Skip to main content
Erschienen in: Endocrine 1/2024

Open Access 18.10.2023 | Review

Cabozantinib for different endocrine tumours: killing two birds with one stone. A systematic review of the literature

verfasst von: Elena Zago, Antonio Galluzzo, Silvia Pradella, Lorenzo Antonuzzo, Mario Maggi, Luisa Petrone, Clotilde Sparano

Erschienen in: Endocrine | Ausgabe 1/2024

Abstract

Purpose

Cabozantinib is an oral multi-tyrosine kinase inhibitor (TKI) that has been approved in Europe for advanced renal cell carcinoma, hepatocellular carcinoma, locally advanced and metastatic medullary thyroid carcinoma (MTC) and radioiodine-refractory differentiated thyroid cancer. Merkel cell carcinoma (MCC) is a rare and highly aggressive cutaneous malignant neuroendocrine tumour that usually presents in sun-exposed skin areas of immunosuppressed patients. Conflicting data exist about cabozantinib for MCC and this TKI is currently under investigation in several onco-endocrine frameworks.

Methods

We herein report a case of an 83-year-old man who was diagnosed with MCC during the treatment of an advanced metastatic MTC. The diagnosis of MCC was established based on clinical, histopathologic evaluation and immunohistochemistry. A systematic review of the literature on cabozantinib use for advanced endocrine and neuroendocrine tumours has been performed.

Results

The patient was initially treated with surgery and adjuvant radiotherapy. Cabozantinib was therefore started to control both MTC and MCC. After 24 months, no sign of local or metastatic MCC relapse was evidenced.

Conclusion

Promising data on cabozantinib treatment for endocrine and neuroendocrine neoplasms is recently emerging in the literature. In our clinical case, we reported that, besides the good response for the MTC, cabozantinib also seems to effectively control metastatic MCC, along with efficient surgery and adjuvant radiotherapy. Further investigations are needed to determine the efficacy and safety of cabozantinib in MCC patients and in off-label endocrine tumours.
Hinweise

Supplementary information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12020-023-03526-0.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

The therapeutic breakthrough in the field of endocrine tumours has been the introduction of tyrosine-kinase inhibitors (TKIs) for several advanced carcinomas. Among the available drugs, cabozantinib (CBZ) has increasingly drawn attention for its wide range of benefits in progressive thyroid cancers and beyond. CBZ is an oral small molecular inhibitor of several tyrosine kinases, such as vascular endothelial growth factor receptor 2 (VEGFR2), tyrosine-protein kinase receptor UFO (AXL), mesenchymal-epidermal transition factor (MET), and rearranged during transfection (RET). Targeting intracellular key pathways is essential to hamper tumour survival, angiogenesis, epithelial-to-mesenchymal transition and other metastatic processes [1]. In particular, while VEGFR inhibition alone can still allow tumour spread, due to the activation of the parallel hypoxia-induced MET signalling, this TKI can concurrently block both pathways, resulting in enhanced disease control (Fig. 1) [1]. As regarding thyroid tumours, CBZ was firstly approved for locally advanced and metastatic medullary thyroid cancer (MTC) and later on, supported by the promising data from phase III COSMIC-311 trial, as second-line therapy for progressive radioiodine-refractory differentiated thyroid cancer (RAIR-DTC) [2, 3]. Out of on-label context, this TKI is currently under consideration for several endocrine tumours, showing interesting results. Although a few data are available, pheochromocytoma/paraganglioma (PPGL), adrenocortical carcinoma (ACC), neuroendocrine neoplasms (NENs) and Merkel cell Carcinoma (MCC) represent orphan diseases, that might benefit from this broad-based targeted therapy. Finally, CBZ represents an effective therapeutic weapon on syndromic tumours within the framework of multiple endocrine neoplasia type 2 (MEN2), a model of endocrine cancers with different primary sites, but with a common molecular signature (i.e. RET mutations).
To support those considerations, we hereby present a rare case of MTC and metachronous MCC, both with long disease control under CBZ.
Starting from this case report a systematic revision of current literature has been performed to summarise the available data or ongoing studies, exploring the use of CBZ in the endocrine tumours of various origins and histotypes.

Patients and methods

The patient gave his informed consent for the present case report.

Electronic literature search strategy

Systematic research of articles published between 2009 and 2023 on MEDLINE (PubMed), Scopus and EMBASE was performed. For search strategy and search terms refer to review protocol (Supplementary File 1). Reference lists were manually screened for further relevant articles. The search strategy was based on the PICO approach (a standardised way of defining research questions, focusing on Patients, Intervention, Comparison, and Outcome) [4]. The review was registered with PROSPERO registration number CRD42023403886 (https://​www.​crd.​york.​ac.​uk/​prospero/​).

Data collection and analysis

Titles and abstracts were first reviewed. Eligible studies were assessed on the basis of their full text and referenced using Zotero Software v6.0.26 (Zotero.org). Preferred Reported Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were followed (see Supplementary File 2) [5].

Results

Case presentation

A 79 years old man was evaluated in our tertiary referral centre in 2018 for persistently high Carcinoembryonic Antigen (CEA) levels (up to 63.3 ng/mL), observed during the follow-up for a history of colorectal adenocarcinoma (pT3pN2aM0, stage IIIB [6]), previously classified as recovered in 2017, after surgery and adjuvant chemotherapy.
An increased glucose uptake in the thyroid right lobe on 18Fluorodeoxyglucose-Positron emission tomography (FDG-PET) was revealed and neck ultrasound confirmed the presence of a 14 mm suspicious nodule on the right thyroid lobe without suspect lymph nodes in the lateral cervical districts. The following fine needle aspiration showed a TIR4/Suspicious for malignancy cytology, according to the Italian classification system for thyroid cytology [7]. Preoperative plasma calcitonin (plCT) levels resulted in 1350 pg/mL, thus suggesting the MTC diagnosis. In May 2018, the patient underwent total thyroidectomy and cervical central lymph node dissection, and the histology confirmed the diagnosis of a stage IVA MTC [pT1b (m), pN1a], according to TNM 2017 edition VIII [8]. The patient was initially classified as M0, due to the absence of distant metastases at computer tomography (CT) and FDG-PET studies. Genetic screenings for Multiple Endocrine Neoplasia 2 (MEN2) and Familial Medullary Thyroid Cancer (FMTC) were negative, but somatic RETM918T mutation was detected in the tumour sample.
After surgery, he didn’t reach the biochemical cure. Due to a higher risk of recurrence [9], he underwent strict radiological and biochemical monitoring and a progressive increase of plCT up to 5864 pg/mL and CEA up to 104.7 ng/mL was observed. A neck ultrasound revealed the presence of right lateral-cervical round lymph nodes suspicious for MCT locoregional metastases. Moreover, the total body CT scan showed some focal hepatic lesions in IV, VIII and VI liver segments, up to about one centimetre. A liver biopsy then confirmed the MTC origin of the metastatic lesion in November 2019.
Due to the quick progression and the multiple metastatic sites, systemic treatment was established and, despite the excellent global performance status according to Eastern Cooperative Oncology Group Performance Status (ECOG PS), considering the patient’s age, his body mass index of 25 kg/m2 and the clinical history, a first-line therapy with vandetanib at the personalised dose of 200 mg daily was started. The patient was systematically evaluated from biochemical, radiological and clinical perspectives, and adverse events (AE) were classified according to Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 [10]. This treatment was poorly tolerated, with a grade (G) 3 asthenia and haematologic toxicity, requiring therapeutic pauses and dose reductions. However, it allowed a morphologic stabilisation of the disease, and a biochemical response with a plCT and CEA nadir of 1280 pg/mL and 61 ng/mL, respectively.
In May 2020, a new-onset right inguinal swelling was detected at the CT scan, during MTC follow-up. Owing to the uncommon tumour progression a focused histological insight was decided and the biopsy revealed a lymph node metastasis from a Merkel Cell Carcinoma (MCC). In fact, immunohistochemistry was positive for CK20, CgA, Neuron Specific Enolase (NSE) and CD56. The following investigations by FDG-PET and a new CT scan revealed a primary homolateral MCC on the right leg with locoregional lymph node metastases up to 27 mm. The biochemical assessment revealed a Chromogranin A (CgA) level of 104.9 ng/mL (range 0.0–76.3 ng/ml). Furthermore, a total body (TB) CT scan performed in September 2020, showed: stable millimetric focal hypodense hepatic lesions in IV, VIII and VI liver segments, but two new onset right lateral-cervical lymph node metastases (with anteroposterior axis of 11 and 9 mm) with contrast enhancement, and some bone rarefaction areas in the 7th and 9th thoracic vertebrae and in the 2nd lumbar vertebra, all suspicious for MTC origin; multiple external iliac and inguinal lymph nodes metastases (the largest lesions presented anteroposterior axes of 27 × 18 mm and 19 × 17 mm, respectively) of MCC origin. The patient underwent surgery on the primary MCC, and adjuvant locoregional radiotherapy (42 Gray on inguinal and external iliac lymph nodes) was performed. The MCC was finally classified as a clinical-stage III, due to the presence of in-transit metastases with lymph node metastases (i.e. N3 category), and accordingly, the risk of recurrence resulted as high with an estimated 5-year overall survival <30% [11]. PlCT, CEA and CgA were 6120 pg/mL, 74.1 ng/mL and 76.5 ng/mL, respectively. Due to the bad endurance and the actual diagnosis, vandetanib was definitely withdrawn.
In November 2020, considering the global oncological history with two aggressive and active endocrine tumours, second-line therapy with CBZ to control both MTC and MCC was started at the dosage of 100 mg daily but soon interrupted for a week and then titrated to 60 mg daily due to several AEs [weight loss (G2), fatigue (G1-G2), dysgeusia (G1), mild hypocalcaemia (G1), elevated blood pressure (G1), and moderate lymphopenia (G3)].
In 2021, three years after the MTC diagnosis, one year after the MCC diagnosis and three months after the switch from vandetanib to CBZ, the following TB-CT scan showed partial response disease: the previously described hepatic lesions, after an initial pseudo-progression (due to the mild swelling by drug-induced necrosis), were no longer evidenced, and the MCC iliac lymph nodes underwent a gradual downsizing, up to the complete response, according to the Response Evaluation Criteria in Solid Tumours (RECIST version 1.1) [12]. Bone metastases showed a < 20% decrease in dimension on PET-FDG and MRI. PlCT was 1010 pg/mL and CgA 137.5 ng/mL (under proton-pump inhibitor therapy).
In 2022, CBZ was again temporarily disrupted due to G4 leucopenia and G1 thrombocytopenia. Infective diseases and concomitant haematologic causes of pancytopenia were excluded. After a break of one month, the patient was in good clinical condition (ECOG PS was 0); blood pressure was controlled without medication, QTc interval was in range and the blood count was stable with only mild lymphopenia (G1), CBZ was started again at the personalised dose of 40 mg daily.
Nowadays, after four years from the MTC and two years from the MCC diagnosis, the patient, aged 83 years old, appears in good general condition (ECOG PS 0) with an excellent tolerance of this treatment, despite his age. Mild leukopenia (G1) is still present, while platelets are within the normal range. At the CT scan, MTC sustained the previous stable disease response, and no sign of local or metastatic relapses of MCC was evidenced, configuring a complete response according to RECIST criteria (Fig. 2). At the present time, plCT is 1680 pg/mL; CEA 54.8 ng/mL and CgA 275 ng/mL. Figure 3 shows the patient’s plCT and CEA monitoring before and during treatment.

