Next Article in Journal
Understanding Secondary Sarcopenia Development in Young Adults Using Pig Model with Chronic Pancreatitis
Previous Article in Journal
Recombinant Subunit Vaccine Candidate against the Bovine Viral Diarrhea Virus
Previous Article in Special Issue
Neuroblastoma—A Review of Combination Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker?

by
Amparo López-Carrasco
1,2,
Isaac Vieco-Martí
1,2,
Sofía Granados-Aparici
1,2,
Delia Acevedo-León
3,
Nuria Estañ-Capell
3,
Raquel Portugal
4,
Jorge Huerta-Aragonés
5,
Adela Cañete
6,
Samuel Navarro
1,2,7 and
Rosa Noguera
1,2,7,*
1
Incliva Biomedical Health Research Institute, 46010 Valencia, Spain
2
CIBER of Cancer (CIBERONC), 28029 Madrid, Spain
3
University Hospital Doctor Peset, 46017 Valencia, Spain
4
General University Hospital of Burgos, 09006 Burgos, Spain
5
General University Hospital Gregorio Marañón, 28007 Madrid, Spain
6
Politechnic and University Hospital La Fe, 46026 Valencia, Spain
7
Pathology Department, Medical School, University of Valencia, 46010 Valencia, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 8733; https://doi.org/10.3390/ijms25168733 (registering DOI)
Submission received: 9 July 2024 / Revised: 2 August 2024 / Accepted: 9 August 2024 / Published: 10 August 2024

Abstract

:
Vitronectin is a glycoprotein present in plasma and the extracellular matrix that is implicated in cell migration. The high amount of vitronectin found in neuroblastoma biopsies has been associated with poor prognosis. Moreover, increased vitronectin levels have been described in the plasma of patients with different cancers. Our aim was to assess vitronectin as a potential circulating biomarker of neuroblastoma prognosis. Vitronectin concentration was quantified using ELISA in culture media of four neuroblastoma cell lines grown in a monolayer and in 3D models, and in the plasma of 114 neuroblastoma patients. Three of the neuroblastoma cell lines secreted vitronectin to culture media when cultured in a monolayer and 3D models. Vitronectin release was higher by neuroblastoma cells cultured in 3D models than in the monolayer and was still elevated when cells were grown in 3D scaffolds with cross-linked vitronectin. Vitronectin secretion occurred independently of cell numbers in cultures. Its concentration in the plasma of neuroblastoma patients ranged between 52.4 and 870 µg/mL (median, 218 µg/mL). A ROC curve was used to establish a cutoff of 361 µg/mL, above which patients over 18 months old had worse prognosis (p = 0.0018). Vitronectin could be considered a new plasma prognostic biomarker in neuroblastoma and warrants confirmation in collaborative studies. Drugs inhibiting vitronectin interactions with cells and/or the extracellular matrix could represent a significant improvement in survival for neuroblastoma patients.

1. Introduction

Vitronectin (VN), also known as serum spreading factor, is a multifunctional glycoprotein present in plasma and the extracellular matrix (ECM) [1]. It contains multiple cell receptor binding sites including integrins, urokinase-type plasminogen activator receptor (uPAR), and plasminogen activator inhibitor-1 (PAI-1) [2]. VN seems to anchor to ECM fibers and proteoglycans, leading to transitory ECM element–cell junctions, cell adhesion, and migration [3,4,5]. Indeed, coating culture surfaces with VN have been used for many years to promote the adhesion and growth of stem cells in their undifferentiated state and to direct differentiation [6,7]. Besides its role in spreading and metastasis, VN is involved in other key steps of cancer such as apoptosis, inflammation, vascular permeability, and vascular endothelial growth factor-induced angiogenesis [8,9,10]. Its association with tumor aggressiveness has been described in several cancers [8,11,12,13,14,15,16,17], including neuroblastoma (NB) [5].
NB originates from the neural crest in the sympathetic nervous system and is one of the most common pediatric solid tumors [18]. The risk of progression in NB patients is defined by several clinical, biological and genetic features [19], and the current survival rate of high-risk (HR) NB is under 50% [20]. Anchoring molecules, such as VN, could become primary targets for pharmacological strategies to increase this survival rate, as has occurred in other cancers [21]. In previous studies, we detected an elevated amount of VN in the cytoplasm of malignant neuroblasts and, adjacent to them, applied immunohistochemistry (IHC) on NB biopsies, which was related to poor outcome of HR-NB [5]. Moreover, we observed that this VN secreted to ECM form tracks, postulating that VN could participate in tumor cell migration [22]. In subsequent studies, our experimental models (orthotopic xenograft VN knock-out mice, and two 3D models: hydrogels (HGs) of methacrylated gelatin plus increasing concentrations of methacrylated alginate, and HGs of polyethylene glycol with and without cross-linked VN) supported the role of this glycoprotein in NB cell dynamics, aggressiveness, and in the clonal selection of segmental chromosomal aberrations (SCAs) [23,24,25,26].
Besides the role of VN in tumor ECM, its presence in blood plasma and in other types of liquid biopsies (ascites and cerebrospinal fluid) has also been studied in several tumor types. Specifically, elevated concentrations of circulating VN have been observed in adult patients with glioma, melanoma, breast, ovarian, endometrium cancer, and in pediatric patients with Hodgkin lymphoma and acute lymphoblastic leukemia [12,14,15,16,27,28,29,30,31,32,33]. VN in plasma is particularly understudied in pediatric solid tumors, including NB [34]. These tumors represent a great clinical challenge due to their aggressiveness and high mortality, and, in many cases, to the difficulty of obtaining appropriate biopsies, given the young age of some patients and the high frequency of intratumor heterogeneity [35].
The aim of the present study was to explore the value of VN detection in liquid biopsy as a novel diagnostic, prognostic, and therapeutic biomarker. We evaluated VN secretion to culture media by different NB cell lines grown in 2D and 3D HGs, and determined whether VN secretion was reflected in the plasma of HR-NB patients.

2. Results

2.1. Higher VN Release in 3D Models

We studied the VN levels in culture media of four NB cell lines (Figure 1A) grown in 2D until reaching confluence and in HGs for two and three weeks. VN release in control HGs (no cells) with cross-linked VN was null at both culture times. However, all but one cell line secreted VN to culture media when grown in 2D and in 3D models. SK-N-BE(2) and SH-SY5Y cell lines secreted VN in all growth conditions. PDX2 showed VN secretion when cultured in 2D and in HGs after three weeks of culture. We were unable to detect VN in culture media of PDX1 in any of the culture conditions. Aside from the abovementioned negative cases, VN concentration was significantly higher in media of the 3D models (median = 11.1 ng/mL/million cells) than in monolayer (median = 1.5 ng/mL/million cells) cultures (p = 0.0052, Figure 1B).

2.2. VN Secretion Independent of Cell Numbers

Pearson’s r correlation showed that the VN concentration secretion to culture media per million of cells and the estimated number of cells in HGs by digital analysis were independent variants. SK-N-BE(2) presented a marked reduction in VN secretion to the culture media after three weeks of culture (mean = 10.1 ng/mL/million cells) compared to two weeks (mean = 19.9 ng/mL/million cells), despite the greater number of cells estimated in the HGs with longer time. The opposite was observed for the SH-SY5Y (mean = 14.2 ng/mL at 3 weeks vs. 7 ng/mL at 2 weeks) and PDX2 (mean = 12.1 ng/mL at 3 weeks vs. 0 ng/mL at 2 weeks) cell lines, which presented an increase in VN secretion to media after three weeks of culture, unrelated to greater cell growth (Figure 1C). SK-N-BE(2) and SH-SY5Y cell lines showed a larger secretion of VN per million cells when cultured in HGs without added VN (mean = 16.3 ng/mL and 12.1 ng/mL, respectively) compared to HGs with added VN (mean = 13.5 ng/mL and 7.8 ng/mL, respectively) at both two and three weeks of culture, despite the increasing trend in cell growth observed in the HGs with added VN, which was more notable in the HGs with three weeks of culture (Figure 1C).

