Next Article in Journal
Stability Characterization of the Novel Anti-Cancer HM-10/10 HDL-Mimetic Peptide
Next Article in Special Issue
Bypass of Abasic Site–Peptide Cross-Links by Human Repair and Translesion DNA Polymerases
Previous Article in Journal
Comparative Analysis of a Human Neutralizing mAb Specific for SARS-CoV-2 Spike-RBD with Cilgavimab and Tixagevimab for the Efficacy on the Omicron Variant in Neutralizing and Detection Assays
Previous Article in Special Issue
Entangled Motifs in Membrane Protein Structures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Brain Pre-Metastatic Niche: Biological and Technical Advancements

1
Department of Neurosurgery, University Hospital, Goethe-University, 60528 Frankfurt am Main, Germany
2
Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany
3
German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
4
Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
5
Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60528 Frankfurt am Main, Germany
6
German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(12), 10055; https://doi.org/10.3390/ijms241210055
Submission received: 28 February 2023 / Revised: 31 May 2023 / Accepted: 5 June 2023 / Published: 13 June 2023
(This article belongs to the Collection Feature Papers in “Molecular Biology”)

Abstract

:
Metastasis, particularly brain metastasis, continues to puzzle researchers to this day, and exploring its molecular basis promises to break ground in developing new strategies for combatting this deadly cancer. In recent years, the research focus has shifted toward the earliest steps in the formation of metastasis. In this regard, significant progress has been achieved in understanding how the primary tumor affects distant organ sites before the arrival of tumor cells. The term pre-metastatic niche was introduced for this concept and encompasses all influences on sites of future metastases, ranging from immunological modulation and ECM remodeling to the softening of the blood–brain barrier. The mechanisms governing the spread of metastasis to the brain remain elusive. However, we begin to understand these processes by looking at the earliest steps in the formation of metastasis. This review aims to present recent findings on the brain pre-metastatic niche and to discuss existing and emerging methods to further explore the field. We begin by giving an overview of the pre-metastatic and metastatic niches in general before focusing on their manifestations in the brain. To conclude, we reflect on the methods usually employed in this field of research and discuss novel approaches in imaging and sequencing.

1. Introduction

Cancer poses the greatest burden of any disease clinically, socially, and economically and is the second cause of death worldwide. Metastasis, the spread of cancer cells from the primary tumor to distant organs, is the leading cause of death in cancer patients [1]. Brain metastasis is a devastating complication of cancer that affects approximately 20% of all cancer patients [2], yet it eludes effective treatment and has been extraordinarily hard to decipher [3]. In recent years, metastasis research in general and brain metastasis research in particular have seen productive surges fueled by advances in both pre-clinical and clinical techniques [4,5,6].
For example, the recognition of the microenvironment as a key factor in the development and progression of cancer metastasis forms the concept of the metastatic niche (MN) [7]. Another key insight was the discovery of the pre-metastatic niche (PMN)—this proposed microenvironment is established by the primary tumor at a distance, ripening the sites of future metastasis before tumor cells reach the final steps of extravasation, migration, and invasion. The PMN is established via the secretion of exosomes, cytokines, growth factors, and other signaling molecules. These factors modify the composition of the extracellular matrix in the target organs via the recruitment of immune and stromal cells. This deliberately generated microenvironment provides a soil in distant organs to accept circulating tumor cells and support the survival, growth, and colonization of cancer cells. The concept has opened up new possibilities for understanding some of the most intriguing questions in cancer biology [8].
Studying the brain PMN has developed into an area of active research with promising discoveries. Our review aims to provide an overview of recent understandings in this area and to discuss possible experimental approaches, leading to more insights in the future.

