Rufini, V.; Calcagni, M.L.; Baum, R.P. Imaging of neuroendocrine tumors. Semin. Nucl. Med. 2006, 36, 228–247. [Google Scholar] [CrossRef]
  9. Castellani, M.R.; Seregni, E.; Maccauro, M.; Chiesa, C.; Aliberti, G.; Orunesu, E.; Bombardieri, E. MIBG for diagnosis and therapy of medullary thyroid carcinoma: Is there still a role? Q. J. Nucl. Med. Mol. Imaging 2008, 52, 430–440. [Google Scholar]
  10. Mato, E.; Matias-Guiu, X.; Chico, A.; Webb, S.M.; Cabezas, R.; Berna, L.; De Leiva, A. Somatostatin and somatostatin receptor subtype gene expression in medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 1998, 83, 2417–2420. [Google Scholar] [CrossRef]
  11. Hofland, L.J.; Lamberts, S.W. Somatostatin receptors and disease: Role of receptor subtypes. Baillieres Clin. Endocrinol. Metab. 1996, 10, 163–176. [Google Scholar] [CrossRef] [Green Version]
  12. Faggiano, A.; Grimaldi, F.; Pezzullo, L.; Chiofalo, M.G.; Caraco, C.; Mozzillo, N.; Angeletti, G.; Santeusanio, F.; Lombardi, G.; Colao, A.; et al. Secretive and proliferative tumor profile helps to select the best imaging technique to identify postoperative persistent or relapsing medullary thyroid cancer. Endocr. Relat. Cancer 2009, 16, 225–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. De Luca, S.; Fonti, R.; Camera, L.; Salvatore, B.; Faggiano, A.; Ciarmiello, A.; Segreto, S.; Colao, A.; Salvatore, M.; Del Vecchio, S. Multimodal imaging with (18)F-FDG-PET/CT and (111)In-Octreotide SPECT in patients with metastatic medullary thyroid carcinoma. Ann. Nucl. Med. 2016, 30, 234–241. [Google Scholar] [CrossRef] [PubMed]
  14. Baudin, E.; Schlumberger, M.; Lumbroso, J.; Travagli, J.P.; Caillou, B.; Parmentier, C. Octreotide scintigraphy in patients with differentiated thyroid carcinoma: Contribution for patients with negative radioiodine scan. J. Clin. Endocrinol. Metab. 1996, 81, 2541–2544. [Google Scholar] [CrossRef] [Green Version]
  15. Kaltsas, G.; Korbonits, M.; Heintz, E.; Mukherjee, J.J.; Jenkins, P.J.; Chew, S.L.; Reznek, R.; Monson, J.P.; Besser, G.M.; Foley, R.; et al. Comparison of somatostatin analog and meta-iodobenzylguanidine radionuclides in the diagnosis and localization of advanced neuroendocrine tumors. J. Clin. Endocrinol. Metab. 2001, 86, 895–902. [Google Scholar] [CrossRef]
  16. Dorr, U.; Wurstlin, S.; Frank-Raue, K.; Raue, F.; Hehrmann, R.; Iser, G.; Scholz, M.; Guhl, L.; Buhr, H.J.; Bihl, H. Somatostatin receptor scintigraphy and magnetic resonance imaging in recurrent medullary thyroid carcinoma: A comparative study. Horm. Metab. Res. Suppl. 1993, 27, 48–55. [Google Scholar]
  17. Behr, T.M.; Behe, M.P. Cholecystokinin-B/Gastrin receptor-targeting peptides for staging and therapy of medullary thyroid cancer and other cholecystokinin-B receptor-expressing malignancies. Semin. Nucl. Med. 2002, 32, 97–109. [Google Scholar] [CrossRef]
  18. Roosenburg, S.; Laverman, P.; van Delft, F.L.; Boerman, O.C. Radiolabeled CCK/gastrin peptides for imaging and therapy of CCK2 receptor-expressing tumors. Amino Acids 2011, 41, 1049–1058. [Google Scholar] [CrossRef] [Green Version]
  19. Maina, T.; Konijnenberg, M.W.; KolencPeitl, P.; Garnuszek, P.; Nock, B.A.; Kaloudi, A.; Kroselj, M.; Zaletel, K.; Maecke, H.; Mansi, R.; et al. Preclinical pharmacokinetics, biodistribution, radiation dosimetry and toxicity studies required for regulatory approval of a phase I clinical trial with (111)In-CP04 in medullary thyroid carcinoma patients. Eur. J. Pharm. Sci. 2016, 91, 236–242. [Google Scholar] [CrossRef]