2.3. Clinical and Genetic Characteristics of NB Patients Included in This Study

Having detected VN in the culture media of the studied aggressive NB cell lines, we next sought to analyze whether the same would occur in the plasma of NB patients. A total of 114 NB patients aged between 0 and 296 months at time of diagnosis (mean = 27.2 ± 35.2) were included in this study, of whom 52 were older than 18 months and 54 showed metastasis at diagnosis. Homogeneous MYCN amplification was detected in twenty and heterogeneous amplification in four NB patients. Typical SCAs (typSCAs) of NB patients were detected in 60 cases, two tumors presented atypical SCAs (atypSCAs) and 33 NB patients showed numeric chromosomal aberrations (NCAs). After 5–204 months of follow-up (mean = 65.1 ± 37.8), 81 patients remained alive (Table 1, Figure S1).

2.4. Discriminatory Power of Plasma VN Levels for NB Patient Outcomes

VN levels in the plasma of the 33 NB patients who died were significantly higher than those obtained in the 81 patients who survived (median = 308 µg/mL vs. 205 µg/mL, respectively, p = 0.0009, Figure S2). In line with the prognostic power of this factor in NB patients, however, age showed a pronounced association with death and is the confounder; only four of the 62 patients younger than 18 months died after the abovementioned follow-up, without significant differences in their plasma VN concentration (Figure 2A,B). Plasma VN levels in NB patients older than 18 months who died (N = 29) were significantly higher than in the ones that survived [N = 23] (median = 324 µg/mL vs. 215 µg/mL, respectively, p = 0.0074, Figure 2A,B). As plasma VN levels had outcome discriminatory power in those patients, a ROC curve was represented to identify the optimal cutoff according to survival (Figure 2C). The ROC curve was significant (p = 0.0026) with AUC = 0.75 (95% CI = 0.61–0.88), and a cutoff of 361 µg/mL showed the best likelihood ratio (10.31). The mean VN concentration previously described in the plasma of healthy controls with similar ages was 367.7 µL/mL [37], so the cutoff is also almost over this value (Figure S3).
Kaplan–Meier curves for overall survival (OS) and event-free survival (EFS) reflected the significantly worse outcome of NB patients older than 18 months with VN plasma levels over the cutoff compared to the patients with VN levels under these values (p = 0.0018 and p = 0.0027, respectively) (Figure 3A,B). Five-year OS was 7% for patients with VN levels over the cutoff and 53% for those under, and five-year EFS were 7% and 45%, respectively.

2.5. Plasma VN Level Association with Stage and Genetic Features of NB

Besides the association of plasma VN levels with outcomes in NB patients older than 18 months, we also studied the potential link of VN concentrations with certain clinical and genetic factors (Table 2). We detected an increase in the amount of patients with metastatic disease at diagnosis and VN levels over the cutoff (p = 0.021), and of NB with SCAs (p = 0.014), specifically with the typical loss of 11q (−11q, p = 0.006).
Cox regression analysis using stepwise forward and backward Wald methods proved that the cutoff VN level of 361 µg/mL had a significant outcome predictive value, independent of other clinical and genetic factors (Table 3).

3. Discussion

The search for new biomarkers that improve and speed up patient diagnosis, better classify them into risk groups, and define new therapeutic strategies is one of the greatest challenges of oncological research in recent years. Circulating biomarkers have the added advantage of being non-invasive compared to biomarkers analyzed on tumor biopsies. Circulating biomarkers currently used in NB diagnosis and prognosis include urine catecholamines, serum lactate dehydrogenase, ferritin, and neuron-specific enolase [38]. Circulating tumor cells (e.g., cell markers such as GD2), specific genetic alterations in cell-free DNA (e.g., MYCN, ALK, C-Circles), mRNAs (e.g., TH, PHOX2B and DCX), and more recently, hypermethylation (e.g., RASSF1A) and extracellular vesicles, also seem of interest for defining prognosis and detecting early relapses and minimal residual disease [38,39,40,41]. These biomarkers are related to the underlying mechanisms influencing NB progression, as an impairment of cell cycle regulation mediated by MYCN and ALK targets [42,43]; telomere maintenance mechanisms, especially alternative telomere elongation, which maintains undefined cell survival and division [41,44]; and neuroblast plasticity cause a partial transition from noradrenergic to migratory and chemoresistant mesenchyme cells [45,46]. Interestingly, these biomarkers are currently sought not only at the level of malignant cells but also in the tumor microenvironment, which along with its important role in tumor progression and metastasis also represents a whole universe of therapeutic targets [40,47,48]. The peculiarities of the tumor microenvironment and especially of the ECM play a key role in clonal selection [23,46,48] and are partly responsible for the different mutation landscape crucial in tumor evolution and resistance to therapies [49]. ECM features, such as a high presence of cross-linked reticuline fibers, low amount of glycosaminoglycans, high quantity of sinusoidal blood and lymphatic vessels and a high expression of territorial VN, have been associated with tumor aggressiveness and poor prognosis [5,50].
In previous studies, we proposed VN as a tissue biomarker based on its high expression by IHC in NB biopsies and several experimental models [5,22,23,24,25,26]. In this work, we went one step further to study whether the NB cells grown in our new 3D models secrete VN to culture media and whether secretion can be detected in the plasma of NB patients.
VN release from NB cells to culture media was higher in 3D models than in monolayer cultures. This reflects the importance of using biomimetic models which reflect not only cell–cell interactions but also cell–scaffold communication. In this sense, although we linked high concentrations of VN to HGs, cells continued to secrete their own VN at considerable levels (although somewhat lower than those of HGs without added VN). A previous study with glioma cells showed an increase in cell migration when cells were cultured with complete human serum, as well as a reduction of motility when the VN of those serums was depleted [32]. Similar effects were described in other publications with several cancer cells, and the authors postulated that VN present in plasma is a key migration-inducing factor and more potent chemoattractant than already known pro-metastatic chemokines or growth factors [33,51]. Moreover, VN glycosylation patterns can impact its ability to interact with ligands and induce advantageous conformational changes, as has been shown in human hepatocellular carcinoma-derived plasmas in comparison to healthy controls, as well as in ovarian cancer ascites [52,53]. Interestingly, no relationship was found between the number of cells and amount of VN secretion in our NB models. Hao et al. described high VN levels in the serum of breast cancer patients compared with healthy controls, and they also pointed to a negative correlation between tumor size and serum VN levels. Their hypothesis was that this could be due to a greater secretion of proteases (such as MMP2) which degrade VN upon tumor progression [54]. Whether this could apply in our 3D models (and to what extent) or whether this reduction in VN secretion could be due to cellular activity focused on other cellular functions, to differences in synthetized VN retention between cells or in scaffolds, or to reduced viability caused by cell overgrowth are also interesting questions for future studies.
High VN levels in plasma have been previously described in glioma, melanoma, hepatic, ovary, endometrium and breast cancer patients associated with poor prognosis [12,14,15,16,29,30,31,32,33]. Children and adolescents with Hodgkin lymphoma who relapsed and pediatric patients with acute lymphoblastic leukemia also showed increased levels in plasma of this glycoprotein [27,28]. Nonetheless, to our best knowledge, only one publication has studied VN plasma levels in pediatric patients with solid tumors, without obtaining significant differences with healthy controls. Only eight plasmas from eight children with NB were included in that publication: one from an initial stage patient and seven from children in advanced stages of illness [34]. The small number of plasmas precluded studying the association of VN levels with tumor aggressiveness and prognosis. Our study includes 114 plasmas from NB patients, a not inconsiderable figure taking into account the small number of annual cases of this rare cancer type. We observed that high plasma VN levels had a prognostic value, independently from clinical and genetic factors of NB applying Cox regression. These findings substantialize plasma VN as a potential circulating prognostic biomarker for NB in patients aged over 18 months. However, mean plasma VN values obtained in NB patients were lower than those reported in healthy controls [37] of the same age (269.5 µg/mL vs. 367.6 µg/mL, respectively; Supplementary Figure S3). As the ELISA kits used for VN detection were different, there may be discrepancies between the values obtained, as has been observed in studies comparing healthy control levels [29,37,55].
High VN expression was observed by our group in ECM of HR-NB biopsies [5] with significant branching in VN patterns, which seemed to form tracks enabling malignant cells to reach blood vessels [22]. VN could intervene in cell adhesion, proteinase secretion and tissue remodeling to achieve a more favorable ECM, leading to tumor progression, migration and survival. In this context, VN has been shown to activate phosphorylation of p42/44 MAPK and AKT in cell lines, and VN serum has been correlated with PI3K and AKT levels [15,33]. Moreover, VN is considered to play an important role in the regulation of endothelial permeability and VEGF signaling, promoting angiogenesis and vascular permeability [9]. Once in the bloodstream and in other fluids, such as lymph, ascites and cerebrospinal fluid, VN could promote circulating tumor cells to disseminate and adhere to distal organs [54,56]. As VN can be produced in the peritoneal cavity, liver, and bones [51,57,58], these could represent suitable premetastatic niches for forming new NB metastatic tumors.