2. General Concepts of the Pre-Metastatic Niche

Past metastasis research focused on the six basic steps of the metastatic cascade: primary tumor growth, tumor intravasation, tumor cell dissemination, tumor cell extravasation, tumor cell adaption/dormancy, and a distant tumor in the metastasis process. In hindsight, this provided relatively simple description of the actual processes [9]. The prevailing model did not account for complex signaling by primary tumors and the interplay between pioneering micrometastases and the extracellular matrix, including the multitude of angiogenetic factors [10].
This dogma was questioned when the initiation of a pre-metastatic niche was observed for the first time. Kaplan et al. showed a mechanism through which bone-marrow-derived stem cells (BMDCs) clustered in future sites of metastasis in the lung and upregulated local fibronectin production to promote the adhesion of circulating tumor cells [11]. Through this mechanism, a fertile soil can be generated by a primary tumor to accept its seeds—this reflects a refinement of the seed and soil concept that was famously proposed by Paget and Fuchs in 1889 [12]. Since then, a number of theoretical PMN mechanisms have been explored for their ability to modify organ sites from a distance (Figure 1). They range from preparing the extracellular matrix by upregulating pro-adhesive factors to the early initiation of angiogenesis [13,14]. Discoveries concerning endothelial cells consistently emerge in the PMN literature [8,15,16,17,18,19]. The endothelial wall is the first barrier to circulating tumor cells blocking intra and extravasation. Hence, an important role of the endothelium in early metastasis is to be expected. The remodeling of endothelial cells and the upregulation of cell adhesion molecules by melanoma and lung carcinoma cells was found to be mediated via hijacking the endothelial signal transducer activator of transcription 3 (STAT3) [20]. Similarly, endothelial adhesion seems to be promoted in estrogen-receptor-negative breast cancer cells via E-selectin [21]. Endothelial cells are also modified through the upregulation of the Notch1 receptor, promoting senescence and inflammation, which are conducive to metastasis [22]. In a spontaneous murine breast cancer model, pre-metastatic lung tissue experienced an influx of immature myeloid cells, which subsequently remodeled the endothelium via matrix metalloproteinase 9 (MMP9) and promoted metastasis—a phenotype that could be reverted via MMP9 deletion [17].
Over the last two decades, the importance of the immune system in cancer was established. This sprouted a fertile research field known as immuno-oncology [9,10,23,24]. For example, the mesenchymal–epithelial–transition is seen as a crucial step in providing a pro-invasive microenvironment and is controlled by a number of immunological factors, including transforming growth factor (TGF)-β [6], which is widely characterized in a pre-metastatic setting [25,26,27]. Immunological modulations, the recruitment of immunosuppressive cells and the ambiguous plasticity of neutrophils have attracted the interests of metastasis researchers in general and the field of PMN scientists in particular [28,29]. In fact, immunological modulation can move in both suppressive and activating directions as cancer makes twofold use of the immune system [30]. Immune escape phenomena are well known in the tumor microenvironment in which regulatory T-cells (Treg) and tumor-associated macrophages (TAM) suppress CD4+ and CD8+ T-cell activation [30,31,32,33]. TAMs create an immuno-suppressive environment via the expression of anti-inflammatory cytokines (TGFb1, IL1 receptor antagonist and IL10) [34], while tumor inflammatory monocytes (TIMs) promote a pro-inflammatory milieu (via IL1β and TNF) [35,36]. Interestingly, profiling TIMs across different metastatic sites and primary tumors revealed significant changes in their pro-inflammatory signatures depending on the origin of the tumor. Functional assays linked the presence of TIMs and TAMs with the exhaustion of cytotoxic and helper T cells in the metastatic site via the CCL20-CCR6 axis. Blocking this axis reverted immunosuppression, reduced T-cell exhaustion and delayed the progression of metastasis in mouse models [35], illustrating the value of modulating the immune microenvironment as an alternative therapeutic strategy to indirectly suppress tumor cell expansion. Single-cell profiling of bone metastasis samples from prostate cancer patients revealed significant changes in the transcriptional signatures of myeloid cells located at the metastatic versus distal vertebral sites [35]. Recently, a pre-metastatic immune modulation of circulating tumor cells was discussed, but the field still lacks conclusive results regarding those effects in the PMN or even the MN [37].
The mechanisms through which specific organs are targeted by primary tumors and how they form niches remain under investigation. Some of the most promising candidates are tumor-derived exosomes, which carry surface proteins with organotropic signatures [38,39]. This allows organotropism in a range of different cancer types via variations in integrin expression in endothelial cells [40]. Moreover, exosomes modulate the initial and subsequent tumor microenvironments of the primary and the metastatic tumors, respectively [41]. Exosomes from pancreatic ductal adenocarcinomas (PDACs) were found to regulate Kupffer cells in the liver, increasing pro-invasive fibronectin production [25]. The upregulation of Rab-proteins (notably RAB1A, RAB5B, RAB7 and RAB27A), which have implicit functions in membrane trafficking and exosome formation, can be found in a range of cancers [42,43,44].
Pro-angiogenic modulation in different tumors and organs by exosomal non-coding RNA (ncRNA) is well documented [14,45,46,47]. Recently, the influence of ncRNA in metastasis and its effect on distant organ sites has been introduced. Through the direct genetic manipulation of target cells and easy packaging in exosomes, ncRNA acts as an early effector and modulator of the pre-metastatic niche [18,48,49,50,51]. The organ specificity of tumor-derived exosomal ncRNA promises to improve targeted therapy and will identify novel therapeutic targets [52,53].