  20. Rottenburger, C.; Nicolas, G.P.; McDougall, L.; Kaul, F.; Cachovan, M.; Vija, A.H.; Schibli, R.; Geistlich, S.; Schumann, A.; Rau, T.; et al. Cholecystokinin 2 Receptor Agonist (177)Lu-PP-F11N for Radionuclide Therapy of Medullary Thyroid Carcinoma: Results of the Lumed Phase 0a Study. J. Nucl. Med. 2020, 61, 520–526. [Google Scholar] [CrossRef]
  21. Otte, A.; Herrmann, R.; Heppeler, A.; Behe, M.; Jermann, E.; Powell, P.; Maecke, H.R.; Muller, J. Yttrium-90 DOTATOC: First clinical results. Eur. J. Nucl. Med. 1999, 26, 1439–1447. [Google Scholar] [CrossRef]
  22. Waldherr, C.; Pless, M.; Maecke, H.R.; Haldemann, A.; Mueller-Brand, J. The clinical value of [90Y-DOTA]-D-Phe1-Tyr3-octreotide (90Y-DOTATOC) in the treatment of neuroendocrine tumours: A clinical phase II study. Ann. Oncol. 2001, 12, 941–945. [Google Scholar] [CrossRef] [PubMed]
  23. Paganelli, G.; Zoboli, S.; Cremonesi, M.; Bodei, L.; Ferrari, M.; Grana, C.; Bartolomei, M.; Orsi, F.; De Cicco, C.; Macke, H.R.; et al. Receptor-mediated radiotherapy with 90Y-DOTA-D-Phe1-Tyr3-octreotide. Eur. J. Nucl. Med. 2001, 28, 426–434. [Google Scholar] [CrossRef] [PubMed]
  24. Bodei, L.; Cremonesi, M.; Zoboli, S.; Grana, C.; Bartolomei, M.; Rocca, P.; Caracciolo, M.; Macke, H.R.; Chinol, M.; Paganelli, G. Receptor-mediated radionuclide therapy with 90Y-DOTATOC in association with amino acid infusion: A phase I study. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 207–216. [Google Scholar] [CrossRef]
  25. Bodei, L.; Handkiewicz-Junak, D.; Grana, C.; Mazzetta, C.; Rocca, P.; Bartolomei, M.; Lopera Sierra, M.; Cremonesi, M.; Chinol, M.; Macke, H.R.; et al. Receptor radionuclide therapy with 90Y-DOTATOC in patients with medullary thyroid carcinomas. Cancer Biother. Radiopharm. 2004, 19, 65–71. [Google Scholar] [CrossRef]
  26. Gao, Z.; Biersack, H.J.; Ezziddin, S.; Logvinski, T.; An, R. The role of combined imaging in metastatic medullary thyroid carcinoma: 111In-DTPA-octreotide and 131I/123I-MIBG as predictors for radionuclide therapy. J. Cancer Res. Clin. Oncol. 2004, 130, 649–656. [Google Scholar] [CrossRef]
  27. Iten, F.; Muller, B.; Schindler, C.; Rochlitz, C.; Oertli, D.; Macke, H.R.; Muller-Brand, J.; Walter, M.A. Response to [90Yttrium-DOTA]-TOC treatment is associated with long-term survival benefit in metastasized medullary thyroid cancer: A phase II clinical trial. Clin. Cancer Res. 2007, 13, 6696–6702. [Google Scholar] [CrossRef] [Green Version]
  28. Budiawan, H.; Salavati, A.; Kulkarni, H.R.; Baum, R.P. Peptide receptor radionuclide therapy of treatment-refractory metastatic thyroid cancer using (90)Yttrium and (177)Lutetium labeled somatostatin analogs: Toxicity, response and survival analysis. Am. J. Nucl. Med. Mol. Imaging 2013, 4, 39–52. [Google Scholar] [PubMed]
  29. Vaisman, F.; Rosado de Castro, P.H.; Lopes, F.P.; Kendler, D.B.; Pessoa, C.H.; Bulzico, D.A.; de Carvalho Leal, D.; Vilhena, B.; Vaisman, M.; Carneiro, M.; et al. Is there a role for peptide receptor radionuclide therapy in medullary thyroid cancer? Clin. Nucl. Med. 2015, 40, 123–127. [Google Scholar] [CrossRef] [PubMed]
  30. Lapa, C.; Werner, R.A.; Schmid, J.S.; Papp, L.; Zsoter, N.; Biko, J.; Reiners, C.; Herrmann, K.; Buck, A.K.; Bundschuh, R.A. Prognostic value of positron emission tomography-assessed tumor heterogeneity in patients with thyroid cancer undergoing treatment with radiopeptide therapy. Nucl. Med. Biol. 2015, 42, 349–354. [Google Scholar] [CrossRef]
