Next Article in Journal
Persistence of Hyper-Ramified Microglia in Porcine Cortical Gray Matter after Mild Traumatic Brain Injury
Previous Article in Journal
Sodium Alginate-Based MgO Nanoparticles Coupled Antibiotics as Safe and Effective Antimicrobial Candidates against Staphylococcus aureus of Houbara Bustard Birds
Previous Article in Special Issue
Insights into Healthcare Professionals’ Perceptions and Attitudes toward Nanotechnological Device Application: What Is the Current Situation in Glioblastoma Research?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Assessment of Hyperosmolar Blood–Brain Barrier Opening in Glioblastoma via Histology with Evans Blue and DCE-MRI

1
UCLouvain, Louvain Drug Research Institute (LDRI), Biomedical Magnetic Resonance Research Group, 1200 Brussels, Belgium
2
UCLouvain, Louvain Drug Research Institute (LDRI), Advanced Drug Delivery and Biomaterials Research Group, 1200 Brussels, Belgium
3
UCLouvain, Louvain Drug Research Institute (LDRI), Nuclear and Electron Spin Technologies (NEST) Platform, 1200 Brussels, Belgium
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(7), 1957; https://doi.org/10.3390/biomedicines11071957
Submission received: 6 June 2023 / Revised: 2 July 2023 / Accepted: 7 July 2023 / Published: 11 July 2023
(This article belongs to the Special Issue Targeted Drug Delivery to Brain)

Abstract

:
Background: While the blood–brain barrier (BBB) is often compromised in glioblastoma (GB), the perfusion and consequent delivery of drugs are highly heterogeneous. Moreover, the accessibility of drugs is largely impaired in the margins of the tumor and for infiltrating cells at the origin of tumor recurrence. In this work, we evaluate the value of methods to assess hemodynamic changes induced by a hyperosmolar shock in the core and the margins of a tumor in a GB model. Methods: Osmotic shock was induced with an intracarotid infusion of a hypertonic solution of mannitol in mice grafted with U87-MG cells. The distribution of fluorescent dye (Evans blue) within the brain was assessed via histology. Dynamic contrast-enhanced (DCE)-MRI with an injection of Gadolinium-DOTA as the contrast agent was also used to evaluate the effect on hemodynamic parameters and the diffusion of the contrast agent outside of the tumor area. Results: The histological study revealed that the fluorescent dye diffused much more largely outside of the tumor area after osmotic shock than in control tumors. However, the study of tumor hemodynamic parameters via DCE-MRI did not reveal any change in the permeability of the BBB, whatever the studied MRI parameter. Conclusions: The use of hypertonic mannitol infusion seems to be a promising method to increase the delivery of compounds in the margins of GB. Nevertheless, the DCE-MRI analysis method using gadolinium-DOTA as a contrast agent seems of limited value for determining the efficacy of opening the BBB in GB after osmotic shock.

1. Introduction

Glioblastoma (GB) is the most common and aggressive malignant tumor of the central nervous system in adults, accounting for more than 50% of gliomas. The current standard of care is to remove as much tumor tissue as possible via surgical resection, followed by adjuvant radiotherapy and chemotherapy to eradicate residual infiltrating tumor cells at the origin of recurrences [1]. Despite this aggressive treatment, the prognosis of patients suffering from GB remains poor, with a high recurrence rate, a median survival time lower than 2 years, and a 5 years survival rate lower than 5% [2,3].
This tumor shows a high proliferation rate, intratumoral and intertumoral heterogeneity, highly invasive and infiltrative cell properties in the adjacent brain parenchyma, and resistance to chemotherapy, making GB a very challenging cancer to treat. Many distinctive features of GB (genetic, cytological, or anatomical) hinder the treatment efficacy and must be overcome. One of the fundamental issues for treating GB is linked to the blood–brain barrier (BBB). This physiological barrier controls the entry of compounds into the brain and significantly inhibits the passage of drugs from the bloodstream to the brain, reducing the effectiveness of chemotherapeutic agents’ systemic delivery [4]. While the BBB is often compromised in GB, the perfusion and consequent delivery of drugs are highly heterogeneous. Indeed, tumoral BBB is characterized by an aberrant distribution of pericytes, a loss of astrocytic end-feet, as well as a loss of tight junction proteins between endothelial cells. Unlike the healthy BBB, the leaky tumoral BBB allows the passage of large molecules between endothelial cells, where the paracellular pathway is damaged [5]. However, tumoral BBB permeability is highly heterogeneous and varies not only between GB but also within the same tumor, with tumor regions (mainly in the margins of the tumor) where the BBB is still intact, which limit the access of drugs to the infiltrating tumor cells at the origin of the tumor recurrence [6,7]. Therefore, the tumoral BBB remains a major hurdle to overcome.
Many efforts have been made in past decades to develop new approaches in order to overcome it and improve the delivery of drugs to the tumor, especially in its margins. One of the strategies that we will examine deeper in this article is osmotic shock. Previous works have shown that hypertonic solution infusion induces a reversible opening of a healthy BBB [8,9]. The mechanism of action of this method is explained in Figure 1.
In this work, our aim was to evaluate whether this strategy can be used for tumoral BBB opening in GB, especially for the margins of the tumor. To do so, we evaluated whether a hyperosmolar shock may change hemodynamic parameters in the core and the margins of the tumor in a GB model. Two methods have been used to evaluate the efficacy of the treatment on perfusion/permeability parameters: a non-invasive one, based on magnetic resonance imaging (MRI) analysis (T1-weighted and T2-weighted imaging as well as dynamic contrast-enhanced (DCE)-MRI), and an invasive one, based on histological analysis with a fluorescent vascular leakage marker (Evans blue).
For the MRI analysis, we determined the core of the tumor through the use of T2-weighted contrast. In addition, T1-weighted images were obtained after the administration of a contrast agent to analyze the diffusion of the contrast agent within the bulk of the tumor and in its margins. Gadolinium-DOTA was used because this hydrophilic contrast agent is unable to cross the intact BBB, offering the possibility to tackle changes in BBB permeability. We defined the tumor margins as the difference between the area of the T1-enhancing region after the injection of the contrast agent and the area depicted by the T2-weighted contrast delineating the core of the tumor. The hypothesis was that, if there is an increase in the permeability of the BBB, the contrast agent should pass the barrier more easily and diffuse much more largely outside of the tumor area into the brain parenchyma. As a consequence, a larger contrast uptake area should be seen in the T1-weighted post-contrast image compared to the T2-weighted anatomical image. In other words, an increase in T1 in the T2 tumor surface ratio would indicate the opening of the BBB. In addition, DCE-MRI analysis gave access to complementary tumor hemodynamic parameters that may be biomarkers of the change in BBB permeability [10].
For the histological analysis, BBB integrity and vascular leakage were assessed by perfusing mice with Evans blue fluorescent dye which binds to plasma albumin with a very high affinity. Albumin cannot cross an intact BBB. However, when compromised, albumin-bound Evans blue can diffuse into the brain parenchyma and be visualized by fluorescence [11].