3. Recent Progress in Understanding the Brain Pre-Metastatic Niche

Empirical evidence for a biological niche that functionally predetermines metastatic seeding in the brain is scarce. One of the most important aspects that sets brain metastasis formation apart from other organ sites is the exclusive entry through the BBB, as there is no lymphatic system capable of introducing metastases in the brain [3,54,55]. Therefore the brain endothelium promises to be the main target for further research into this topic (Figure 2), and its possible role in cerebral metastases has been long appreciated [56].
An early event in brain metastatic colonization represents the activation of platelets through the von Willebrand factor (VWF) produced by platelets and cerebral endothelial cells. VWF fibers promote platelet clustering and support tumor cell arrest in vessels before extravasation. Interestingly, accumulations of VWF fibers were observed at similar extents in vessels in brain metastatic and perimetastatic regions, as well as in cerebral vessels in tumor-bearing mice without established brain metastasis, indicating a role in premetastatic niche formation [57]. Moreover, glial cells, in particular astrocytes and microglia, are active mediators of cerebral endothelial cell remodeling and are recruited to sites of future metastasis, where they undergo phenotypic switching. In melanoma brain metastasis, astrocyte recruitment involves the upregulation of neuroinflammatory and astrogliotic genes (most notably CCL2, CCl17, CXCL10, IL-23 and GFAP), leading to the formation of a glia scar and BBB disruption—a process well documented in other neuroinflammatory diseases [58,59,60,61,62] (Figure 2). Rodrigues et al. demonstrated that brain tropic breast cancer cells (231-BrT) shed exosomes containing high levels of CEMIP (cell migration-inducing and hyaluronan-binding protein), which increased cytokine production in the microglia with the effect of neuroinflammation and increased the leakiness of the neurovasculature. Although an effect on metastasis formation could be seen in vivo after pre-treatment with exosomes, cellular knockdown of CEMIP also abrogated metastasis. Hence, one could argue that the tumor-growth-suppressing functions of CEMIP significantly confounded the findings [63] (Figure 2). In overt breast cancer metastasis, reactive SIP3 (sphingosine-1 phosphate receptor 3) signaling from astrocytes in the neuroinflammatory response has been shown to regulate the vascular–tumor interface in overt breast cancer metastasis, highlighting a sustained importance of neuroinflammation throughout the metastatic process [64]. Neuroinflammation and glial activation were long regarded as host defense responses leading to tumor cell eradication. However, it is increasingly recognized that the interaction between metastatic tumor cells and microglia in the vascular niche leads to microglial reprogramming and functional cooption. This results in tumor-promoting functions across the stages of metastatic colonization, including vascular remodeling, tumor extravasation and outgrowth [65,66,67,68]. Likewise, functional cooption and the blockading of astrocytes’ anti-cancer functions by metastatic tumor cells at the neurovascular interface have been described and associated with increased brain metastatic potential and the instigation of neuroinflammatory responses [69,70,71]. Accordingly, neuroinflammatory responses have been documented in a number of human brain metastasis samples [72,73].