  31. Salavati, A.; Puranik, A.; Kulkarni, H.R.; Budiawan, H.; Baum, R.P. Peptide Receptor Radionuclide Therapy (PRRT) of Medullary and Nonmedullary Thyroid Cancer Using Radiolabeled Somatostatin Analogues. Semin. Nucl. Med. 2016, 46, 215–224. [Google Scholar] [CrossRef] [PubMed]
  32. Beukhof, C.M.; Brabander, T.; van Nederveen, F.H.; van Velthuysen, M.F.; de Rijke, Y.B.; Hofland, L.J.; Franssen, G.J.H.; Froberg, L.A.C.; Kam, B.L.R.; Visser, W.E.; et al. Peptide receptor radionuclide therapy in patients with medullary thyroid carcinoma: Predictors and pitfalls. BMC Cancer 2019, 19, 325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Parghane, R.V.; Naik, C.; Talole, S.; Desmukh, A.; Chaukar, D.; Banerjee, S.; Basu, S. Clinical utility of (177) Lu-DOTATATE PRRT in somatostatin receptor-positive metastatic medullary carcinoma of thyroid patients with assessment of efficacy, survival analysis, prognostic variables, and toxicity. Head Neck 2020, 42, 401–416. [Google Scholar] [CrossRef]
  34. Satapathy, S.; Mittal, B.R.; Sood, A.; Verma, R.; Panda, N. Efficacy and safety of concomitant 177Lu-DOTATATE and low-dose capecitabine in advanced medullary thyroid carcinoma: A single-centre experience. Nucl. Med. Commun. 2020, 41, 629–635. [Google Scholar] [CrossRef]
  35. Caplin, M.E.; Mielcarek, W.; Buscombe, J.R.; Jones, A.L.; Croasdale, P.L.; Cooper, M.S.; Burroughs, A.K.; Hilson, A.W. Toxicity of high-activity 111In-Octreotide therapy in patients with disseminated neuroendocrine tumours. Nucl. Med. Commun. 2000, 21, 97–102. [Google Scholar] [CrossRef]
  36. Valkema, R.; De Jong, M.; Bakker, W.H.; Breeman, W.A.; Kooij, P.P.; Lugtenburg, P.J.; De Jong, F.H.; Christiansen, A.; Kam, B.L.; De Herder, W.W.; et al. Phase I study of peptide receptor radionuclide therapy with [In-DTPA]octreotide: The Rotterdam experience. Semin. Nucl. Med. 2002, 32, 110–122. [Google Scholar] [CrossRef]
  37. Buscombe, J.R.; Caplin, M.E.; Hilson, A.J. Long-term efficacy of high-activity 111in-pentetreotide therapy in patients with disseminated neuroendocrine tumors. J. Nucl. Med. 2003, 44, 1–6. [Google Scholar]
  38. Pasieka, J.L.; McEwan, A.J.; Rorstad, O. The palliative role of 131I-MIBG and 111In-octreotide therapy in patients with metastatic progressive neuroendocrine neoplasms. Surgery 2004, 136, 1218–1226. [Google Scholar] [CrossRef]
  39. Roman, S.; Lin, R.; Sosa, J.A. Prognosis of medullary thyroid carcinoma: Demographic, clinical, and pathologic predictors of survival in 1252 cases. Cancer 2006, 107, 2134–2142. [Google Scholar] [CrossRef] [PubMed]
  40. Parghane, R.V.; Ostwal, V.; Ramaswamy, A.; Bhandare, M.; Chaudhari, V.; Talole, S.; Shrikhande, S.V.; Basu, S. Long-term outcome of “Sandwich” chemo-PRRT: A novel treatment strategy for metastatic neuroendocrine tumors with both FDG- and SSTR-avid aggressive disease. Eur. J. Nucl. Med. Mol. Imaging 2020. [Google Scholar] [CrossRef]
  41. Claringbold, P.G.; Brayshaw, P.A.; Price, R.A.; Turner, J.H. Phase II study of radiopeptide 177Lu-octreotate and capecitabine therapy of progressive disseminated neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 302–311. [Google Scholar] [CrossRef]