2. Materials and Methods

2.1. Orthotopic U-87MG Mouse Model

All experiments were performed in accordance with the European Directive 2010/63/EU and following the Belgian national regulation guidelines, and were approved by the ethical committee for animal care by the Faculty of Medicine of the Université Catholique de Louvain (2019/UCL/MD/004). Water and food were given ad libitum. Animal body weight was constantly monitored throughout the experiment.
Six-week-old female NMRI nude mice (Janvier, France) were anesthetized via intraperitoneal injection of ketamine/xylazine (100 and 13 mg/kg, respectively) and fixed on a stereotaxic frame. In 2 µL of native EMEM (ATCC), 4 × 104 cells of U-87MG (ATCC) were injected into the right frontal lobe using an infusion syringe pump (Harvard Apparatus, Holliston, MA, USA) mounted with a Hamilton syringe (26S gauge needle). The injection coordinates were 2.1 mm lateral and 0.5 mm posterior from the bregma, and 2.6 mm deep from the outer border of the cranium [12]. The tumor size monitoring was performed via MRI (see Section 2.3).

2.2. Mannitol-Induced Hyperosmotic BBB Disruption

When the tumor size reached 7 ± 1 mm3, osmotic shock was induced with an intracarotid infusion of a hypertonic solution of mannitol 25% v/m (the control group received NaCl 0.9%). Mice were anesthetized with isoflurane mixed with air (2.5% for induction, 1.5% for maintenance). The common carotid artery (CCA) bifurcation was isolated. A polyethylene microcatheter (PE 10, inner diameter 0.28 mm and outer diameter 0.61 mm, BD IntramedicTM, Sparks, NV, USA) was inserted into the CCA via a small arteriotomy and moved into the internal carotid artery for an infusion of warm and filtered mannitol 25% or normal saline (200 µL/min for 1 min) [13].

2.3. MRI

MRI was performed using a 11.7 T Bruker Biospec MRI system (Bruker, Ettlingen, Germany) equipped with a 1H quadrature transmit/receive birdcage coil (21 mm inner diameter, RAPID Biomedical, Rimpar, Germany). Mice were anesthetized with isoflurane mixed with air (2.5% for induction, 1.5% for maintenance). Animals were covered with a heating blanket and their temperature was monitored. A pressure pad was used to monitor the respiration rate.
Anatomical images were obtained using T2-weighted rapid acquisition with a refocused echo (RARE) sequence (echo time = 30 ms; repetition time = 2500 ms; number of slices = 25; field of view = 20 mm × 20 mm; matrix size = 200 × 200; resolution = 0.1 mm × 0.1 mm; slice thickness = 0.3 mm; acquisition time = 5 min 20 s; averages = 8). Tumor volume was determined from a manually drawn region of interest (ROI) using Paravision 6.0.1 software (Bruker BioSpin) on day 14 following tumor induction, and then daily until the tumor size reached 7 ± 1 mm3.
For DCE-MRI acquisition, T1-weighted gradient echo images were obtained via a fast low-angle shot (FLASH) sequence (echo time = 1.4 ms; repetition time = 11.719 ms; flip angle = 10.0°; field of view = 20 mm × 20 mm; matrix size = 128 × 128; resolution = 0.156 mm × 0.156 mm; slice thickness = 0.9 mm; averages = 1; total acquisition time = 22 min 40 s). A set of 450 scans with a temporal resolution of 3.02 s was acquired, with Gadolinium-DOTA (Dotarem® 0.5 mol/mL; Guerbet, Villepinte, France) administered intravenously at a dose of 0.29 mmol/kg after the 10th scan over 5 s. DCE-MRI acquisition was performed 24 h before (day 0), 5 min after (day 1), and 24 h (day 2) after the injection of mannitol 25% or saline.

2.4. Histological Study

Evans blue dye (EB: 2% in normal saline; Alfa Aesar, Haverhill, MA, USA) was intravenously injected (3 mL/kg) 5 min after Mannitol or NaCl intra-carotid injection. Thirty minutes later, the mice were euthanized and intracardially perfused with paraformaldehyde (PFA) 4% to discard all the remaining dye in the blood vessels and fix the tissue. Brains were then removed, fixed overnight in PFA 4%, cryoprotected in sucrose 20%, included in optimal cutting temperature compound (OCT, Sakura Finetek, Alphen aan den Rijn, The Netherlands), and kept at −80 °C. Cryostat 30 µm sections were counterstained with diamidino-2-phenylindole (DAPI, Thermofisher, Waltham, MA, USA) and examined under a fluorescence microscope slide scanner (Panoramic 250 Flash III, 3DHistech, Budapest, Hungary) with DAPI and Cyanine 5 (Cy5) filter [14,15].