Paracrine secretions have been found to be sources of pre-metastatic signaling. For example, neural progenitor cells (NPC) were transformed into astrocytes in a co-culture with breast cancer cells via paracrine communication through transforming growth factor-β (TGF-β) family factor BMP-2 (bone morphogenic protein) [74] (Figure 2). As in vivo experiments require a strong ethical justification, co-culture data provide a good starting point to gauge individual research objectives and establish more elaborate animal experiments. In another case, the placental growth factor (PLGF) secreted by small-cell lung cancer (SCLC) has been shown to mediate the disassembly of brain endothelial tight-junctions and promote brain metastasis both in vitro and in patient samples, but the study lacks the in vivo data vital to drawing conclusions with respect to the PMN [75] (Figure 2). The role of vascular endothelial growth factor (VEGF) in metastasis is generally well understood, and its disruptive effects on the BBB have been documented both in vivo and in vitro [76,77,78]. VEGF-A-containing exosomes secreted by glioblastoma cells have been shown to open the BBB and promote angiogenesis [79], suggesting a similar role in metastasizing cancer (Figure 2). Vascular leakiness as an influence of estrogenic regulation has also been proposed in breast-cancer-associated metastasis but lacks experimental confirmation [80].
Finally, exosomal ncRNA has been shown to have a direct influence on future sites of metastasis. Its role in brain metastasis is mostly unexplored, but a few promising insights have been gained already. ncRNA was shown to downregulate the expression of tight junction proteins and promote cerebral metastasis in a murine NSCLC (non-small-cell lung cancer) model and was similarly proposed in an in vitro model of human breast cancer cells [81,82]. Fong et al. showed that the miR-122 secreted by breast cancer cells alters glucose metabolism in astrocytes, which favors the metabolism of metastatic cells (Figure 2). They were able to isolate the exosomes via gradient separation from a number of different breast cancer cell lines and conduct mechanistic experiments in vitro. By pre-conditioning mice with exosomes and through the subsequent injection of tumor cells, a strong effect on metastatic growth could be shown in vivo [83]. This study exemplifies the necessity for an observation of the proposed mechanisms isolated from confounders, such as circulating tumor cells, to convincingly show a real pre-metastatic effect. In another investigation on breast cancer exosomes by the same group which used comparable methodology, miR-105 was able to destroy endothelial tight junctions, and its upregulation led to more brain metastasis in a murine model [19] (Figure 2). This multitude of effects implies that more significant discoveries lie ahead for studying exosomal RNA in brain metastasis. Specific RNA-targeted therapeutics have also been discussed in the context of brain metastasis, as they have been for primary tumor RNA therapeutics, but to date, no suitable candidates could be discovered [84,85].