4. Materials and Methods

4.1. Two-Dimensional Cell Cultures

SK-N-BE(2) and SH-SY5Y NB cell lines were acquired from American Type Culture Collection (ATCC, Masassas, VA, USA). Both cell lines were expanded in IMDM medium (Gibco, Life Technologies, Waltham, MA, USA) supplemented with 10% FBS, 1% insulin/transferrin and 1% Penicillin/streptomycin at 37 °C in 5% CO2 atmosphere. Patient-derived xenograft 1 (PDX1) and PDX2 cells were previously established as described in [36]. Both cell lines were expanded in DMEM-high glucose GlutaMAX™ medium, supplemented with 1% B-27™ without Vitamin A (Gibco, Life Technologies, Waltham, MA, USA), 20 ng/mL of EGF and FGF, and 1% Penicillin/streptomycin. Culture medium was replaced every 2–3 days.

4.2. Three-Dimensional Hydrogel Construction

Three-dimensional HGs were based on previous work [59]. Briefly, sf (Sigma Aldrich, Merck, USA) was mixed with previously synthetized and lyophilized GTA (Sigma Aldrich, Merck, St. Louis, MO, USA) in unsupplemented IMDM cell culture medium to obtain 4% w/v solutions. The ratio of sf/GTA was 75:25. VN (PrepoTech, Rocky Hill, NJ, USA) resuspended in dPBS was added to half of the solution, with a final concentration of 0.4 mg/mL. dPBS was added in the remaining solution to construct HGs without VN. Horseradish peroxidase (20 U/mL) was also added to the mix. Prior to gelification, 1.25 × 105 of commercial NB cells, or 2.5 × 105 of PDX cells, previously cultured in 2D, were resuspended in the solution per each HG. To start the polymerization process, 2 µL of hydrogen peroxide (0.01%) was placed in the center of each well in a 24-well plate and mixed by fast and smooth pipetting with aliquots of 60 μL of the mix solution. HGs were incubated at 37 °C for 1 h to complete gelification. Subsequently, 2 mL of growth culture medium was added. Culture media were replaced and collected for ELISA every three days. We kept the 3D models for two and three weeks.

4.3. Patients and Samples

Blood plasmas from 114 NB patients diagnosed between 2003 and 2023 were included in this study. Peripheral blood samples were collected at the time of primary tumor biopsies. All samples were sent to the Spanish Reference Centre for NB Molecular and Pathological studies (Department of Pathology, University of Valencia-INCLIVA) and stored in our biobank (reference B.0000339 29/01/2015). Histopathological data of the NBs were provided by the reference group pathologist. Clinical data were provided by the attending pediatric oncologist when possible, or by the Reference Centre for NB Clinical Studies. These included outcome data of EFS (defined as length of time from diagnosis to any progression, death or to the date of last contact) and OS (defined as length of time from diagnosis until death or last medical check-up in surviving patients). Genetic data were compiled from our internal database NeuPAT [60]. All clinical-biological data are shown in Table 1. This study was approved by the Research Ethics Committee of the Clinic Hospital of Valencia (No. 2020/025, Act: 372, 30 September 2021). Participants or their family members/legal guardians provided written informed consent for the research studies performed in our laboratory.

4.4. Plasma VN Determination

Peripheral blood samples of NB patients were collected in sterile EDTA tubes. Samples were centrifuged at 2000× g for 10 min to separate plasma. Plasma samples were stored at −80 °C until assayed. VN levels were measured using a sandwich colorimetric enzyme-linked immunosorbent assay kit (Human Vitronectin ELISA Kit, Novus Biologicals, Centennial, CO, USA) technique according to the manufacturer’s instructions. Briefly, the human VN ELISA kit (Novus Biologicals, LLC 10771 E Easter Ave Centennial, CO 80112, USA) recognizes both recombinant and natural human VN with an assay sensitivity of 15.19 pg/mL. Samples were loaded and incubated with biotinylated antibody, and HRP reaction, absorbance reading and results calculation steps were performed using a Triturus automated analyzer (Grífols, Barcelona, Spain). To bring VN levels within the detection ranges of the kit, plasma samples of patients were diluted between 1:1000 and 1:5000 before assay. For VN level determination in media of 2D cell cultures, 8 mL of medium from confluent T75 cell culture flasks were centrifugated at 1200 rpm for 5 min to remove cellular debris. Next, media were concentrated 16 times with SpeedVac™ (Thermo Fisher Scientific, Newington, NH, USA). The number of cells of 2D cultures were determined by a TC20 Automated Cell Counter (Bio Rad, Hercules, CA, USA) after mixing with trypan blue for comparison with the VN level secreted to culture media. For 3D cell cultures, media were collected from two HG replicates (4 mL) together, for each composition and culture time. Next, they were centrifugated as before and concentrated eight times with SpeedVac™. With this step, we reduced the variability intrinsic to HG replicates and achieved the minimum VN level for ELISA detection. As culture media was collected with each change (every 3 days), we analyzed VN levels with ELISA at several time points. However, in this study, VN concentration was obtained from culture media of the two HGs together, relative to the total cell count from the same two HGs at the 2-week and 3-week time points (Figure 1C, no error bars applicable). The HGs were formalin-fixed and paraffine-embedded at 2 and 3 weeks, and a 3 µm section was stained with hematoxylin and eosin. Slides were scanned with Ventana iScan (Roche, Basel, Switzerland). The number of cells per section was determined with StarDist extension for QuPath and scaled to the total volume of HGs (28 mm3). The results were multiplied by the dilution or concentration factor, and those from culture media were also scaled to the number of cells (per millions) in the cultures.