2.5. Image Processing and Statistical Analyses

DCE-MRI data were analyzed using in-house software written in Matlab (version 9.6). Regions of interest (ROIs) were manually delineated. We considered ROI T1 as the delineation of the entire tumor area using T1-weighted images with Gd-DOTA as the contrast agent, ROI T2 as the delineation of the tumor bulk area using T2-weighted anatomical images, and ROI Delta as the ROI T1 from which we subtracted ROI T2 in order to cover the margins of the tumor. In this way, we were able to study the hemodynamic parameters for the whole tumor region but also for the margins of the tumor.
The hemodynamic parameters were computed using a two-compartmental model, the extended Tofts model [16]. This model takes into account the contribution of the vascular compartment, which is not negligible in tumors.
C t t = v p × C p t + K t r a n s 0 t C p τ e k e p ( t τ ) d τ
where Ktrans is the volume transfer constant between blood plasma and extravascular extracellular space (EES) [min−1], vp is the blood plasma volume per unit volume of tissue, and kep is the flux rate constant between EES and blood plasma [min−1] [17]. Ve is the EES volume per unit volume of tissue, calculated as follows:
v e = K t r a n s / k e p
We were also interested in AUC60 and AUC90 corresponding to the area under the curve (AUC) of contrast agent concentration as a function of time from 0 to 60 or to 90 s.
Histological images were analyzed using Qupath (version 0.3.2) [18]. The tumor ROI and the Evans blue diffusion ROI were manually delineated.
For statistical analyses, two-way ANOVA tests (Tukey’s test) and t-tests were performed using GraphPad Prism (version 9.1.2), with p-values < 0.05 (*), p < 0.01 (**), p < 0.001 (***), and p < 0.0001 (****) considered as the levels of significance. The results are presented as mean ± standard deviation (SD).

3. Results

3.1. Histological Study

BBB integrity was assessed by perfusing mice with EB dye, a fluorescent vascular leakage marker. On the brain sections treated with osmotic shock, the fluorescent dye diffused more largely outside of the tumor area than on the brain sections of the untreated tumors (Figure 2). We observed that the surface stained with EB was about two times larger than the tumor surface area (1.93 ± 0.43, mean ± SD, n = 4) in the control mice (treated with saline). The EB stained/tumor surface ratio in the group of mice receiving osmotic shock (3.282 ± 0.74; mean ± SD, n = 4) was significantly higher (p < 0.0001) than in the control group receiving saline, a result consistent with an opening of the BBB.

3.2. MRI Studies

3.2.1. T1/T2 Tumor Surface Ratio

BBB permeability was additionally evaluated using MRI. Here, we determined the anatomical tumor size using T2-weighted contrast images and diffusion of the tracer inside and outside of the tumor using T1-weighted images after the administration of Gd-DOTA used as a contrast agent (Figure 3). There was no significant difference (p > 0.05) between the T1/T2 surfaces ratio of the treated (1.68 ± 0.24, mean ± SD, n = 7) and untreated group (1.63 ± 0.21, mean ± SD, n = 5). These results indicate that this MRI assessment was unable to detect any difference in the BBB permeability between these two groups.

3.2.2. DCE-MRI

To further investigate BBB disruption, we also performed DCE-MRI to provide hemodynamic parameters such as contrast agent efflux transfer constant (Ktrans), contrast agent reflux transfer constant (kep), the intravascular volume fraction (vp), the extravascular volume fraction (ve), and the area under the curve of contrast agent concentration as a function of time from 0 to 60 or to 90 s (AUC60 and AUC90). These parameters were analyzed in two different regions of interest (ROIs) of the tumor: the ROI T1 corresponding to the whole tumor region, and the ROI Delta corresponding to the margin tumor area.
We did not observe any significant difference in the tumor hemodynamic parameters between tumors in mice treated with mannitol (n = 7) or saline (n = 5), whatever the studied parameter and the studied ROI (Figure 4 and Figure 5).