  42. Ballal, S.; Yadav, M.P.; Damle, N.A.; Sahoo, R.K.; Bal, C. Concomitant 177Lu-DOTATATE and Capecitabine Therapy in Patients With Advanced Neuroendocrine Tumors: A Long-term-Outcome, Toxicity, Survival, and Quality-of-Life Study. Clin. Nucl. Med. 2017, 42, e457–e466. [Google Scholar] [CrossRef] [PubMed]
  43. Yadav, M.P.; Ballal, S.; Bal, C. Concomitant (177)Lu-DOTATATE and capecitabine therapy in malignant paragangliomas. EJNMMI Res. 2019, 9, 13. [Google Scholar] [CrossRef]
  44. Engelbach, M.; Gorges, R.; Forst, T.; Pfutzner, A.; Dawood, R.; Heerdt, S.; Kunt, T.; Bockisch, A.; Beyer, J. Improved diagnostic methods in the follow-up of medullary thyroid carcinoma by highly specific calcitonin measurements. J. Clin. Endocrinol. Metab. 2000, 85, 1890–1894. [Google Scholar] [CrossRef] [PubMed]
  45. Gronlund, B.; Hogdall, C.; Hilden, J.; Engelholm, S.A.; Hogdall, E.V.; Hansen, H.H. Should CA-125 response criteria be preferred to response evaluation criteria in solid tumors (RECIST) for prognostication during second-line chemotherapy of ovarian carcinoma? J. Clin. Oncol. 2004, 22, 4051–4058. [Google Scholar] [CrossRef] [PubMed]
  46. Barbet, J.; Campion, L.; Kraeber-Bodere, F.; Chatal, J.F.; Group, G.T.E.S. Prognostic impact of serum calcitonin and carcinoembryonic antigen doubling-times in patients with medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2005, 90, 6077–6084. [Google Scholar] [CrossRef] [Green Version]
  47. Bodei, L.; Mueller-Brand, J.; Baum, R.P.; Pavel, M.E.; Horsch, D.; O’Dorisio, M.S.; O’Dorisio, T.M.; Howe, J.R.; Cremonesi, M.; Kwekkeboom, D.J.; et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2013, 40, 800–816. [Google Scholar] [CrossRef]
  48. Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 Trial of (177)Lu-Dotatate for Midgut Neuroendocrine Tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef]
  49. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A.; ESMO Guidelines Committee. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-updagger. Ann. Oncol. 2019, 30, 1856–1883. [Google Scholar] [CrossRef] [Green Version]
  50. Wells, S.A., Jr.; Asa, S.L.; Dralle, H.; Elisei, R.; Evans, D.B.; Gagel, R.F.; Lee, N.; Machens, A.; Moley, J.F.; Pacini, F.; et al. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid 2015, 25, 567–610. [Google Scholar] [CrossRef]
  51. Erba, P.A.; Maecke, H.; Mikolajczak, R.; Decristoforo, C.; Zaletel, K.; Maina-Nock, T.; Peitl, P.K.; Garnuszek, P.; Froberg, A.; Goebel, G.; et al. A novel CCK2/gastrin receptor-localizing radiolabeled peptide probe for personalized diagnosis and therapy of patients with progressive or metastatic medullary thyroid carcinoma: A multicenter phase I GRAN-T-MTC study. Pol. Arch. Intern Med. 2018, 128, 791–795. [Google Scholar] [CrossRef]
  52. Kaloudi, A.; Nock, B.A.; Krenning, E.P.; Maina, T.; De Jong, M. Radiolabeled gastrin/CCK analogs in tumor diagnosis: Towards higher stability and improved tumor targeting. Q. J. Nucl. Med. Mol. Imaging 2015, 59, 287–302. [Google Scholar] [PubMed]
  53. Sauter, A.W.; Mansi, R.; Hassiepen, U.; Muller, L.; Panigada, T.; Wiehr, S.; Wild, A.M.; Geistlich, S.; Behe, M.; Rottenburger, C.; et al. Targeting of the Cholecystokinin-2 Receptor with the Minigastrin Analog (177)Lu-DOTA-PP-F11N: Does the Use of Protease Inhibitors Further Improve In Vivo Distribution? J. Nucl. Med. 2019, 60, 393–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. PRRT targeting SSRs with 90Y and 177Lu-SSA in MTC.