4. Differences between the PMN and MN

The pre-metastatic and metastatic niches are parts of the early phases of metastasis formation and although one precedes the other conceptually, they share considerable overlap in both mechanisms and effectors (Table 1) [7,8]. It can be assumed that the underlying molecular processes occur simultaneously and interdependently.
However, the pre-metastatic niche refers to the microenvironment that is created in a distant organ by the primary tumor prior to cancer cell infiltration. It provides a supportive environment by inducing vascular disruption and promoting extravasation, angiogenesis, inflammation and the early remodeling of the extracellular matrix via the recruitment of stromal cells [8].
The transition to the metastatic niche is fluent and begins after circulating cancer cells arrive in this pre-established tumor microenvironment. The establishment of the metastatic niche can be characterized by pro-survival signals of the pioneering micro-metastatic cells and the proliferation and initiation of angiogenesis. The goal of the first micro-metastasis is to foster self-survival and form stable growth and invasion [7].

5. Discussion

5.1. Experimentally Defining the PMN

The most obvious challenge in defining any characteristics of a hypothetical pre-metastatic niche is to capture the right time in the metastatic cascade in order to identify a causal link between a secreted factor from the primary tumor and the colonization of a specific organ [55]. Different approaches have been used to address this causality. One of the earliest methods described involves flow-cytometric measurements of GFP-tagged BMDCs and DsRed-tagged B16 murine melanoma cells at different time points in the lung lysates of mice previously injected orthotopically with B16 cells. The dynamic changes in cell type in the microenvironment show an influx of BMDCs days before the arrival of tumor cells, suggesting the formation of hospitable pre-metastatic conditions [11]. This method can show the dynamic composition of a proposed niche and clearly delineates its temporal component, yet it requires strong candidate factors previously identified for a successful experiment and does not necessarily prove a causal relationship. Moreover, analyzing organs at multiple time points can become very resource-intensive and will require a strong justification from an ethical perspective.
Probing a causal link between the formation of a pre-metastatic niche by a primary tumor and subsequent cell-specific homing was previously carried out in a challenging experiment [13]. In this setup, a primary tumor was implanted, which either secreted possible pre-metastatic niche-forming factors but did not metastasize itself or was resected before metastases could occur. In a second step, a metastasizing tumor cell line was injected into the bloodstream, and the metastasis pattern was analyzed against a non-secreting primary tumor control. Any differences could be attributed to the primary tumor and its niche-forming capabilities. This approach can enable the functional characterization of pre-metastatic mechanisms and by cross-linking different cell lines, it can describe ubiquitous niches across tumor entities. It is necessary to have a good understanding of the time points at which the primary tumor metastasizes, which is why this approach is probably limited to well-established tumor models. As is the case with any possible approach to a proposed pre-metastatic niche, a tumor model with reliable spontaneous metastasis is required.

5.2. Further Investigation Strategies for the PMN in the Brain

The exploration of pre-metastatic changes in the brain is challenging due to difficulties in obtaining spontaneous CNS metastases. In most cases, this necessitates the generation of brain-tropic cell lines (BrM) from existing tumor models. To obtain these specific cell lines, multiple rounds of tumor cell inoculation and in vivo selection from positive brains are performed [100]. The generated cell lines have the potential to reliably generate brain metastasis, but one could argue that the biology then becomes highly selected and diverges too far from the original tumor. To highlight specific differences between the parental and BrM cell lines, a comprehensive experimental setup should include the parallel cells. A comprehensive list of available selected cell lines is available online [101], rendering the first endeavors into the realm of in vivo brain metastatic research more approachable.
Arguably, the most restricting factor in studying pre-metastatic mechanisms in the brain is defining a suitable time point during the metastasis process and keeping the process as close to the natural biology as possible. As brain metastases are not as abundant in the course of the disease as visceral filliae, the humane endpoint of any experiment might be reached before meaningful conclusions can be drawn. To get ahead of the disease, approaches detecting the earliest micrometastases are needed (Figure 3).
The most pragmatic solution involves tumor inoculation/injection and the temporally spaced sacrifice of recipient animals, followed by measurements of the resident tumor cells in the brain tissues. This was performed by thin-slicing tissues and histopathological analyses [102]. Plotting the metastatic cells found in the tissue over these time points identified the first temporal occurrence of micrometastasis. This allows for the prior time point to be defined as the pre-metastatic niche. The benefits of this approach are the spatial-temporal reconstruction of the pre-metastatic niche and the identification of histological hotspots for tumor extravasation. On the contrary, biological variances in tumor growth and the metastasis process will make a clear division between a true pre-metastatic setting and overt metastasis difficult.