Systematic literature revision

We screened 1913 papers in the primary search. One manuscript has been manually added. After the exclusion of duplicates, studies published in other languages than English, studies involving the paediatric population and studies with different topics, 426 citations were screened for eligibility criteria on an abstract basis. 119 articles were finally analysed with a full-text review, and 26 studies were included in the analysis (Table 1). Figure 4 provides a flowchart of the literature search.
Table 1
List of included studies
Condition
PI
PY
Study design
Phase
Enrolled patients
Dose (mg/daily)
DTC
Cabanillas
2014
Clinical Trial
I
15
140
DTC
Shah
2015
Clinical Trial
II
25
60 or 80 (if tolerated)
DTC
Konda
2017
Prospective analysis on clinical trial
II
25
60 or 80 (if tolerated)
DTC
Brose
2021
Clinical Trial
III
187
60
DTC
Taylor
2022
Clinical Trial
Ib
102
40 or 60 (plus atezolizumab)
MTC
Elisei
2013
Clinical Trial
III
330
140
MTC
Bentzien
2013
Preclinical
preclinical
biochemical assays/mice model
N/A
MTC
Rinciog
2014
Adjusted indirect comparison
indirect comparison
N/A
N/A
MTC
Sherman
2016
Correlative analysis
indirect comparison
330
140
MTC
Schlumberger
2017
Clinical Trial
III
330
140
MTC
Koehler
2021
Real World Comparative Study
N/A
48
N/A
MTC
Capdevila
2022
Clinical Trial
IV
247
60 vs 140
MTC (+ other cancers)
Kurzrock
2011
Clinical Trial
I
37, (85)
MTD (175)
MCC
Rabinowits
2018
Clinical Trial
II
8
60 (40 or 20 if not tolerated)
MCC
Tarabadkar
2018
Case Series
N/A
5
60 reduced to 40
PPGL
Roman
2017
Review
N/A
N/A
N/A
PPGL
Jimenez
2018
Expert Opinion
N/A
N/A
N/A
PPGL
Economides
2020
Case Report
N/A
1
60/40
GU Tract Tumours
Apolo
2020
Clinical Trial
I
54
40
ACC
Phan
2015
Preclinical
N/A
biochemical assays/mice model
N/A
ACC
Kroiss
2020
Retrospective cohort study
N/A
16
60/100/140/20
ACC
Miller
2020
Retrospective cohort study
N/A
15
N/A
NEN
Sennino
2012
Preclinical
preclinical
biochemical assays/mice model
N/A
NEN
Reuther
2016
Preclinical
preclinical
biochemical assays/mice model
N/A
NEN
Chan
2018
Clinical Trial
II
35
60
NEN
Grillo
2018
Review
N/A
N/A
N/A
PI Principal Investigator, PY Publication Year, DTC Differentiated Thyroid Cancer, MTC Medullary Thyroid Carcinoma, MCC Merkel Cell Carcinoma, PPGL Pheochromocytoma and Paraganglioma, GU genitourinary, ACC Adrenocortical Carcinoma, NEN Neuroendocrine Neoplasm, N/A not applicable, MTD maximum tolerated dose

Thyroid cancers

Use of CBZ in MTC

MTC is a rare neuroendocrine disease arising from parafollicular C-cells of the thyroid, thus producing CT. It encompasses about 2–5% of all thyroid cancers, occurring as part of MEN2 syndrome in about 25% of cases and sporadically in 75% [13]. RET mutations can be detected at a germline level in familial forms, as well as at a somatic level in up to 50–90% of histological samples, according to literature results [14]. Irrespective of RET mutation status, MTC discloses poor prognosis in the event of distant metastases at diagnosis or during the follow-up [13]. At early stages, complete surgical resection represents a curative treatment; while the management of advanced and progressive MTC remains challenging [15]. Cytotoxic chemotherapy and radiotherapy showed limited efficacy, due to different mechanisms, including higher expression of Multi Drug Resistant 1 (MDR-1) proteins and enhancement of Heat Shock Protein pathways [16]. By reason of the low PDL1 expression on C-cells, disappointing results have been observed with immune-checkpoint inhibitors [17]. Besides, experience with peptide receptor radionuclide therapy (PRRT) is still limited, and an objective response is observed in only 10–15% of patients [18]. The best results in the treatment of advanced and progressive MTC came from TKI drugs. In particular, vandetanib and CBZ (Cometriq®), had been approved by the United States Food and Drug Administration (FDA) in 2011 and 2021, and by the European Medicine Agency (EMA) in 2012 and 2022, respectively. From a molecular perspective, in vitro and in vivo studies on mouse models demonstrated that, besides the robust MET and VEGFR2 antagonism, CBZ inhibits both the wild type and the mutated forms of RET, preventing MTC cells and xenograft from growing and metastasising [19]. As regards clinical trials, in phase I (Study of XL184 (Cabozantinib) in Adults With Advanced Malignancies) and III trials (EXAM trial), CBZ showed an acceptable safety profile (Table 2), a longer progression-free survival (PFS) (11.2 months versus 4.0 months in the placebo arm, p < 0.001) and overall survival (OS) (26.6 versus 21.1 months, although the difference did not reach statistical significance, p = 0.24) [20, 21]. These promising results were obtained mainly in patients with RETM918T mutation (which was present in almost 65–70% of the tumours) [22]. In fact, according to the exploratory analysis of this study, RETM918T subgroup achieved the greatest observed PFS benefit for CBZ versus placebo (HR, 0.15; 95% CI, 0.08–0.28; P < 0.0001) [23]. Furthermore, an indirect treatment comparison between CBZ and vandetanib showed a positive trend in favour of CBZ in terms of PFS [24]. As regarding adverse effects, CBZ showed a dose-dependent relation but the EXAMINER trial failed to prove the non-inferiority of the 60 mg/day tablets versus the 140 mg/day capsules [25]. Moreover, Koehler et al. study found that both PFS and OS were significantly longer in younger patients experiencing more than five AEs, thus suggesting a dose-related correlation with the tumour response [26]. In conclusion, in MTC, CBZ appeared to be effective in prolonging survival, although many patients often do not receive full dosage or need a discontinuation of the treatment due to poor tolerance or toxicities (Table 2).
Table 2
Cabozantinib related adverse events [21, 34, 83]
Frequent AEs (>50%)
All grades (%)
G3 (%)
Common AEs (50-10%)
All grades (%)
G3 (%)
Not common AEs (<10%)
All grades (%)
G3 (%)
Cardiovascular
hypertension
33–53
8.4–20
   
pulmonary embolism
3.3
0
Dermatological
palmar-plantar erythrodyesthesia
40–53
13–12
rash
19.2–23
0.9
   
mucositis
20–53
0–3.3
dry skin
19.2–20.1
0–2.8
   
Gastrointestinal
diarrhoea
52–87
7.3–21
dysgeusia
26–45
0.1–0.5
   
decreased appetite
49–80
7–13
constipation
26–34
0-1
   
nausea
43–80
0–6.9
stomatitis
19–30
1.4–2.6
   
vomiting
24–60
2.3–7
      
Constitutional
fatigue
41–80
7–13
arthralgia
14
2.3
   
weight decrease
35–73
3.4–13
myalgia
7–27
1
   
   
insomnia
10.7
0
   
   
pruritus
13.6
0.5
   
   
headache
13–18.2
0.4–0.5
   
   
alopecia
16.4
0
   
   
hair colour changes
33–34
0.5
   
Renal
   
proteinuria
47
7
   
Lab alteration
incresed AST
10–100
7–3.2
hypophosphatemia
20
13
increased ALP
3
0
increased ALT
24–73
7-5
hyponatremia
13–33
7–27
   
increased LDH
73
7
      
hypocalcemia
23–60
13-0
      
hypokalemia
47
13-3.8
      
Haematologic
   
anaemia
20–31
7
   
   
leukopenia
27
7
   
Respiratory tract
   
pneumonia
4.2–13
13
   
   
dyspnoea
13.6–21
2.3–3.5
   
   
dysphonia
21–27
0–10.1
   
Thyroid
   
thyroid disfunction
14
0
   
AE Adverse Event, AST Aspartate Aminotransferase, ALT Alanine Aminotransferase, LDH Lactate Dehydrogenase, ALP Alkaline Phosphatase