Table 1. PRRT targeting SSRs with 90Y and 177Lu-SSA in MTC.
ReferencesDemographicsDisease DataTreatment Schedule and Follow-Up
AuthorsYearType of StudySubjects nAge-Years Mean (Median) (Range)F/M nMutation Status nDisease Status at BaselineSubjects with Metastases %RadioisotopeDose/Cycle GBqCycles nCumulative Dose-GBq Mean (Median) (Range)Follow-Up Months Mean (Median) (Range)
Otte et al. [21]1999NS264.5 (64.5) (62–67)2/0NSNS2 (100%)90Y1.6–2.949.4 (9.4) (9.2–9.6)2 (2) (2–2)
Waldherr et al. [22]2001NS1255.8 (60.0) (24–72)5/71 MEN2 11 NSProgressiveNS90YNS1–48.1 (9.1) (1.7–14.0)NS
Paganelli et al. [23]2001NS351.0 (55.0) (34–64)2/1NSNS2 (67%)90Y1.835.5 (5.5) (5.5–5.5)11 (12) (10–12)
Bodei et al. [24]2003NS845.4 (46.5) (31–67)1/7NSProgressive in 3/8 patients8 (100%)90Y2.9–4.827.9 (8.5) (5.9–9.6)19 (21) (4–26)
Bodei et al. [25]2004Retrospective2151.4 (53.0) (31–78)8/13NSProgressive21 (100%)90Y2.2–5.1 (max)2–810.7 (10.4) (7.6–19.2)(3–40)
Gao et al. [26]2004NS157.00/1NSProgressive1 (100%)90Y3.3413.2NS
Iten et al. [27]2007Prospective31(56.7) (24–77)10/212 MEN2 (1 additional MEN1), 28 NSProgressive31 (100%)90Y3.7 (GBq/m2)1–5(12.6) (1.7–29.6)(12) (1–107)
Budiawan et al. [28]2013NS859.1 (61.0) (40–76)4/4NSProgressive8 (100%)90Y and/or 177LuNS1–3NSNS
Vaisman et al. [29]2015Prospective7NSNSNSProgressiveNS177Lu7.4429.6 (29.6) (29.6–29.6)(8–12)
Lapa et al. [30]2015Retrospective449.0 (44.0) (33–75)2/22 MEN2 2NSNS4 (100%)177LuNS2–531.4 (31.4) (23.7–39.0) 2 patients NSNS
Salavati et al. [31]2016NS2847.9 (26–72)14/14NSNS28 (100%)90Y and/or 177LuNS≤ 5NSNS
Beukhof et al. [32]2019Retrospective10(63.0) (19–75)6/46 sporadic 4 NSProgressive in 8/10 patients10 (100%)177LuNS4 (mean)(27.8–29.6)NS
Parghane et al. [33]2020Retrospective43(48.0) (25–80)8/35NSProgressive43 (100%)177Lu5.5 (mean)1–618.5 (5.55–33.3)(20) (8–78)
Satapathy et al. [34]2020Retrospective846.0 (47.5) (22–70)5/38 sporadicProgressive or advanced or inoperable8 (100%)177Lu *6.0–7.41–419.1 (20.9) (6.4–27.8)40 (34) (14–69) 1 patient NS
-1999–2020-186-67/112-Progressive or Advanced or Inoperable 142/149 (95.3%)166/167 (99.4%)-----
ReferencesRadiographic ResponseBiochemical ResponseTreatment OutcomeTreatment Toxicity
Authors, YearSubjects Suitable for Evaluative nResponse CriteriaPD n (%)SD n (%)PR n (%)CR n (%)Subjects
Suitable for Evaluative n
Response Criteria (Calcitonin, CT)ResponsePFS MonthsOS MonthsDiscontinuation n (%)Grade III/IV Nephrotoxicity n (%)Grade III/IV Haemotoxicity n (%)
Otte et al., 1999 [21]2NS0 (0%)2 (100%)0 (0%)0 (0%)0--NSNS0 (0%)0 (0%)0 (0%)