Diverse methods have been described to define the pre-metastatic niche in individual animals to make the results more consistent. Recently, it was shown that mCherry transcripts of subdermally inoculated mCherry-expressing melanoma cells could be measured in the CSF of tumor bearing mice and were an accurate predictor of incipient micrometastases in a spontaneous metastasis model [58]. The clear advantage of this approach is the early detection of micro metastases in a living animal, which could not be rendered otherwise. Although technically elegant, the experiment will probably only work with a limited selection of tumor entities that shed cells into the CSF. In addition, one could argue that the timeframe of a pre-metastatic niche has passed when the detection of cells in the CSF is possible, as the BBB must have then been breached already.
Intravital imaging has been fruitful in recent times and with the advancement of technology, the visualization of tumor cells in multiple organs in vivo has become possible [103]. The fixed position of the brain inside the skull also allows for methods not otherwise possible in visceral organs. Through a cranial window, blood vessels and the fate of individual tumor cells can be traced with great precision over multiple weeks. This enables a very precise in vivo depiction of cellular metastasis, although it is restricted to superficial metastasis. Furthermore, fruitful results can only be expected from experimental metastasis models (vascular tumor cell injection) as spontaneous metastasis models are too slow and unreliable [99,104,105]. The method is probably also limited in its widespread use as technical requirements are high and surgical expertise is necessary.
Advanced imaging techniques can be used to visualize aspects of the pre-metastatic niche. Emerging metastases can be detected by tissue-wide or whole-body in vivo luminescence and fluorescence, with new detection methods allowing for the highly sensitive detection of metastases, even though the resolution might not suffice for single cells or micro metastases [106,107,108,109,110,111]. Moreover, sophisticated tissue-clearing methods, combined with state-of-the-art open-top light-sheet microscopy for the rapid volumetric imaging of whole organs, has the potential to revolutionize the field [112,113,114,115]. Today, it is possible to clear and stain whole mouse bodies and create a pan-optic visualization down to single-cell resolution [116,117,118]. Additionally, expansion microscopy, the water-based polymer swelling of organs, can push histological tissue analysis to subcellular resolution with standard equipment [119,120,121]. Expanding on these imaging modalities, SPECT (single-photon-emission computed tomography)-based methods allow for the differential tracking of tumor cells and macromolecules in vivo and have been used to characterize tumor–immune interactions in a pre-metastatic setting [122,123,124,125]. Similarly, the method of choice to non-invasively image metastatic growths in vivo is high field magnetic resonance imaging (MRI) [106,110]. Here, ferritin heavy chain (FH1) tagging allows for the imaging of exosomes and other subcellular structures in pre-metastatic and metastatic niches [126]. Adapting these imaging methods opens up new dimensions for researching the early phases of the metastasis process.
Beyond imaging, next generation and single-cell sequencing have equipped researchers with powerful tools that can be used to understand complex cellular interactions. Both allow for the analysis of individual cells rather than bulk tissue. This provides a comprehensive view of the cellular compositions and molecular signatures of metastases and distant organ sites. As the PMN and MN are composed of a diverse range of cells, each with their own unique molecular profiles and functions, it is especially important to be able to work with high spatial and temporal resolutions [127,128,129]. Contrasting the high resolution of novel sequencing techniques are technical shortcomings that still limit data interpretation. For instance, a single sequencing run can only display a momentary snapshot of the expression profile and could possibly miss crucial abnormalities. In addition, the characterization of the genotype alone would probably not suffice to describe highly specialized microenvironments such as the pre-metastatic niche. In this regard, so called multiomic approaches have recently been established, combining transcriptional sequencing with protein and protein–interaction analyses [130]. Similarly, in a technological push to combine imaging and genomics, MERFISH, Visium and GeoMX now position relevant molecular signatures into spatial contexts, a process known as spatial transcriptomics [131,132,133,134]. Applying these revolutionary methods can provide unprecedented insights into the individual steps of the brain metastatic cascade, especially with the development of high-throughput methods that can uncover genetic processes even in fixed pathology samples and circulating tumor cells [130,135,136]. The possibility of cutting edge RNA sequencing penetrating even paraformaldehyde-fixed samples allows researchers to utilize large databases of patient samples that have been accumulated in pathology departments [136].