Use of CBZ in RAIR-DTC

DTCs, which account for about 90% of all thyroid cancers, usually have favourable long-term prognosis following surgery±radioiodine treatment, with mortality rates ranging from 0.3–0.5/100,000 population [27, 28].
However, two-thirds of patients with recurrent or metastatic DTC are RAIR with a 10-year survival rate of only 10% after diagnosis, resulting in a challenge for physicians [29].
While indolent and oligometastatic RAIR-DTC can be followed up with suppressive levothyroxine dose alone; symptomatic, progressive low-burden disease may benefit from locoregional therapies i.e., surgery, external radiotherapy, or thermal ablation. The major challenge lies in rapidly progressive and inoperable cases or high-burden metastatic RAIR-DTC, which requires systemic therapies. In those patients, the treatment with cytotoxic chemotherapy showed disappointing results, with selective benefits only in patients who fail to respond to TKIs (data are mainly anecdotal) [30]. Hence, TKIs are considered in the event of disease progression, mainly after surgical and radiation therapy approaches [31]. Until fairly recently, lenvatinib, a wide spectrum anti-angiogenetic TKI, was the only available option for advanced RAIR-DTC and, at the time of progression to this drug, the therapeutic chances were poor [32]. In 2021 and 2022, CBZ (Cabometyx®) was finally approved by FDA and EMA for RAIR-DTC patients, who disclosed progressive disease after prior treatments with VEGFR inhibitors. This endorsement was based on the promising data from the COSMIC-311 study: a multicentral, randomised, double-blind trial in which patients, with locally advanced or metastatic RAIR-DTC, progressing during or after treatment with at least one VEGFR-targeting TKI, were treated with either CBZ 60 mg orally once daily or placebo [3]. The median PFS was 11.0 months [95% confidence interval (CI), 7.4–13.8] in the CBZ arm compared with 1.9 months (95% CI, 1.9–3.7) in the control arm [3]. The final analysis of COSMIC-311 with longer follow-up confirmed the superiority of CBZ versus placebo with a manageable safety profile [3]. The PFS benefit was consistent with the interim analysis and irrespective of prior VEGFR-targeted therapy [33]. Phase I and II trials that preceded the COSMIC trial showed that CBZ presented a safety profile similar to other multitargeted VEGFR and clinical and statistically significant activity in DTC patients who have progressed on prior VEGFR targeted therapies [3436]. Furthermore, Konda et al. performed a prospective analysis as part of a multicenter International Thyroid Oncology Group phase II study of CBZ in patients with RAIR-DTC with bone metastases who progressed on prior VEGFR‐targeted therapy: CBZ was associated with a highly significant reduction in all bone turnover markers (even when they were within normal limits at baseline) and a minor decreases in bone lesion uptake was noted on Sodium Fluoride PET in four out of six patients [37]. Finally, CBZ provided the same efficacy advantage when in monotherapy and when associated with immune-checkpoint inhibitors such as nivolumab and ipilimumab, while it showed a durable response and high rate of disease control when in combination with atezolizumab [38, 39]. In 2018, Brose et al. presented the first study documenting CBZ anti-tumoral activity in patients with RAIR-DTC as a first-line therapy [40]. A phase I trial on the simultaneous use of CBZ and PD-1 inhibitors in HIV-positive patients is now recruiting (Table 3 - https://​clinicaltrials.​gov).
Table 3
List of active trials
Condition
Identifier
Status
Study Start Date
Study Completion Date
Ph
EP
Primary Objectives
Drugs
RAIR- DTC
NCT05660954
not yet recruiting
April, 2023
June, 2025
II
41
Gene expression
cabozantinib
DTC
NCT03914300
active, not recruiting
July 15, 2019
January 15, 2024
II
27
ORR
cabozantinib, nivolumab and ipilimumab
RAIR- DTC
NCT02041260
active, not recruiting
January 2014
May 2022
II
43
AEs
cabozantinib
Advanced DTC (+others)
NCT04514484
recruiting
November 4, 2020
November 2, 2025
I
18
DLTs
cabozantinib and nivolumab
DTC (+others)
NCT03170960
active, not recruiting
September 5, 2017
August 2024
Ib
1732
MTD
cabozantinib and atezolizumab
PPGL
NCT02302833
recruiting
February 17, 2015
December 31, 2023
II
22
ORR
cabozantinib
ACC
NCT03612232
recruiting
June 4, 2019
April 30, 2025
II
37
PFS
cabozantinib
ACC
NCT03370718
active, not recruiting
February 26, 2018
February 28, 2026
II
18
PFS
cabozantinib
NEN
NCT03375320
recruiting
July 18, 2018
October 1, 2025
III
395
PFS
cabozantinib
NEC
NCT04079712
active, not recruiting
January 14, 2020
October 1, 2023
II
30
ORR
cabozantinib, nivolumab, and ipilimumab
NEN
NCT04412629
recruiting
November 24, 2020
July 31, 2025
II
32
ORR
cabozantinib
NEN
NCT04893785
recruiting
June 15, 2021
December 2024
II
35
ORR
cabozantinib, temozolomide
NEN
NCT04427787
recruiting
June 20, 2020
November 2023
II
69
ORR
cabozantinib, lanreotide
NEN
NCT05249114
recruiting
December 28, 2022
December 1, 2031
Ib
90
MTD
cabozantinib, PRRT
Carcinoid Tumours
NCT04197310
active, not recruiting
December 26, 2019
December 26, 2023
II
35
ORR
cabozantinib, nivolumab
NEN, PPGL, ATC, Adenocarcinoma
NCT04400474
active, not recruiting
October 7, 2020
March 2024
II
93
ORR
cabozantinib, atezolizumab
NEN
NCT05048901
recruiting
September 17, 2021
December 31, 2025
I/II
49
MTD, PFS
cabozantinib
NEN G3
NCT04524208
recruiting
March 1, 2021
June 30, 2023
II
40
DCR
cabozantinib
NEN G3
NCT05289856
recruiting
March 28, 2022
December 1, 2025
II
30
DCR
cabozantinib, avelumab
RAIR Radioiodine-Refractory, DTC Differentiated Thyroid Carcinoma, PPGL Pheochromocytoma and Paraganglioma, ACC Adrenocortical Carcinoma, NEN Neuroendocrine Neoplasm, ATC Anaplastic Thyroid Carcinoma, NEC Neuroendocrine Carcinoma, Ph Phase, ER Enrolled Patients, DLTs Dose Limiting Toxicities, ORR Overall Response Rate, PFS Progression Free Survival, MTD Maximum Tolerated Dose, DCT Disease Control Rate, G grade

Use of CBZ in MCC

MCC is a rare, highly aggressive neuroendocrine skin tumour that usually develops in sun-exposed areas [41]. Despite major advantages in understanding its carcinogenesis process, the MCC origin has not been completely understood: on one hand, Merkel cell precursor has been speculated as a source of MCC (normal Merkel Cells are terminally differentiated, thus allegedly not able to devolve into cancer); on the other hand, pro-B/pre-B lymphocytes, fibroblasts, dermal mesenchymal stem cells or aberrant keratinocytes have also been suspected as oncological precursors [42].
Elderly subjects with a story of sun exposition and/or immunosuppressant therapy are at higher risk for this disease [43, 44]. Local and regional lymph node metastasis can occur at the early stages, and the long-term prognosis is poor, dropping to 14% at five years from the diagnosis in the event of metastatic disease [11, 45]. Whether possible, first-line therapy encompasses a wide tumour excision, along with sentinel lymph node dissection and/or lymphadenectomy. High-dose adjuvant radiotherapy (50–60 Gray) to the primary site is usually delivered during the post-operative takeover, but this treatment also seems beneficial for locoregional control in patients with unresectable tumours [45, 46]. The historically favoured systemic therapy consisted of carboplatin/cisplatin-etoposide chemotherapy, while any second lines were based on anthracyclines, cyclophosphamide, vincristine, bleomycin, and 5-fluorouracil regimens [47]. Although cytotoxic chemotherapy has an objective response rate of >50%, responses are rarely durable with a median PFS of only three months [44]. Moreover, several safety concerns are well-known, making the traditional regimens mostly reserved for palliative care, i.e. patients with advanced or locally recurrent MCC [48]. During the last few years, thanks to the advancement in immunotherapy, different molecules have been proposed: immune-checkpoint inhibitors (PD-1 and PDL-1 ligand inhibitors) are the favoured agents. In particular, first-line therapy with avelumab showed a PFS in up to 72% of the cases in an international multicentre phase II trial (JAVELIN Merkel 200) [49]. Despite the success of immune therapy, a considerable proportion of patients with metastatic MCC do not respond to PD-1/PD-L1 blockade [49]. In this setting, TKIs have been proposed due to their capability of targeting angiogenesis and tumour-spreading processes as MCC expresses VEGF-A, VEGF-C, VEGF-R2, and platelet-derived growth factor (PDGF)-b in 91, 75, 88 and 72%, respectively [50, 51]. Despite these promising data, very little literature regarding anti-VEGF therapy on MCC is available: Tarabadkar described five cases of successful use of anti-VEGF TKIs such as pazopanib and CBZ in patients with advanced MCC [52]. In particular, a single case report of a 3.5-year PFS in a patient receiving CBZ after platinum-based chemotherapy has been described. Nonetheless, Rabinowits et al. failed to demonstrate the efficacy of single-agent CBZ and the 8 patients-trial prematurely failed, because of the lack of efficacy and toxicity [53]. However, all the included patients have been already pre-treated with heavy chemotherapy regimens - such as platin-etoposide - and received CBZ as a second or further line, which could have affected the safety and the tolerance to this drug. After all, no trial on CBZ in MCC is currently ongoing, since the phase II study on CBZ in recurrent MCC (NCT02036476), failed for lack of recruital.

Use of CBZ in PPGLs

Pheochromocytomas and paragangliomas (PPGLs) represent a rare group of neoplasia issued from chromaffin cells. In particular, pheochromocytomas arise from the adrenal medulla, while extra-adrenal lesions develop from the extra-adrenal paraganglia.
Surgical removal is the therapy of choice in PPGLs with either a curative intent in non-metastatic disease or, in metastatic disease, to minimise symptoms derived from catecholamine excess and from compression of the surrounding structures [54]. Surgical-related morbidity, however, needs to be carefully evaluated in particular in head and neck non-functioning PPGL. In fact, the closeness to cranial nerves and vases often induces to favour a non-surgical approach, such as watchful waiting or radiosurgery (gamma-knife/cyberknife), in the event of compression of the surrounding structures [55]. Aside from radiotherapeutic options, the only officially approved treatment for advanced tumours is the high-specific iodine-131-meta-iodobenzylguanidine [(131 I)MIBG] therapy (Ultratrace), while chemotherapy and different targeted therapies are widely used outside controlled clinical trials [5658]. Recently, TKIs have been in the spotlight, due to their ability to inhibit tumour neoangiogenesis. Furthermore, they also seemed to play a role in the bone microenvironment, by interfering with bone turnover, thus reducing eventual pain [59].
So far, the best studied TKI is sunitinib, which seems to have better efficacy in RET and SDHx mutated patients: in a large prospective phase II multicentre study, the total disease control rate (including stable disease or partial response) in patients with germline mutations (SDHA, SDHB, RET) was 83% (95% CI: 61–95%) [60]. At the moment, sunitinib is under investigation in the first randomised placebo-controlled phase II clinical trial in advanced PPGLs (FIRSTMAPPP, NCT01371202). Despite the promising data on sunitinib, other TKI such as CBZ have not yet been completely studied. In fact, in the literature, there is a case report of a 32-year-old male patient suffering from an advanced metastatic paraganglioma, showing a progressive disease when enrolled in a CBZ-based trial as a monotherapy treatment. Nonetheless, he showed prolonged disease control with a significant tumour reduction and decrease in metastatic burden when enrolled in the study by Apolo (started on CBZ plus nivolumab) [61, 62]. A phase II clinical trial on CBZ in monotherapy is now recruiting patients with unresectable metastatic paraganglioma or pheochromocytoma (Table 3 - https://​clinicaltrials.​gov). Preliminary results on patients treated with 60 mg daily (titrated down to 40 or 20 mg depending on the tolerability) showed tumour mass shrinkage and stability of the disease [63]. Moreover, a phase II trial on the combination therapy (CBZ plus atezolizumab) in patients suffering from advanced and progressive neoplasms of the endocrine system is ongoing, although not actively recruiting (Table 3 - https://​clinicaltrials.​gov).

