Next Article in Journal
High Glucose Induces Late Differentiation and Death of Human Oral Keratinocytes
Next Article in Special Issue
Modulation of Spheroid Forming Capacity and TRAIL Sensitivity by KLF4 and Nanog in Gastric Cancer Cells
Previous Article in Journal
Prophylactic Effects of Purple Shoot Green Tea on Cytokine Immunomodulation through Scavenging Free Radicals and NO in LPS-Stimulated Macrophages
Previous Article in Special Issue
The Expression and Prognostic Significance of VEGF and CXCR4 in Gastric Cancer: Correlation with Angiogenesis, Lymphangiogenesis and Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Circulating Angiogenic Markers in Gastroenteropancreatic Neuroendocrine Neoplasms: A Systematic Review

by
Irina Sandra
1,2,
Irina Mihaela Cazacu
2,3,
Vlad Mihai Croitoru
2,3,
Mariana Mihaila
4,*,
Vlad Herlea
3,5,
Mircea Mihai Diculescu
1,6,
Simona Olimpia Dima
3,7,8,† and
Adina Emilia Croitoru
2,3,†
1
Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 020022 Bucharest, Romania
2
Department of Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
3
Faculty of Medicine, “Titu Maiorescu” University of Medicine and Pharmacy, 020022 Bucharest, Romania
4
Department of Internal Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
5
Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania
6
Department of Gastroenterology and Hepatology, Fundeni Clinical Institute, 022328 Bucharest, Romania
7
Department of General Surgery, Fundeni Clinical Institute, 022328 Bucharest, Romania
8
Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Curr. Issues Mol. Biol. 2022, 44(9), 4001-4014; https://doi.org/10.3390/cimb44090274
Submission received: 5 July 2022 / Revised: 26 August 2022 / Accepted: 30 August 2022 / Published: 4 September 2022
(This article belongs to the Special Issue Molecular-Based Approaches in Therapy for Gastrointestinal Cancers)

Abstract

:
Background: Neuroendocrine neoplasms are a heterogeneous group of tumors that raise challenges in terms of diagnosis, treatment and monitoring. Despite continuous efforts, no biomarker has showed satisfying accuracy in predicting outcome or response to treatment. Methods: We conducted a systematic review to determine relevant circulating biomarkers for angiogenesis in neuroendocrine tumors. We searched three databases (Pubmed, Embase, Web of Science) using the keywords “neuroendocrine” and “biomarkers”, plus specific biomarkers were searched by full and abbreviated name. From a total of 2448 publications, 11 articles met the eligibility criteria. Results: VEGF is the most potent and the most studied angiogenic molecule, but results were highly controversial. Placental growth factor, Angiopoietin 2 and IL-8 were the most consistent markers in predicting poor outcome and aggressive disease behavior. Conclusions: There is no robust evidence so far to sustain the use of angiogenic biomarkers in routine practice, although the results show promising leads.

1. Introduction

Neuroendocrine neoplasms (NENs) comprise a group of genetically and clinically diverse epithelial tumors derived from the neuroendocrine system, with primary sites located in most organs [1]. In 50% of cases, primary tumors are located in the gastrointestinal tract and pancreas [2]. Their presence is considered uncommon, even though the age-adjusted incidence has doubled in the last two decades from 2.48 to 5.86 per 100,000/year, mostly due to improved detection and awareness [2,3].
Until recently, NENs were an obscure disorder, due to the scarcity of knowledge and limited therapeutic options. The rapid growth of interest in recent decades led to remarkable progress, with many countries developing networks of multidisciplinary experts dedicated to the management of NENs, starting with diagnosis.
Apart from its low prevalence, NENs feature intriguing particularities that stand out from other malignancies. Around 40% of these tumors retained their original capacity to synthesize and excrete bioactive molecules, such as insulin, gastrin, glucagon or somatostatin, and are associated with symptoms of variable degrees due to hormone overproduction (Neuroendocrine Tumor of the Gastrointestinal Tract: Introduction, 2021). Another distinctive characteristic is the wide range of behaviors, from completely dormant tumors to aggressive progression and metastasis. It appears that pancreatic neuroendocrine neoplasms (pNENs) exhibit a slightly worse outcome than other gastrointestinal NENs (GEP-NENs), except insulinomas [4]. Furthermore, neuroendocrine tumors benefit from a rich vascularization network compared to other solid tumors [5]. This interesting feature has been proven useful in clinical practice for diagnostic and therapeutic purposes and shows promising perspectives to extend its role as a non-invasive predictive marker.
Monitoring progression or response to treatment might be challenging, especially in slowly progressive well-differentiated NENs, where radiologic evaluation can be inconclusive. Currently, the most commonly used predictive markers in routine practice are disease stage, tumor cell differentiation and proliferation, quantified by Ki-67 [6]. The last one was included in the three available NEN staging systems (WHO, ENETS and AJCC). However, clinical experience has proven countless times that individuals belonging to the same disease stage evolve differently and respond variably to therapy. One of the major research directions in the field of NENs is identifying non-invasive and reliable biomarkers to circumvent this issue. Initially, efforts were focused on predicting survival and tumor progression (prognostic biomarker). Gradually, studies oriented towards response to treatment [7] and, eventually, to acquired loss of response (predictive biomarker) [8].
The aim of this study was to conduct a systematic review of the literature to determine relevant circulant biomarkers of angiogenesis in neuroendocrine tumors.

2. Materials and Methods

This systematic review was carried out according to the PRISMA standards. We searched in three databases using the keywords “neuroendocrine” in title or abstract AND specific biomarkers by full name and abbreviated form in full text: vascular endothelial growth factor (VEGF), angiopoietin (Ang), platelet-derived growth factor (PDGFR), basic fibroblast growth factor (bFGF), placental growth factor (PlGF), interleukin (IL)-8 and circulating endothelial cells.
Due to the scarcity of the results, a broader search was conducted using the keywords “neuroendocrine” AND “biomarker” to ensure that all relevant literature was covered. From the search results, only articles including gastroenteropancreatic NEN were selected. The search was last updated on 29 March 2022. In total, 2448 publications were primarily retrieved in PubMed, Scopus and Web of Science. We added 13 after manually searching major scientific journals and from other references. After excluding 503 duplicates, 1958 titles were screened for eligibility and 352 articles were assessed for full-text evaluation. Only human studies were accepted and most articles were excluded due to irrelevancy. We focused mainly on circulating biomarkers; therefore, studies that included only tissular markers or gene expression analysis were excluded. No cut-off point was set for the date of publishing due to the paucity of articles. At the end of the process, 11 articles were included. The search strategy is detailed in Figure 1.