5.3. Conclusive Remarks

The experimental approaches outlined in this manuscript, applied in symphony with each other, will aid in gaining a more comprehensive understanding of the metastatic and pre-metastatic niches and their role in the development and progression of brain metastasis. The elusive nature of the pre-metastatic niche, especially in the cerebral environment, asks for sophisticated and well validated methods. The technological advancements discussed herein, namely, experimental high-resolution microscopy and multiomic approaches, represent powerful tools that are critical for the advancement of metastatic research. Given that no single method of investigation will resolve the pre-metastatic niche in both spatial and temporal dimensions simultaneously, an interdisciplinary effort to integrate evolving methods is essential. This will identify novel drug targets for bench-to-bedside clinical trials.

Author Contributions

Conceptualization, M.G., T.B. and M.C.; writing—original draft preparation, M.G., T.B., K.D.K., H.M. and L.S.; writing—review and editing, all authors; visualization, M.G. and L.S. All authors have read and agreed to the published version of the manuscript.

Funding

T.B., L.S., K.D.K., H.M. and M.C. were supported in part by the LOEWE Center Frankfurt Cancer Institute (FCI), funded by the Hessen State Ministry for Higher Education, Research, and the Arts [III L 5-519/03/03.001-(0015)].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

All figures were created with BioRender.com, accessed on 3 June 2023.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. J. Epidemiol. Glob. Health 2019, 9, 217–222. [Google Scholar] [CrossRef] [Green Version]
  2. Sacks, P.; Rahman, M. Epidemiology of Brain Metastases. Neurosurg. Clin. 2020, 31, 481–488. [Google Scholar] [CrossRef]
  3. Boire, A.; Brastianos, P.K.; Garzia, L.; Valiente, M. Brain Metastasis. Nat. Rev. Cancer 2020, 20, 4–11. [Google Scholar] [CrossRef] [PubMed]
  4. Kotecha, R.; Gondi, V.; Ahluwalia, M.S.; Brastianos, P.K.; Mehta, M.P. Recent Advances in Managing Brain Metastasis. F1000Research 2018, 7, F1000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Valiente, M.; Ahluwalia, M.S.; Boire, A.; Brastianos, P.K.; Goldberg, S.B.; Lee, E.Q.; Le Rhun, E.; Preusser, M.; Winkler, F.; Soffietti, R. The Evolving Landscape of Brain Metastasis. Trends Cancer 2018, 4, 176–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ganesh, K.; Massagué, J. Targeting Metastatic Cancer. Nat. Med. 2021, 27, 34–44. [Google Scholar] [CrossRef]
  7. Celià-Terrassa, T.; Kang, Y. Metastatic Niche Functions and Therapeutic Opportunities. Nat. Cell Biol. 2018, 20, 868–877. [Google Scholar] [CrossRef] [Green Version]
  8. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-Metastatic Niches: Organ-Specific Homes for Metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
  9. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  10. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  11. Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-Positive Haematopoietic Bone Marrow Progenitors Initiate the Pre-Metastatic Niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Paget, S. The Distribution of Secondary Growths in Cancer of the Breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef] [Green Version]
  13. Seubert, B.; Grünwald, B.; Kobuch, J.; Cui, H.; Schelter, F.; Schaten, S.; Siveke, J.T.; Lim, N.H.; Nagase, H.; Simonavicius, N.; et al. Tissue Inhibitor of Metalloproteinases (TIMP)-1 Creates a Premetastatic Niche in the Liver through SDF-1/CXCR4-Dependent Neutrophil Recruitment in Mice: Seubert, Grünwald et Al. Hepatology 2015, 61, 238–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles Released from Human Renal Cancer Stem Cells Stimulate Angiogenesis and Formation of Lung Premetastatic Niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [Green Version]
  15. Hiratsuka, S.; Ishibashi, S.; Tomita, T.; Watanabe, A.; Akashi-Takamura, S.; Murakami, M.; Kijima, H.; Miyake, K.; Aburatani, H.; Maru, Y. Primary Tumours Modulate Innate Immune Signalling to Create Pre-Metastatic Vascular Hyperpermeability Foci. Nat. Commun. 2013, 4, 1853. [Google Scholar] [CrossRef] [Green Version]