4.5. Statistical Analysis

Statistical analysis was carried out using GraphPad Prism 8 (Graphpad Software, Boston, MA, USA) and SPSS 28.0 (SPSS Inc., Chicago, IL, USA). The Kolmogorov–Smirnov test was carried out to investigate data distribution. Non-paired T-tests were applied to compare VN release in 2D and 3D cultures, and Pearson’s r correlation was used to compare VN secretion to culture media and HGs cell count. For patient cohort analysis, Mann–Whitney and Kruskal–Wallis tests were applied to compare survival and plasma VN levels between the age groups, while Spearman’s r correlation tests were performed to study associations between plasma VN levels and patients’ clinical/molecular features. The receiver operating characteristic (ROC) curve was performed to assess discriminatory ability with an estimated area under curve (AUC). A suitable cutoff value, based on the best likelihood ratio, was selected from ROC curves to obtain the optimal sensitivity and specificity. Survival curves were made using the Kaplan–Meier method, and Cox regression analysis was employed to assess OS- and EFS-related predictors (Wald forward and backward stepwise methods). Patient age at diagnosis was the confounding variant (age > 18 months old at diagnosis is associated with poor prognosis [19], and healthy controls of more than 12 months had more VN in plasma than younger controls [37]); therefore, only patients > 18 months old were included in ROC, Kaplan–Meier and Cox analysis.

5. Conclusions

This study and our previous research with patient biopsies and other experimental models all suggest that VN (expressed by tumor cells and detected in plasma) could be closely linked to tumor aggressiveness and poor prognosis in NB patients, appearing to play a role in different steps of cancer progression. Herein, we provide a proof of principle of VN secretion by malignant neuroblasts as a novel predictive circulating biomarker. Further work is needed to refine the estimation of the reference VN. Collaborative studies including more plasma samples from patients and healthy controls must be conducted for VN levels to be considered alongside classical prognosis factors in therapeutic decision making, which would represent a step forward in precision medicine. In combination with classic therapies, drugs such as Cilengitide [61,62,63,64] (which binds to integrins, acting as a competitive inhibitor of their binding to VN and is currently being tested by our group in 3D models) and fibrinogen analogs (which seem to quench VN chemoattraction [33,51]) could bring a significant improvement regarding the survival of patients with NB and other cancers.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25168733/s1.

Author Contributions

A.L.-C., I.V.-M. and S.G.-A. carried out the in vitro experiments and the culture media collection; J.H.-A. and R.P. collected blood samples of some NB patients; A.L.-C. prepared plasma samples; D.A.-L. and N.E.-C. performed the ELISAs; A.C. provided pediatric clinical data; A.L.-C. wrote the original draft of the manuscript; S.N. carried out histopathological analyses, reviewed the paper and suggested draft changes; R.N. elaborated the concept and designed the study, acquired funding, reviewed the paper and suggested draft changes. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by FIS (Institute of Health Carlos III, Madrid/ERDF, PI20/01107) and CIBERONC (CB16/12/00484). FNB (Fundación Neuroblastoma) and Fundación CRIS (2023/188) fund AL-C contract. The funders were not involved in the research process or the preparation and submission of this article.

Institutional Review Board Statement

This study was approved by the Research Ethics Committee of the Clinic Hospital of Valencia (No. 2020/025, Act: 372, 30 September 2021).

Informed Consent Statement

Participants or their family members/legal guardians provided written informed consent for histological and molecular studies performed in our laboratory.

Data Availability Statement

The datasets used and analyzed in the current study are available from the corresponding author upon reasonable request.