3. Results and Discussion

3.1. Overview of the Selected Articles

We included 11 articles, most of them originating in Europe. Seven studies were prospective and one was designed with a retrospective and a prospective phase; seven studies included a control group and one was a placebo-controlled clinical trial. Four of the selected studies were clinical trials: two with Sunitinib (phase II and phase IV) [9,10], one with Everolimus (RADIANT 3, phase III) [11] and one with Pazopanib (PAZONET, phase II) [12]. Four studies were published more than 10 years ago. All studies included pNENs and contained previously treated patients. Some studies included extra-digestive tumor sites [12,13,14,15]. Two clinical trials were conducted exclusively on pNENs [9,11]. Biomarkers of interest were: VEGF, angiopoietin, bFGF, PlGF, IL-8, endostatin, stromal cell-derived factor-1 (SDF-1a) and circulating endothelial cells. All studies focused on progression-free survival (PFS), overall survival (OS) and response to treatment as primary or secondary endpoints. Further details on selected studies are summarized in Table 1 and Table 2.

3.2. The Current State of Biomarkers in NENs and Future Perspectives

To date, a plethora of markers has been studied and some correlated more or less ideally with tumor outcome. Classic radiological, clinical and pathological features (such as tumor grade, differentiation degree, proliferation status, neurovascular invasion, functionality and tumor spreading) are invaluable parameters to provide a basic overview and select the appropriate strategy in routine practice [6]. Assessment of common serum NEN markers is equally endorsed by guidelines [20], but the results must be interpreted with caution.
Even though the granin family consists of at least three members (chromogranin A, B and C), all secreted by neuroendocrine cells, only chromogranin A (CgA) has proven its utility in the diagnosis of NENs [21]. Its dynamic also correlates significantly with progression [22], survival [23] and response to treatment [24], making it the most valuable serological biomarker in NENs at the moment. Still, multiple conditions are falsely associated with an up to 10–15-fold increase in CgA, of which the most frequent are hypergastrinemia, renal insufficiency and use of proton-pump inhibitors [25]. Except for serotonin and its metabolite 5-hydroxyindoloacetic acid (5HIAA) in evaluating carcinoid syndrome [26], other serological biomarkers (for example: neuron-specific enolase, pancreatic polypeptide, N-terminal probrain natriuretic peptide) are inferior to CgA as diagnostic or predictive biomarkers [26,27,28]. Incremental progress has been noted in functional imaging as well, culminating in the emerging field of theranostics, which currently refines scoring systems that aim to better predict staging and clinical outcome [29]. Response to somatostatin analogs (SSAs) and peptide receptor radionuclide therapy is classically predicted using radiolabeled SSAs. Moreover, dynamic contrast-enhanced magnetic resonance imaging has shown promising leads in predicting response to anti-angiogenic treatment, even determining optimal biological drug dose [30].
Looking forward, we are on the verge of an inflection point where modern techniques, such as genome-wide expression profiling or liquid biopsy, are becoming more accessible. Next-generation sequencing has provided not only insights into the pathophysiology of NENs, but also potential targetable mutations and prognostic markers [31,32]. NETest (Wren Laboratories, Branford, CT, USA) is a polymerase chain reaction (PCR)-based multianalyte algorithmic assay using blood or tissue samples [33] that shows the most promising results so far in terms of predicting diagnosis, outcome and response to treatment [33,34,35], with the potential to detect tumor progression up to 2 years before radiological changes [36]. Circulating tumor cells, free circulating DNA and microRNA testing has been extrapolated in NENs and shown some potential, although further research is required [37,38,39,40]. Despite encouraging evidence, no biomarker has yet ideally met the need to accurately predict tumor behavior.

3.3. Angiogenesis in Neuroendocrine Tumors

Hypervascularization is a hallmark of NENs that has been explored for diagnostic and therapeutic purposes. Neoangiogenesis in NEN is based on several adaptive features that are specific to tumor cells in order to sustain the need of the rapidly developing cells. Notably, while normal tissue relies on the intricate mechanisms of sprouting and intussusception (the division of a preexisting vessel), tumor tissue has developed other pathological models in response to a hypoxic environment, of which the most important are [41]:
  • − Vessel co-option: preexisting vessels are “hijacked” to serve tumor cells.
  • − Vasculogenic mimicry: tumor cells build blood channels similar to the endothelium [42].
These mechanisms not only sustain a “comfortable” environment for tumor growth, but they are also responsible, at least in part, for eluding anti-angiogenic therapy [43]. These models are more relevant in high-grade NENs, where hypervascularization is triggered mostly by hypoxia through signaling pathways common with other malignancies [44,45].
In well-differentiated NENs, neoangiogenesis appears to be triggered by the autocrine/paracrine secretion of vascular endothelial growth factor (VEGF), regardless of tissular oxygenation. This function is inherited from normal neuroendocrine cells that constitutively synthesize VEGF and release it into the blood flow [42]. This phenomenon has been better described in pNENs as “neuroendocrine paradox”, where low-grade tumors with less aggressive behavior bear the richest vascularization [46].
Further unexpected behavior was observed in experimental mouse models showing that liver metastases do not rely on active angiogenesis until advanced stages when a certain tumor volume is reached [46]. This raises a question regarding the propitious timing for anti-angiogenic treatment [42].

3.4. The Vascular Endothelial Growth Factor Family

The vascular endothelial growth factor family and their receptors (VEGF-VEGFR/Neurolipins) represent the main proangiogenic molecules. VEGFA (also referred to as VEGF) is the first detected angiogenic molecule and the most potent [47]. It binds to VEGFR-2, resulting in the most efficient signaling to neurolipins that potentiate this angiogenic effect and to VEGFR-1 (or Flt-1) for a weaker activity [43], to the point that a reversed effect was reported by trapping its ligand [48]
Since an overwhelming proportion of angiogenesis is governed by VEGF and its signaling pathways [43], it is natural that most studies address the members of this particular family. Pavel et al. [13] were the first to analyze circulating angiogenic markers in NENs. The group consisted of 38 patients with digestive and extra-digestive primary site, mostly low grade and metastatic. VEGF levels were significantly higher in NENs compared to healthy controls, irrespective of site, grade or functionality status. On a short-term period, the dynamics of VEGF were as reliable as CgA (p < 0.0001) in predicting disease progression (p < 0.011), even though fewer samples were available for serial analysis. It is reasonable to infer that results were hardly impacted by treatment, as most pre-treated patients were on octreotide and mean VEGF levels were similar between treated and naïve patients. However, the negative influence of SSAs on VEGF levels was reported [49,50]. VEGF serum levels did not correlate with OS.
Worse outcome in patients with high VEGF levels was not confirmed in a larger study by Berkovic et al. [16]. Higher median levels of VEGF were reported in GEP-NENs, but inversely correlated with histologic differentiation and aggressiveness (notably with Ki-67 rather than grade). This is suggestive of the “neuroendocrine paradox” described in pNENs [42] but OS was not assessed in the study. Interestingly, while circulating VEGF levels were increased in metastatic disease, the highest levels were noted in lymph node metastases and the lowest in liver metastases. Berkovic et al. broke new ground with a multi-directional approach in studying the role of VEGF in GEP-NENs. They were the first to study the role of VEGF 1154A/G polymorphism in GEP-NENs. Higher VEGF serum levels were associated with the -1154G allele, thus, inferring a potential involvement of VEGF-1154 SNP (single nucleotide polymorphism) in VEGF expression, in line with the ones generated by other cancers [51,52]. However, further research is required, as the correlation was statistically significant only in non-functional non-pancreatic gastrointestinal NENs.
Five more studies included VEGF (Grande et al., Yao et al., Zurita et al., Melen-Muncha et al., Figueroa-Vega et al.), but no statistical significance was demonstrated concerning the study objectives, although two of the clinical trials involved large sample sizes.
Three clinical trials analyzed VEGFR (Zurita et al., Yao et al., Grande et al.). Zurita et al. [10] were the first to report predictive value for better survival in pancreatic (and not intestinal) well-differentiated NEN patients with high levels of VEGFR-2 at baseline (p = 0.01). No significant correlation with PFS was found. In carcinoid tumors, VEGFR-3 predicted poor outcome in terms of PFS (p = 0.006) and OS (p = 0.047). Yao et al. and Grande et al. did not report clinical correlations with VEGF receptors. Jiménez-Fonseca et al. [9] additionally tested 14 SNP variants in nine genes, including VEGFR-3. None of them were associated with outcome after applying multivariate analysis. Three other VEGFR-3 SNP variants correlated with shorter OS (rs307826 and rs307821, Zurita et al.) and shorter PFS (rs307821, Grande et al.).