4. Discussion

While the BBB is generally disrupted at the site of the tumor bulk in glioblastoma, the BBB remains intact in the infiltrating part of the tumor margins, that is, at the origin of subsequent tumor recurrence [19,20,21,22]. This feature limits the conventional systemic delivery of many chemotherapy drugs and allows residual tumor cells to escape to cytotoxic treatments [23]. Several strategies have emerged to overcome this limited accessibility of drugs to residual tumor cells by temporarily and reversibly opening the BBB. Among the strategies, our present study has been focused on the use of the intraarterial infusion of a hypertonic solution. In preclinical studies, EB staining is often used to assess BBB permeability/integrity, including in glioblastoma models [24,25,26]. While this histological tool is highly effective in preclinical models, it requires the sacrifice of the animal and is obviously useless for clinical applications. To monitor treatment-induced changes in BBB permeability, DCE-MRI is particularly attractive as contrast-enhanced MRI is systematically used for the characterization of brain tumors in patients. Previous studies have shown that contrast-enhanced MRI was useful in preclinical models to assess changes in BBB permeability and to evaluate the effect of strategies opening the BBB [13,27,28]. Of note, most studies focused on the BBB opening in the brain parenchyma. A few examples evaluated ultrasound-mediated BBB opening in glioblastomas [29,30,31], but, as far as we know, none evaluated the ability of DCE-MRI to monitor osmotic-shock-induced BBB opening in brain tumors. In our study, particular attention was paid to the ability of the treatment to enlarge the delivery of compounds into the margins of the tumor. For this purpose, we compared areas accessible to BBB-impermeable agents with the anatomical areas of the tumors, with or without osmotic treatment.
In the histological studies, we observed that the area accessible to EB was 93% larger than the anatomic area in the group of mice receiving saline (control mice) (Figure 2). This suggests that the BBB is already permeable in regions surrounding the bulk tumor in the U-87MG mouse glioblastoma model. The diffusion of EB significantly increased after the infusion of 20% mannitol solution in the carotid artery: the accessible area to the dye was 3.28 times the area covered by the bulk tumor, as defined via DAPI staining (Figure 2). This increased diffusion capability could be particularly interesting to exploit in order to increase the delivery of anticancer drugs, whatever their mode of action.
To mimic the endpoints measured via histology in the MRI study, we compared T2-weighted images (corresponding to the anatomical image of the bulk tumor) and T1-weighted images after Gd-DOTA administration (corresponding to areas accessible by the BBB-impermeable contrast agent). In the group of mice receiving saline (control mice), the area accessible to Gd-DOTA visualized in the T1-weighted images was 68% larger than the bulk area of the tumor seen in the T2-weighted images (Figure 3). The area accessible to Gd-DOTA was not significantly altered for the group receiving the mannitol infusion (Figure 3). In contrast to the EB assay, non-invasive MRI using Gd-DOTA thus seemed unable to tackle these changes in BBB permeability in the surrounding margins of the tumor. In addition to the focus on diffusion patterns outside of the tumor bulk area, we sought to identify possible changes in intratumoral and marginal hemodynamics. For this purpose, we interrogated possible changes in Ktrans, kep, ve, vp, AUC60, and AUC90 in these tumor regions. There were no significant relative changes in these parameters compared to the basal values recorded one day before the treatment (Figure 4).
The differences in the results obtained through both methods regarding the assessment of BBB opening deserve discussion. It is first important to note that the values recorded via histology or MRI cannot be superimposed. First is the invasive nature of histology in terms of repeating the measurement on the same animal: the histological images obtained in the control and treated mice came from different cohorts of animals, while the MR images were measured longitudinally on the same animals. More importantly, it is important to realize that tracers differ in their molecular and biodistribution properties. Fluorescent EB (960 Da, molecular weight) strongly binds to albumin (68 kDa), while Gd-DOTA (580 Da) is a highly hydrophilic compound that does not bind to albumin. These properties mean that they have differences in their ability to cross vessel fenestrations. Gd-DOTA can diffuse easily in the extracellular compartment of normal vessels, except for those located in the BBB due to inter-endothelial tight junctions. In contrast, due to its binding to albumin, EB remains in the vessels, is unable to cross small fenestrations in the vascular wall, and can only cross large fenestrations. As osmotic shock induces a change in tight junctions between endothelial cells, we can assume that the observed change in permeability would be more pronounced for larger molecules (complex EB-albumin) than for smaller molecules (Gd-DOTA) that were already able to cross small fenestrations of the damaged tumoral BBB. Comparative studies have indeed reported differences in glioblastoma accumulation for small Gd complexes and radiolabeled albumin measured via PET [32]. In the future, it could be interesting to test the size-dependent ability to report on BBB opening using larger molecular entities or gadolinium-based contrast agents with high affinity for albumin, such as gadobenate (Gd-BOPTA), that would better mimic the distribution behavior of EB [33]. Moreover, it is important to notice that both methods present differences in terms of sensitivity and resolution. Indeed, the optical fluorescent method has a higher sensitivity of detection of probes informing on BBB permeability and much higher spatial resolution than the MRI method.
Regarding the DCE-MRI study, it is likely that the changes in hemodynamic parameters could be too subtle to be detected through the use of the pharmacokinetic model (the extended Tofts model). This model is a two-compartmental model widely used to study hemodynamic parameters in tumors [17,34]. It takes into account the contribution of the vascular compartment which is not negligible in tumors. Several studies have used this model to describe variations in hemodynamic parameters (Ktrans, kep, ve, and vp) after the disruption of the BBB [35,36,37,38,39]. Here, however, we were unable to tackle any variation in these parameters using the extended Tofts model. Measuring subtle BBB leakage using DCE-MRI presents unique challenges [40,41,42]. Among the kinetic models described in the literature, the Patlak model has been reported to detect subtle changes in hemodynamics associated with BBB disruption [40,41]. This model seems to be appropriate for studying low-level blood-brain barrier leakage where the back-flux from the interstitium to the capillaries is negligible [42,43,44,45]. In the tumor core of the GB, the BBB is damaged and the extended Tofts model seems to be more appropriate as the back-flux is not negligible. However, in further studies, it could be interesting to apply the Patlak model in the margins of the tumor where the BBB is less fenestrated and where the back-flux from the interstitium can be neglected. The parameters AUC60 and AUC90 that are model-independent could have potentially hinted a change in contrast agent uptake. While there was a trend for an increased AUC60 and AUC90, the changes were not significant.
It is important to highlight that the anesthetic regimen used in the present study may have played a role in our assessment of hemodynamics. We used isoflurane because previous studies have shown that this mode of anesthesia preserved the oxygenation in most tissues and peripheric tumors [46,47]. However, in the brain, several reports have suggested that isoflurane presents a vasodilatory effect that can increase baseline cerebral blood flow. This effect of isoflurane may decrease the vasodilatory reserve that can be recruited, as reported in functional MRI studies [48,49,50,51]. Therefore, isoflurane could have potentially affected our ability to see hemodynamic changes at the brain tumor level. However, we should notice that isoflurane was used for both EB and MRI assessments. As we observed, regarding the permeabilization of the BBB using EB, it is unlikely that our inability to see this phenomenon using MRI was due to the sole effect of isoflurane.

Author Contributions

Conceptualization, J.C., V.P. and B.G.; methodology, J.C., N.J., B.U., K.V., V.P. and B.G.; investigation, J.C., N.J., K.V. and B.U.; software, N.J.; validation, N.J., V.P. and B.G.; resources, N.J., V.P. and B.G.; data curation, J.C.; writing—original draft preparation, J.C.; writing—review and editing, J.C., V.P. and B.G.; funding acquisition, V.P. and B.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FNRS-Televie, grant numbers 7.4620.19 and 7.6505.21.

Institutional Review Board Statement

The study was approved by the ethical committee for animal care of the Health Sector of the Université catholique de Louvain (approved protocol number 2019/UCL/MD/004) and was conducted in accordance with the Belgian Law of 29 May 2013, regarding the protection of laboratory animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data will be provided upon e-mail request.