Use of CBZ in ACC

Adrenocortical carcinoma (ACC) is a rare tumour arising from the adrenocortical gland, usually producing steroid hormones [64]. The prognosis is often poor since patients at low risk of recurrence are the minority and they can also frequently further develop recurrent disease [65]. In localised stages, complete surgical resection is the treatment of choice, while mitotane is the only approved drug that showed efficacy in prolonging recurrence-free survival when administrated as adjuvant treatment in patients at higher risk of recurrence [66]. Other adjuvant therapies, including chemotherapy, had been under investigation: although the European Society of Endocrinology did not reach a definitive consensus on adjuvant use of cytotoxic drugs for ACC, based on phase III trial, the combination of etoposide, doxorubicin and cisplatin (EDP) with mitotane (EDP-M) is recommended in patients with very high risk of recurrence [67]. Although the rates of response and PFS were significantly better with EDP-M treated patients, the study failed to show a significant difference in terms of overall survival [67]. In this setting, TKIs have been considered as a second-line therapy for patients who showed disease progression under EDP-M [68]. The rationale for the use of TKIs is based on the study of Pnan et al. who demonstrated an increased phosphorylation of c-MET in the immunohistochemistry of ACC tissue samples compared to adrenocortical adenomas and normal cortex, thus speculating that targeting c-MET could provide a breakthrough in the management of these aggressive diseases [69]. Moreover, an in vivo experiment in a mouse model confirmed that the knockout of c-MET inhibited adrenocortical carcinoma growth, tumour-related angiogenesis, chemotherapy resistance and cell survival, further suggesting this molecular pathway as a valuable therapeutic target for adrenocortical carcinoma [69]. Based on those promising preclinical data, a retrospective cohort study on CBZ use in patients with progressive disease under mitotane or cytotoxic chemotherapy regimens has been performed: best response was partial response in three out of 16 subjects, stable disease in five and progressive disease in eight patients. Although eight subjects (50% of the patients) showed tumour progression, this result was considered favourable (PFS was 16 weeks and OS was 58 weeks), compared to other treatments that failed to demonstrate a prolonged overall survival [68, 70]. A single institution experience showed a stable disease lasting six months in one patient treated with CBZ after previously progressing through chemotherapy and pembrolizumab [71].
To date, there is a phase II active trial actively recruiting patients suffering from unresectable or metastatic ACC in order to assess the efficacy and safety of CBZ (Table 3- www.​ClinicalTrials.​gov).

Use of CBZ in NENs

Neuroendocrine neoplasms (NENs) are a heterogeneous group of neoplasms originating from neuroendocrine cells localised in different organs, mostly within the digestive tract and in the lungs [72]. Therapeutic options rely on the tumour’s primary site, morphology, grade and stage, encompassing the “watchful and wait” attitude, surgical resection, locoregional treatment, or systemic therapies, including somatostatin receptor analogues, targeted therapies or traditional chemotherapy, in the event of advanced and progressive disease not responding to other treatments [7375]. As regards multi-TKIs, sunitinib is the only one that obtained EMA and FDA approval for pancreatic NENs, while other TKIs are currently under evaluation [76]. In particular, Reuther and colleagues found out that, while c-MET inhibition alone is not sufficient to exert direct antitumor or antimigratory effects in NEN cells, the multi-TKIs CBZ, as well as tivantinib, disclose therapeutic efficacy, probably due to the suppression of other molecular pathways [77]. Preclinical studies showed a reduced invasion and metastatic activity in mice with pancreatic NEN treated with CBZ: in particular, the simultaneous inhibition of VEGFR and MET pathways resulted in a smaller tumour mass and no metastases for CBZ-treated mice, compared to only anti-VEGF-treated mice [78]. The multi-TKI CBZ has also shown promising antitumor activity in a preliminary report of a phase II trial for pancreatic and extra-pancreatic NENs, showing a PFS of 21.8 months and 31.4 months in patients with pancreatic and non-pancreatic NENs, respectively [79]. Based on those results, five clinical trials are actively recruiting while three more trials are still active but not recruiting (Table 3 - www.​ClinicalTrials.​gov) [80].

Discussion

To the best of our knowledge, very few data on CBZ in endocrine and neuroendocrine tumours are available so far, while it is widely agreed that TKI improved MTC and differentiated thyroid cancer’s natural history. In the present case report, CBZ, along with a multimodal therapeutic approach, including surgery and adjuvant radiotherapy, showed a 24-month PFS from both MTC and MCC in an 83-year-old man. Of note, this patient disclosed two highly aggressive endocrine tumours, i.e. IV-stage MTC and a high-recurrence risk MCC with an estimated 5-year survival of <30%. For these reasons, it was necessary to provide a broad-spectrum treatment, able to control both these diseases. CBZ appeared as the better compromise, even if its efficacy on MCC is still uncertain. In fact, literature data are scant and contrasting, but it is worth noting that the failed focused trial on CBZ for MCC [53] was significantly biased by the heavy systemic chemotherapy treatments performed by the included patients, at odds with the present patient’s history.
Although globally safe and well-tolerated, CBZ-related adverse events often need a tailored approach with personalized doses and frequent clinical, biochemical and radiological follow-ups. In the present experience, CBZ, as a second-line therapy, was interrupted for some days or weeks when the patient showed lower tolerance, and the dose was also adjusted up to 40 mg daily without affecting the PFS.
Different neoplasms with higher metastatic load are effectively controlled under targeted therapies and many endocrine tumours include multi-targeted or selective TKI in their protocols. These treatments can produce different adverse events, i.e. variable rates of myelosuppression and immunodepression, along with anorexia, fatigue, nausea, anomalies in liver function test, hypertension, thrombocytopenia, palmar-plantar erythrodysesthesia, QTc prolongation, notably in elderly subjects [22]. As a matter of fact, despite the high prevalence of cancer in elderly people, very few data on antineoplastic therapy effects in this specific population are available [81]. Clinical trials, in fact, usually include a very selected population and, as a consequence, the experience with targeted therapy is limited in the geriatric population. On the other hand, it is well-known that about 30–50% of elderly patients will experience severe side effects of antineoplastic therapy, which is further increased when combined regimens are used [82].
Rare endocrine diseases are often orphan diseases but with overlapping effectiveness of some drugs. It is probably the case of CBZ, which has been tested with benefits in the thyroid cancer field of both follicular and neuroendocrine origins. Moreover, a somewhat therapeutic potential also emerged in other advanced endocrine tumours, although the conclusion on its effectiveness is often biased by the presence of former heavy treatment regimes. However, MCC, adrenal, chromaffin and neuroendocrine tumours have been or currently are under investigation for this TKI. The endocrine tumour framework is wide and complex, however, several touchpoints in the molecular pathways and therapies emerged from preclinical models to real practice. We are aware that streamlining the therapeutic approach in this various disease spectrum is probably not fully viable, since each tumour retains its peculiarities and uniqueness. However, to speed up the insight into new drugs’ effectiveness on different and rare endocrine tumours, a new clinical trial concept is desired, switching between a tumour-specific study mindset to a comprehensive and wide-inclusive endocrine tumour model.

Supplementary information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12020-023-03526-0.

Compliance with ethical standards

Conflict of interest

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

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

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

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

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

Jetzt bestellen und 100 € sparen!