Acknowledgments

The authors thank the Spanish Society of Paediatric Haemato-Oncology (SEHOP) and Desireé Ramal from University and Polytechnic Hospital la Fe de Valencia for patient data management. We also thank Ruth Collado, Ruth Panadero, Laura Martínez, Marta Gómez, Marta Valls, and Karina Parra-Haro—who are/were part of our Translational Research Group in Pediatric Solid Tumors of INCLIVA—for their technical support; the oncopediatricians for collaborating with us in obtaining plasma from NB patients; Kathryn Davies for the English correction; and we are especially thankful to the NB patients and legal guardians for participating in this study.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Schvartz, I.; Seger, D.; Shaltiel, S. Vitronectin . Int. J. Biochem. Cell Biol. 1999, 31, 539–544. [Google Scholar] [CrossRef] [PubMed]
  2. Waltz, D.A.; Natkin, L.R.; Fujita, R.M.; Wei, Y.; Chapman, H.A. Plasmin and plasminogen activator inhibitor type 1 promote cellular motility by regulating the interaction between the urokinase receptor and vitronectin. J. Clin. Investig. 1997, 100, 58–67. [Google Scholar] [CrossRef] [PubMed]
  3. Madsen, C.D.; Ferraris, G.M.S.; Andolfo, A.; Cunningham, O.; Sidenius, N. uPAR-induced cell adhesion and migration: Vitronectin provides the key. J. Cell Biol. 2007, 177, 927–939. [Google Scholar] [CrossRef] [PubMed]
  4. Leavesley, D.I.; Kashyap, A.S.; Croll, T.; Sivaramakrishnan, M.; Shokoohmand, A.; Hollier, B.G.; Upton, Z. Vitronectin-Master controller or micromanager? IUBMB Life 2013, 65, 807–818. [Google Scholar] [CrossRef] [PubMed]
  5. Burgos-Panadero, R.; Noguera, I.; Cañete, A.; Navarro, S.; Noguera, R. Vitronectin as a molecular player of the tumor microenvironment in neuroblastoma. BMC Cancer 2019, 19, 479. [Google Scholar] [CrossRef] [PubMed]
  6. Kawase, E.; Nakatsuji, N. Development of substrates for the culture of human pluripotent stem cells. Biomater. Sci. 2023, 11, 2974–2987. [Google Scholar] [CrossRef] [PubMed]
  7. Braam, S.R.; Zeinstra, L.; Litjens, S.; Ward-van Oostwaard, D.; van den Brink, S.; van Laake, L.; Lebrin, F.; Kats, P.; Hochstenbach, R.; Passier, R.; et al. Recombinant Vitronectin Is a Functionally Defined Substrate That Supports Human Embryonic Stem Cell Self-Renewal via αVβ5 Integrin. Stem Cells 2008, 26, 2257–2265. [Google Scholar] [CrossRef] [PubMed]
  8. Uhm, J.H.; Dooley, N.P.; Kyritsis, A.P.; Rao, J.S.; Gladson, C.L. Vitronectin, a Glioma-derived Extracellular Matrix Protein, Protects Tumor Cells from Apoptotic Death 1. Clin. Cancer Res. 1999, 5, 1587–1594. [Google Scholar] [PubMed]
  9. Li, R.; Ren, M.; Chen, N.; Luo, M.; Zhang, Z.; Wu, J. Vitronectin Increases Vascular Permeability by Promoting VE-Cadherin Internalization at Cell Junctions. PLoS ONE 2012, 7, e37195. [Google Scholar] [CrossRef]
  10. Reilly, J.T.; Nash, G. Vitronectin (serum spreading factor): Its localisation in normal and fibrotic tissue. J. Clin. Pathol. 1988, 41, 1269–1272. [Google Scholar] [CrossRef]
  11. Heyman, L.; Leroy-Dudal, J.; Fernandes, J.; Seyer, D.; Dutoit, S.; Carreiras, F. Mesothelial vitronectin stimulates migration of ovarian cancer cells. Cell Biol. Int. 2010, 34, 493–502. [Google Scholar] [CrossRef] [PubMed]
  12. Radwan, A.F.; Ismael, O.E.; Fawzy, A.; El-Mesallamy, H.O. Evaluation of Serum Integrin αvβ3 & Vitronectin in the Early Diagnosis of Breast Cancer. Clin. Lab. 2019, 65, 1185–1193. [Google Scholar] [CrossRef]
  13. Edwards, S.; Lalor, P.F.; Tuncer, C.; Adams, D.H. Vitronectin in human hepatic tumours contributes to the recruitment of lymphocytes in an avb3-independent manner. Br. J. Cancer 2006, 95, 1545–1554. [Google Scholar] [CrossRef] [PubMed]
  14. Ciereszko, A.; Dietrich, M.A.; Słowińska, M.; Nynca, J.; Ciborowski, M.; Kisluk, J.; Michalska-Falkowska, A.; Reszec, J.; Sierko, E.; Nikliński, J. Identification of protein changes in the blood plasma of lung cancer patients subjected to chemotherapy using a 2D-DIGE approach. PLoS ONE 2019, 14, e0223840. [Google Scholar] [CrossRef] [PubMed]
  15. Bera, A.; Subramanian, M.; Karaian, J.; Eklund, M.; Radhakrishnan, S.; Gana, N.; Rothwell, S.; Pollard, H.; Hu, H.; Shriver, C.D.; et al. Functional role of vitronectin in breast cancer. PLoS ONE 2020, 15, e0242141. [Google Scholar] [CrossRef] [PubMed]
  16. Tas, F.; Karabulut, S.; Bilgin, E.; Tastekin, D.; Duranyildiz, D. Clinical significance of serum fibronectin and vitronectin levels in melanoma patients. Melanoma Res. 2014, 24, 475–479. [Google Scholar] [CrossRef] [PubMed]
  17. Stahl, A.; Mueller, B.M. Melanoma cell migration on vitronectin: Regulation by components of the plasminogen activation system. Int. J. Cancer 1997, 71, 116–122. [Google Scholar] [CrossRef]
  18. Cheung, N.-K.V.; Dyer, M.A. Neuroblastoma: Developmental biology, cancer genomics and immunotherapy. Nat. Rev. Cancer 2013, 13, 397–411. [Google Scholar] [CrossRef] [PubMed]
  19. Cohn, S.L.; Pearson, A.D.J.; London, W.B.; Monclair, T.; Ambros, P.F.; Brodeur, G.M.; Faldum, A.; Hero, B.; Iehara, T.; Machin, D.; et al. The International Neuroblastoma Risk Group (INRG) classification system: An INRG task force report. J. Clin. Oncol. 2009, 27, 289–297. [Google Scholar] [CrossRef]
  20. Von Stedingk, K.; De Preter, K.; Vandesompele, J.; Noguera, R.; Øra, I.; Koster, J.; Versteeg, R.; Påhlman, S.; Lindgren, D.; Axelson, H. Individual patient risk stratification of high-risk neuroblastomas using a two-gene score suited for clinical use. Int. J. Cancer 2015, 137, 868–877. [Google Scholar] [CrossRef]
  21. R Schmidmaier, P.B. ANTI-ADHESION evolves to a promising therapeutic concept in oncology. Curr. Med. Chem. 2008, 15, 978–990. [Google Scholar] [CrossRef] [PubMed]
  22. Vicente-Munuera, P.; Burgos-Panadero, R.; Noguera, I.; Navarro, S.; Noguera, R.; Escudero, L.M. The topology of vitronectin: A complementary feature for neuroblastoma risk classification based on computer-aided detection. Int. J. Cancer 2020, 146, 553–565. [Google Scholar] [CrossRef] [PubMed]
  23. López-Carrasco, A.; Martín-Vañó, S.; Burgos-Panadero, R.; Monferrer, E.; Berbegall, A.P.; Fernández-Blanco, B.; Navarro, S.; Noguera, R. Impact of extracellular matrix stiffness on genomic heterogeneity in MYCN-amplified neuroblastoma cell line. J. Exp. Clin. Cancer Res. 2020, 39, 226. [Google Scholar] [CrossRef] [PubMed]
  24. Monferrer, E.; Sanegre, S.; Martín-Vañó, S.; García-Lizarribar, A.; Burgos-Panadero, R.; López-Carrasco, A.; Navarro, S.; Samitier, J.; Noguera, R. Digital image analysis applied to tumor cell proliferation, aggressiveness, and migration-related protein synthesis in neuroblastoma 3d models. Int. J. Mol. Sci. 2020, 21, 8676. [Google Scholar] [CrossRef] [PubMed]
  25. Monferrer, E.; Martín-Vañó, S.; Carretero, A.; García-Lizarribar, A.; Burgos-Panadero, R.; Navarro, S.; Samitier, J.; Noguera, R. A three-dimensional bioprinted model to evaluate the effect of stiffness on neuroblastoma cell cluster dynamics and behavior. Sci. Rep. 2020, 10, 6370. [Google Scholar] [CrossRef] [PubMed]
  26. Monferrer, E.; Dobre, O.; Trujillo, S.; González Oliva, M.A.; Trubert-Paneli, A.; Acevedo-León, D.; Noguera, R.; Salmeron-Sanchez, M. Vitronectin-based hydrogels recapitulate neuroblastoma growth conditions. Front. Cell Dev. Biol. 2022, 10, 988699. [Google Scholar] [CrossRef] [PubMed]
  27. Repetto, O.; Caggiari, L.; De Zorzi, M.; Elia, C.; Mussolin, L.; Buffardi, S.; Pillon, M.; Muggeo, P.; Casini, T.; Steffan, A.; et al. Quantitative Plasma Proteomics to Identify Candidate Biomarkers of Relapse in Pediatric/Adolescent Hodgkin Lymphoma. Int. J. Mol. Sci. 2022, 23, 9911. [Google Scholar] [CrossRef]
  28. Braoudaki, M.; Lambrou, G.I.; Vougas, K.; Karamolegou, K.; Tsangaris, G.T.; Tzortzatou-Stathopoulou, F. Protein biomarkers distinguish between high- and low-risk pediatric acute lymphoblastic leukemia in a tissue specific manner. J. Hematol. Oncol. 2013, 6, 52. [Google Scholar] [CrossRef] [PubMed]
  29. Kadowaki, M.; Sangai, T.; Nagashima, T.; Sakakibara, M.; Yoshitomi, H.; Takano, S.; Sogawa, K.; Umemura, H.; Fushimi, K.; Nakatani, Y.; et al. Identification of vitronectin as a novel serum marker for early breast cancer detection using a new proteomic approach. J. Cancer Res. Clin. Oncol. 2011, 137, 1105–1115. [Google Scholar] [CrossRef]
  30. Turan, T.; Torun, M.; Atalay, F.; Gönenç, A.; Turk, J. Endometrial ve Ovaryum Kanserinde Tanısal Değer Yönünden Serum Biyobelirteci Olarak Vitronektin, Solubl Epitel-Kaderin ve TGF-β1’in Değerlendirilmesi Assessment of Vitronectin, Soluble Epithelial-Cadherin and TGF-β1 as a Serum Biomarker with Predictive Value for Endometrial and Ovarian Cancers. Pharm. Sci. 2017, 14, 141–147. [Google Scholar] [CrossRef]
  31. Ortega-Martínez, I.; Gardeazabal, J.; Erramuzpe, A.; Sanchez-Diez, A.; Cortés, J.; García-Vázquez, M.D.; Pérez-Yarza, G.; Izu, R.; Luís Díaz-Ramón, J.; de la Fuente, I.M.; et al. Vitronectin and dermcidin serum levels predict the metastatic progression of AJCC I–II early-stage melanoma. Int. J. Cancer 2016, 139, 1598–1607. [Google Scholar] [CrossRef] [PubMed]