3.5. Placental Growth Factor

Placental growth factor (PlGF) has a similar structure to VEGF, but it becomes active only in pathological conditions [53]. It appears to bind selectively to VEGFR-1 and neurolipins, but not to VEGFR-2, thus, increasing the availability of VEGF for its most efficient pathway [54]. Hilfenhaus et al. [19] conducted the most comprehensive study on PlGF regarding the function, the prognostic value and the potential of a single marker as a therapeutic target, with encouraging results. The study was designed to benefit from a multi-level approach: clinical analysis in serum and tissue samples collected retrospectively and prospectively, in vitro analysis using a panel of cell lines confirmed with PlGF receptors and in vivo analysis on mouse models. The study showed compelling clinical and preclinical evidence for the pivotal role of PlGF in the tumorigenesis of well-differentiated NENs. Higher levels were reported in NENs and correlated unexpectedly with grading, but not with metastatic disease. Further analysis confirmed the role of PlGF as a prognostic marker of poor outcome (indicative of shorter survival in pNENs and shorter time to progression in intestinal NENs).
An in vitro experiment not only confirmed the role of PlGF in tumor aggressiveness by stimulating proliferation and cell migration but also hypothesized that PlGF might be useful in monitoring the short-term response to angiogenic treatment (levels raised significantly in response to Sunitinib). Finally, in vivo experiment on mouse models inoculated with pNENs xenografts showed important reduction in tumor volume after PlGF inhibition, thus, revealing the opportunity of a new therapeutic target.
In a classical trial RADIANT 3, a panel of markers was assessed in a large group of well-differentiated pNENs treated with Everolimus (PlGF, VEGF-A, VEGFR1, VEGFR2, bFGF). After applying multivariate analysis, only PlGF emerged as an independent prognostic factor for disease progression.

3.6. Angiopoietin-Tie-2

Angiopoietin (Ang)-Tie: Ang-1 acts as an agonist to Tie-2, promoting vascular stability. Ang-2 is expressed in pathological conditions in response to proangiogenic molecules and functions as a context-depending antagonist, competing for the same receptor Tie-2 [55].
Srirajaskanthan et al. [14] focused on analyzing two of the most relevant exponents of the angiopoietin family (Ang-1 and Ang-2) in a group of well-differentiated NENs having different primary sites. They were the first to demonstrate higher levels of Ang-2 in NENs compared to healthy controls (p < 0.001), proportional to tumor burden (p = 0.014, one-way ANOVA test), with a sensitivity close to that of CgA. Despite short follow-up (6 months), Kaplan–Meier curves showed a statistically significant correlation with a shorter time to disease progression (p = 0.03). No particular pattern was noticed in Ang-1 levels, with the caveat that the group was highly heterogenous in terms of tumor site, previous treatment and tumor extension. Ang-2 was also assessed in nine tissue samples; however, the small sample size and intra-tumoral variation in staining hampered the conclusions.
Detjen et al. [17] extended research on clinical and physiological implications of Ang-2 in NENs in a holistic manner, implying circulant levels, tissue immunohistochemical staining and in situ hybridization and preclinical mouse model using orthotopic xenografts. Clinical findings were of particular interest. Prognostic potential for unfavorable outcome in high-serum Ang-2 group (confirmed also in the previous study) was extended to survival (median of 13 months versus undefined in low/medium Ang-2, p = 0.0003). It appears that Ang-2 is more reliable in predicting poor survival than the presence of metastases. Furthermore, expression patterns of Ang-2 mRNA and receptor Tie-2 in a small subset of tissue samples provided insights into the physiology of the Ang-2–Tie-2 axis in NENs, in favor of a de novo autocrine or paracrine secretion of Ang-2. Finally, the experimental mouse model highlighted the stimulating effect of Ang-2 on angiogenesis and metastasis in the subgroup of pNENs. On the contrary, Ang-1 levels did not correlate with Ang-2, nor did they provide significant clinical correlations.
Controversial data are brought by Figueroa-Vega et al. [18], regarding Ang-1, Ang-2 and Tie-2 expression in blood and tissue samples. Higher levels of all three analyzed markers were reported in metastatic GEP-NENs when compared to healthy controls; however, neither was effective in predicting the risk of metastasis, vascular invasion and response to SSAs. Histological expression of these markers equally failed to demonstrate significant relevance, most probably due to the small sample size. The original aspect of this study design is the assessment of Tie-2-expressing monocytes in well-differentiated GEP-NENs, which brings valuable knowledge on the physiology and potential therapeutic relevance of the Ang-Tie-2 axis. Tie-2-expressing monocytes were significantly increased in tumor cells (p < 0.05), with enhanced chemotaxis, demonstrated in vitro in response to Ang-2 but not to Ang-1, nor in the presence of blocking antiTie-2 antibodies.
Melen-Mucha et al. [15] assessed a larger panel of markers in plasma from 36 well-differentiated GEP NENs patients. The particular choice of using plasma samples is not arbitrary, as platelets are known to artifact the results. Very few markers showed statistically significant relevance. Only Tie-2 levels were elevated in NENs compared with healthy controls (p < 0.001), proving to be as effective as CgA as diagnosis markers. Tie-2 did not correlate with the outcome of the disease. Elevated levels of Ang-2 were once again confirmed in metastatic disease (p < 0.05).