Anhänge
Literatur
1.
Zurück zum Zitat F.M. Yakes, J. Chen, J. Tan, K. Yamaguchi, Y. Shi, P. Yu, F. Qian, F. Chu, F. Bentzien, B. Cancilla, J. Orf, A. You, A.D. Laird, S. Engst, L. Lee, J. Lesch, Y.-C. Chou, A.H. Joly, Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Mol. Cancer Ther. 10, 2298–2308 (2011). https://doi.org/10.1158/1535-7163.MCT-11-0264CrossRefPubMed F.M. Yakes, J. Chen, J. Tan, K. Yamaguchi, Y. Shi, P. Yu, F. Qian, F. Chu, F. Bentzien, B. Cancilla, J. Orf, A. You, A.D. Laird, S. Engst, L. Lee, J. Lesch, Y.-C. Chou, A.H. Joly, Cabozantinib (XL184), a Novel MET and VEGFR2 Inhibitor, Simultaneously Suppresses Metastasis, Angiogenesis, and Tumor Growth. Mol. Cancer Ther. 10, 2298–2308 (2011). https://​doi.​org/​10.​1158/​1535-7163.​MCT-11-0264CrossRefPubMed
3.
Zurück zum Zitat M.S. Brose, B. Robinson, S.I. Sherman, J. Krajewska, C.-C. Lin, F. Vaisman, A.O. Hoff, E. Hitre, D.W. Bowles, J. Hernando, L. Faoro, K. Banerjee, J.W. Oliver, B. Keam, J. Capdevila, Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 22, 1126–1138 (2021). https://doi.org/10.1016/S1470-2045(21)00332-6CrossRefPubMed M.S. Brose, B. Robinson, S.I. Sherman, J. Krajewska, C.-C. Lin, F. Vaisman, A.O. Hoff, E. Hitre, D.W. Bowles, J. Hernando, L. Faoro, K. Banerjee, J.W. Oliver, B. Keam, J. Capdevila, Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 22, 1126–1138 (2021). https://​doi.​org/​10.​1016/​S1470-2045(21)00332-6CrossRefPubMed
5.
Zurück zum Zitat A. Liberati, D.G. Altman, J. Tetzlaff, C. Mulrow, P.C. Gotzsche, J.P.A. Ioannidis, M. Clarke, P.J. Devereaux, J. Kleijnen, D. Moher, The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ 339, b2700–b2700 (2009). https://doi.org/10.1136/bmj.b2700CrossRefPubMedPubMedCentral A. Liberati, D.G. Altman, J. Tetzlaff, C. Mulrow, P.C. Gotzsche, J.P.A. Ioannidis, M. Clarke, P.J. Devereaux, J. Kleijnen, D. Moher, The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ 339, b2700–b2700 (2009). https://​doi.​org/​10.​1136/​bmj.​b2700CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat M.B. Amin, F.L. Greene, S.B. Edge, C.C. Compton, J.E. Gershenwald, R.K. Brookland, L. Meyer, D.M. Gress, D.R. Byrd, D.P. Winchester, The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a popula-based to a more “personalized” approach to cancer staging: The Eighth Edition AJCC Cancer Staging Manual, CA. Cancer J. Clin. 67, 93–99 (2017). https://doi.org/10.3322/caac.21388CrossRef M.B. Amin, F.L. Greene, S.B. Edge, C.C. Compton, J.E. Gershenwald, R.K. Brookland, L. Meyer, D.M. Gress, D.R. Byrd, D.P. Winchester, The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a popula-based to a more “personalized” approach to cancer staging: The Eighth Edition AJCC Cancer Staging Manual, CA. Cancer J. Clin. 67, 93–99 (2017). https://​doi.​org/​10.​3322/​caac.​21388CrossRef
7.
Zurück zum Zitat F. Nardi, F. Basolo, A. Crescenzi, et al., Italian consensus for the classification and reporting of thyroid cytology. J. Endocrinol. Investig. 37, 593–599 (2014)CrossRef F. Nardi, F. Basolo, A. Crescenzi, et al., Italian consensus for the classification and reporting of thyroid cytology. J. Endocrinol. Investig. 37, 593–599 (2014)CrossRef
8.
Zurück zum Zitat M. Tuttle, L.F. Morris, B. Haugen, J. Shah, J.A. Sosa, E. Rohren, R.M. Subramaniam, J.L. Hunt, N.D. Perrier, M.B. Amin, S.B. Edge, F. Greene, D. Byrd, R.K. Brookland, M.K. Washington, C.C. Compton, K.R. Hess, D.C. Sullivan, J.M. Jessup, Thyroid-differentiated and anaplastic carcinoma (Chapter 73), Springer International Publishing, n.dA. M. Tuttle, L.F. Morris, B. Haugen, J. Shah, J.A. Sosa, E. Rohren, R.M. Subramaniam, J.L. Hunt, N.D. Perrier, M.B. Amin, S.B. Edge, F. Greene, D. Byrd, R.K. Brookland, M.K. Washington, C.C. Compton, K.R. Hess, D.C. Sullivan, J.M. Jessup, Thyroid-differentiated and anaplastic carcinoma (Chapter 73), Springer International Publishing, n.dA.
10.
Zurück zum Zitat Common Terminology Criteria for Adverse Events (CTCAE), (2017) Common Terminology Criteria for Adverse Events (CTCAE), (2017)
11.
Zurück zum Zitat M.-L. Gauci, C. Aristei, J.C. Becker, A. Blom, V. Bataille, B. Dreno, V. Del Marmol, A.M. Forsea, M.C. Fargnoli, J.-J. Grob, F. Gomes, A. Hauschild, C. Hoeller, C. Harwood, N. Kelleners-Smeets, R. Kaufmann, A. Lallas, J. Malvehy, D. Moreno-Ramirez, K. Peris, G. Pellacani, P. Saiag, A.J. Stratigos, R. Vieira, I. Zalaudek, A.C.J. Van Akkooi, P. Lorigan, C. Garbe, C. Lebbé, Diagnosis and treatment of Merkel cell carcinoma: European consensus-based interdisciplinary guideline – Update 2022. Eur. J. Cancer 171, 203–231 (2022). https://doi.org/10.1016/j.ejca.2022.03.043CrossRefPubMed M.-L. Gauci, C. Aristei, J.C. Becker, A. Blom, V. Bataille, B. Dreno, V. Del Marmol, A.M. Forsea, M.C. Fargnoli, J.-J. Grob, F. Gomes, A. Hauschild, C. Hoeller, C. Harwood, N. Kelleners-Smeets, R. Kaufmann, A. Lallas, J. Malvehy, D. Moreno-Ramirez, K. Peris, G. Pellacani, P. Saiag, A.J. Stratigos, R. Vieira, I. Zalaudek, A.C.J. Van Akkooi, P. Lorigan, C. Garbe, C. Lebbé, Diagnosis and treatment of Merkel cell carcinoma: European consensus-based interdisciplinary guideline – Update 2022. Eur. J. Cancer 171, 203–231 (2022). https://​doi.​org/​10.​1016/​j.​ejca.​2022.​03.​043CrossRefPubMed
12.
Zurück zum Zitat E.A. Eisenhauer, P. Therasse, J. Bogaerts, L.H. Schwartz, D. Sargent, R. Ford, J. Dancey, S. Arbuck, S. Gwyther, M. Mooney, L. Rubinstein, L. Shankar, L. Dodd, R. Kaplan, D. Lacombe, J. Verweij, New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009). https://doi.org/10.1016/j.ejca.2008.10.026CrossRefPubMed E.A. Eisenhauer, P. Therasse, J. Bogaerts, L.H. Schwartz, D. Sargent, R. Ford, J. Dancey, S. Arbuck, S. Gwyther, M. Mooney, L. Rubinstein, L. Shankar, L. Dodd, R. Kaplan, D. Lacombe, J. Verweij, New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009). https://​doi.​org/​10.​1016/​j.​ejca.​2008.​10.​026CrossRefPubMed
13.
Zurück zum Zitat S.A. Wells, S.L. Asa, H. Dralle, R. Elisei, D.B. Evans, R.F. Gagel, N. Lee, A. Machens, J.F. Moley, F. Pacini, F. Raue, K. Frank-Raue, B. Robinson, M.S. Rosenthal, M. Santoro, M. Schlumberger, M. Shah, S.G. Waguespack, Revised American Thyroid Association Guidelines for the Management of Medullary Thyroid Carcinoma: The American Thyroid Association Guidelines Task Force on Medullary Thyroid Carcinoma. Thyroid 25, 567–610 (2015). https://doi.org/10.1089/thy.2014.0335CrossRefPubMedPubMedCentral S.A. Wells, S.L. Asa, H. Dralle, R. Elisei, D.B. Evans, R.F. Gagel, N. Lee, A. Machens, J.F. Moley, F. Pacini, F. Raue, K. Frank-Raue, B. Robinson, M.S. Rosenthal, M. Santoro, M. Schlumberger, M. Shah, S.G. Waguespack, Revised American Thyroid Association Guidelines for the Management of Medullary Thyroid Carcinoma: The American Thyroid Association Guidelines Task Force on Medullary Thyroid Carcinoma. Thyroid 25, 567–610 (2015). https://​doi.​org/​10.​1089/​thy.​2014.​0335CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat A. Hegde, A.Y. Andreev-Drakhlin, J. Roszik, L. Huang, S. Liu, K. Hess, M. Cabanillas, M.I. Hu, N.L. Busaidy, S.I. Sherman, R. Dadu, E.G. Grubbs, S.M. Ali, J. Lee, Y.Y. Elamin, G.R. Simon, G.R. Blumenschein, V.A. Papadimitrakopoulou, D. Hong, F. Meric-Bernstam, J. Heymach, V. Subbiah, Responsiveness to immune checkpoint inhibitors versus other systemic therapies in RET-aberrant malignancies, ESMO Open. 5 (2020). https://doi.org/10.1136/esmoopen-2020-000799. A. Hegde, A.Y. Andreev-Drakhlin, J. Roszik, L. Huang, S. Liu, K. Hess, M. Cabanillas, M.I. Hu, N.L. Busaidy, S.I. Sherman, R. Dadu, E.G. Grubbs, S.M. Ali, J. Lee, Y.Y. Elamin, G.R. Simon, G.R. Blumenschein, V.A. Papadimitrakopoulou, D. Hong, F. Meric-Bernstam, J. Heymach, V. Subbiah, Responsiveness to immune checkpoint inhibitors versus other systemic therapies in RET-aberrant malignancies, ESMO Open. 5 (2020). https://​doi.​org/​10.​1136/​esmoopen-2020-000799.
19.