  32. Fukushima, Y.; Tamura, M.; Nakagawa, H.; Itoh, K. Induction of glioma cell migration by vitronectin in human serum and cerebrospinal fluid. J. Neurosurg. 2007, 107, 578–585. [Google Scholar] [CrossRef] [PubMed]
  33. Schneider, G.; Suszynska, M.; Kakar, S.; Ratajczak, M.Z. Vitronectin in the ascites of human ovarian carcinoma acts as a potent chemoattractant for ovarian carcinoma: Implication for metastasis by cancer stem cells. J. Cancer Stem Cell Res. 2016, 4, e1005. [Google Scholar] [CrossRef] [PubMed]
  34. Tugcu, D.; Devecioglu, O.; Unuvar, A.; Ekmekci, H.; Ekmekci, O.B.; Anak, S.; Ozturk, G.; Akcay, A.; Aydogan, G. Plasma Levels of Plasminogen Activator Inhibitor Type 1 and Vitronectin in Children with Cancer. Clin. Appl. Thromb. Hemost. 2016, 22, 28–33. [Google Scholar] [CrossRef] [PubMed]
  35. López-Carrasco, A.; Berbegall, A.P.; Martín-Vañó, S.; Blanquer-Maceiras, M.; Castel, V.; Navarro, S.; Noguera, R. Intra-Tumour Genetic Heterogeneity and Prognosis in High-Risk Neuroblastoma. Cancers 2021, 13, 5173. [Google Scholar] [CrossRef] [PubMed]
  36. Braekeveldt, N.; Wigerup, C.; Gisselsson, D.; Mohlin, S.; Merselius, M.; Beckman, S.; Jonson, T.; Börjesson, A.; Backman, T.; Tadeo, I.; et al. Neuroblastoma patient-derived orthotopic xenografts retain metastatic patterns and geno-and phenotypes of patient tumours. Int. J. Cancer 2015, 136, E252–E261. [Google Scholar] [CrossRef]
  37. Newall, F.; Johnston, L.; Ignjatovic, V.; Summerhayes, R.; Monagle, P. Age-related plasma reference ranges for two heparin-binding proteins—vitronectin and platelet factor 4. Int. J. Lab. Hematol. 2009, 31, 683–687. [Google Scholar] [CrossRef] [PubMed]
  38. Trigg, R.M.; Shaw, J.A.; Turner, S.D. Opportunities and challenges of circulating biomarkers in neuroblastoma. Open Biol. 2019, 9, 190056. [Google Scholar] [CrossRef]
  39. van Zogchel, L.M.; van Wezel, E.M.; van Wijk, J.; Stutterheim, J.; Bruins, W.S.; Zappeij-Kannegieter, L.; Slager, T.J.; Schumacher-Kuckelkorn, R.; Verly, I.R.; van der Schoot, C.E.; et al. Hypermethylated RASSF1A as Circulating Tumor DNA Marker for Disease Monitoring in Neuroblastoma. JCO Precis. Oncol. 2020, 4, 291–306. [Google Scholar] [CrossRef]
  40. Filipe, E.C.; Chitty, J.L.; Cox, T.R. Charting the unexplored extracellular matrix in cancer. Int. J. Exp. Pathol. 2018, 99, 58–76. [Google Scholar] [CrossRef]
  41. Chen, Y.Y.; Dagg, R.; Zhang, Y.; Lee, J.H.Y.; Lu, R.; La Rotta, N.M.; Sampl, S.; Korkut-Demirbaş, M.; Holzmann, K.; Lau, L.M.S.; et al. The c-circle biomarker is secreted by alternative-lengthening-of-telomeres positive cancer cells inside exosomes and provides a blood-based diagnostic for alt activity. Cancers 2021, 13, 5369. [Google Scholar] [CrossRef] [PubMed]
  42. Nian, Z.; Wang, D.; Wang, H.; Liu, W.; Ma, Z.; Yan, J.; Cao, Y.; Li, J.; Zhao, Q.; Liu, Z. Single-cell RNA-seq reveals the transcriptional program underlying tumor progression and metastasis in neuroblastoma. Front. Med. 2024. [Google Scholar] [CrossRef]
  43. Li, H.L.; Dong, L.L.; Jin, M.J.; Li, Q.Y.; Wang, X.; Jia, M.Q.; Song, J.; Zhang, S.Y.; Yuan, S. A Review of the Regulatory Mechanisms of N-Myc on Cell Cycle. Molecules 2023, 28, 1141. [Google Scholar] [CrossRef]
  44. Akter, J.; Kamijo, T. How do telomere abnormalities regulate the biology of neuroblastoma? Biomolecules 2021, 11, 1112. [Google Scholar] [CrossRef] [PubMed]
  45. Gautier, M.; Thirant, C.; Delattre, O.; Janoueix-Lerosey, I. Plasticity in neuroblastoma cell identity defines a noradrenergic-to-mesenchymal transition (Nmt). Cancers 2021, 13, 2904. [Google Scholar] [CrossRef] [PubMed]
  46. Unraveled, N.; Lazic, D.; Kromp, F.; Rifatbegovic, F.; Repiscak, P.; Kirr, M.; Mivalt, F.; Halbritter, F.; Bernkopf, M.; Bileck, A.; et al. Landscape of Bone Marrow Metastasis in Human Multiplex Imaging. Cancers 2021, 13, 4311. [Google Scholar] [CrossRef]
  47. Hahn, W.C.; Bader, J.S.; Braun, T.P.; Califano, A.; Clemons, P.A.; Druker, B.J.; Ewald, A.J.; Fu, H.; Jagu, S.; Kemp, C.J.; et al. An expanded universe of cancer targets. Cell 2021, 184, 1142–1155. [Google Scholar] [CrossRef] [PubMed]
  48. Neophytou, C.M.; Panagi, M.; Stylianopoulos, T.; Papageorgis, P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers 2021, 13, 2053. [Google Scholar] [CrossRef]
  49. Schulte, M.; Köster, J.; Rahmann, S.; Schramm, A. Cancer evolution, mutations, and clonal selection in relapse neuroblastoma. Cell Tissue Res. 2018, 372, 263–268. [Google Scholar] [CrossRef]
  50. Tadeo, I.; Berbegall, A.P.; Castel, V.; García-Miguel, P.; Callaghan, R.; Påhlman, S.; Navarro, S.; Noguera, R. Extracellular matrix composition defines an ultra-high-risk group of neuroblastoma within the high-risk patient cohort. Br. J. Cancer 2016, 115, 480–489. [Google Scholar] [CrossRef]
  51. Schneider, G.; Bryndza, E.; Poniewierska-Baran, A.; Serwin, K.; Suszynska, M.; Sellers, Z.P.; Merchant, M.L.; Kaliappan, A.; Ratajczak, J.; Kucia, M.; et al. Evidence that vitronectin is a potent migration-enhancing factor for cancer cells chaperoned by fibrinogen: A novel view of the metastasis of cancer cells to low-fibrinogen lymphatics and body cavities. Oncotarget 2016, 7, 69829–69843. [Google Scholar] [CrossRef] [PubMed]
  52. Lee, H.-J.; Cha, H.-J.; Lim, J.-S.; Lee, S.H.; Song, S.Y.; Kim, H.; Hancock, W.S.; Yoo, J.S.; Paik, Y.-K. Abundance-Ratio-Based Semiquantitative Analysis of Site-Specific N-Linked Glycopeptides Present in the Plasma of Hepatocellular Carcinoma Patients. J. Proteome Res. 2014, 13, 2328–2338. [Google Scholar] [CrossRef] [PubMed]
  53. Benachour, H.; Leroy-Dudal, J.; Agniel, R.; Wilson, J.; Briand, M.; Carreiras, F.; Gallet, O. Vitronectin (Vn) glycosylation patterned by lectin affinity assays-A potent glycoproteomic tool to discriminate plasma Vn from cancer ascites Vn. J. Mol. Recognit. 2018, 31, e2690. [Google Scholar] [CrossRef] [PubMed]
  54. Hao, W.; Zhang, X.; Xiu, B.; Yang, X.; Hu, S.; Liu, Z.; Duan, C.; Jin, S.; Ying, X.; Zhao, Y.; et al. Vitronectin: A promising breast cancer serum biomarker for early diagnosis of breast cancer in patients. Tumor Biol. 2016, 37, 8909–8916. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, M.H.; Lu, C.; Sun, J.; Chen, X.D.; Dai, J.X.; Cai, J.Y.; Chen, X.L. Diagnostic and prognostic value of serum vitronectin levels in human glioma. J. Neurol. Sci. 2016, 371, 54–59. [Google Scholar] [CrossRef] [PubMed]
  56. Gladson, C.L.; Cheresht, D.A. Glioblastoma Expression of Vitronectin and the av/83 Integrin. Adhesion Mechanism for Transformed Glial Cells. J. Clin. Investig. 1991, 88, 1924–1932. [Google Scholar] [CrossRef]
  57. Mikuła-Pietrasik, J.; Uruski, P.; Tykarski, A.; Książek, K. The peritoneal “soil” for a cancerous “seed”: A comprehensive review of the pathogenesis of intraperitoneal cancer metastases. Cell. Mol. Life Sci. 2018, 75, 509–525. [Google Scholar] [CrossRef]
  58. Kobayashi, J.; Yamada, S.; Kawasaki, H. Distribution of vitronectin in plasma and liver tissue: Relationship to chronic liver disease. Hepatology 1994, 20, 1412–1417. [Google Scholar] [CrossRef]
  59. Hasturk, O.; Jordan, K.E.; Choi, J.; Kaplan, D.L. Enzymatically crosslinked silk and silk-gelatin hydrogels with tunable gelation kinetics, mechanical properties and bioactivity for cell culture and encapsulation. Biomaterials 2019, 232, 119720. [Google Scholar] [CrossRef] [PubMed]
  60. Villamón, E.; Piqueras, M.; Meseguer, J.; Blanquer, I.; Berbegall, A.P.; Tadeo, I.; Hernández, V.; Navarro, S.; Noguera, R. NeuPAT: An intranet database supporting translational research in neuroblastic tumors. Comput. Biol. Med. 2013, 43, 219–228. [Google Scholar] [CrossRef]
  61. Stoks, M.; Vieco-Martí, I.; Noguera, I.; Sánchez-Sánchez, M.; Burgos-Panadero, R.; Navarro, S.; Noguera, R. Digital image analysis workflows for evaluation of cell behavior and tumor microenvironment to aid therapeutic assessment in high-risk neuroblastoma. Comput. Biol. Med. 2023, 164, 107364. [Google Scholar] [CrossRef] [PubMed]
  62. Burgos-Panadero, R.; El Moukhtari, S.H.; Noguera, I.; Rodríguez-Nogales, C.; Martín-Vañó, S.; Vicente-Munuera, P.; Cañete, A.; Navarro, S.; Blanco-Prieto, M.J.; Noguera, R. Unraveling the extracellular matrix-tumor cell interactions to aid better targeted therapies for neuroblastoma. Int. J. Pharm. 2021, 608, 121058. [Google Scholar] [CrossRef]
  63. Kurozumi, K.; Ichikawa, T.; Onishi, M.; Fujii, K.; Date, I. Cilengitide treatment for malignant glioma: Current status and future direction. Neurol. Med. Chir. 2012, 52, 539–547. [Google Scholar] [CrossRef] [PubMed]
  64. Reardon, D.A.; Nabors, L.B.; Stupp, R.; Mikkelsen, T. Cilengitide: An integrin-targeting arginine-glycine-aspartic acid peptide with promising activity for glioblastoma multiforme. Expert Opin. Investig. Drugs 2008, 17, 1225–1235. [Google Scholar] [CrossRef] [PubMed]