Waldherr et al., 2001 [22]12WHO7 (58%)5 (42%)0 (0%)0 (0%)0--NS (TTP: mean 8, median 10, range 3–14)NS0 (0%)0 (0%)NS
Paganelli et al., 2001 [23]3WHO0 (0%)3 (100%)0 (0%)0 (0%)0--NSNS0 (0%)0 (0%)NS
Bodei et al., 2003 [24]7WHO2 (29%)3 (43%)1 (14%)1 (14%)0--NSNS0 (0%)0 (0%)7 (88%)
Bodei et al., 2004 [25]21SWOG7 (33%)12 (57%)0 (0%)2 (10%)21PD (increase ≥ 25% in basal value)
SD (none of the others)
PR (decrease ≥ 50% in basal value)
CR (<15 pg/mL)
12 (57%)
3 (14%)
5 (24%)
1 (5%)
NSNS0 (0%)NS1 (5%)
Gao et al., 2004 [26]1WHO0 (0%)1 (100%)0 (0%)0 (0%)1Pre-therapy and Post-therapy values10,461 pg/mL; 3414 pg/mLNS (TTP: 6)NS0 (0%)0 (0%)0 (0%)
Iten et al., 2007 [27]0-----31Post-PRRT Prolongation of CT Doubling Time (≥100%)Response = 18/31 (58%)NS16 (median) 1-107 (range)2 (6%): kidney toxicity1 (3%)1 (3%)
Budiawan et al., 2013 [28]0-----0--NSNS0 (0%)0 (0%)0 (0%)
Vaisman et al., 2015 [29]7RECIST 1.11 (14%)3 (43%)3 (43%)0 (0%)0--NSNS0 (0%)0 (0%)0 (0%)
Lapa et al., 2015 [30]4RECIST 1.1 “in most cases”4 (100%)0 (0%)0 (0%)0 (0%)0--NSNSNSNSNS
Salavati et al., 2016 [31]0-----0--NS24 in PD (median) 36 in SD (median) 72 in PR (median)NSNSNS
Beukhof et al., 2019 [32]10RECIST 1.16 (60%)4 (40%)0 (0%)0 (0%)0--8 (median) 4–144 (range)14 (median) 5–144 (range)0 (0%)0 (0%)0 (0%)
Parghane et al., 2020 [33]43RECIST 1.116 (37%)25 (58%)2 (5%)0 (0%)43PD (increase ≥ 30% in basal value)
SD (none of the others)
PR (decrease ≥ 50% in basal value)
CR (<15 pg/mL)
21 (49%)
4 (9%)
13 (30%)
5 (12%)
24 (median)26 (median)0 (0%)0 (0%)0 (0%)
Satapathy et al., 2020 [34]7RECIST 1.11 (14%)6 (86%)0 (0%)0 (0%)5Increase ≥ 25% in basal value None of the others Decrease ≥ 50% in basal value CT < 15 pg/mL2 (40%)
2 (40%)
(20%)
0 (0%)
NSNS0 (0%)0 (0%)0 (0%)
-117-44 (37.6%)64 (54.7%)6 (5.1%)3 (2.6%)101----2/154 (1.3%)1/133 (0.8%)9/139 (6.5%)
PRRT: peptide receptor radionuclide therapy; SSR: somatostatin receptor; 90Y: 90 yttrium; 177Lu: lutetium; SSA: somatostatin analogue; MTC: medullary thyroid carcinoma; F: female; M: male; GBq: GigaBecquerel; NS: not specified; MEN: multiple endocrine neoplasia; PD: progressive disease; SD: stable disease; PR: partial response; CR: complete response; PFS: progression-free survival; OS: overall survival; WHO: World Health Organization; TTP: time-to-progression; SWOG: Southwest Oncology Group; RECIST: Response Evaluation Criteria In Solid Tumours. * Concomitant low-dose oral capecitabine therapy (1.250 mg/m2/day from day 0 to day 14 of each PRRT therapy).
Table 2. PRRT targeting SSRs with 111In-SSA in MTC.
Table 2. PRRT targeting SSRs with 111In-SSA in MTC.