Zurück zum Zitat F. Bentzien, M. Zuzow, N. Heald, A. Gibson, Y. Shi, L. Goon, P. Yu, S. Engst, W. Zhang, D. Huang, L. Zhao, V. Vysotskaia, F. Chu, R. Bautista, B. Cancilla, P. Lamb, A.H. Joly, F.M. Yakes, In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid 23, 1569–1577 (2013). https://doi.org/10.1089/thy.2013.0137CrossRefPubMedPubMedCentral F. Bentzien, M. Zuzow, N. Heald, A. Gibson, Y. Shi, L. Goon, P. Yu, S. Engst, W. Zhang, D. Huang, L. Zhao, V. Vysotskaia, F. Chu, R. Bautista, B. Cancilla, P. Lamb, A.H. Joly, F.M. Yakes, In vitro and in vivo activity of cabozantinib (XL184), an inhibitor of RET, MET, and VEGFR2, in a model of medullary thyroid cancer. Thyroid 23, 1569–1577 (2013). https://​doi.​org/​10.​1089/​thy.​2013.​0137CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat R. Kurzrock, S.I. Sherman, D.W. Ball, A.A. Forastiere, R.B. Cohen, R. Mehra, D.G. Pfister, E.E.W. Cohen, L. Janisch, F. Nauling, D.S. Hong, C.S. Ng, L. Ye, R.F. Gagel, J. Frye, T. Müller, M.J. Ratain, R. Salgia, Activity of XL184 (Cabozantinib), an Oral Tyrosine Kinase Inhibitor, in Patients With Medullary Thyroid Cancer. J. Clin. Oncol. 29, 2660–2666 (2011). https://doi.org/10.1200/JCO.2010.32.4145CrossRefPubMedPubMedCentral R. Kurzrock, S.I. Sherman, D.W. Ball, A.A. Forastiere, R.B. Cohen, R. Mehra, D.G. Pfister, E.E.W. Cohen, L. Janisch, F. Nauling, D.S. Hong, C.S. Ng, L. Ye, R.F. Gagel, J. Frye, T. Müller, M.J. Ratain, R. Salgia, Activity of XL184 (Cabozantinib), an Oral Tyrosine Kinase Inhibitor, in Patients With Medullary Thyroid Cancer. J. Clin. Oncol. 29, 2660–2666 (2011). https://​doi.​org/​10.​1200/​JCO.​2010.​32.​4145CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat M. Schlumberger, R. Elisei, S. Müller, P. Schöffski, M. Brose, M. Shah, L. Licitra, J. Krajewska, M.C. Kreissl, B. Niederle, E.E.W. Cohen, L. Wirth, H. Ali, D.O. Clary, Y. Yaron, M. Mangeshkar, D. Ball, B. Nelkin, S. Sherman, Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann. Oncol. 28, 2813–2819 (2017). https://doi.org/10.1093/annonc/mdx479CrossRefPubMedPubMedCentral M. Schlumberger, R. Elisei, S. Müller, P. Schöffski, M. Brose, M. Shah, L. Licitra, J. Krajewska, M.C. Kreissl, B. Niederle, E.E.W. Cohen, L. Wirth, H. Ali, D.O. Clary, Y. Yaron, M. Mangeshkar, D. Ball, B. Nelkin, S. Sherman, Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann. Oncol. 28, 2813–2819 (2017). https://​doi.​org/​10.​1093/​annonc/​mdx479CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat S.I. Sherman, D.O. Clary, R. Elisei, M.J. Schlumberger, E.E.W. Cohen, P. Schöffski, L.J. Wirth, M. Mangeshkar, D.T. Aftab, M.S. Brose, Correlative analyses of RET and RAS mutations in a phase 3 trial of cabozantinib in patients with progressive, metastatic medullary thyroid cancer. Cancer 122, 3856–3864 (2016). https://doi.org/10.1002/cncr.30252CrossRefPubMed S.I. Sherman, D.O. Clary, R. Elisei, M.J. Schlumberger, E.E.W. Cohen, P. Schöffski, L.J. Wirth, M. Mangeshkar, D.T. Aftab, M.S. Brose, Correlative analyses of RET and RAS mutations in a phase 3 trial of cabozantinib in patients with progressive, metastatic medullary thyroid cancer. Cancer 122, 3856–3864 (2016). https://​doi.​org/​10.​1002/​cncr.​30252CrossRefPubMed
25.
Zurück zum Zitat J. Capdevila, A. Klochikhin, S. Leboulleux, P. Isaev, C. Badiu, B. Robinson, B.G.M. Hughes, B. Keam, F. Parnis, R. Elisei, P. Gajate, H.K. Gan, E. Kapiteijn, L. Locati, M. Mangeshkar, L. Faoro, J. Krajewska, B. Jarzab, A Randomized, Double-Blind Noninferiority Study to Evaluate the Efficacy of the Cabozantinib Tablet at 60 mg per Day Compared with the Cabozantinib Capsule at 140 mg per Day in Patients with Progressive, Metastatic Medullary Thyroid Cancer. Thyroid 32, 515–524 (2022). https://doi.org/10.1089/thy.2022.0027CrossRefPubMedPubMedCentral J. Capdevila, A. Klochikhin, S. Leboulleux, P. Isaev, C. Badiu, B. Robinson, B.G.M. Hughes, B. Keam, F. Parnis, R. Elisei, P. Gajate, H.K. Gan, E. Kapiteijn, L. Locati, M. Mangeshkar, L. Faoro, J. Krajewska, B. Jarzab, A Randomized, Double-Blind Noninferiority Study to Evaluate the Efficacy of the Cabozantinib Tablet at 60 mg per Day Compared with the Cabozantinib Capsule at 140 mg per Day in Patients with Progressive, Metastatic Medullary Thyroid Cancer. Thyroid 32, 515–524 (2022). https://​doi.​org/​10.​1089/​thy.​2022.​0027CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat V.F. Koehler, P. Adam, K. Frank-Raue, F. Raue, E. Berg, E. Hoster, S. Allelein, M. Schott, M. Kroiss, C. Spitzweg; on Behalf of the German Study Group for Rare Malignant Tumors of the Thyroid and Parathyroid Glands, Real-World Efficacy and Safety of Cabozantinib and Vandetanib in Advanced Medullary Thyroid Cancer. Thyroid 31, 459–469 (2021). https://doi.org/10.1089/thy.2020.0206CrossRefPubMed V.F. Koehler, P. Adam, K. Frank-Raue, F. Raue, E. Berg, E. Hoster, S. Allelein, M. Schott, M. Kroiss, C. Spitzweg; on Behalf of the German Study Group for Rare Malignant Tumors of the Thyroid and Parathyroid Glands, Real-World Efficacy and Safety of Cabozantinib and Vandetanib in Advanced Medullary Thyroid Cancer. Thyroid 31, 459–469 (2021). https://​doi.​org/​10.​1089/​thy.​2020.​0206CrossRefPubMed
28.
Zurück zum Zitat H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, CA. Cancer J. Clin. 71, 209–249 (2021). https://doi.org/10.3322/caac.21660CrossRef H. Sung, J. Ferlay, R.L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, CA. Cancer J. Clin. 71, 209–249 (2021). https://​doi.​org/​10.​3322/​caac.​21660CrossRef
31.
Zurück zum Zitat B.R. Haugen, E.K. Alexander, K.C. Bible, G.M. Doherty, S.J. Mandel, Y.E. Nikiforov, F. Pacini, G.W. Randolph, A.M. Sawka, M. Schlumberger, K.G. Schuff, S.I. Sherman, J.A. Sosa, D.L. Steward, R.M. Tuttle, L. Wartofsky, 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 26, 1–133 (2016). https://doi.org/10.1089/thy.2015.0020CrossRefPubMedPubMedCentral B.R. Haugen, E.K. Alexander, K.C. Bible, G.M. Doherty, S.J. Mandel, Y.E. Nikiforov, F. Pacini, G.W. Randolph, A.M. Sawka, M. Schlumberger, K.G. Schuff, S.I. Sherman, J.A. Sosa, D.L. Steward, R.M. Tuttle, L. Wartofsky, 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 26, 1–133 (2016). https://​doi.​org/​10.​1089/​thy.​2015.​0020CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat M. Schlumberger, M. Tahara, L.J. Wirth, B. Robinson, M.S. Brose, R. Elisei, M.A. Habra, K. Newbold, M.H. Shah, A.O. Hoff, A.G. Gianoukakis, N. Kiyota, M.H. Taylor, S.-B. Kim, M.K. Krzyzanowska, C.E. Dutcus, B. De Las Heras, J. Zhu, S.I. Sherman, Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. N. Engl. J. Med. 372, 621–630 (2015). https://doi.org/10.1056/NEJMoa1406470CrossRefPubMed M. Schlumberger, M. Tahara, L.J. Wirth, B. Robinson, M.S. Brose, R. Elisei, M.A. Habra, K. Newbold, M.H. Shah, A.O. Hoff, A.G. Gianoukakis, N. Kiyota, M.H. Taylor, S.-B. Kim, M.K. Krzyzanowska, C.E. Dutcus, B. De Las Heras, J. Zhu, S.I. Sherman, Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. N. Engl. J. Med. 372, 621–630 (2015). https://​doi.​org/​10.​1056/​NEJMoa1406470CrossRefPubMed
33.
Zurück zum Zitat M.S. Brose, J.A. Krajewska, F. Vaisman, A.O. Hoff, E. Hitre, J. Oliver, D.S. Williamson, N. Berry, J. Capdevila Castillon, 1653P Cabozantinib (C) vs placebo (P) in patients (pts) with radioiodine-refractory (RAIR) differentiated thyroid cancer (DTC) who progressed after prior VEGFR-targeted therapy: Outcomes by duration of prior lenvatinib (L) treatment. Ann. Oncol. 33, S1297–S1298 (2022). https://doi.org/10.1016/j.annonc.2022.07.1733CrossRef M.S. Brose, J.A. Krajewska, F. Vaisman, A.O. Hoff, E. Hitre, J. Oliver, D.S. Williamson, N. Berry, J. Capdevila Castillon, 1653P Cabozantinib (C) vs placebo (P) in patients (pts) with radioiodine-refractory (RAIR) differentiated thyroid cancer (DTC) who progressed after prior VEGFR-targeted therapy: Outcomes by duration of prior lenvatinib (L) treatment. Ann. Oncol. 33, S1297–S1298 (2022). https://​doi.​org/​10.​1016/​j.​annonc.​2022.​07.​1733CrossRef
35.
Zurück zum Zitat M.H. Shah, J. De souza, L. Wirth, M.E. Menefee, S. Liu, S. Geyer, J. Wright, M. Villalona, M. Cabanillas, Cabozantinib in patients with radioiodine-refractory differentiated thyroid cancer who progressed on prior VEGFR-targeted therapy: Results of NCI-and itog-sponsored multicenter phase II clinical trial. Thyroid 25, A31–A32 (2015). https://doi.org/10.1089/thy.2015.29004.abstractsCrossRef M.H. Shah, J. De souza, L. Wirth, M.E. Menefee, S. Liu, S. Geyer, J. Wright, M. Villalona, M. Cabanillas, Cabozantinib in patients with radioiodine-refractory differentiated thyroid cancer who progressed on prior VEGFR-targeted therapy: Results of NCI-and itog-sponsored multicenter phase II clinical trial. Thyroid 25, A31–A32 (2015). https://​doi.​org/​10.​1089/​thy.​2015.​29004.​abstractsCrossRef
38.
Zurück zum Zitat B. Konda, E. Sherman, E. Massarelli, B. Xia, J. Muzaffar, J. Morris, M. Ryder, A. Ho, M. Agulnik, L. Wei, R. Jacob, J. Wright, H. Streicher, W. Carson, M. Shah, Cabozantinib in combination with nivolumab and ipilimumab in patients with radioiodine (rai)-refractory differentiated thyroid cancer (dtc) whose cancer progressed after one prior vascular endothelial growthfactor receptor (vegfr)-targeted therapy: interim results of a multicenter phase 2 national cancer institute (nci)-international thyroid oncology group (itog) trial (nci#10240). Thyroid 32, A98 (2022). https://doi.org/10.1089/thy.2022.29140.lb.abstractsCrossRef B. Konda, E. Sherman, E. Massarelli, B. Xia, J. Muzaffar, J. Morris, M. Ryder, A. Ho, M. Agulnik, L. Wei, R. Jacob, J. Wright, H. Streicher, W. Carson, M. Shah, Cabozantinib in combination with nivolumab and ipilimumab in patients with radioiodine (rai)-refractory differentiated thyroid cancer (dtc) whose cancer progressed after one prior vascular endothelial growthfactor receptor (vegfr)-targeted therapy: interim results of a multicenter phase 2 national cancer institute (nci)-international thyroid oncology group (itog) trial (nci#10240). Thyroid 32, A98 (2022). https://​doi.​org/​10.​1089/​thy.​2022.​29140.​lb.​abstractsCrossRef