Figure 1. VN secretion by NB cells to culture media. (A) Origin and main clinical-molecular characteristics of the four NB cell lines. All presented aggressive features as derived from stage 4 patients, being MYCN-amplified or ALK-mutated and having SCAs. * For more details of PDX origin, see ref. [36]. (B) Comparison of VN levels secreted to culture media of 2D (monolayer) and 3D HG cultures by NB cell lines in which VN detection was positive by ELISA (p-value = 0.0052). (C) Concentration of VN secreted by the four NB cell lines to culture media of the 3D HGs in ng/mL per million cells (left Y-axis scale); blue and orange bars represent HGs without (NO-VN) and with cross-linked VN (VN), respectively; light colors refer to 2 weeks (2W) of culture and dark colors to 3W; number of cells calculated with digital analysis in two HGs from which we collected the culture media, measured in millions of cells, as shown on the right Y-axis scale (dots inside the bars).
Figure 1. VN secretion by NB cells to culture media. (A) Origin and main clinical-molecular characteristics of the four NB cell lines. All presented aggressive features as derived from stage 4 patients, being MYCN-amplified or ALK-mutated and having SCAs. * For more details of PDX origin, see ref. [36]. (B) Comparison of VN levels secreted to culture media of 2D (monolayer) and 3D HG cultures by NB cell lines in which VN detection was positive by ELISA (p-value = 0.0052). (C) Concentration of VN secreted by the four NB cell lines to culture media of the 3D HGs in ng/mL per million cells (left Y-axis scale); blue and orange bars represent HGs without (NO-VN) and with cross-linked VN (VN), respectively; light colors refer to 2 weeks (2W) of culture and dark colors to 3W; number of cells calculated with digital analysis in two HGs from which we collected the culture media, measured in millions of cells, as shown on the right Y-axis scale (dots inside the bars).
Ijms 25 08733 g001
Figure 2. VN levels in plasma of NB patients. (A) Descriptive statistics of patient numbers and VN levels detected by ELISA in the plasma of all patients younger and older than 18 months of the patients who remained alive and of those who died. Median VN values are highlighted in bold format and the red font points out the lower levels of VN in alive than in dead patients older than 18 months. (B) Graphic representation of VN levels in the mentioned groups. VN concentration was significantly different between alive and dead patients older than 18 months (p-value = 0.0074). (C) The ROC curve obtained for patients older than 18 months was significant (p-value = 0.0026) and allowed us to establish a cutoff of 361 µL/mL (red arrow), over which patients showed a poor prognosis.
Figure 2. VN levels in plasma of NB patients. (A) Descriptive statistics of patient numbers and VN levels detected by ELISA in the plasma of all patients younger and older than 18 months of the patients who remained alive and of those who died. Median VN values are highlighted in bold format and the red font points out the lower levels of VN in alive than in dead patients older than 18 months. (B) Graphic representation of VN levels in the mentioned groups. VN concentration was significantly different between alive and dead patients older than 18 months (p-value = 0.0074). (C) The ROC curve obtained for patients older than 18 months was significant (p-value = 0.0026) and allowed us to establish a cutoff of 361 µL/mL (red arrow), over which patients showed a poor prognosis.
Ijms 25 08733 g002
Figure 3. Survival probabilities of NB patients older than 18 months according to VN levels. Kaplan–Meier curves for (A) overall survival (OS) and (B) event-free survival (EFS) reflected the significantly lower survival of NB patients with VN plasma levels over the cutoff (p = 0.0018 and p = 0.0027, respectively). Five-year OS for patients with VN levels under and over the cutoff was 53% and 7%, respectively, and EFS was 45% and 7%, respectively.
Figure 3. Survival probabilities of NB patients older than 18 months according to VN levels. Kaplan–Meier curves for (A) overall survival (OS) and (B) event-free survival (EFS) reflected the significantly lower survival of NB patients with VN plasma levels over the cutoff (p = 0.0018 and p = 0.0027, respectively). Five-year OS for patients with VN levels under and over the cutoff was 53% and 7%, respectively, and EFS was 45% and 7%, respectively.
Ijms 25 08733 g003
Table 1. Patient cohort characteristics and tumor features, grouped by relationship with VN plasma level.
Table 1. Patient cohort characteristics and tumor features, grouped by relationship with VN plasma level.
Total CohortVN > 361 µg/mLVN < 361 µg/mL
Mean ± SDN%Mean ± SDN%Mean ± SDN%
Total 114100% 2421% 9079%
OSDead65.1 ± 37.83329%53.4 ± 31.01458%68.3 ± 38.91921%
Alive8171%1042%7179%
EFSEvent52.7 ± 39.74640%46.0 ± 53.41458%54.5 ± 40.63236%
Non-event6860%1042%5864%
Age>18 months27.2 ± 35.25246%39.4 ± 57.71427%23.9 ± 25.23873%
≤18 months6254%1016%5284%
StageStage 4 5447% 1563% 3943%
Non-stage 4 5851% 938% 5157%
Unknown 22% 28% 00%
MYCNStatusAmplified 2018% 625% 1416%
Het-amplified 44% 00% 44%
Non-amplified 9079% 1875% 7280%
Chromosomic profileTypical SCA 6053% 1771% 4348%
Atypical SCA 22% 14% 11%
NCA 3329% 313% 3033%
Unknown 1917% 313% 1618%
−1pPresent 2925% 833% 2123%
Absent 6658% 1354% 5359%
Unknown 1917% 313% 1618%
+1qPresent 98% 313% 67%
Absent 8675% 1875% 6876%
Unknown 1917% 313% 1618%
+2pPresent 2320% 729% 1618%
Absent 7263% 1458% 5864%
Unknown 1917% 313% 1618%
−3pPresent 1311% 521% 89%
Absent 8272% 1667% 6673%
Unknown 1917% 313% 1618%
−4pPresent 109% 28% 89%
Absent 8575% 1979% 6673%
Unknown 1917% 313% 1618%
−11qPresent 2925% 1250% 1719%
Absent 6658% 938% 5763%
Unknown 1917% 313% 1618%
−17qPresent 4943% 1458% 3539%
Absent 4640% 729% 3943%
Unknown 1917% 313% 1618%
OS, overall survival; EFS, event-free survival; Het-amplified, heterogeneously amplified; SCA, segmental chromosomal alterations; NCA, numerical chromosomal alteration. OS, EFS, and age means and standard deviations (SDs) are expressed in months.
Table 2. VN level by cutoff is associated with death, stage, genomic profile, and 11q loss in NB patients older than 18 months.
Table 2. VN level by cutoff is associated with death, stage, genomic profile, and 11q loss in NB patients older than 18 months.
Correlation of Clinic-Molecular Features of NB Patients Older than 18 Months and VN Levels
Plasma VN LevelsSpearman r (95% CI)p-Value (Two-Tailed)
>361 µg/mL<361 µg/mL
Death (yes/no)13/116/220.363 (0.186–0.517)0.000 *
Relapse (yes/no)13/118/200.179 (−0.010–0.356)0.1572
Unknown/metastatic/localized1/10/323/150.217 (0.003–0.392)0.021 *
MYCN status (MNA/MNNA)4/1011/270.115 (−0.077–0.298)0.226
Genomic profile (unknow/SCAs/NCAs)2/12/06/29/30.248 (0.046–0.430)0.014 *
−1p (unknown/present/absent)2/6/67/17/140.157 (−0.050–0.351)0.125
+1q (unknown/present/absent)2/2/108/6/24−0,005 (−0.211–0.201)0.959
+2p (unknown/present/absent)2/5/77/11/200.138 (−0.073–0.330)0.192
−3p (unknown/present/absent)2/4/88/7/230.126 (−0.083–0.324)0.222
−4p (unknown/present/absent)2/2/108/7/23−0.024 (−0.229 to 0.183)0.819
−11q (unknown/present/absent)2/9/37/16/150.277 (0.076–0.456)0.006 *
+17q (unknown/present/absent)2/9/38/22/80.176 (−0.032–0.369)0.087
MYCN status: MNNA, MYCN non-amplified; MYCN heterogeneously amplified; MNA, MYCN amplified; NCA, numerical chromosomal alteration; SCA, segmental chromosomal alterations. Significant p-values (<0.05) are highlighted in bold format and with asterisks. The p-value that indicates the significant association of VN level and death is highlighted in red.
Table 3. Cox regression tests results after applying stepwise forward and backward Wald methods: last step.
Table 3. Cox regression tests results after applying stepwise forward and backward Wald methods: last step.
VariableBSEWaldExp (B)p-Value
OS Wald (forward stepwise) method
VN cutoff1.3290.4478.8443.7780.003 *
OS Wald (backward stepwise) method
VN cutoff1.2630.4497.9223.5360.005 *
MYCN status0.8400.4603.3352.3160.068
EFS Wald (forward stepwise) method
−4p−1.4710.7523.8250.2300.050
VN cutoff1.4600.4729.5754.3070.002 *
EFS Wald (backward stepwise) method
−4p−1.4710.7523.8250.2300.050
VN cutoff1.4600.4729.5754.3070.002 *
VN levels dichotomized by cutoff in NB patients older than 18 months, MYCN status (MYCN status: MNNA, MYCN non-amplified; hetMNA, MYCN heterogeneously amplified; MNA, MYCN amplified) and loss of 4p chromosomic region were the variables maintained in the last step of the Cox regression tests according to EFS and OS; however, only VN levels showed significant p-values. Other variables without significant results included in the Cox regression analysis were age, stage (localized/metastatic), genomic profile (NCAs, chromosomal numeric aberrations/SCAs, chromosomal segmental aberrations) and typical SCAs (−1p, +1q, +2p, 3p−, −4p, −11q, +17q). B: beta coefficient; SE: standard error. Coefficients Exp (B)  >  1 indicate that high values of this parameter increase its probability of being an independent poor prognostic factor. *: Significant p-values (< 0.05).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