ReferencesDemographicsDisease DataTreatment Schedule and Follow-Up
AuthorsYearType of StudySubjects nAge-Years Mean (Median) (Range)F/M nMutation Status nDisease Status at BaselineSubjects with Metastases %RadioisotopeDose/Cycle GBqCycles nCumulative Dose-GBq Mean (Median) (Range)Follow-Up Months Mean (Median) (Range)
Caplin et al. [35]2000Prospective146.01/0NSInoperable1 (100%)111In2.8 (mean)411.4NS
Valkema et al. [36]2002Prospective655.0 (57.5) (28–77)NSNSProgressive6 (100%)111InNSNS42.5 (28.7) (5.8–87.3)9.5 (5.6) (1–27)
Buscombe et al. [37] 2003Retrospective252.0 (52.0) (46–58)1/1NSProgressive2 (100%)111InNS3–410.9 (10.9) (10.5–11.4)18 (18) (18–18)
Pasieka et al. [38]2004NS146.00/1NSProgressive1 (100%)111In5.9 (mean)211.89
-2000–2004-10-2/2-Progressive or advanced or inoperable 10/10 (100.0%)10/10 (100.0%)-----
ReferencesRadiographic ResponseBiochemical ResponseTreatment OutcomeTreatment Toxicity
Authors, YearSubjects
Suitable for Evaluation n
Response CriteriaPD n (%)SD n (%)PR n (%)CR n (%)Subjects
Suitable for Evaluation n
Response Criteria (Calcitonin, CT)ResponsePFS MonthsOS MonthsDiscontinuation n (%)Grade III/IV Nephrotoxicity n (%)Grade III/IV Haemotoxicity n (%)
Caplin et al., 2000 [35]0-----1Normalisation1 (100%)NSNS0 (0%)0 (0%)0 (0%)
Valkema et al., 2002 [36]5SWOG3 (60%)2 (40%)0 (0%)0 (0%)5Markers (NS criteria) PD SD2 (40%) 3 (60%)NSNSNS0 (0%)NS
Buscombe et al., 2003 [37]2RECIST0 (0%)0 (0%)0 (0%)2 (100%)0--NSNS0 (0%)0 (0%)0 (0%)
Pasieka et al., 2004 [38]1“WHO modified” **1 (100%)0 (0%)0 (0%)0 (0%)1PD (increase ≥ 25% in basal value)
NO RESPONSE (none of the others)
PR (decrease ≥ 50% in basal value)
CR (normalisation)
1 (100%)
0 (0%)
0 (0%)
0 (0%)
NSNS0 (0%)0 (0%)0 (0%)
-8-4 (50.0%)2 (25.0%)0 (0%)2 (25.0%)7----0/4 (0.0%)0/10 (0.0%)0/4 (0.0%)
PRRT: peptide receptor radionuclide therapy, SSR: somatostatin receptor, 111In: 111 indium; SSA: somatostatin analogue, MTC: medullary thyroid carcinoma, F: female, M: male, GBq: GigaBecquerel, NS: not specified, PD: progressive disease, SD: stable disease, PR: partial response, CR: complete response, PFS: progression-free survival, OS: overall survival, SWOG: Southwest Oncology Group, RECIST: Response Evaluation Criteria In Solid Tumours, and WHO: World Health Organization. ** “WHO modified”, PD described as the “appearance of new lesions or increase of 25% or more in size of existing lesions”.
Table 3. PRRT targeting CCK2Rs in MTC.
Table 3. PRRT targeting CCK2Rs in MTC.
ReferencesDemographicsDisease DataTreatment Schedule and Follow-Up
AuthorsYearType of StudySubjects nAge-Years Mean (Median) (Range)F/M nMutation Status nDisease Status at BaselineSubjects with Metastases %RadioisotopeDose/Cycle mCi/m2Cycles nCumulative Dose-GBq Mean (Median) (Range)Follow-Up Months Mean (Median) (Range)
Behr and Behe [17]2002NS8NSNSNSProgressive and advanced8 (100%)90Y30–50≤ 4NSNS
ReferencesRadiographic ResponseBiochemical ResponseTreatment OutcomeTreatment Toxicity
Authors, YearSubjects
Suitable for Evaluation n
Response CriteriaPD n (%)SD n (%)PR n (%)CR n (%)Subjects
Suitable for Evaluation n
Response Criteria (Calcitonin, CT)ResponsePFS MonthsOS MonthsDiscontinuation n (%)Grade III/IV Nephrotoxicity n (%)Grade III/IV Haemotoxicity n (%)
Behr and Behe, 2002 [17]NSNS----0--NSNS0 (0%)0 (0%)3 (38%)
PRRT: peptide receptor radionuclide therapy, CCK2R: cholecystokinin 2 receptor, MTC: medullary thyroid carcinoma, F: female, M: male, GBq: GigaBecquerel; NS: not specified, 90Y: 90 yttrium; PD: progressive disease, SD: stable disease, PR: partial response, CR: complete response, PFS: progression-free survival, and OS: overall survival.
Table 4. PRRT targeting SSRs and CCK2Rs in MTC: registered clinical trials.
Table 4. PRRT targeting SSRs and CCK2Rs in MTC: registered clinical trials.