39.
Zurück zum Zitat M. Taylor, G. Daniels, K. Thein, Y. Loriot, S. Khan, J. Goldschmidt, L. Lebellec, J. Sarantopoulos, A. Vozy, Z. Castel-Ajgal, S. Andrianova, R. Sudhagoni, R. Levytskyy, R. Dadu, Cabozantinib in combination with atezolizumab as a first-line therapy in patients with radioiodine-refractory (rair) differentiated thyroid cancer (dtc): results from cohort 18 of the phase 1b cosmic-21 study. Thyroid 32, A3 (2022). https://doi.org/10.1089/thy.2022.29138.abstractsCrossRef M. Taylor, G. Daniels, K. Thein, Y. Loriot, S. Khan, J. Goldschmidt, L. Lebellec, J. Sarantopoulos, A. Vozy, Z. Castel-Ajgal, S. Andrianova, R. Sudhagoni, R. Levytskyy, R. Dadu, Cabozantinib in combination with atezolizumab as a first-line therapy in patients with radioiodine-refractory (rair) differentiated thyroid cancer (dtc): results from cohort 18 of the phase 1b cosmic-21 study. Thyroid 32, A3 (2022). https://​doi.​org/​10.​1089/​thy.​2022.​29138.​abstractsCrossRef
40.
Zurück zum Zitat M.S. Brose, S. Shenoy, N. Bhat, A.K. Harlacker, R.K. Yurtal, Z.A. Posey, D.M. Torrente, C. Grande, C.M. Squillante, A. Troxel, M. Yarchoan, A phase II trial of cabozantinib (CABO) for the treatment of radioiodine (RAI)-refractory differentiated thyroid carcinoma (DTC) in the first-line setting, J. Clin. Oncol. 36 (2018). https://doi.org/10.1200/JCO.2018.36.15-suppl.6088 M.S. Brose, S. Shenoy, N. Bhat, A.K. Harlacker, R.K. Yurtal, Z.A. Posey, D.M. Torrente, C. Grande, C.M. Squillante, A. Troxel, M. Yarchoan, A phase II trial of cabozantinib (CABO) for the treatment of radioiodine (RAI)-refractory differentiated thyroid carcinoma (DTC) in the first-line setting, J. Clin. Oncol. 36 (2018). https://​doi.​org/​10.​1200/​JCO.​2018.​36.​15-suppl.​6088
47.
Zurück zum Zitat P. Nghiem, S. Bhatia, E.J. Lipson, W.H. Sharfman, R.R. Kudchadkar, A.S. Brohl, P.A. Friedlander, A. Daud, H.M. Kluger, S.A. Reddy, B.C. Boulmay, A.I. Riker, M.A. Burgess, B.A. Hanks, T. Olencki, K. Margolin, L.M. Lundgren, A. Soni, N. Ramchurren, C. Church, S.Y. Park, M.M. Shinohara, B. Salim, J.M. Taube, S.R. Bird, N. Ibrahim, S.P. Fling, B. Homet Moreno, E. Sharon, M.A. Cheever, S.L. Topalian, Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J. Clin. Oncol. 37, 693–702 (2019). https://doi.org/10.1200/JCO.18.01896CrossRefPubMedPubMedCentral P. Nghiem, S. Bhatia, E.J. Lipson, W.H. Sharfman, R.R. Kudchadkar, A.S. Brohl, P.A. Friedlander, A. Daud, H.M. Kluger, S.A. Reddy, B.C. Boulmay, A.I. Riker, M.A. Burgess, B.A. Hanks, T. Olencki, K. Margolin, L.M. Lundgren, A. Soni, N. Ramchurren, C. Church, S.Y. Park, M.M. Shinohara, B. Salim, J.M. Taube, S.R. Bird, N. Ibrahim, S.P. Fling, B. Homet Moreno, E. Sharon, M.A. Cheever, S.L. Topalian, Durable Tumor Regression and Overall Survival in Patients With Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J. Clin. Oncol. 37, 693–702 (2019). https://​doi.​org/​10.​1200/​JCO.​18.​01896CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat H.L. Kaufman, Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after ≥1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial, 6(1), 7 (2018). H.L. Kaufman, Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after ≥1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial, 6(1), 7 (2018).
51.
Zurück zum Zitat M. Brunner, D. Thurnher, J. Pammer, S. Geleff, G. Heiduschka, C.M. Reinisch, P. Petzelbauer, B.M. Erovic, Expression of VEGF-A/C, VEGF-R2, PDGF-a/b, c-kit, EGFR, Her-2/Neu, Mcl-1 and Bmi-1 in Merkel cell carcinoma. Mod. Pathol. 21(7), 876–84 (2008)CrossRefPubMed M. Brunner, D. Thurnher, J. Pammer, S. Geleff, G. Heiduschka, C.M. Reinisch, P. Petzelbauer, B.M. Erovic, Expression of VEGF-A/C, VEGF-R2, PDGF-a/b, c-kit, EGFR, Her-2/Neu, Mcl-1 and Bmi-1 in Merkel cell carcinoma. Mod. Pathol. 21(7), 876–84 (2008)CrossRefPubMed
54.
56.
Zurück zum Zitat D.A. Pryma, B.B. Chin, R.B. Noto, J.S. Dillon, S. Perkins, L. Solnes, L. Kostakoglu, A.N. Serafini, M.H. Pampaloni, J. Jensen, T. Armor, T. Lin, T. White, N. Stambler, S. Apfel, V.A. DiPippo, S. Mahmood, V. Wong, C. Jimenez, Efficacy and Safety of High-Specific-Activity 131 I-MIBG Therapy in Patients with Advanced Pheochromocytoma or Paraganglioma. J. Nucl. Med. 60, 623–630 (2019). https://doi.org/10.2967/jnumed.118.217463CrossRefPubMedPubMedCentral D.A. Pryma, B.B. Chin, R.B. Noto, J.S. Dillon, S. Perkins, L. Solnes, L. Kostakoglu, A.N. Serafini, M.H. Pampaloni, J. Jensen, T. Armor, T. Lin, T. White, N. Stambler, S. Apfel, V.A. DiPippo, S. Mahmood, V. Wong, C. Jimenez, Efficacy and Safety of High-Specific-Activity 131 I-MIBG Therapy in Patients with Advanced Pheochromocytoma or Paraganglioma. J. Nucl. Med. 60, 623–630 (2019). https://​doi.​org/​10.​2967/​jnumed.​118.​217463CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat N.D. Niemeijer, G. Alblas, L.T. van Hulsteijn, O.M. Dekkers, E.P.M. Corssmit, Chemotherapy with cyclophosphamide, vincristine and dacarbazine for malignant paraganglioma and pheochromocytoma: systematic review and meta-analysis. Clin. Endocrinol. (Oxf.). 81, 642–651 (2014). https://doi.org/10.1111/cen.12542CrossRefPubMed N.D. Niemeijer, G. Alblas, L.T. van Hulsteijn, O.M. Dekkers, E.P.M. Corssmit, Chemotherapy with cyclophosphamide, vincristine and dacarbazine for malignant paraganglioma and pheochromocytoma: systematic review and meta-analysis. Clin. Endocrinol. (Oxf.). 81, 642–651 (2014). https://​doi.​org/​10.​1111/​cen.​12542CrossRefPubMed
61.
Zurück zum Zitat M.P. Economides, A.Y. Shah, C. Jimenez, M.A. Habra, M. Desai, M.T. Campbell, A Durable Response With the Combination of Nivolumab and Cabozantinib in a Patient With Metastatic Paraganglioma: A Case Report and Review of the Current Literature. Front. Endocrinol. 11, 594264 (2020). https://doi.org/10.3389/fendo.2020.594264CrossRef M.P. Economides, A.Y. Shah, C. Jimenez, M.A. Habra, M. Desai, M.T. Campbell, A Durable Response With the Combination of Nivolumab and Cabozantinib in a Patient With Metastatic Paraganglioma: A Case Report and Review of the Current Literature. Front. Endocrinol. 11, 594264 (2020). https://​doi.​org/​10.​3389/​fendo.​2020.​594264CrossRef
62.
Zurück zum Zitat A.B. Apolo, R. Nadal, D.M. Girardi, S.A. Niglio, L. Ley, L.M. Cordes, S.M. Steinberg, O. Sierra Ortiz, J. Cadena, C. Diaz, M. Mallek, N.N. Davarpanah, R. Costello, J.B. Trepel, M.-J. Lee, M.J. Merino, M.H. Bagheri, P. Monk, W.D. Figg, J.L. Gulley, P.K. Agarwal, V. Valera, H.J. Chalfin, J. Jones, H. Streicher, J.J. Wright, Y.M. Ning, H.L. Parnes, W.L. Dahut, D.P. Bottaro, P.N. Lara, B. Saraiya, S.K. Pal, M.N. Stein, A. Mortazavi, Phase I Study of Cabozantinib and Nivolumab Alone or With Ipilimumab for Advanced or Metastatic Urothelial Carcinoma and Other Genitourinary Tumors. J. Clin. Oncol. 38, 3672–3684 (2020). https://doi.org/10.1200/JCO.20.01652CrossRefPubMedPubMedCentral A.B. Apolo, R. Nadal, D.M. Girardi, S.A. Niglio, L. Ley, L.M. Cordes, S.M. Steinberg, O. Sierra Ortiz, J. Cadena, C. Diaz, M. Mallek, N.N. Davarpanah, R. Costello, J.B. Trepel, M.-J. Lee, M.J. Merino, M.H. Bagheri, P. Monk, W.D. Figg, J.L. Gulley, P.K. Agarwal, V. Valera, H.J. Chalfin, J. Jones, H. Streicher, J.J. Wright, Y.M. Ning, H.L. Parnes, W.L. Dahut, D.P. Bottaro, P.N. Lara, B. Saraiya, S.K. Pal, M.N. Stein, A. Mortazavi, Phase I Study of Cabozantinib and Nivolumab Alone or With Ipilimumab for Advanced or Metastatic Urothelial Carcinoma and Other Genitourinary Tumors. J. Clin. Oncol. 38, 3672–3684 (2020). https://​doi.​org/​10.​1200/​JCO.​20.​01652CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat M. Fassnacht, O.M. Dekkers, T. Else, E. Baudin, A. Berruti, R.R. de Krijger, H.R. Haak, R. Mihai, G. Assie, M. Terzolo, European Society of Endocrinology Clinical Practice Guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Network for the Study of Adrenal Tumors. Eur. J. Endocrinol. 179, G1–G46 (2018). https://doi.org/10.1530/EJE-18-0608CrossRefPubMed M. Fassnacht, O.M. Dekkers, T. Else, E. Baudin, A. Berruti, R.R. de Krijger, H.R. Haak, R. Mihai, G. Assie, M. Terzolo, European Society of Endocrinology Clinical Practice Guidelines on the management of adrenocortical carcinoma in adults, in collaboration with the European Network for the Study of Adrenal Tumors. Eur. J. Endocrinol. 179, G1–G46 (2018). https://​doi.​org/​10.​1530/​EJE-18-0608CrossRefPubMed
66.