Acknowledgments

The authors would like to thank Caroline Bouzin from the 2IP imaging platform of the Institut de Recherches Expérimentales et Cliniques (IRECs) of UCLouvain for the technical assistance regarding the Qupath software.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cha, G.D.; Kang, T.; Baik, S.; Kim, D.; Choi, S.H.; Hyeon, T.; Kim, D.-H. Advances in drug delivery technology for the treatment of glioblastoma multiforme. J. Control. Release 2020, 328, 350–367. [Google Scholar] [CrossRef] [PubMed]
  2. Lukas, R.V.; Wainwright, D.A.; Ladomersky, E.; Sachdev, S.; Sonabend, A.M.; Stupp, R. Newly Diagnosed Glioblastoma: A Review on Clinical Management. Oncology 2019, 33, 91–100. [Google Scholar] [PubMed]
  3. Hottinger, A.F.; Stupp, R.; Homicsko, K. Standards of care and novel approaches in the management of glioblastoma multiforme. Chin. J. Cancer 2014, 33, 32–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Obermeier, B.; Daneman, R.; Ransohoff, R.M. Development, maintenance and disruption of the blood-brain barrier. Nat. Med. 2013, 19, 1584–1596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
  6. Lockman, P.R.; Mittapalli, R.K.; Taskar, K.S.; Rudraraju, V.; Gril, B.; Bohn, K.A.; Adkins, C.E.; Roberts, A.; Thorsheim, H.R.; Gaasch, J.A.; et al. Heterogeneous blood-tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer. Clin. Cancer Res. 2010, 16, 5664–5678. [Google Scholar] [CrossRef] [Green Version]
  7. Osswald, M.; Blaes, J.; Liao, Y.; Solecki, G.; Gömmel, M.; Berghoff, A.S.; Salphati, L.; Wallin, J.J.; Phillips, H.S.; Wick, W.; et al. Impact of Blood-Brain Barrier Integrity on Tumor Growth and Therapy Response in Brain Metastases. Clin. Cancer Res. 2016, 22, 6078–6087. [Google Scholar] [CrossRef] [Green Version]
  8. Rapoport, S.I.; Hori, M.; Klatzo, I. Testing of a hypothesis for osmotic opening of the blood-brain barrier. Am. J. Physiol. 1972, 223, 323–331. [Google Scholar] [CrossRef] [Green Version]
  9. Rapoport, S.I. Advances in osmotic opening of the blood-brain barrier to enhance CNS chemotherapy. Expert Opin. Investig. Drugs 2001, 10, 1809–1818. [Google Scholar] [CrossRef]
  10. Shukla, G.; Alexander, G.S.; Bakas, S.; Nikam, R.; Talekar, K.; Palmer, J.D.; Shi, W. Advanced magnetic resonance imaging in glioblastoma: A review. Chin. Clin. Oncol. 2017, 6, 40. [Google Scholar] [CrossRef] [Green Version]
  11. Ahishali, B.; Kaya, M. Evaluation of Blood-Brain Barrier Integrity Using Vascular Permeability Markers: Evans Blue, Sodium Fluorescein, Albumin-Alexa Fluor Conjugates, and Horseradish Peroxidase. Methods Mol. Biol. 2021, 2367, 87–103. [Google Scholar] [CrossRef] [PubMed]
  12. Bianco, J.; Bastiancich, C.; Joudiou, N.; Gallez, B.; des Rieux, A.; Danhier, F. Novel model of orthotopic U-87 MG glioblastoma resection in athymic nude mice. J. Neurosci. Methods 2017, 284, 96–102. [Google Scholar] [CrossRef] [PubMed]
  13. Chu, C.; Liu, G.; Janowski, M.; Bulte, J.W.M.; Li, S.; Pearl, M.; Walczak, P. Real-Time MRI Guidance for Reproducible Hyperosmolar Opening of the Blood-Brain Barrier in Mice. Front. Neurol. 2018, 9, 921. [Google Scholar] [CrossRef] [PubMed]
  14. Nicaise, C.; Mitrecic, D.; Demetter, P.; De Decker, R.; Authelet, M.; Boom, A.; Pochet, R. Impaired blood-brain and blood-spinal cord barriers in mutant SOD1-linked ALS rat. Brain Res. 2009, 1301, 152–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. del Valle, J.; Camins, A.; Pallàs, M.; Vilaplana, J.; Pelegrí, C. A new method for determining blood-brain barrier integrity based on intracardiac perfusion of an Evans Blue-Hoechst cocktail. J. Neurosci. Methods 2008, 174, 42–49. [Google Scholar] [CrossRef] [PubMed]
  16. Sourbron, S.P.; Buckley, D.L. Classic models for dynamic contrast-enhanced MRI. NMR Biomed. 2013, 26, 1004–1027. [Google Scholar] [CrossRef]
  17. Tofts, P.S.; Brix, G.; Buckley, D.L.; Evelhoch, J.L.; Henderson, E.; Knopp, M.V.; Larsson, H.B.; Lee, T.Y.; Mayr, N.A.; Parker, G.J.; et al. Estimating kinetic parameters from dynamic contrast-enhanced T(1)-weighted MRI of a diffusable tracer: Standardized quantities and symbols. J. Magn. Reson. Imaging 1999, 10, 223–232. [Google Scholar] [CrossRef]
  18. Bankhead, P.; Loughrey, M.B.; Fernández, J.A.; Dombrowski, Y.; McArt, D.G.; Dunne, P.D.; McQuaid, S.; Gray, R.T.; Murray, L.J.; Coleman, H.G.; et al. QuPath: Open source software for digital pathology image analysis. Sci. Rep. 2017, 7, 16878. [Google Scholar] [CrossRef] [Green Version]
  19. Van Tellingen, O.; Yetkin-Arik, B.; de Gooijer, M.C.; Wesseling, P.; Wurdinger, T.; de Vries, H.E. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist. Updates 2015, 19, 1–12. [Google Scholar] [CrossRef]
  20. Kane, J.R. The Role of Brain Vasculature in Glioblastoma. Mol. Neurobiol. 2019, 56, 6645–6653. [Google Scholar] [CrossRef]
  21. Zhang, D.Y.; Dmello, C.; Chen, L.; Arrieta, V.A.; Gonzalez-Buendia, E.; Kane, J.R.; Magnusson, L.P.; Baran, A.; James, C.D.; Horbinski, C.; et al. Ultrasound-mediated Delivery of Paclitaxel for Glioma: A Comparative Study of Distribution, Toxicity, and Efficacy of Albumin-bound Versus Cremophor Formulations. Clin. Cancer Res. 2020, 26, 477–486. [Google Scholar] [CrossRef]
  22. Gould, A.; Zhang, D.; Arrieta, V.A.; Stupp, R.; Sonabend, A.M. Delivering albumin-bound paclitaxel across the blood-brain barrier for gliomas. Oncotarget 2021, 12, 2474–2475. [Google Scholar] [CrossRef] [PubMed]
  23. Gebhardt, B.J.; Dobelbower, M.C.; Ennis, W.H.; Bag, A.K.; Markert, J.M.; Fiveash, J.B. Patterns of failure for glioblastoma multiforme following limited-margin radiation and concurrent temozolomide. Radiat. Oncol. 2014, 9, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Dong, Q.; He, L.; Chen, L.; Deng, Q. Opening the Blood-Brain Barrier and Improving the Efficacy of Temozolomide Treatments of Glioblastoma Using Pulsed, Focused Ultrasound with a Microbubble Contrast Agent. BioMed Res. Int. 2018, 2018, 6501508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Stegmayr, C.; Oliveira, D.; Niemietz, N.; Willuweit, A.; Lohmann, P.; Galldiks, N.; Shah, N.J.; Ermert, J.; Langen, K.J. Influence of Bevacizumab on Blood-Brain Barrier Permeability and O-(2-(18)F-Fluoroethyl)-l-Tyrosine Uptake in Rat Gliomas. J. Nucl. Med. 2017, 58, 700–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Leten, C.; Struys, T.; Dresselaers, T.; Himmelreich, U. In vivo and ex vivo assessment of the blood brain barrier integrity in different glioblastoma animal models. J. Neurooncology 2014, 119, 297–306. [Google Scholar] [CrossRef] [PubMed]
  27. Rhine, W.D.; Benaron, D.A.; Enzmann, D.R.; Chung, C.; Gonzales-Mendez, R.; Sayre, J.R.; Stevenson, D.K. Gd-DTPA MR detection of blood-brain barrier opening in rats after hyperosmotic shock. J. Comput. Assist. Tomogr. 1993, 17, 563–566. [Google Scholar] [CrossRef] [PubMed]
  28. Blanchette, M.; Pellerin, M.; Tremblay, L.; Lepage, M.; Fortin, D. Real-time monitoring of gadolinium diethylenetriamine penta-acetic acid during osmotic blood-brain barrier disruption using magnetic resonance imaging in normal wistar rats. Neurosurgery 2009, 65, 344–350; discussion 350-341. [Google Scholar] [CrossRef]
  29. Ahmed, M.H.; Hernández-Verdin, I.; Quissac, E.; Lemaire, N.; Guerin, C.; Guyonnet, L.; Zahr, N.; Mouton, L.; Santin, M.; Petiet, A.; et al. Low-Intensity Pulsed Ultrasound-Mediated Blood-Brain Barrier Opening Increases Anti-Programmed Death-Ligand 1 Delivery and Efficacy in Gl261 Mouse Model. Pharmaceutics 2023, 15, 455. [Google Scholar] [CrossRef]
  30. Wang, J.; Li, Z.; Pan, M.; Fiaz, M.; Hao, Y.; Yan, Y.; Sun, L.; Yan, F. Ultrasound-mediated blood-brain barrier opening: An effective drug delivery system for theranostics of brain diseases. Adv. Drug Deliv. Rev. 2022, 190, 114539. [Google Scholar] [CrossRef]
  31. Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M.; et al. Enhancing glioblastoma treatment using cisplatin-gold-nanoparticle conjugates and targeted delivery with magnetic resonance-guided focused ultrasound. Nanomedicine 2018, 14, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  32. Brady, M.; Raghavan, R.; Sampson, J. Determinants of Intraparenchymal Infusion Distributions: Modeling and Analyses of Human Glioblastoma Trials. Pharmaceutics 2020, 12, 895. [Google Scholar] [CrossRef] [PubMed]
  33. Cavagna, F.M.; Maggioni, F.; Castelli, P.M.; Daprà, M.; Imperatori, L.G.; Lorusso, V.; Jenkins, B.G. Gadolinium chelates with weak binding to serum proteins. A new class of high-efficiency, general purpose contrast agents for magnetic resonance imaging. Investig. Radiol. 1997, 32, 780–796. [Google Scholar] [CrossRef] [PubMed]
  34. Sourbron, S.P.; Buckley, D.L. On the scope and interpretation of the Tofts models for DCE-MRI. Magn. Reson. Med. 2011, 66, 735–745. [Google Scholar] [CrossRef]
  35. Oghabian, M.A.; Fatemidokht, A.; Haririchian, M.H. Quantification of Blood-Brain-Barrier Permeability Dysregulation and Inflammatory Activity in MS Lesions by Dynamic-Contrast Enhanced MR Imaging. Basic Clin. Neurosci. 2022, 13, 117–128. [Google Scholar] [CrossRef]
  36. Oh, S.S.; Lee, E.H.; Kim, J.H.; Seo, Y.B.; Choo, Y.J.; Park, J.; Chang, M.C. The Use of Dynamic Contrast-Enhanced Magnetic Resonance Imaging for the Evaluation of Blood-Brain Barrier Disruption in Traumatic Brain Injury: What Is the Evidence? Brain Sci. 2021, 11, 775. [Google Scholar] [CrossRef]
  37. Yoen, H.; Yoo, R.E.; Choi, S.H.; Kim, E.; Oh, B.M.; Yang, D.; Hwang, I.; Kang, K.M.; Yun, T.J.; Kim, J.H.; et al. Blood-Brain Barrier Disruption in Mild Traumatic Brain Injury Patients with Post-Concussion Syndrome: Evaluation with Region-Based Quantification of Dynamic Contrast-Enhanced MR Imaging Parameters Using Automatic Whole-Brain Segmentation. Korean J. Radiol. 2021, 22, 118–130. [Google Scholar] [CrossRef]
  38. Inglese, M.; Ordidge, K.L.; Honeyfield, L.; Barwick, T.D.; Aboagye, E.O.; Waldman, A.D.; Grech-Sollars, M. Reliability of dynamic contrast-enhanced magnetic resonance imaging data in primary brain tumours: A comparison of Tofts and shutter speed models. Neuroradiology 2019, 61, 1375–1386. [Google Scholar] [CrossRef] [Green Version]
  39. Keil, V.C.; Gielen, G.H.; Pintea, B.; Baumgarten, P.; Datsi, A.; Hittatiya, K.; Simon, M.; Hattingen, E. DCE-MRI in Glioma, Infiltration Zone and Healthy Brain to Assess Angiogenesis: A Biopsy Study. Clin. Neuroradiol. 2021, 31, 1049–1058. [Google Scholar] [CrossRef]
  40. Heye, A.K.; Thrippleton, M.J.; Armitage, P.A.; Valdés Hernández, M.D.C.; Makin, S.D.; Glatz, A.; Sakka, E.; Wardlaw, J.M. Tracer kinetic modelling for DCE-MRI quantification of subtle blood-brain barrier permeability. Neuroimage 2016, 125, 446–455. [Google Scholar] [CrossRef] [Green Version]
  41. Heye, A.K.; Culling, R.D.; Valdés Hernández Mdel, C.; Thrippleton, M.J.; Wardlaw, J.M. Assessment of blood-brain barrier disruption using dynamic contrast-enhanced MRI. A systematic review. NeuroImage Clin. 2014, 6, 262–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Manning, C.; Stringer, M.; Dickie, B.; Clancy, U.; Valdés Hernandez, M.C.; Wiseman, S.J.; Garcia, D.J.; Sakka, E.; Backes, W.H.; Ingrisch, M.; et al. Sources of systematic error in DCE-MRI estimation of low-level blood-brain barrier leakage. Magn. Reson. Med. 2021, 86, 1888–1903. [Google Scholar] [CrossRef] [PubMed]
  43. Rosenberg, G.A.; Wallin, A.; Wardlaw, J.M.; Markus, H.S.; Montaner, J.; Wolfson, L.; Iadecola, C.; Zlokovic, B.V.; Joutel, A.; Dichgans, M.; et al. Consensus statement for diagnosis of subcortical small vessel disease. J. Cereb. Blood Flow Metab. 2016, 36, 6–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Thrippleton, M.J.; Backes, W.H.; Sourbron, S.; Ingrisch, M.; van Osch, M.J.P.; Dichgans, M.; Fazekas, F.; Ropele, S.; Frayne, R.; van Oostenbrugge, R.J.; et al. Quantifying blood-brain barrier leakage in small vessel disease: Review and consensus recommendations. Alzheimer’s Dement. J. 2019, 15, 840–858. [Google Scholar] [CrossRef] [PubMed]
  45. Wong, S.M.; Jansen, J.F.A.; Zhang, C.E.; Staals, J.; Hofman, P.A.M.; van Oostenbrugge, R.J.; Jeukens, C.; Backes, W.H. Measuring subtle leakage of the blood-brain barrier in cerebrovascular disease with DCE-MRI: Test-retest reproducibility and its influencing factors. J. Magn. Reson. Imaging 2017, 46, 159–166. [Google Scholar] [CrossRef] [PubMed]
  46. Baudelet, C.; Gallez, B. Effect of anesthesia on the signal intensity in tumors using BOLD-MRI: Comparison with flow measurements by Laser Doppler flowmetry and oxygen measurements by luminescence-based probes. Magn. Reson. Imaging 2004, 22, 905–912. [Google Scholar] [CrossRef]
  47. Gallez, B. Oxygenation Status in Normal Tissues, Pathological Tissues and Malignant Tumors: A pO2 Database Based on Electron Paramagnetic Resonance (EPR) Oximetry Measurements. Appl. Magn. Reson. 2021, 52, 1395–1450. [Google Scholar] [CrossRef]
  48. Schroeter, A.; Schlegel, F.; Seuwen, A.; Grandjean, J.; Rudin, M. Specificity of stimulus-evoked fMRI responses in the mouse: The influence of systemic physiological changes associated with innocuous stimulation under four different anesthetics. Neuroimage 2014, 94, 372–384. [Google Scholar] [CrossRef]
  49. Grandjean, J.; Schroeter, A.; Batata, I.; Rudin, M. Optimization of anesthesia protocol for resting-state fMRI in mice based on differential effects of anesthetics on functional connectivity patterns. Neuroimage 2014, 102 Pt 2, 838–847. [Google Scholar] [CrossRef]
  50. Wu, T.; Grandjean, J.; Bosshard, S.C.; Rudin, M.; Reutens, D.; Jiang, T. Altered regional connectivity reflecting effects of different anaesthesia protocols in the mouse brain. Neuroimage 2017, 149, 190–199. [Google Scholar] [CrossRef] [Green Version]
  51. Munting, L.P.; Derieppe, M.P.P.; Suidgeest, E.; Denis de Senneville, B.; Wells, J.A.; van der Weerd, L. Influence of different isoflurane anesthesia protocols on murine cerebral hemodynamics measured with pseudo-continuous arterial spin labeling. NMR Biomed. 2019, 32, e4105. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Model for the opening of the inter-endothelial tight junctions via carotid infusion of a hypertonic solution. During infusion, endothelial cells dehydrate, water leaves the brain, leading to vasodilation, and the endothelial cell cytoskeleton contracts. Tension is exerted at tight junctional areas due to each of these factors, leading to a reversible opening of the junctions [9].
Figure 1. Model for the opening of the inter-endothelial tight junctions via carotid infusion of a hypertonic solution. During infusion, endothelial cells dehydrate, water leaves the brain, leading to vasodilation, and the endothelial cell cytoskeleton contracts. Tension is exerted at tight junctional areas due to each of these factors, leading to a reversible opening of the junctions [9].
Biomedicines 11 01957 g001
Figure 2. Assessment of diffusion of contrast agent outside of the tumor area via histology with Evans blue dye. Representative histological images of brain sections from a control mouse (treated with NaCl 0.9%, top row) and a mouse treated with osmotic shock (mannitol 25%, bottom row). The tumor area is encircled in green and the area of diffusion of EB is delineated in orange. The right panel shows the ratio of the surface stained with EB on the tumor surface for mice treated with osmotic shock (n = 4) and control mice (n = 4). The fluorescent dye diffused more largely outside of the tumor area after osmotic shock than in untreated tumors. The results are expressed as means ± SD. **** p < 0.0001.
Figure 2. Assessment of diffusion of contrast agent outside of the tumor area via histology with Evans blue dye. Representative histological images of brain sections from a control mouse (treated with NaCl 0.9%, top row) and a mouse treated with osmotic shock (mannitol 25%, bottom row). The tumor area is encircled in green and the area of diffusion of EB is delineated in orange. The right panel shows the ratio of the surface stained with EB on the tumor surface for mice treated with osmotic shock (n = 4) and control mice (n = 4). The fluorescent dye diffused more largely outside of the tumor area after osmotic shock than in untreated tumors. The results are expressed as means ± SD. **** p < 0.0001.
Biomedicines 11 01957 g002
Figure 3. Assessment of diffusion of contrast agent outside of the tumor area using MRI. MRI images of brains from control mice (receiving NaCl 0.9%, top row) and mice treated with osmotic shock (receiving mannitol 25%, bottom row), with T2-weighted contrast (left) and T1-weighted contrast after administration of Gd-DOTA (right). The T2-weighted images allow the anatomical delineation of the tumor (encircled in green) and the post-contrast T1-weighted images allow the assessment of vascular leakage (encircled in red). The right panel shows the ratio between the T1 and T2 surface areas. Using this procedure, no significant difference in BBB permeability was observed between tumors receiving saline (n = 5) or mannitol (n = 7). The results are expressed as means ± SD.
Figure 3. Assessment of diffusion of contrast agent outside of the tumor area using MRI. MRI images of brains from control mice (receiving NaCl 0.9%, top row) and mice treated with osmotic shock (receiving mannitol 25%, bottom row), with T2-weighted contrast (left) and T1-weighted contrast after administration of Gd-DOTA (right). The T2-weighted images allow the anatomical delineation of the tumor (encircled in green) and the post-contrast T1-weighted images allow the assessment of vascular leakage (encircled in red). The right panel shows the ratio between the T1 and T2 surface areas. Using this procedure, no significant difference in BBB permeability was observed between tumors receiving saline (n = 5) or mannitol (n = 7). The results are expressed as means ± SD.
Biomedicines 11 01957 g003
Figure 4. Hemodynamic parameters measured via DCE-MRI study. (a) Relative changes in tumor hemodynamic parameters (Ktrans, kep, vp, ve, AUC60, and AUC90) between mice treated with osmotic shock (dark red) and control mice (dark blue) using ROI T1; (b) relative changes in tumor hemodynamic parameters between mice treated with osmotic shock (light red) and control mice (light blue) using ROI Delta corresponding to the margin tumor area; note that the changes observed in the margins (b) are rather comparable to the changes observed in the core of the tumor (a), with the variability being generally larger in the tumor margins; (c) tumor hemodynamic parameter values between mice treated with osmotic shock using ROI Delta (light red) and using ROI T1 (dark red), and control mice using ROI Delta (light blue) and ROI T1 (dark blue). DCE-MRI acquisition was performed 24 h before (day 0), 5 min after (day 1), and 24 h (day 2) after the injection of mannitol 25% or saline. There was no significant difference in the tumor hemodynamic parameters between tumors receiving mannitol (n = 7) and saline (n = 5), whatever the studied parameter and ROI. The results are expressed as means ± SD.
Figure 4. Hemodynamic parameters measured via DCE-MRI study. (a) Relative changes in tumor hemodynamic parameters (Ktrans, kep, vp, ve, AUC60, and AUC90) between mice treated with osmotic shock (dark red) and control mice (dark blue) using ROI T1; (b) relative changes in tumor hemodynamic parameters between mice treated with osmotic shock (light red) and control mice (light blue) using ROI Delta corresponding to the margin tumor area; note that the changes observed in the margins (b) are rather comparable to the changes observed in the core of the tumor (a), with the variability being generally larger in the tumor margins; (c) tumor hemodynamic parameter values between mice treated with osmotic shock using ROI Delta (light red) and using ROI T1 (dark red), and control mice using ROI Delta (light blue) and ROI T1 (dark blue). DCE-MRI acquisition was performed 24 h before (day 0), 5 min after (day 1), and 24 h (day 2) after the injection of mannitol 25% or saline. There was no significant difference in the tumor hemodynamic parameters between tumors receiving mannitol (n = 7) and saline (n = 5), whatever the studied parameter and ROI. The results are expressed as means ± SD.
Biomedicines 11 01957 g004
Figure 5. DCE-MRI parametric maps. Illustration of typical Ktrans (a) and vp (b) maps obtained from an untreated (NaCl 0.9%) and a treated mouse (mannitol 25%) each with Gd-DOTA. DCE-MRI acquisition was performed 24 h before (day 0), 5 min after (day 1), and 24 h (day 2) after the injection of mannitol 25% or saline.
Figure 5. DCE-MRI parametric maps. Illustration of typical Ktrans (a) and vp (b) maps obtained from an untreated (NaCl 0.9%) and a treated mouse (mannitol 25%) each with Gd-DOTA. DCE-MRI acquisition was performed 24 h before (day 0), 5 min after (day 1), and 24 h (day 2) after the injection of mannitol 25% or saline.
Biomedicines 11 01957 g005
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

Conq, J.; Joudiou, N.; Ucakar, B.; Vanvarenberg, K.; Préat, V.; Gallez, B. Assessment of Hyperosmolar Blood–Brain Barrier Opening in Glioblastoma via Histology with Evans Blue and DCE-MRI. Biomedicines 2023, 11, 1957. https://doi.org/10.3390/biomedicines11071957

AMA Style

Conq J, Joudiou N, Ucakar B, Vanvarenberg K, Préat V, Gallez B. Assessment of Hyperosmolar Blood–Brain Barrier Opening in Glioblastoma via Histology with Evans Blue and DCE-MRI. Biomedicines. 2023; 11(7):1957. https://doi.org/10.3390/biomedicines11071957

Chicago/Turabian Style

Conq, Jérôme, Nicolas Joudiou, Bernard Ucakar, Kevin Vanvarenberg, Véronique Préat, and Bernard Gallez. 2023. "Assessment of Hyperosmolar Blood–Brain Barrier Opening in Glioblastoma via Histology with Evans Blue and DCE-MRI" Biomedicines 11, no. 7: 1957. https://doi.org/10.3390/biomedicines11071957

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