López-Carrasco, A.; Vieco-Martí, I.; Granados-Aparici, S.; Acevedo-León, D.; Estañ-Capell, N.; Portugal, R.; Huerta-Aragonés, J.; Cañete, A.; Navarro, S.; Noguera, R. Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker? Int. J. Mol. Sci. 2024, 25, 8733. https://doi.org/10.3390/ijms25168733

AMA Style

López-Carrasco A, Vieco-Martí I, Granados-Aparici S, Acevedo-León D, Estañ-Capell N, Portugal R, Huerta-Aragonés J, Cañete A, Navarro S, Noguera R. Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker? International Journal of Molecular Sciences. 2024; 25(16):8733. https://doi.org/10.3390/ijms25168733

Chicago/Turabian Style

López-Carrasco, Amparo, Isaac Vieco-Martí, Sofía Granados-Aparici, Delia Acevedo-León, Nuria Estañ-Capell, Raquel Portugal, Jorge Huerta-Aragonés, Adela Cañete, Samuel Navarro, and Rosa Noguera. 2024. "Vitronectin Levels in the Plasma of Neuroblastoma Patients and Culture Media of 3D Models: A Prognostic Circulating Biomarker?" International Journal of Molecular Sciences 25, no. 16: 8733. https://doi.org/10.3390/ijms25168733

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

Article Metrics

Back to TopTop