Zurück zum Zitat T. Massimo, A. Alberto, F. Martin, D. Fulvia, T. Libuse, C.P. Antonio, R. Ruth, B. Lisa, S. Paola, G. Erika, R. Giuseppe, B. Enrico, P. Mauro, S. Wolfgang, H. Stefanie, K. Ann-Cathrin, A. Emanuela, A. Bruno, L. Paola, L. Gaetano, M. Massimo, B. Paolo, M. Franco, A. Bruno, D. Luigi, B. Alfredo, Adjuvant Mitotane Treatment for Adrenocortical Carcinoma, N. Engl. J. Med. 356:2372–2380 (2007) T. Massimo, A. Alberto, F. Martin, D. Fulvia, T. Libuse, C.P. Antonio, R. Ruth, B. Lisa, S. Paola, G. Erika, R. Giuseppe, B. Enrico, P. Mauro, S. Wolfgang, H. Stefanie, K. Ann-Cathrin, A. Emanuela, A. Bruno, L. Paola, L. Gaetano, M. Massimo, B. Paolo, M. Franco, A. Bruno, D. Luigi, B. Alfredo, Adjuvant Mitotane Treatment for Adrenocortical Carcinoma, N. Engl. J. Med. 356:2372–2380 (2007)
67.
Zurück zum Zitat M. Fassnacht, M. Terzolo, B. Allolio, E. Baudin, H. Haak, A. Berruti, S. Welin, C. Schade-Brittinger, A. Lacroix, B. Jarzab, H. Sorbye, D.J. Torpy, V. Stepan, D.E. Schteingart, W. Arlt, M. Kroiss, S. Leboulleux, P. Sperone, A. Sundin, I. Hermsen, S. Hahner, H.S. Willenberg, A. Tabarin, M. Quinkler, C. de la Fouchardière, M. Schlumberger, F. Mantero, D. Weismann, F. Beuschlein, H. Gelderblom, H. Wilmink, M. Sender, M. Edgerly, W. Kenn, T. Fojo, H.-H. Müller, B. Skogseid, Combination Chemotherapy in Advanced Adrenocortical Carcinoma. N. Engl. J. Med. 366, 2189–2197 (2012). https://doi.org/10.1056/NEJMoa1200966CrossRefPubMed M. Fassnacht, M. Terzolo, B. Allolio, E. Baudin, H. Haak, A. Berruti, S. Welin, C. Schade-Brittinger, A. Lacroix, B. Jarzab, H. Sorbye, D.J. Torpy, V. Stepan, D.E. Schteingart, W. Arlt, M. Kroiss, S. Leboulleux, P. Sperone, A. Sundin, I. Hermsen, S. Hahner, H.S. Willenberg, A. Tabarin, M. Quinkler, C. de la Fouchardière, M. Schlumberger, F. Mantero, D. Weismann, F. Beuschlein, H. Gelderblom, H. Wilmink, M. Sender, M. Edgerly, W. Kenn, T. Fojo, H.-H. Müller, B. Skogseid, Combination Chemotherapy in Advanced Adrenocortical Carcinoma. N. Engl. J. Med. 366, 2189–2197 (2012). https://​doi.​org/​10.​1056/​NEJMoa1200966CrossRefPubMed
68.
Zurück zum Zitat A. Berruti, P. Sperone, A. Ferrero, A. Germano, A. Ardito, A.M. Priola, S. De Francia, M. Volante, F. Daffara, D. Generali, S. Leboulleux, P. Perotti, E. Baudin, M. Papotti, M. Terzolo, Phase II study of weekly paclitaxel and sorafenib as second/third-line therapy in patients with adrenocortical carcinoma. Eur. J. Endocrinol. 166, 451–458 (2012). https://doi.org/10.1530/EJE-11-0918CrossRefPubMed A. Berruti, P. Sperone, A. Ferrero, A. Germano, A. Ardito, A.M. Priola, S. De Francia, M. Volante, F. Daffara, D. Generali, S. Leboulleux, P. Perotti, E. Baudin, M. Papotti, M. Terzolo, Phase II study of weekly paclitaxel and sorafenib as second/third-line therapy in patients with adrenocortical carcinoma. Eur. J. Endocrinol. 166, 451–458 (2012). https://​doi.​org/​10.​1530/​EJE-11-0918CrossRefPubMed
69.
Zurück zum Zitat L.M. Phan, E. Fuentes-Mattei, W. Wu, G. Velazquez-Torres, K. Sircar, C.G. Wood, T. Hai, C. Jimenez, G.J. Cote, L. Ozsari, M.-C. Hofmann, S. Zheng, R. Verhaak, L. Pagliaro, M.A. Cortez, M.-H. Lee, S.-C.J. Yeung, M.A. Habra, Hepatocyte Growth Factor/cMET Pathway Activation Enhances Cancer Hallmarks in Adrenocortical Carcinoma. Cancer Res. 75, 4131–4142 (2015). https://doi.org/10.1158/0008-5472.CAN-14-3707CrossRefPubMedPubMedCentral L.M. Phan, E. Fuentes-Mattei, W. Wu, G. Velazquez-Torres, K. Sircar, C.G. Wood, T. Hai, C. Jimenez, G.J. Cote, L. Ozsari, M.-C. Hofmann, S. Zheng, R. Verhaak, L. Pagliaro, M.A. Cortez, M.-H. Lee, S.-C.J. Yeung, M.A. Habra, Hepatocyte Growth Factor/cMET Pathway Activation Enhances Cancer Hallmarks in Adrenocortical Carcinoma. Cancer Res. 75, 4131–4142 (2015). https://​doi.​org/​10.​1158/​0008-5472.​CAN-14-3707CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat R. Garcia-Carbonero, H. Sorbye, E. Baudin, E. Raymond, B. Wiedenmann, B. Niederle, E. Sedlackova, C. Toumpanakis, M. Anlauf, J.B. Cwikla, M. Caplin, D. O”Toole, A. Perren; all other Vienna Consensus Conference participants, ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology 103, 186–194 (2016). https://doi.org/10.1159/000443172CrossRefPubMed R. Garcia-Carbonero, H. Sorbye, E. Baudin, E. Raymond, B. Wiedenmann, B. Niederle, E. Sedlackova, C. Toumpanakis, M. Anlauf, J.B. Cwikla, M. Caplin, D. O”Toole, A. Perren; all other Vienna Consensus Conference participants, ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology 103, 186–194 (2016). https://​doi.​org/​10.​1159/​000443172CrossRefPubMed
74.
Zurück zum Zitat M. Pavel, D. O’Toole, F. Costa, J. Capdevila, D. Gross, R. Kianmanesh, E. Krenning, U. Knigge, R. Salazar, U.-F. Pape, K. Öberg; all other Vienna Consensus Conference participants, ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology 103, 172–185 (2016). https://doi.org/10.1159/000443167CrossRefPubMed M. Pavel, D. O’Toole, F. Costa, J. Capdevila, D. Gross, R. Kianmanesh, E. Krenning, U. Knigge, R. Salazar, U.-F. Pape, K. Öberg; all other Vienna Consensus Conference participants, ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology 103, 172–185 (2016). https://​doi.​org/​10.​1159/​000443167CrossRefPubMed
75.
Zurück zum Zitat J.C. Yao, M. Hassan, A. Phan, C. Dagohoy, C. Leary, J.E. Mares, E.K. Abdalla, J.B. Fleming, J.-N. Vauthey, A. Rashid, D.B. Evans, One Hundred Years After “Carcinoid”: Epidemiology of and Prognostic Factors for Neuroendocrine Tumors in 35,825 Cases in the United States. J. Clin. Oncol. 26, 3063–3072 (2008). https://doi.org/10.1200/JCO.2007.15.4377CrossRefPubMed J.C. Yao, M. Hassan, A. Phan, C. Dagohoy, C. Leary, J.E. Mares, E.K. Abdalla, J.B. Fleming, J.-N. Vauthey, A. Rashid, D.B. Evans, One Hundred Years After “Carcinoid”: Epidemiology of and Prognostic Factors for Neuroendocrine Tumors in 35,825 Cases in the United States. J. Clin. Oncol. 26, 3063–3072 (2008). https://​doi.​org/​10.​1200/​JCO.​2007.​15.​4377CrossRefPubMed
76.
Zurück zum Zitat E. Raymond, L. Dahan, J.-L. Raoul, Y.-J. Bang, I. Borbath, C. Lombard-Bohas, J. Valle, P. Metrakos, D. Smith, A. Vinik, J.-S. Chen, D. Hörsch, P. Hammel, B. Wiedenmann, E. Van Cutsem, S. Patyna, D.R. Lu, C. Blanckmeister, R. Chao, P. Ruszniewski, Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N. Engl. J. Med. 364, 501–513 (2011). https://doi.org/10.1056/NEJMoa1003825CrossRefPubMed E. Raymond, L. Dahan, J.-L. Raoul, Y.-J. Bang, I. Borbath, C. Lombard-Bohas, J. Valle, P. Metrakos, D. Smith, A. Vinik, J.-S. Chen, D. Hörsch, P. Hammel, B. Wiedenmann, E. Van Cutsem, S. Patyna, D.R. Lu, C. Blanckmeister, R. Chao, P. Ruszniewski, Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N. Engl. J. Med. 364, 501–513 (2011). https://​doi.​org/​10.​1056/​NEJMoa1003825CrossRefPubMed
77.
Zurück zum Zitat C. Reuther, V. Heinzle, M. Spampatti, G. Vlotides, E. De Toni, G. Spöttl, J. Maurer, S. Nölting, B. Göke, C.J. Auernhammer, Cabozantinib and Tivantinib, but Not INC280, Induce Antiproliferative and Antimigratory Effects in Human Neuroendocrine Tumor Cells in vitro: Evidence for “Off-Target” Effects Not Mediated by c-Met Inhibition. Neuroendocrinology 103, 383–401 (2016). https://doi.org/10.1159/000439431CrossRefPubMed C. Reuther, V. Heinzle, M. Spampatti, G. Vlotides, E. De Toni, G. Spöttl, J. Maurer, S. Nölting, B. Göke, C.J. Auernhammer, Cabozantinib and Tivantinib, but Not INC280, Induce Antiproliferative and Antimigratory Effects in Human Neuroendocrine Tumor Cells in vitro: Evidence for “Off-Target” Effects Not Mediated by c-Met Inhibition. Neuroendocrinology 103, 383–401 (2016). https://​doi.​org/​10.​1159/​000439431CrossRefPubMed
83.
Zurück zum Zitat M. Smith, J. De Bono, C. Sternberg, S. Le Moulec, S. Oudard, U. De Giorgi, M. Krainer, A. Bergman, W. Hoelzer, R. De Wit, M. Bögemann, F. Saad, G. Cruciani, A. Thiery-Vuillemin, S. Feyerabend, K. Miller, N. Houédé, S. Hussain, E. Lam, J. Polikoff, A. Stenzl, P. Mainwaring, D. Ramies, C. Hessel, A. Weitzman, K. Fizazi, Phase III Study of Cabozantinib in Previously Treated Metastatic Castration-Resistant Prostate Cancer: COMET-1. J. Clin. Oncol. 34, 3005–3013 (2016). https://doi.org/10.1200/JCO.2015.65.5597CrossRefPubMed M. Smith, J. De Bono, C. Sternberg, S. Le Moulec, S. Oudard, U. De Giorgi, M. Krainer, A. Bergman, W. Hoelzer, R. De Wit, M. Bögemann, F. Saad, G. Cruciani, A. Thiery-Vuillemin, S. Feyerabend, K. Miller, N. Houédé, S. Hussain, E. Lam, J. Polikoff, A. Stenzl, P. Mainwaring, D. Ramies, C. Hessel, A. Weitzman, K. Fizazi, Phase III Study of Cabozantinib in Previously Treated Metastatic Castration-Resistant Prostate Cancer: COMET-1. J. Clin. Oncol. 34, 3005–3013 (2016). https://​doi.​org/​10.​1200/​JCO.​2015.​65.​5597CrossRefPubMed
Metadaten
Titel
Cabozantinib for different endocrine tumours: killing two birds with one stone. A systematic review of the literature
verfasst von
Elena Zago
Antonio Galluzzo
Silvia Pradella
Lorenzo Antonuzzo
Mario Maggi
Luisa Petrone
Clotilde Sparano
Publikationsdatum
18.10.2023
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 1/2024
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-023-03526-0

Weitere Artikel der Ausgabe 1/2024

Endocrine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

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