3.7. Other Molecules

IL-8 is a pro-inflammatory chemokine that exerts a strong angiogenic effect as a chemotactic and growth factor for endothelial cells [56]. It has been studied in numerous cancers [57,58,59], including adenocarcinomas of the pancreas [60] but data on NENs are scarce. Pavel et al. [13] demonstrated that low baseline levels of IL-8 correlated not only with disease progression (p < 0.008), but also with poor survival (p = 0.009) [13]. Notably, none of the non-survivors at 2 years had baseline levels below the detection limit. One particular strength of the study conducted by Pavel et al. is leveraging the use of ultra-sensitive ELISA to observe higher levels of IL-8 also in stable disease (p < 0.037), which was not obvious otherwise. Moreover, in two clinical trials, low baseline IL-8 predicted a better response to Sunitinib in carcinoids (Jiménez-Fonseca et al.) and pNENs (Zurita et al.). High baseline levels were also prognostic for shorter PFS and OS (Zurita et al.). Moreover, Jiménez-Fonseca et al. observed, during follow-up, increasing levels of IL-8 in patients resistant to Sunitinib, suggesting that it may be a part of the mechanism of resistance to treatment.
Stromal-cell-derived factor 1a (SDF1a) is a ubiquitous cytokine that recruits endothelial progenitor cells during angiogenesis [61]. Zurita et al. assessed SDF-1a in a subgroup of 28 patients and correlated high levels with poor outcome in terms of progression and survival. As such, 22 out of 28 died during follow-up.
Zurita et al. also demonstrated that CD14+ monocyte co-expressing VEGFR-1 or CXCR4 significantly lowered during treatment with Sunitinib (p = 0.04 and p = 0.03, respectively), suggesting that they serve as predictors of response.
bFGF was analyzed in two studies (Pavel et al., Yao et al.) but the results failed to show any clinical correlations.
In Pavel et al.’s study, angiogenin failed to demonstrate any prognostic value, although angiogenin was significantly higher in NENs compared to controls (p < 0.003) [13].
In spite of sustained endeavor toward research, there is no compelling evidence to sustain the use of angiogenic markers in clinical practice for prognostic or predictive purposes. The majority of studies face several limitations that impact the reliability of the results. It is worth reiterating that a small sample size is the major issue in studying such rare diseases. Except for two clinical trials, only one study in our selection exceeded 100 patients. At the same time, the population is usually heterogenous in terms of primary site, staging and grading, and the majority of patients have already benefited from at least one treatment.
Another issue regards the lack of data on G3 NENs for two main reasons. Most studies focused on well-differentiated tumors G1 or G2 for understandable reasons (only two studies included G3). Secondly, since all studies were conducted before the most recent WHO classification (2017), well-differentiated G3 tumors were not part of the picture.

4. Conclusions

Although data are not mature yet, there are still some promising leads in the field of angiogenic markers. Statistically significant correlations with tumor behavior or outcome were reported for markers, such as PlGF, Ang-2 or IL-8, in at least two independent studies, suggesting attractive leads for deeper research. In theory, all therapeutically targetable molecules additionally motivate further effort. For future perspectives, it is possible that a meta-analysis would overcome the limitations of size and heterogeneity. Moreover, since single predictors are insufficient, the next step in the research should include multianalyte biomarkers.