ClinicalTrials.gov IdentifierRadiopharmaceutical under StudyTrial NameStudy PhaseMedical Condition under InvestigationAssigned InterventionPrimary OutcomeEstimated Enrolment, nEstimated Study Completion DateTrial Status
NCT00002947111In-DTPA-D-Phe-OctreotideA Phase I Study of (111In-DTPA-D-Phe)-Octreotide in Patients with Refractory Malignancies Expressing Somatostatin ReceptorsPhase 1Refractory Malignancies Expressing Somatostatin Receptors *4 cycled of 111In-DTPA-D-Phe-Octreotide Determine the MTD, toxic effects, and the preliminary antitumor activity of indium in 111 pentetreotide.35 NATerminated (reason: NS)
NCT03647657177Lu-PP-F11N177Lu-PP-F11N in Combination With Sacubitril for Receptor Targeted Therapy and Imaging of Metastatic Thyroid Cancer (Lumed Phase 0/B)Early Phase 1Medullary Thyroid CancerIntravenous application of 2 × 1 GBq 177Lu-PP-F11N with and without co-medication with Sacubitril Tumour radiation doses [time frame: measurement up to 72 h after each injection of 177Lu-PP-F11N]; evaluation of the radiation doses in tumour tissue from MTC after injection of 177Lu-PP-F11N alone and in combination with Sacubitril 8 October 2021Recruiting
NCT02088645177Lu-PP-F11N177Lu-PP-F11N for Receptor Targeted Therapy and Imaging (Theranostics) of Metastatic Medullary Thyroid Cancer—a Pilot and a Phase I StudyPhase 1 **Medullary Thyroid CancerPhase 1: intravenous application of 3 x max. 15 GBq 177Lu-PP-F11N. All patients with or without Physiogel, depending on the results of the phase 0 study **Phase 1: MTD (time frame: up to 9 months) **Phase 1:
12–18 **
March 2022Recruiting
NCT04106843177-Lu-DOTA-Tyr3-OctreotateA Phase II Study to Evaluate the Effects of 177Lu-DOTATATE in Patients with Unresectable and Progressive Rare Metastatic Endocrine Carcinomas: Medullary Thyroid Cancer, Parathyroid Carcinoma, Pituitary Carcinoma, and Malignant Pheochromocytoma/ParagangliomaPhase 2Medullary Thyroid Cancer, Parathyroid Carcinoma, Pituitary Carcinoma, and Malignant Pheochromocytoma/Paraganglioma177Lu-DOTATATE intravenously (IV) over 30 min every 8–16 weeks. Treatment continues for up to 52 weeks in the absence of disease progression or unacceptable toxicityRadiographic Response (time frame: up to 52 weeks)50January 2022Not yet recruiting
PRRT: peptide receptor radionuclide therapy, SSR: somatostatin receptor, CCK2R: cholecystokinin 2 receptor, MTC: medullary thyroid carcinoma, MTD: maximum tolerated dose, DTPA: diethylenetriaminepentacetic acid, and NA: not applicable. * Including head and neck cancers. ** The study includes a phase 0 whose results have already been published [20].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grossrubatscher, E.; Fanciulli, G.; Pes, L.; Sesti, F.; Dolci, C.; de Cicco, F.; Colao, A.; Faggiano, A.; NIKE Group. Advances in the Management of Medullary Thyroid Carcinoma: Focus on Peptide Receptor Radionuclide Therapy. J. Clin. Med. 2020, 9, 3507. https://doi.org/10.3390/jcm9113507

AMA Style

Grossrubatscher E, Fanciulli G, Pes L, Sesti F, Dolci C, de Cicco F, Colao A, Faggiano A, NIKE Group. Advances in the Management of Medullary Thyroid Carcinoma: Focus on Peptide Receptor Radionuclide Therapy. Journal of Clinical Medicine. 2020; 9(11):3507. https://doi.org/10.3390/jcm9113507

Chicago/Turabian Style

Grossrubatscher, Erika, Giuseppe Fanciulli, Luca Pes, Franz Sesti, Carlotta Dolci, Federica de Cicco, Annamaria Colao, Antongiulio Faggiano, and NIKE Group. 2020. "Advances in the Management of Medullary Thyroid Carcinoma: Focus on Peptide Receptor Radionuclide Therapy" Journal of Clinical Medicine 9, no. 11: 3507. https://doi.org/10.3390/jcm9113507

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop