Vol 75, No 4 (2024)
Original paper
Published online: 2024-07-24

open access

Page views 453
Article views/downloads 191
Get Citation

Connect on Social Media

Connect on Social Media

Angiogenic biomarkers of response to treatment with peptide receptor radionuclide therapy in neuroendocrine tumours

Janusz Strzelczyk1, Monika Wójcik-Giertuga1, Karolina Makulik1, Violetta Rosiek1, Grzegorz Kamiński2, Dariusz Kajdaniuk3, Beata Kos-Kudła1
Pubmed: 39279310
Endokrynol Pol 2024;75(4):412-418.

Abstract

Background: Neuroendocrine tumours (NETs) are a heterogeneous group of tumours, which is characterized by rich vascularization. The role of angiogenesis in NETs has been widely researched. Peptide receptor radionuclide therapy (PRRT) is an effective treatment method for patients with disease progression in NETs. Due to the heterogeneousness of NETs, the response to treatment varies. Currently, the finding of efficient markers helpful in assessing the response to treatment in NETs is crucial. The aim of this study was to assess chromogranin A (CgA) and angiogenic factors in gastro-entero-pancreatic (GEP) and broncho-pulmonary (BP) NET patients treated with PRRT. Material and Methods: The study group included 40 patients with GEP NETs and BP NETs who completed four cycles of PRRT. Serum levels of CgA and angiogenic factors such as vascular endothelial growth factor (VEGF), its receptors (VEGF-R1, VEGF-R2, VEGF-R3), were assessed before and after four cycles of PRRT. All tests were determined using ELISA. Results: The concentration of CgA, VEGF-R1 and VEGF-R2 decreased significantly, whereas VEGF-R3 increased significantly after PRRT. PRRT did not affect VEGF, it was similar before and after the radioisotope treatment. Based on AUROC, only for VEGF-R1 AUC was a consequence of 0.7 which can be considered as a good response to PRRT treatment. Conclusions: VEGF-R1 may be a potential biomarker useful in assessing the effectiveness of PRRT in NET patients.

Original paper

Endokrynologia Polska

DOI: 10.5603/ep.100241

ISSN 0423–104X, e-ISSN 2299–8306

Volume/Tom 75; Number/Numer 4/2024

Submitted: 14.04.2024

Accepted: 27.05.2024

Early publication date: 24.07.2024

Angiogenic biomarkers of response to treatment with peptide receptor radionuclide therapy in neuroendocrine tumours

Janusz Strzelczyk1Monika Wójcik-Giertuga1Karolina Makulik1Violetta Rosiek1Grzegorz Kamiński2Dariusz Kajdaniuk3Beata Kos-Kudła1
1Department of Endocrinology and Neuroendocrine Tumours, Chair of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
2Department of Endocrinology and Radioisotope Therapy, Military Institute of Medicine — National Research Institute, Warsaw, Poland
3Department of Pathophysiology, Chair of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland

Janusz Strzelczyk, Department of Endocrinology and Neuroendocrine Tumours, Chair of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland; e-mail: janusz.strzelczyk@sum.edu.pl

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially

Abstract
Introduction: Neuroendocrine tumours (NETs) are a heterogeneous group of tumours, which is characterised by rich vascularisation. The role of angiogenesis in NETs has been widely researched. Peptide receptor radionuclide therapy (PRRT) is an effective treatment method for patients with disease progression in NETs. Due to the heterogeneity of NETs, the response to treatment varies. Currently, the finding of efficient markers helpful in assessing the response to treatment in NETs is crucial. The aim of this study was to assess chromogranin A (CgA) and angiogenic factors in gastro-entero-pancreatic (GEP) and broncho-pulmonary (BP) NET patients treated with PRRT.
Material and methods: The study group included 40 patients with GEP NETs and BP NETs, who completed 4 cycles of PRRT. Serum levels of CgA and angiogenic factors such as vascular endothelial growth factor (VEGF) and its receptors (VEGF-R1, VEGF-R2, VEGF-R3) were assessed before and after 4 cycles of PRRT. All tests were determined using ELISAs.
Results: The concentration of CgA, VEGF-R1, and VEGF-R2 decreased significantly, whereas VEGF-R3 increased significantly after PRRT. PRRT did not affect VEGF it was similar before and after the radioisotope treatment. Based on AUROC, only VEGF-R1 exhibited good performance in distinguishing between NET patients before and after PRRT; the area under the curve (AUC) was 0.7.
Conclusions: VEGF-R1 is a potential biomarker for assessment of the effectiveness of PRRT in NET patients. (Endokrynol Pol 2024; 75 (4): 412–418)
Key words: neuroendocrine tumour (NET); peptide receptor radionuclide therapy (PRRT); radioligand therapy (RLT); angiogenic markers; vascular endothelial growth factor (VEGF); vascular endothelial growth factor receptor (VEGF-R)

Introduction

Neuroendocrine tumours (NETs) are rare, heterogeneous tumours originating from the diffuse endocrine system (DES). Most NETs (70%) are located in the gastrointestinal tract (gastroenteropancreatic neuroendocrine neoplasms; GEP-NETs) [1–3], and secondly (20%) in the respiratory system (bronchopulmonary neuroendocrine tumours; BP-NETs) [4]. Over 12 years of observation it was shown that the incidence of NETs increased ~2-fold [5]. NETs are richly vascularised; hence, the importance of angiogenesis has been widely studied in these tumours [6–8]. Vascular endothelial growth factor (VEGF) is the main factor associated with the angiogenesis process, and it binds to the receptors VEGFR-1, VEGFR-2, and VEGFR-3 [9, 10]. VEGF (also known as VEGF-A) is part of a family of growth factors that also includes VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PLGF) [11]. Studies have proven that both VEGF and VEGF-R may play an important role in disease progression [12, 13]. According to the literature, VEGF acts as a factor in increasing vascular permeability and as a mitogen specific for endothelial cells, and therefore it might initiate angiogenesis in malignant tumours [11]. The VEGF pathway is responsible for maintaining cancer cell autonomy primarily through autocrine signalling [14]. Angiogenesis plays a key role in the process of tumour initiation, which facilitates the spread of cancer cells, including through the so-called vascular co-option, in which existing vessels are “hijacked”, or by building endothelium-like blood channels by tumour cells (vasogenic mimicry) [15]. Although the primary treatment method for NETs is surgery, other forms of systemic therapy are also used [1, 2, 16]. Among other things, increased expression of somatostatin receptors in somatostatin receptor scintigraphy (SRI) allows patients to be qualified for targeted therapy with isotopically labelled somatostatin analogues (PRRT peptide receptor radionuclide therapy) [17]. PRRT is most often the second line of treatment in the disease progression in patients with advanced, unresectable G1/G2 NETs showing overexpression of somatostatin receptors [2, 17, 18]. The effectiveness of this therapy has been confirmed in numerous studies, where a significant reduction in the risk of disease progression was confirmed [19]. PRRT may also be a method of neoadjuvant treatment, enabling surgical intervention in initially inoperable NETs [20]. PRRT acts through somatostatin receptors in the tumour (SST-2), the expression of which also varies significantly and thus induces DNA damage in cells, which affects the different therapeutic responses [21]. There are reports that many mechanisms related to the tumour microenvironment, such as hypoxia, the composition of the extracellular matrix, or the presence of tumour-associated fibroblasts, may affect the final effect of treatment, including being responsible for resistance to PRRT therapy [22]. The biochemical diagnosis of NETs includes the determination of non-specific markers, such as chromogranin A (CgA), belonging to the granin family, i.e. acidic glycoproteins [23]. CgA is currently the most used marker for monitoring patients with NETs, also treated PRRT, but it is still not a perfect biomarker for assessing the response to treatment [1, 23–25]. According to our current knowledge, there are no data on the assessment of angiogenic factors as potential biomarkers useful in monitoring patients treated with PRRT. The aim of the study was to assess the concentration of CgA and angiogenesis factors (VEGF, VEGFR-1, VEGFR-2, VEGFR-3) in a group of patients with NETs before and after PRRT treatment, and to assess whether angiogenic factors could be useful in assessing the effectiveness of PRRT.

Material and methods

Patients

The study included patients under the care of the Department of Endocrinology and Neuroendocrine Tumours, the European Neuroendocrine Tumour Society (ENETS) Centre of Excellence in Katowice, with histopathologically confirmed advanced neuroendocrine tumours (NET G1/G2 with Ki-67 < or = 20%) in IV clinical stage according to the Tumor–Nodes–Metastases (TNM) American Joint Committee on Cancer(AJCC)/Union for International Cancer Control (UICC) classification and those qualified for radioisotope therapy using yttrium-90 (90Y)/lutetium-177 (177Lu)-DOTA-0-Tyr3-Octreotate (DOTATATE).

The radioisotope treatment was applied in standard 4-course protocols. Patients received lutetium (7.4 GBq of [177Lu]Lu-DOTA-TATE) (LutaPol®, Polatom, Otwock, Poland) or tandem therapy (1.85 GBq [90Y]Y-DOTA-TATE + 1.85 GBq [177Lu]Lu-DOTA-TATE) (ItraPol®, Polatom, Otwock, Poland and LutaPol®, Polatom, Otwock, Poland). During 8–12-week intervals, long-lasting somatostatin analogues were also administered: octreotide (30 mg) or lanreotide (120 mg) every 4 weeks.

Laboratory parameters were assessed before PRRT treatment (3–6 months) and after PRRT treatment (2–6 months). The stage of the disease and the differentiation of the tumour were assessed based on the current TNM staging and grading system for NET classifications according to the World Health Organisation (WHO) 2019 criteria [2]. Disease status was assessed according to radiological Response Evaluation Criteria in Solid Tumours (RECIST) 1.1 criteria. The exclusion criteria from the study were the presence or suspicion of another malignant tumour, advanced heart failure, and renal failure in stages IV and V. The study was conducted in accordance with ethical standards and approved by the Bioethics Committee of the Silesia Medical University in Katowice.

Enzyme-linked immunosorbent assay (ELISA)

Serum levels of VEGF, VEGFR-1, and VEGFR-2 were determined using the Quantikine Human Immunoassay (R&D Systems) and VEGFR-3 by Platinum ELISA (Bioscience), according to the manufacturer’s protocol. Blood specimens were collected during hospitalisations. Fasting blood samples at 8.00 a.m. from an arm vein were gathered. Until the analysis, the serum was stored at –80oC. The serum levels of Cg were determined using a uQuant (Bio-Tek).

Statistical analyses

Statistical analyses were carried out using STATISTICA version 13.36.0 (StatSoft) software. The distribution of the data was determined by the Kolmogorov-Smirnov test. Data are presented as median and interquartile ranges for nonparametric data. The comparison of CgA, VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 concentrations between the NET patients before and after PRRT (naïve and receiving PRRT) was performed using the Wilcoxon test for paired samples. To investigate the prognostic value of CgA, VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 in predicting PRRT response in NET patients, receiver operating characteristic (ROC) curves were plotted, and the sensitivity, specificity, and area under the curve (AUC) were calculated. For correlation analysis, p values and correlation coefficients (r) were calculated using Spearman’s correlation test. Results were considered significant at p < 0.05.

Results

Clinical characteristics of the study group

The demographic, biochemical, and clinical characteristics of the participants recruited for the study are presented in Table 1. The NET patient cohort consisted of 47.5% males and 52.5% females, with a median age of 54 years. All patients were diagnosed with well-differentiated NETs; 45% each of NET G1 and NET G2. All of them had advanced disease IV stages of TNM (100% of NET patients had distant metastases) at the time of PRRT starting. The most common primary site location was the pancreas (37.5%). Of these patients 42.5% had carcinoid syndrome.

Table 1. Clinical characteristics of the neuroendocrine tumour (NET) patients/the study group

Value

Study group (n = 40)

Age [years]

Mean [range]

54 (25–71)

Sex

Male

Female

19 (47.5%)

21 (52.5%)

Localization

GEP-NET

Pancreas

Small bowel

Rectum

Unknown primary site

BP-NET

15 (37.5%)

13 (32.5%)

1 (2.5%)

7 (17.5%)

4 (10%)

Tumour grade

GEP-NET

G1

G2

BP-NET

Typical

Atypical

36 (90%)

18 (45%)

18 (45%)

4 (10%)

1 (2.5%)

3 (7.5%)

Stage

IV

40 (100%)

Carcinoid syndrome

Yes

No

17 (42.5%)

23 (57.5%)

Kind of treatment

SSA

Yes

No

Previous surgery

Yes

No

PRRT

Yes

No

40 (100%)

0 (0%)

19 (47.5)

21 (52.5)

40 (100%)

0 (0%)

Disease stage after PRRT*

SD

PD

PR

20 (50%)

6 (15%)

14 (35%)

CgA and angiogenic factors: VEGF, VEGF-R1, VEGF-R2, and VEGF-R3

In the next step, we used the Wilcoxon signed rank test to test 2 dependent samples (before and after PRRT), and thus we analysed whether there was a significant difference between the levels of these biomarkers (CgA, VEGF, VEGF-R1, VEGF-R2, and VEGF-R3). The Wilcoxon test showed that these differences were statistically significant (p < 0.05), comparing CgA, VEGF, VEGF-R1, VEGF-R2, and VEGF-R3 in NET patients before and after PRRT, in all biomarkers, except VEGF levels. As shown in Table 2.

Table 2. The Wilcoxon matched pairs test of chromogranin A (CgA), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 1 (VEGF-R1), vascular endothelial growth factor receptor 2 (VEGF-R2), and vascular endothelial growth factor receptor 3 (VEGF-R3) for patients with neuroendocrine tumours (NET) treated with peptide receptor radionuclide therapy (PRRT)

Matched pairs of variables

z

p

CgA before and CgA after

2.25

0.02

VEGF before and VEGF after

0.58

0.56

VEGF R1 before and VEGF R1 after

3.14

< 0.01

VEGF R2 before and VEGF R2 after

2.54

0.01

VEGF R3 before and VEGF R3 after

2.16

0.03

Stratifying tumour marker levels by the differences before and after PRRT identified that individuals after PRRT exhibited significantly lower levels of CgA (1390.03 ± 3248.09 (233.94 [68.82–1200.25])), VEGF-R1 (78.03 ± 29.69 (72.40 [60.70–88.00])), and VEGF-R2 (6635.36 ± 1714.11 (6284.00 [5298.00–7601.00])) than those before PRRT CgA (2151.11 ± 3811.02 (429.97 [79.41–2533.33])), VEGF-R1 (96.62 ± 36.55 (90.30 [71.45–100.50]), and VEGF-R2 (7537.05 ± 1884.36 (7277.00 [6032.00–8849.00]), respectively) (p < 0.05) (Tab. S1 in supplementary material).

Levels of VEGF did not differ significantly between NET patients before PRRT versus NET patients after PRRT (p > 0.05) (Tab. 2 and Tab. S1 in Supplementary Material).

Given the concentration of VEGF-R3, in the group of patients before PRRT, VEGF-R3 concentrations were significantly lower than those after treatment (Tab. S1 in Supplementary Materials).

In the third step, the ROC analysis and AUC were used to assess the capacity of these biomarkers to predict PRRT response based on biomarker level changes. AUC analyses could differentiate PRRT-non-treated from PRRT-treated NET patients for VEGF-R1, VEGF-R2, and VEGF-R3. The authors differentiated the AUC value only between pre- and post-PRRT treatment.