Author Contributions

Conception and design of the article: I.S., S.O.D., A.E.C.; Literature search: I.S., I.M.C., V.M.C.; Interpretation of the relevant literature: I.S., M.M.D., S.O.D., A.E.C.; Article Draft: I.S., I.M.C., V.M.C.; Revision of the article for intellectual content: M.M.D., S.O.D., A.E.C., M.M., V.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Nagtegaal, I.D.; Odze, R.D.; Klimstra, D.; Paradis, V.; Rugge, M.; Schirmacher, P.; Washington, K.; Carneiro, F.; Cree, I. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020, 76, 182–188. [Google Scholar] [CrossRef] [PubMed]
  2. Dasari, A.; Shen, C.; Halperin, D.; Zhao, B.; Zhou, S.; Xu, Y.; Shih, T.; Yao, J.C. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017, 3, 1335–13342. [Google Scholar] [CrossRef] [PubMed]
  3. Hallet, J.; Law, C.H.L.; Cukier, M.; Saskin, R.; Liu, N.; Singh, S. Exploring the rising incidence of neuroendocrine tumors: A population-based analysis of epidemiology, metastatic presentation, and outcomes. Cancer 2015, 121, 589–597. [Google Scholar] [CrossRef] [PubMed]
  4. Neuroendocrine Tumor of the Gastrointestinal Tract: Introduction. Cancer.net. 2021. Available online: https://www.cancer.net/cancer-types/neuroendocrine-tumor-gastrointestinal-tract/introduction (accessed on 2 May 2022).
  5. Rodallec, M.; Vilgrain, V.; Couvelard, A.; Rufat, P.; O’Toole, D.; Barrau, V.; Sauvanet, A.; Ruszniewski, P.; Menu, Y. Endocrine Pancreatic tumours and helical CT: Contrast enhancement is correlated with microvascular density, histoprognostic factors and survival. Pancreatology 2006, 6, 77–85. [Google Scholar] [CrossRef]
  6. Gao, Y.; Gao, H.; Wang, G.; Yin, L.; Xu, W.; Peng, Y.; Wu, J.; Jiang, K.; Miao, Y. A meta-analysis of prognostic factor of pancreatic neuroendocrine neoplasms. Sci. Rep. 2018, 8, 7271. [Google Scholar] [CrossRef] [PubMed]
  7. Ebos, J.M.L.; Lee, C.R.; Christensen, J.G.; Mutsaers, A.J.; Kerbel, R.S. Multiple circulating proangiogenic factors induced by sunitinib malate are tumor-independent and correlate with antitumor efficacy. Proc. Natl. Acad. Sci. USA 2007, 104, 17069–17074. [Google Scholar] [CrossRef]
  8. Beyens, M.; Vandamme, T.; Peeters, M.; van Camp, G.; de Beeck, K.O. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocr. Relat. Cancer 2019, 26, R109–R130. [Google Scholar] [CrossRef] [PubMed]
  9. Jiménez-Fonseca, P.; Martín, M.N.; Carmona-Bayonas, A.; Calvo, A.; Fernández-Mateos, J.; Redrado, M.; Capdevila, J.; Lago, N.M.; Lacasta, A.; Muñarrizet, J.; et al. Biomarkers and polymorphisms in pancreatic neuroendocrine tumors treated with sunitinib. Oncotarget 2018, 9, 36894–36905. [Google Scholar] [CrossRef] [PubMed]
  10. Zurita, A.J.; Khajavi, M.; Wu, H.K.; Tye, L.; Huang, X.; Kulke, M.H.; Lenz, H.; Meropol, N.; Carley, W.; DePrimo, S.; 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, 1199–1205. [Google Scholar] [CrossRef]
  11. Yao, J.C.; Pavel, M.; Lombard-Bohas, C.; van Cutsem, E.; Voi, M.; Brandt, U.; He, W.; Chen, D.; Capdevilla, J.; De Vries, E.; et al. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: Overall survival and circulating biomarkers from the randomized, Phase III RADIANT-3 study. J. Clin. Oncol. 2016, 34, 3906–3913. [Google Scholar] [CrossRef]
  12. Grande, E.; Capdevila, J.; Castellano, D.; Teulé, A.; Durán, I.; Fuster, J.; Sevilla, I.; Escudero, P.; Sastre, J.; Sastre, J.; et al. Pazopanib in pretreated advanced neuroendocrine tumors: A phase II, open-label trial of the Spanish Task Force Group for Neuroendocrine Tumors (GETNE). Ann. Oncol. 2015, 26, 1987–1993. [Google Scholar] [CrossRef]
  13. Pavel, M.E.; Hassler, G.; Baum, U.; Hahn, E.G.; Lohmann, T.; Schuppan, D. Circulating of angiogenic cytokines can predict tumour progression and prognosis in neuroendocrine carcinomas. Clin. Endocrinol. 2005, 62, 434–443. [Google Scholar] [CrossRef] [PubMed]
  14. Srirajaskanthan, R.; Dancey, G.; Hackshaw, A.; Luong, T.; Caplin, M.E.; Meyer, T. Circulating angiopoietin-2 is elevated in patients with neuroendocrine tumours and correlates with disease burden and prognosis. Endocr. Relat. Cancer 2009, 16, 967–976. [Google Scholar] [CrossRef] [PubMed]
  15. Melen-Mucha, G.; Niedziela, A.; Mucha, S.; Motylewska, E.; Lawnicka, H.; Komorowski, J.; Stepien, H. Elevated peripheral blood plasma concentrations of tie-2 and angiopoietin 2 in patients with neuroendocrine tumors. Int. J. Mol. Sci. 2012, 13, 1444–1460. [Google Scholar] [CrossRef] [PubMed]
  16. Cigrovski Berković, M.; Čačev, T.; Catela Ivković, T.; Marout, J.; Ulamec, M.; Zjačić-Rotkvić, V.; Kapitanović, S. High VEGF serum values are associated with locoregional spread of gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Mol. Cell Endocrinol. 2016, 425, 61–68. [Google Scholar] [CrossRef]
  17. Detjen, K.M.; Rieke, S.; Deters, A.; Schulz, P.; Rexin, A.; Vollmer, S.; Hauff, P.; Wiedenmann, B.; Pavel, M.; Scholz, A. Angiopoietin-2 promotes disease progression of neuroendocrine tumors. Clin. Cancer Res. 2010, 16, 420–429. [Google Scholar] [CrossRef]
  18. Figueroa-Vega, N.; Díaz, Á.; Adrados, M.; Álvarez-Escolá, C.; Paniagua, A.; Aragonés, J.; Martín-Pérez, E.; Leskela, S.; Moreno-Otero, R.; González-Amaro, R.; et al. The association of the angiopoietin/Tie-2 system with the development of metastasis and leukocyte migration in neuroendocrine tumors. Endocr. Relat. Cancer 2010, 17, 897–908. [Google Scholar] [CrossRef]
  19. Hilfenhaus, G.; Göhrig, A.; Pape, U.F.; Neumann, T.; Jann, H.; Zdunek, D.; Hess, G.; Stassen, J.M.; Wiedenmann, B.; Detjen, K.; et al. Placental growth factor supports neuroendocrine tumor growth and predicts disease prognosis in patients. Endocr. Relat. Cancer 2013, 20, 305–319. [Google Scholar] [CrossRef]
  20. Oberg, K.; Couvelard, A.; Delle Fave, G.; Gross, D.; Grossman, A.; Jensen, R.T.; Pape, U.F.; Perren, A.; Rindi, G.; Ruszniewski, P.; et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: Biochemical markers. Neuroendocrinology 2017, 105, 201–211. [Google Scholar] [CrossRef]
  21. Yang, X.; Yang, Y.; Li, Z.; Cheng, C.; Yang, T.; Wang, C.; Liu, L.; Liu, S. Diagnostic value of circulating chromogranin A for neuroendocrine tumors: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0124884. [Google Scholar] [CrossRef] [Green Version]
  22. Hofland, J.; Zandee, W.T.; de Herder, W.W. Role of biomarker tests for diagnosis of neuroendocrine tumours. Nat. Rev. Endocrinol. 2018, 14, 656–669. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, Y.H.; Yang, Q.C.; Lin, Y.; Xue, L.; Chen, M.H.; Chen, J. Chromogranin A as a marker for diagnosis, treatment, and survival in patients with gastroenteropancreatic neuroendocrine neoplasm. Medicine 2014, 93, e247. [Google Scholar] [CrossRef] [PubMed]
  24. Yin, X.; Wang, X.; Shen, B.; Jing, Y.; Li, Q.; Cai, M.C.; Gu, Z.; Yang, Q.; Zhang, Z.; Liu, J.; et al. A VEGF-dependent gene signature enriched in mesenchymal ovarian cancer predicts patient prognosis. Sci. Rep. 2016, 6, 31079. [Google Scholar] [CrossRef] [PubMed]
  25. D’Herbomez, M.; Coppin, L.; Bauters, C.; Rouaix-Emery, N.; Carnaille, B.; Do Cao, C. Les marqueurs biologiques des tumeurs endocrines. Ann. Biol. Clin. 2016, 74, 669–679. [Google Scholar] [CrossRef]
  26. Ferolla, P.; Faggiano, A.; Mansueto, G.; Avenia, N.; Cantelmi, M.G.; Giovenali, P.; Del Basso De Caro, M.L.; Milone, F.; Scarpelli, G.; Masone, F.; et al. The biological characterization of neuroendocrine tumors: The role of neuroendocrine markers. J. Endocrinol. Investig. 2008, 31, 277–286. [Google Scholar] [CrossRef]
  27. Nobels, F.R.; Kwekkeboom, D.J.; Coopmans, W.; Schoenmakers, C.H.; Lindemans, J.; De Herder, W.W.; Krenning, E.P.; Bouillon, R.; Lamberts, S.W.J. Chromogranin A as serum marker for neuroendocrine neoplasia: Comparison with neuron-specific enolase and the alpha-subunit of glycoprotein hormones. J. Clin. Endocrinol. Metab. 1997, 82, 2622–2628. [Google Scholar]
  28. Yao, J.C.; Pavel, M.; Phan, A.T.; Kulke, M.H.; Hoosen, S.; St Peter, J.; Cherfi, A.; Öberg, K.E. Chromogranin A and neuron-specific enolase as prognostic markers in patients with advanced pNET treated with everolimus. J. Clin. Endocrinol. Metab. 2011, 96, 3741–3749. [Google Scholar] [CrossRef]
  29. Zhang, P.; Yu, J.; Li, J.; Shen, L.; Li, N.; Zhu, H.; Zhai, S.; Zhang, Y.; Yang, Z.; Lu, M. Clinical and prognostic value of PET/CT imaging with combination of (68)Ga-DOTATATE and (18)F-FDG in gastroenteropancreatic neuroendocrine neoplasms. Contrast Media Mol. Imaging 2018, 26, 2340389. [Google Scholar] [CrossRef]
  30. Drevs, J.; Schneider, V. The use of vascular biomarkers and imaging studies in the early clinical development of anti-tumour agents targeting angiogenesis. J. Intern. Med. 2006, 260, 517–529. [Google Scholar] [CrossRef]
  31. Vijayvergia, N.; Boland, P.M.; Handorf, E.; Gustafson, K.S.; Gong, Y.; Cooper, H.S.; Sheriff, F.; Astsaturov, I.; Cohen, S.J.; Engstrom, P.F. Molecular profiling of neuroendocrine malignancies to identify prognostic and therapeutic markers: A Fox Chase Cancer Center pilot study. Br. J. Cancer 2016, 115, 564–570. [Google Scholar] [CrossRef]
  32. Chakravarty, D.; Gao, J.; Phillips, S.M.; Kundra, R.; Zhang, H.; Wang, J.; Rudolph, J.E.; Yaeger, R.; Soumerai, T.; Nissan, M.H.; et al. OncoKB: A precision oncology knowledge base. JCO Precis. Oncol. 2017, 1, 1–6. [Google Scholar] [CrossRef] [PubMed]
  33. Modlin, I.; Drozdov, I.; Kidd, M. A multitranscript blood neuroendocrine tumor molecular signature to identify treatment efficacy and disease progress. J. Clin. Oncol. 2013, 31, A4137. [Google Scholar] [CrossRef]
  34. Malczewska, A.; Kos-Kudła, B.; Kidd, M.; Drozdov, I.; Bodei, L.; Matar, S.; Oberg, K.; Modlin, I.M. The clinical applications of a multigene liquid biopsy (NETest) in neuroendocrine tumors. Adv. Med. Sci. 2020, 65, 18–29. [Google Scholar] [CrossRef] [PubMed]
  35. Öberg, K.; Califano, A.; Strosberg, J.R.; Ma, S.; Pape, U.; Bodei, L.; Kaltsas, G.; Toumpanakis, C.; Goldenring, J.R.; Frilling, A.; et al. A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Ann. Oncol. 2020, 31, 202–212. [Google Scholar] [CrossRef] [PubMed]
  36. Takayanagi, D.; Cho, H.; Machida, E.; Kawamura, A.; Takashima, A.; Wada, S.; Tsunoda, T.; Kohno, T.; Shiraishi, K. Update on epidemiology, diagnosis, and biomarkers in gastroenteropancreatic neuroendocrine neoplasms. Cancers 2022, 14, 1119. [Google Scholar] [CrossRef]
  37. Khan, M.S.; Kirkwood, A.; Tsigani, T.; Garcia-Hernandez, J.; Hartley, J.A.; Caplin, M.E.; Meyer, T. Circulating tumor cells as prognostic markers in neuroendocrine tumors. J. Clin. Oncol. 2013, 31, 365–372. [Google Scholar] [CrossRef]
  38. Boons, G.; Vandamme, T.; Peeters, M.; Beyens, M.; Driessen, A.; Janssens, K.; Zwaenepoel, K.; Roeyen, G.; Van Camp, G.; De Beeck, K.O. Cell-free DNA from metastatic pancreatic neuroendocrine tumor patients contains tumor-specific mutations and copy number variations. Front. Oncol. 2018, 8, 467. [Google Scholar] [CrossRef]
  39. Roldo, C.; Missiaglia, E.; Hagan, J.P.; Falconi, M.; Capelli, P.; Bersani, S.; Calin, G.A.; Volinia, S.; Liu, C.G.; Scarpa, A.; et al. MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J. Clin. Oncol. 2006, 24, 4677–4684. [Google Scholar] [CrossRef]
  40. Schneider, B.P.; Shen, F.; Miller, K.D. Pharmacogenetic biomarkers for the prediction of response to antiangiogenic treatment. Lancet Oncol. 2012, 13, e427–e436. [Google Scholar] [CrossRef]
  41. Ping, Y.F.; Bian, X.W. Consice review: Contribution of cancer stem cells to neovascularization. Stem Cells 2011, 29, 888–894. [Google Scholar] [CrossRef]
  42. Scoazec, J.Y. Angiogenesis in neuroendocrine tumors: Therapeutic applications. Neuroendocrinology 2013, 97, 45–56. [Google Scholar] [CrossRef] [PubMed]
  43. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef] [PubMed]
  44. Cives, M.; Ghayouri, M.; Morse, B.; Brelsford, M.; Black, M.; Rizzo, A.; Meeker, A.; Strosberg, J. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumors. Endocr. Relat. Cancer 2016, 23, 759–767. [Google Scholar] [CrossRef] [PubMed]
  45. Ren, B.; Bart Rose, J.; Liu, Y.; Jaskular-Sztul, R.; Contreras, C.; Beck, A.; Chen, H. Heterogeneity of vascular endothelial cells, de novo arteriogenesis and therapeutic implications in pancreatic neuroendocrine tumors. J. Clin. Med. 2019, 8, 1980. [Google Scholar] [CrossRef]
  46. Poncet, G.; Villaume, K.; Walter, T.; Pourreyron, C.; Theillaumas, A.; Lépinasse, F.; Hervieu, V.; Cordier-Bussat, M.; Scoazec, J.Y.; Roche, C. Angiogenesis and tumor progression in neuroendocrine digestive tumors. J. Surg. Res. 2009, 154, 68–77. [Google Scholar] [CrossRef]
  47. Ferrara, N. From the discovery of vascular endothelial growth factor to the introduction of avastin in clinical trials—An interview with Napoleone Ferrara by Domenico Ribatti. Int. J. Dev. Biol. 2011, 55, 383–388. [Google Scholar]
  48. Fischer, C.; Mazzone, M.; Jonckx, B.; Carmeliet, P. FLT1 and its ligands VEGFB and PlGF: Drug targets for anti-angiogenic therapy? Nat. Rev. Cancer 2008, 8, 942–956. [Google Scholar] [CrossRef]
  49. Mentlein, R.; Eichler, O.; Forstreuter, F.; Held-Feindt, J. Somatostatin inhibits the production of vascular endothelial growth factor in human glioma cells. Int. J. Cancer 2001, 92, 545–550. [Google Scholar] [CrossRef] [PubMed]
  50. Cascinu, S.; del Ferro, E.; Ligi, M.; Staccioli, M.P.; Giordani, P.; Catalano, V.; Agostinelli, R.; Muretto, P.; Catalano, G. Inhibition of vascular endothelial growth factor by octreotide in colorectal cancer patients. Cancer Investig. 2001, 19, 8–12. [Google Scholar] [CrossRef]
  51. McCarron, S.L.; Edwards, S.; Evans, P.R.; Gibbs, R.; Dearnaley, D.P.; Dowe, A.; Southgate, C.; Easton, D.F.; Eeles, R.A.; Howell, W.M. Influence of cytokine gene polymorphisms on the development of prostate cancer. Cancer Res. 2002, 62, 3369–3372. [Google Scholar]
  52. Yamamori, M.; Sakaeda, T.; Nakamura, T.; Okamura, N.; Tamura, T.; Aoyama, N.; Kamigaki, T.; Ohno, M.; Shirakawa, T.; Gotoh, A.; et al. Association of VEGF genotype with mRNA level in colorectal adenocarcinomas. Biochem. Biophys. Res. Commun. 2004, 325, 144–150. [Google Scholar] [CrossRef] [PubMed]
  53. De Falco, S. The discovery of placenta growth factor and its biological activity. Exp. Mol. Med. 2012, 44, 1–9. [Google Scholar] [CrossRef] [PubMed]
  54. Pozas, J.; Román, M.S.; Alonso-Gordoa, T.; Pozas, M.; Caracuel, L.; Carrato, A.; Molina-Cerrillo, J. Targeting angiogenesis in pancreatic neuroendocrine tumors: Resistance mechanisms. Int. J. Mol. Sci. 2019, 20, 4949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Coxon, A.; Bready, J.; Min, H.; Kaufman, S.; Leal, J.; Yu, D.; Lee, T.A.; Sun, J.R.; Estrada, J.; Bolon, B.; et al. Context-dependent role of angiopoietin-1 inhibition in the suppression of angiogenesis and tumor growth: Implications for AMG 386, an angiopoietin-1/2-neutralizing peptibody. Mol. Cancer Ther. 2010, 9, 2641–2651. [Google Scholar] [CrossRef] [PubMed]
  56. Li, A.; Dubey, S.; Varney, M.L.; Dave, B.J.; Singh, R.K. IL-8 directly enhanced endothelial cell survival, proliferation, and matrix metalloproteinases production and regulated angiogenesis. J. Immunol. 2003, 170, 3369–3376. [Google Scholar] [CrossRef]
  57. Sales, K.J.; Sutherland, J.R.; Jabbour, H.N.; Katz, A.A. Seminal plasma induces angiogenic chemokine expression in cervical cancer cells and regulates vascular function. Biochim. Biophys. Acta 2012, 1823, 1789–1795. [Google Scholar] [CrossRef]
  58. Maynard, J.P.; Ertunc, O.; Kulac, I.; Baena-Del Valle, J.A.; De Marzo, A.M.; Sfanos, K.S. IL8 expression is associated with prostate cancer aggressiveness and androgen receptor loss in primary and metastatic prostate cancer. Mol. Cancer Res. 2020, 18, 153–165. [Google Scholar] [CrossRef]
  59. Filimon, A.; Preda, I.A.; Boloca, A.F.; Negroiu, G. Interleukin-8 in melanoma pathogenesis, prognosis and therapy—An integrated view into other neoplasms and chemokine networks. Cells 2022, 11, 120. [Google Scholar] [CrossRef]
  60. Hussain, F.; Wang, J.; Ahmed, R.; Guest, S.K.; Lam, E.W.F.; Stamp, G.; El-Bahrawy, M. The expression of IL-8 and IL-8 receptors in pancreatic adenocarcinomas and pancreatic neuroendocrine tumours. Cytokine 2010, 49, 134–140. [Google Scholar] [CrossRef]
  61. Zheng, H.; Fu, G.; Dai, T.; Huang, H. Migration of endothelial progenitor cells mediated by stromal cell-derived factor-1α/CXCR4 via PI3K/Akt/eNOS signal transduction pathway. J. Cardiovasc. Pharmacol. 2007, 50, 274–280. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA diagram for systematic review.
Figure 1. PRISMA diagram for systematic review.
Cimb 44 00274 g001
Table 1. Study design.
Table 1. Study design.
Author, Journal, YearCountryStudy TypeEnrollment PeriodFollow-Up
(Months)
Control GroupSample sizeEndpointsBiomarkersSourceTechnique
(for Circulating Markers)
Pavel et al., Clin Endocrinol,
2005 [13]
GermanyretrospectiveJune 1999–July 200224yes38Progression and OS VEGF, IL-8, bFGF, AngiogeninbloodELISA, usELISA
Berković et al., Mol Cell Endocrinol,
2016 [16]
CroatiaprospectiveNANAyes145Correlation with DP; The role of VEGF 1154 SNPs in VEGF expressionVEGF,
VEGF 1154A/G
polymorphism
blood tissueELISA
Srirajaskanthan et al., Endoc Rel Cancer,
2009 [14]
UKprospective July 2007–March 20086 yes47Correlation with DP and PFSAng-1,
Ang-2
blood tissueELISA
Derjen et al., Clin Cancer Res,
2010 [17]
Germanyprospective1998–200559 yes42Correlation with DP and OS; Physiological implications of Ang-2Ang-2blood tissue xenograftELISA
N Figueroa- Vega et al., Endocr Rel Cancer, 2010 [18]SpainretrospectiveNANAyes47Correlation with DP and response to treatment. Physiological implications of Ang 1 and 2 -Tie 2 axisTie-2, VEGF Ang-1, Ang-2,
TEM
blood tissueELISAFACS
Melen-Mucha et al., Int. J. Mol. Sci,
2012 [15]
PolandretrospectiveMay 2008–February 2011 NAyes36Correlation with DPVEGF, Ang-1,
Ang-2, Tie-2,
Endostatin,
osteopontin
bloodELISA
Hilfenhaus et al.,
Endocr Rel Cancer,
2013 [19]
Germanyretrospective &
prospective
Retrospective:
1998–2012 Prospective: May 2009 December 2012
NAyes175Expression, function, prognostic value and potential therapeutic targetPlGFblood tissue
in vitro xenograft
ELISA
Jiménez-Fonseca et al., Oncotarget,
2018 [9]
Spain prospectivemulticenterphase IV
clinical trial with Sunitinib
November 2012—
February 2015
51No43Correlation with OS, PFS, response to treatment, adverse events panel of 14 SNPs,
HGF, IL-6, IL-8,
TIMP1,
sE-selectin,
osteopontin
bloodmultiplex bead assays
Zurita et al., BJC, 2015 [10]USAprospectivemulticenterphase II
clinical trial with Sunitinib
March 2003–November 2005NANo105Correlation with OS, PFS, response to treatment, adverse events VEGF-A,
VEGFR-2,
VEGFR-3,
IL-8, SDF-1α, Circulating myelomonocytic and endothelial cells
bloodELISA FACS
Yao et al., J of Clin Oncol, 2016 “RADIANT-3” [11] (extension phase)Multi- basedprospective, randomized, placebo-controlled, phase III, initially double-blind then open-label clinical trial with EverolimusJuly 2007–
March 2014
NAInitially double blindPlacebo: 203 Everolimus: 207Correlation with OSPlGF, VEGF-A,
VEGFR1, VEGFR2,
bFGF
bloodELISA
Grande et al., Annals of Oncology, 2015 [12] “PAZONET”Spainprospectiveopen-label, phase II
clinical trial with Pazopanib
January 2011–
March 2012
17No44Correlation with OS, PFS, response to treatment, adverse events VEGF-A, VEGFR-2,
CTCs, CECs,
cytochrome P450 3A5,
VEGFR3 SNPs
blood tissueELISA Cell Search
NA = non-available information; PFS = progression free survival; OS = overall survival; DP = disease phenotype; FACS = flow cytometry analysis; ELISA = enzyme-linked immunosorbent assay; usELISA = ultrasensitive ELISA; VEGF = vascular endothelial growth factor; VEGFR = vascular endothelial growth factor receptor; Ang = angiopoietin; TEM = TIE-2 expressing monocytes; PlGF = placental growth factor; HGF = hepatocyte growth factor; TIMP1 = tissue inhibitor of metalloproteinase-1; bFGF = basic fibroblast growth factor; CTSs = circulant tumor cells; CECs = circulant endothelial cells; SNPs = Single nucleotide polymorphisms.
Table 2. Study characteristics.
Table 2. Study characteristics.
AuthorPrimary
site
MetastaticPrevious treatmentConclusions
Pavel et al.,
Clin Endocrinol, 2005 [13]
pNENs: 11
GI-NENs: 13
Other: 14
37
(97%)
pretreated: 22
treatment naïve: 16
↑ VEGF levels correlated with PD ↓ baseline IL-8 correlated with PD and ↓ OS
Berković et al.,
Mol Cell Endocrinol, 2016 [16]
pNENs: 65
GI-NENs: 80
58
(40%)
NA↑ VEGF in GEP-NENs, particularly in lymph node metastases and secretory status.
Srirajaskanthan, et al.,
Endoc Rel Cancer,
2009 [14]
pNENs: 17
GI-NENs: 22
Other: 8
NApretreated: 43
treatment naïve: 4
↑ Ang-2 in NETs proportional to tumor burden and prognostic of poorer outcome
Detjen et al.,
Clin Cancer Res, 2010 [17]
pNENs: 25
GI-NENs: 15
Unknown: 2
28
(66%)
NA↑ Ang-2 in metastatic NETs and prognostic for ↓ OS
Figueroa-Vega et al.,
Endocr Rel Cancer, 2010 [18]
pNENs: 23
GI-NENs: 12
Other: 12
28NA↑ Ang-1, Ang-2 and Tie-2 in GEP NENs, without prognostic relevance;
Ang-2 stimulates TEM recruitment at tumor site
Melen-Mucha et al., Int. J. Mol. Sci, 2012 [15]pNENs: 2
GI-NENs: 12
Other: 22
27
(75%)
pretreated: 14
(with SSA)
treatment naïve: 22
↑Tie-2 in NET↑ Ang-2 in metastatic disease
Hilfenhaus et al.,
Endocr Rel Cancer, 2013 [19]
pNENs: 118
GI-NENs: 57
155pretreated: 85
naïve: 90
↑ PlGF in NET. Correlation with grading, not metastases.
↑ VEGFR1 in metastatic disease. ↑ PlGF predicted ↓ OS in pNETs (not confirmed in multivariate analysis) and shorter time to progression in GI-NETs.
In vivo: significant reduction in tumor volume after PlGF inhibition
Jiménez-Fonseca et al., Oncotarget, 2018 [9]All pNEN42
(97%)
pretreated: 25
treatment naïve: 18
2 VEGFR-3 SNP (rs307826 and rs307821) associated with ↓ OS↓ IL-8 associated to better objective response
Zurita et al., BJC, 2015 [10]pNENs: 66
Carcinoid: 39
NANA↑ VEGFR-2 predicted ↑ OS in pNETs↓ IL8 predicted response to treatment in carcinoid↑ VEGFR-3 and IL-8 correlated with ↓ PFS and ↓ OS in carcinoid↑ SDF-1α predicted ↑ PFS and ↓ OS
Yao et al., J of Clin Oncol, 2016 [11] “RADIANT-3” (extension phase)All pNENNANAPlGF is an independent prognostic factor for PD
Grande et al., Annals of Oncology, 2015 [12] “PAZONET”pNENs: 18
GI-NENs: 15
Other: 11
NAallNo significant correlation with circulant biomarkers was noted;
VEGFR3rs307821 correlated with ↓ PFS in GEP NET
pNENs = pancreatic neuroendocrine neoplasms; GI-NENs = gastro-intestinal neuroendocrine; GEP-NENs = gastro-entero-pancreatic neoplasia; NETs = (well differentiatied) neuroendocrine tumors; ↑ = increased; ↓ = decreased; PD = progressive disease; OS = overall survival; NA = information not available; VEGF = vascular endothelial growth factor; VEGFR = vascular endothelial growth factor receptor; Ang = angiopoietin; TEM = TIE-2 expressing monocytes; SNPs = Single nucleotide polymorphisms; PlGF = placental growth factor; SSA = somatostatin analogues.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sandra, I.; Cazacu, I.M.; Croitoru, V.M.; Mihaila, M.; Herlea, V.; Diculescu, M.M.; Dima, S.O.; Croitoru, A.E. Circulating Angiogenic Markers in Gastroenteropancreatic Neuroendocrine Neoplasms: A Systematic Review. Curr. Issues Mol. Biol. 2022, 44, 4001-4014. https://doi.org/10.3390/cimb44090274

AMA Style

Sandra I, Cazacu IM, Croitoru VM, Mihaila M, Herlea V, Diculescu MM, Dima SO, Croitoru AE. Circulating Angiogenic Markers in Gastroenteropancreatic Neuroendocrine Neoplasms: A Systematic Review. Current Issues in Molecular Biology. 2022; 44(9):4001-4014. https://doi.org/10.3390/cimb44090274

Chicago/Turabian Style

Sandra, Irina, Irina Mihaela Cazacu, Vlad Mihai Croitoru, Mariana Mihaila, Vlad Herlea, Mircea Mihai Diculescu, Simona Olimpia Dima, and Adina Emilia Croitoru. 2022. "Circulating Angiogenic Markers in Gastroenteropancreatic Neuroendocrine Neoplasms: A Systematic Review" Current Issues in Molecular Biology 44, no. 9: 4001-4014. https://doi.org/10.3390/cimb44090274

Article Metrics

Back to TopTop