Based on AUROC analysis (Tab. S2 in Supplementary Material), we noted that the highest statistically significant AUROC for differentiating NET patients before PRRT from NET patients after PRRT had VEGF-R1 (0.70) (p < 0.01), and their accuracy for differentiating these patient groups was 70%. Also, VEGF-R2 and VEGF-R3 could differentiate both NET patient groups. Although significant (p < 0.05), it should be noted that their AUCs < 0.7 indicate that they are poor predictive PRRT-response markers. The results are shown in Figure 1.

178346.png
Figure 1. Receiver operating characteristic (ROC) curves for differentiating PRRT-non-treated from PRRT-treated patients with neuroendocrine tumours (NET) (PRRT peptide receptor radionuclide therapy); A. The area under the receiver operating characteristic (AUROC) for all markers in NET patients before PRRT and after PRRT; B. The AUROC for vascular endothelial growth factor receptor 1 (VEGF-R1) levels in NET patients before PRRT and after PRRT. The AUROC (blue curve) for differentiating NET patients before PRRT from NET patients after PRRT was 0.70 [95% confidence interval (CI): 0.59–0.82. p < 0.01]. A maximum AUC = 1 identifies an ideal (perfect) differentiation between these groups. The diagonal red line (AUC = 0.5) corresponds to chance discrimination. The VEGF-R1 AUC = 0.65 (blue curve) and p < 0.01 indicate that it is a good biomarker, and it can differentiate NET patients before PRRT from NET patients after PRRT

Discussion

PRRT therapy is most often the second line of treatment in patients with advanced, unresectable G1/G2 NETs with disease progression [2]. Nowadays, the response to PRRT therapy can be assessed using anatomical imaging, i.e. in tests such as computed tomography/magnetic resonance imaging (CT/MRI) (according to the RECIST 1.1 criteria), by scintigraphic tests (the use of [68Ga]Ga-DOTA-TATE PET/CT is of particular importance), and assessment of the concentration of non-specific markers in the blood, such as CgA [26]. The [68Ga]Ga-DOTA-TATE PET/CT scan shows the greatest sensitivity in detecting bone changes in contrast to anatomical imaging, but it is still unknown how to interpret changes in radiotracer uptake after treatment, because reduced tracer uptake may indicate a lower number of somatostatin receptors (SSTR) for various reasons (including disease progression or response to therapy) [26]). Moreover, due to the nature of NETs, which tend to grow slowly, it is not entirely clear whether the RECIST 1.1 scale is the appropriate parameter [26]. CgA is a commonly used biomarker in the clinical practice for monitoring patients with NETs, also treated with PRRT. However, this is a non-specific biomarker that can be influenced by many factors [27]. Evaluation of the multi-gene biomarker NETest in the blood of NETs has shown that it is significantly superior to CgA, and the use of predictive genes (PPQ) can accurately determine which patients will benefit from PRRT therapy and then monitor the disease, but the availability of this biomarker is currently limited [28]. Effective biomarkers are still being sought to assess the response to the treatment in NET patients [2].

The role of VEGF and VEGF-R has been confirmed in numerous studies in malignant tumours [11, 29], including NETs [5, 30–32]. According to Bates et al., epithelial-mesenchymal transition (EMT), a process that facilitates the progression of cancer in colon cancer, is associated with significant expression of VEGF and VEGF-R1, and blocking VEGF-R1 is associated with massive apoptosis in cells which were in EMT [14]. There are many reports in the literature on the correlation of angiogenic factors with metastatic disease in NETs [15]. Hansel et al. examined 19 primary well-differentiated pancreatic NETs and 7 liver metastases to determine the expression of VEGF-A and its family member VEGF-C by immunolabeling analysis. The investigators showed that VEGF-C showed low to moderate expression in primary pancreatic NETs with significantly increased expression in liver metastases, while increased expression of VEGFR-2 and VEGFR-3 suggested a possible role in autocrine and paracrine tumourigenesis processes [33]. A summary of current research on the importance of angiogenic factors in GEP-NETs is presented in the work by Irina Sandra et al. [15]. According to Pavel et al. VEGF may correlate with disease progression in NETs [12]. Similarly, Berkovi´c et al. reported that VEGF is also increased in the case of GEP-NETs, especially hormonally active ones, and with lymph node metastases [34]. Similar observations were reported by the authors for VEGF-R1, which was increased in the setting of metastatic disease in NETs [13], and VEGFR-2 which may allow the prediction of overall survival (OS) in the case of pancreatic NETs [35]. In our study, we showed that in all patients included in the study in IV clinical stage, both VEGF-R1 and VEGF-R2 were increased before PRRT and significantly decreased after PRRT, while VEGF concentrations did not show statistically significant differences. Similarly, in some studies, no statistically significant differences were found between VEGF concentrations in the group of patients with NENs and in the control group [36]. As is known, tissue hypoxia associated with flow stasis in damaged vessels increases the concentration of VEGF, which is a factor promoting tumour growth and progression in malignant tumours [37]. It is worth mentioning, however, that paradoxically in the case of NETs, the angiogenesis seems to be independent of tissue hypoxia, and this phenomenon has been presented as the so-called “neuroendocrine paradox”, in which highly differentiated NETs with a low degree of malignancy are characterised by the richest vascularisation, and therefore the density of the vascular network corresponds to the degree of differentiation rather than the degree of aggressiveness of the tumour [15]. Highly differentiated NETs can synthesise and constitutively secrete VEGF into the bloodstream, while in low-differentiated NETs this process is not constant [38].

Attempts to use the assessment of angiogenic factors in assessing treatment effectiveness have been studied for other therapies in NETs. In clinical practice, therapies currently used in NETs are related to angiogenesis pathways. In the first-line treatment of advanced or metastatic, slowly growing, well-differentiated G1/G2 NETs, somatostatin analogues (SSAs) are primarily used [1, 39]. SSAs have their place in the treatment of NETs due to their antiangiogenic effect directly through the presence of somatostatin receptors on endothelial cells, as well as indirectly by inhibiting the secretion of growth factors [40, 41]. However, Rosiek et al. showed that angiogenesis factors (VEGF and VEGF-R1) seem to have limited use in assessing the effectiveness of SSA treatment in NETs [42]. The study observed a decrease in VEGF concentration and an increase in VEGF-R1 concentration during treatment, while VEGF-R1 showed the best effectiveness in differentiating patients with NETs from healthy individuals [42]. Another study assessing the effectiveness of SSAs treatment in patients with NENs showed that the greatest decrease in VEGF-R2 occurred after 2 years of SSAs treatment, although, as the authors emphasise, the tested angiogenic factors (VEGF-R2, VEGF-R3 and vascular cell adhesion molecule 1 (VCAM-1) are not effective in monitoring patients treated with SSAs [43]. In our work, we confirmed that VEGF-R1 and VEGF-R2 decreased significantly after PRRT treatment, but only VEGF-R1 is a potential biomarker that can be used to assess the effectiveness of PRRT treatment. In the advanced stage of pancreatic NETs G1/G2 disease, 2 drugs with anti-angiogenic properties are also used: a selective m-TOR pathway inhibitor everolimus, and a tyrosine kinase receptor inhibitor sunitinib [44, 45]. In the randomised phase III RADIANT-3 clinical trial, everolimus treatment also led to a significant and progressive reduction in VEGF-R2 [45]. The role of sunitinib in the angiogenesis process has also been studied to assess the effectiveness of therapy and monitor patients [46]. Similarly, one study found that the mean plasma VEGF-R2 concentration was reduced after treatment [38, 47]. Likewise, in another study in NETs with metastatic disease, after 28 days of sunitinib administration, VEGFR-2 and VEGF-R3 levels decreased by30% in approximately 60% and 70% of all patients, respectively, and returned to baseline values after 2 weeks of treatment break [38, 48]. It is not clear why VEGF-R3 increased after radioisotope therapy in our study. However, some authors emphasised that serum VEGF levels significantly correlated with VEGF-R3 in colorectal cancer [49]. Other authors confirmed that VEGF-R3 is not an effective marker in assessing patients treated with SSAs in NETs [43].

Considering the complexity and heterogeneity of NETs treated with PRRT [50], the importance of molecular imaging phenotyping for effective PRRT therapy and an individual approach to treatment is emphasised [51]. Gianetta et al. report that the inflammatory process associated with tumour-associated neutrophils (TAN) promotes the disease progression through the high expression of pro-angiogenic factors, such as VEGF [38]. Ohlendorf et al. also tried to assess whether cancer-related inflammatory markers might play a role in patients with GEP-NETs treated with PRRT and showed that, although these parameters showed significant heterogeneity, they were higher in patients not responding to PRRT therapy [52].

There are no data in the literature regarding the assessment of angiogenic factors in patients treated with PRRT. In our opinion, further prospective studies are still needed to precisely assess these parameters in a larger group of NET patients.

Conclusions

Although the concentrations of CgA, VEGF-R1, and VEGF-R2 decreased significantly after PRRT therapy, only VEGF-R1 is a potential biomarker in assessing the effectiveness of PRRT treatment. In the case of progressive patients with NETs undergoing PRRT treatment, the assessment of angiogenic factors seems important, but further prospective studies are still needed to precisely assess these parameters in NET patients.

Advantages of the study

According to the available literature (PubMed Database), this is one of the first studies evaluating angiogenic factors in NET patients treated with PRRT.

Limitations of the study

This study has potential limitations: it is limited by the small number of patients and limited follow-up time.

Funding

This work was supported by the Ministry of Science and Higher Education: KNW1-130/N/8/K.

Author contributions

Conceptualisation, J.S, K.M.; Methodology, J.S, M.W.-G., V.R. Formal Analysis, J.S., V.R.; Investigation, J.S., M.W.-G., K.M.; Resources, J.S., K.M., M.W.G., Writing Original Draft Preparation, J.S; M.W.-G., K.M., Writing Review & Editing, V.R., G.K., D.K., B.K-K.; Visualization, M.W-G., V.R.; Supervision, B.K.-K.; Project Administration, J.S.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the Ethics Committee of the Medical University of Silesia (approval codes: PCN/022/KB1/97/I/II/19/20). All patients and controls signed the informed consent.

Informed consent statement

Informed consent was obtained from all subjects involved in the study.

Data availability statement

The data used to support the findings of this research are available upon request from the corresponding author, Janusz Strzelczyk: janusz.strzelczyk@sum.edu.pl.

Conflicts of interest

The Authors declare no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

References

  1. Pavel M, Öberg K, Falconi M, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020; 31(7): 844–860, doi: 10.1016/j.annonc.2020.03.304, indexed in Pubmed: 32272208.
  2. Kos-Kudła B, Foltyn W, Malczewska A, et al. Update of the diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours) [Aktualizacja zaleceń ogólnych dotyczących postępowania diagnostyczno-terapeutycznego w nowotworach neuroendokrynnych układu pokarmowego (rekomendowane przez Polską Sieć Guzów Neuroendokrynnych)]. Endokrynol Polska. 2022; 73(3): 387–454, doi: 10.5603/ep.a2022.0049, indexed in Pubmed: 36059171.
  3. Dasari A, Shen C, Halperin D, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol. 2017; 3(10): 1335–1342, doi: 10.1001/jamaoncol.2017.0589, indexed in Pubmed: 28448665.
  4. Gustafsson BI, Kidd M, Chan A, et al. Bronchopulmonary neuroendocrine tumors. Cancer. 2008; 113(1): 5–21, doi: 10.1002/cncr.23542, indexed in Pubmed: 18473355.
  5. Bowen KA, Silva SR, Johnson JN, et al. Gastroent Surg 09 Expression VEGFR GI-NET. 2010; 13(10): 1773–1780, doi: 10.1007/s11605-009-0958-8.An.
  6. Dasari A, Hamilton EP, Falchook GS, et al. A dose escalation/expansion study evaluating dose, safety, and efficacy of the novel tyrosine kinase inhibitor surufatinib, which inhibits VEGFR 1, 2, & 3, FGFR 1, and CSF1R, in US patients with neuroendocrine tumors. Invest New Drugs. 2023; 41(3): 421–430, doi: 10.1007/s10637-023-01359-2, indexed in Pubmed: 37074571.
  7. Dasari A, Shen C, Halperin D, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol. 2017; 3(10): 1335–1342, doi: 10.1001/jamaoncol.2017.0589, indexed in Pubmed: 28448665.
  8. Das S, Phillips S, Lee CL, et al. Efficacy and toxicity of anti-vascular endothelial growth receptor tyrosine kinase inhibitors in patients with neuroendocrine tumours - A systematic review and meta-analysis. Eur J Cancer. 2023; 182: 43–52, doi: 10.1016/j.ejca.2022.12.031, indexed in Pubmed: 36738541.
  9. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000; 407(6801): 249–257, doi: 10.1038/35025220, indexed in Pubmed: 11001068.
  10. Carmeliet P. VEGF as a key mediator of angiogenesis in cancer. Oncology. 2005; 69 Suppl 3: 4–10, doi: 10.1159/000088478, indexed in Pubmed: 16301830.
  11. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013; 13(12): 871–882, doi: 10.1038/nrc3627, indexed in Pubmed: 24263190.
  12. Pavel ME, Hassler G, Baum U, et al. Circulating of Angiogenic Cytokines Can Predict Tumour Progression and Prognosis in Neuroendocrine Carcinomas. Clin. Endocrinol. 2005; 62(4): 434–443, doi: 101111/j.1365-2265.2005.02238, indexed in Pubmed: 15807874.
  13. Hilfenhaus G, Göhrig A, Pape UF, et al. Placental growth factor supports neuroendocrine tumor growth and predicts disease prognosis in patients. Endocr Relat Cancer. 2013; 20(3): 305–319, doi: 10.1530/ERC-12-0223, indexed in Pubmed: 23463017.
  14. Bates RC, Goldsmith JD, Bachelder RE, et al. Flt-1-dependent survival characterizes the epithelial-mesenchymal transition of colonic organoids. Curr Biol. 2003; 13(19): 1721–1727, doi: 10.1016/j.cub.2003.09.002, indexed in Pubmed: 14521839.
  15. Sandra I, Cazacu IM, Croitoru VM, et al. Circulating Angiogenic Markers in Gastroenteropancreatic Neuroendocrine Neoplasms: A Systematic Review. Curr Issues Mol Biol. 2022; 44(9): 4001–4014, doi: 10.3390/cimb44090274, indexed in Pubmed: 36135186.
  16. Panzuto F, Massironi S, Partelli S, et al. Gastro-entero-pancreatic neuroendocrine neoplasia: The rules for non-operative management. Surg Oncol. 2020; 35: 141–148, doi: 10.1016/j.suronc.2020.08.015, indexed in Pubmed: 32877883.
  17. Ambrosini V, Kunikowska J, Baudin E, et al. Consensus on molecular imaging and theranostics in neuroendocrine neoplasms. Eur J Cancer. 2021; 146: 56–73, doi: 10.1016/j.ejca.2021.01.008, indexed in Pubmed: 33588146.
  18. Kwekkeboom DJ, Teunissen JJ, Bakker WH, et al. Radiolabeled somatostatin analog [177Lu-DOTA0,Tyr3]octreotate in patients with endocrine gastroenteropancreatic tumors. J Clin Oncol. 2005; 23(12): 2754–2762, doi: 10.1200/JCO.2005.08.066, indexed in Pubmed: 15837990.
  19. Strosberg J, El-Haddad G, Wolin E, et al. NETTER-1 Trial Investigators. Phase 3 Trial of Lu-Dotatate for Midgut Neuroendocrine Tumors. N Engl J Med. 2017; 376(2): 125–135, doi: 10.1056/NEJMoa1607427, indexed in Pubmed: 28076709.
  20. Sowa-Staszczak A, Pach D, Chrzan R, et al. Peptide receptor radionuclide therapy as a potential tool for neoadjuvant therapy in patients with inoperable neuroendocrine tumours (NETs). Eur J Nucl Med Mol Imaging. 2011; 38(9): 1669–1674, doi: 10.1007/s00259-011-1835-8, indexed in Pubmed: 21559978.
  21. Feijtel D, Doeswijk GN, Verkaik NS, et al. Inter and intra-tumor somatostatin receptor 2 heterogeneity influences peptide receptor radionuclide therapy response. Theranostics. 2021; 11(2): 491–505, doi: 10.7150/thno.51215, indexed in Pubmed: 33391488.
  22. Feijtel D, de Jong M, Nonnekens J. Peptide Receptor Radionuclide Therapy: Looking Back, Looking Forward. Curr Top Med Chem. 2020; 20(32): 2959–2969, doi: 10.2174/1568026620666200226104652, indexed in Pubmed: 32101125.
  23. Glinicki P, Jeske W. [Chromogranin A (CgA) - characteristic of the currently available laboratory methods and conditions which can influence the results]. Endokrynol Pol. 2009; 60(5): 415–419, indexed in Pubmed: 19885814.
  24. Plöckinger U, Rindi G, Arnold R, et al. European Neuroendocrine Tumour Society. Guidelines for the diagnosis and treatment of neuroendocrine gastrointestinal tumours. A consensus statement on behalf of the European Neuroendocrine Tumour Society (ENETS). Neuroendocrinology. 2004; 80(6): 394–424, doi: 10.1159/000085237, indexed in Pubmed: 15838182.
  25. Glinicki P, Kapuścińska R, Jeske W. The differences in chromogranin A (CgA) concentrations measured in serum and in plasma by IRMA and ELISA methods. Endokrynol Pol. 2010; 61(4): 346–350, indexed in Pubmed: 20806177.
  26. Huizing DMV, Aalbersberg EA, Versleijen MWJ, et al. Early response assessment and prediction of overall survival after peptide receptor radionuclide therapy. Cancer Imaging. 2020; 20(1): 57, doi: 10.1186/s40644-020-00335-w, indexed in Pubmed: 32778165.
  27. Gut P, Czarnywojtek A, Fischbach J, et al. Chromogranin A - unspecific neuroendocrine marker. Clinical utility and potential diagnostic pitfalls. Arch Med Sci. 2016; 12(1): 1–9, doi: 10.5114/aoms.2016.57577, indexed in Pubmed: 26925113.
  28. Malczewska A, Kos-Kudła B, Kidd M, et al. The clinical applications of a multigene liquid biopsy (NETest) in neuroendocrine tumors. Adv Med Sci. 2020; 65(1): 18–29, doi: 10.1016/j.advms.2019.10.002, indexed in Pubmed: 31841822.
  29. Mulder BG, Koller M, Duiker EW, et al. Intraoperative Molecular Fluorescence Imaging of Pancreatic Cancer by Targeting Vascular Endothelial Growth Factor: A Multicenter Feasibility Dose-Escalation Study. J Nucl Med. 2023; 64(1): 82–89, doi: 10.2967/jnumed.121.263773, indexed in Pubmed: 35680414.
  30. Zhang J, Jia Z, Li Q, et al. Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors. Cancer. 2007; 109(8): 1478–1486, doi: 10.1002/cncr.22554, indexed in Pubmed: 17340592.
  31. Terris B, Scoazec JY, Rubbia L, et al. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology. 1998; 32(2): 133–138, doi: 10.1046/j.1365-2559.1998.00321.x, indexed in Pubmed: 9543669.
  32. Silva SR, Bowen KA, Rychahou PG, et al. VEGFR-2 expression in carcinoid cancer cells and its role in tumor growth and metastasis. Int J Cancer. 2011; 128(5): 1045–1056, doi: 10.1002/ijc.25441, indexed in Pubmed: 20473929.
  33. Hansel DE, Rahman A, Hermans J, et al. Liver metastases arising from well-differentiated pancreatic endocrine neoplasms demonstrate increased VEGF-C expression. Mod Pathol. 2003; 16(7): 652–659, doi: 10.1097/01.MP.0000077416.68489.50, indexed in Pubmed: 12861060.
  34. Cigrovski Berković M, Čačev T, Catela Ivković T, et al. High VEGF serum values are associated with locoregional spread of gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Mol Cell Endocrinol. 2016; 425: 61–68, doi: 10.1016/j.mce.2016.01.013, indexed in Pubmed: 26805636.
  35. Zurita AJ, Khajavi M, Wu HK, et al. Circulating cytokines and monocyte subpopulations as biomarkers of outcome and biological activity in sunitinib-treated patients with advanced neuroendocrine tumours. Br J Cancer. 2015; 112(7): 1199–1205, doi: 10.1038/bjc.2015.73, indexed in Pubmed: 25756398.
  36. Strzelczyk J, Wójcik-Giertuga M, Cuber P, et al. Assessment of the Concentration of Endogenous Factors Regulating Angiogenesis, VASH-1 and VEGF-A, in the Blood Serum of Patients with Neuroendocrine Neoplasms. Biomed Res Int. 2022; 2022: 9084393, doi: 10.1155/2022/9084393, indexed in Pubmed: 35372578.
  37. Leong A, Kim M. The Angiopoietin-2 and TIE Pathway as a Therapeutic Target for Enhancing Antiangiogenic Therapy and Immunotherapy in Patients with Advanced Cancer. Int J Mol Sci. 2020; 21(22), doi: 10.3390/ijms21228689, indexed in Pubmed: 33217955.
  38. Giannetta E, La Salvia A, Rizza L, et al. Are Markers of Systemic Inflammatory Response Useful in the Management of Patients With Neuroendocrine Neoplasms? Front Endocrinol (Lausanne). 2021; 12: 672499, doi: 10.3389/fendo.2021.672499, indexed in Pubmed: 34367064.
  39. Halfdanarson TR, Strosberg JR, Tang L, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020; 49(7): 863–881, doi: 10.1097/MPA.0000000000001597, indexed in Pubmed: 32675783.
  40. Dasgupta P. Somatostatin analogues: multiple roles in cellular proliferation, neoplasia, and angiogenesis. Pharmacol Ther. 2004; 102(1): 61–85, doi: 10.1016/j.pharmthera.2004.02.002, indexed in Pubmed: 15056499.
  41. García de la Torre N, Wass JAH, Turner HE. Antiangiogenic effects of somatostatin analogues. Clin Endocrinol (Oxf). 2002; 57(4): 425–441, doi: 10.1046/j.1365-2265.2002.01619.x, indexed in Pubmed: 12354124.
  42. Rosiek V, Janas K. Assessment of VEGF and VEGF R1 serum levels in patients with neuroendocrine neoplasms before and after treatment with first-generation somatostatin analogues. Endokrynol Pol. 2022; 73(3): 612–618, doi: 10.5603/EP.a2022.0032, indexed in Pubmed: 36059176.
  43. Rosiek V, Janas K, Kos-Kudła B. Association between Biomarkers (VEGF-R2, VEGF-R3, VCAM-1) and Treatment Duration in Patients with Neuroendocrine Tumors Receiving Therapy with First-Generation Somatostatin Analogues. Biomedicines. 2023; 11(3), doi: 10.3390/biomedicines11030842, indexed in Pubmed: 36979820.
  44. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011; 364(6): 501–513, doi: 10.1056/NEJMoa1003825, indexed in Pubmed: 21306237.
  45. Yao JC, Shah MH, Ito T, et al. RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT-3) Study Group. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011; 364(6): 514–523, doi: 10.1056/NEJMoa1009290, indexed in Pubmed: 21306238.
  46. Mateo J, Heymach JV, Zurita AJ. Circulating biomarkers of response to sunitinib in gastroenteropancreatic neuroendocrine tumors: current data and clinical outlook. Mol Diagn Ther. 2012; 16(3): 151–161, doi: 10.2165/11632590-000000000-00000, indexed in Pubmed: 22515658.
  47. Faivre S, Delbaldo C, Vera K, et al. Safety, pharmacokinetic, and antitumor activity of SU11248, a novel oral multitarget tyrosine kinase inhibitor, in patients with cancer. J Clin Oncol. 2006; 24(1): 25–35, doi: 10.1200/JCO.2005.02.2194, indexed in Pubmed: 16314617.
  48. Bello CD, Friece C, Smeraglia J, et al. Analysis of Circulating Biomarkers of Sunitinib Malate in Patients With Unresectable Neuroendocrine Tumors (NET): VEGF, IL-8, and Soluble VEGF Receptors 2 and 3. J Clin Oncol. 2006; 24(18_Suppl), doi: https://doi.org/10.1200/jco.2006.24.18_suppl.4045.
  49. Goulart A, Ferreira C, Rodrigues A, et al. The correlation between serum vascular endothelial growth factor (VEGF) and tumor VEGF receptor 3 in colorectal cancer. Ann Surg Treat Res. 2019; 97(1): 15–20, doi: 10.4174/astr.2019.97.1.15, indexed in Pubmed: 31297348.
  50. Durma AD, Saracyn M, Kołodziej M, et al. Epidemiology of Neuroendocrine Neoplasms and Results of Their Treatment with [Lu]Lu-DOTA-TATE or [Lu]Lu-DOTA-TATE and [Y]Y-DOTA-TATE-A Six-Year Experience in High-Reference Polish Neuroendocrine Neoplasm Center. Cancers (Basel). 2023; 15(22), doi: 10.3390/cancers15225466, indexed in Pubmed: 38001726.
  51. Iravani A, Parihar AS, Akhurst T, et al. Molecular imaging phenotyping for selecting and monitoring radioligand therapy of neuroendocrine neoplasms. Cancer Imaging. 2022; 22(1): 25, doi: 10.1186/s40644-022-00465-3, indexed in Pubmed: 35659779.
  52. Ohlendorf F, Werner RA, Henkenberens C, et al. Predictive and Prognostic Impact of Blood-Based Inflammatory Biomarkers in Patients with Gastroenteropancreatic Neuroendocrine Tumors Commencing Peptide Receptor Radionuclide Therapy. Diagnostics (Basel). 2021; 11(3), doi: 10.3390/diagnostics11030504, indexed in Pubmed: 33809226.