  16. Hiratsuka, S.; Goel, S.; Kamoun, W.S.; Maru, Y.; Fukumura, D.; Duda, D.G.; Jain, R.K. Endothelial Focal Adhesion Kinase Mediates Cancer Cell Homing to Discrete Regions of the Lungs via E-Selectin up-Regulation. Proc. Natl. Acad. Sci. USA 2011, 108, 3725–3730. [Google Scholar] [CrossRef] [Green Version]
  17. Yan, H.H.; Pickup, M.; Pang, Y.; Gorska, A.E.; Li, Z.; Chytil, A.; Geng, Y.; Gray, J.W.; Moses, H.L.; Yang, L. Gr-1+CD11b+ Myeloid Cells Tip the Balance of Immune Protection to Tumor Promotion in the Premetastatic Lung. Cancer Res. 2010, 70, 6139–6149. [Google Scholar] [CrossRef] [Green Version]
  18. Liu, Y.; Gu, Y.; Han, Y.; Zhang, Q.; Jiang, Z.; Zhang, X.; Huang, B.; Xu, X.; Zheng, J.; Cao, X. Tumor Exosomal RNAs Promote Lung Pre-Metastatic Niche Formation by Activating Alveolar Epithelial TLR3 to Recruit Neutrophils. Cancer Cell 2016, 30, 243–256. [Google Scholar] [CrossRef] [Green Version]
  19. Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-Secreted MiR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [Green Version]
  20. Kim, K.-J.; Kwon, S.-H.; Yun, J.-H.; Jeong, H.-S.; Kim, H.-R.; Lee, E.H.; Ye, S.-K.; Cho, C.-H. STAT3 Activation in Endothelial Cells Is Important for Tumor Metastasis via Increased Cell Adhesion Molecule Expression. Oncogene 2017, 36, 5445–5459. [Google Scholar] [CrossRef]
  21. Kang, S.-A.; Hasan, N.; Mann, A.P.; Zheng, W.; Zhao, L.; Morris, L.; Zhu, W.; Zhao, Y.D.; Suh, K.S.; Dooley, W.C.; et al. Blocking the Adhesion Cascade at the Premetastatic Niche for Prevention of Breast Cancer Metastasis. Mol. Ther. 2015, 23, 1044–1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wieland, E.; Rodriguez-Vita, J.; Liebler, S.S.; Mogler, C.; Moll, I.; Herberich, S.E.; Espinet, E.; Herpel, E.; Menuchin, A.; Chang-Claude, J.; et al. Endothelial Notch1 Activity Facilitates Metastasis. Cancer Cell 2017, 31, 355–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Coussens, L.M.; Werb, Z. Inflammation and Cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  24. van Weverwijk, A.; de Visser, K.E. Mechanisms Driving the Immunoregulatory Function of Cancer Cells. Nat. Rev. Cancer 2023, 23, 193–215. [Google Scholar] [CrossRef]
  25. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic Cancer Exosomes Initiate Pre-Metastatic Niche Formation in the Liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef] [PubMed]
  26. Bragado, P.; Estrada, Y.; Parikh, F.; Krause, S.; Capobianco, C.; Farina, H.G.; Schewe, D.M.; Aguirre-Ghiso, J.A. TGF-Β2 Dictates Disseminated Tumour Cell Fate in Target Organs through TGF-β-RIII and P38α/β Signalling. Nat. Cell Biol. 2013, 15, 1351–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ghajar, C.M.; Peinado, H.; Mori, H.; Matei, I.R.; Evason, K.J.; Brazier, H.; Almeida, D.; Koller, A.; Hajjar, K.A.; Stainier, D.Y.R.; et al. The Perivascular Niche Regulates Breast Tumor Dormancy. Nat. Cell Biol. 2013, 15, 807–817. [Google Scholar] [CrossRef]
  28. El-Kenawi, A.; Hänggi, K.; Ruffell, B. The Immune Microenvironment and Cancer Metastasis. Cold Spring Harb. Perspect. Med. 2020, 10, a037424. [Google Scholar] [CrossRef]
  29. Xiong, S.; Dong, L.; Cheng, L. Neutrophils in Cancer Carcinogenesis and Metastasis. J. Hematol. Oncol. 2021, 14, 173. [Google Scholar] [CrossRef]
  30. Kitamura, T.; Qian, B.-Z.; Pollard, J.W. Immune Cell Promotion of Metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef] [Green Version]
  31. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific Recruitment of Regulatory T Cells in Ovarian Carcinoma Fosters Immune Privilege and Predicts Reduced Survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
  32. Kryczek, I.; Zou, L.; Rodriguez, P.; Zhu, G.; Wei, S.; Mottram, P.; Brumlik, M.; Cheng, P.; Curiel, T.; Myers, L.; et al. B7-H4 Expression Identifies a Novel Suppressive Macrophage Population in Human Ovarian Carcinoma. J. Exp. Med. 2006, 203, 871–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated Monocytes in Peritumoral Stroma of Hepatocellular Carcinoma Foster Immune Privilege and Disease Progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sanjabi, S.; Zenewicz, L.A.; Kamanaka, M.; Flavell, R.A. Anti-Inflammatory and pro-Inflammatory Roles of TGF-Beta, IL-10, and IL-22 in Immunity and Autoimmunity. Curr. Opin. Pharmacol. 2009, 9, 447–453. [Google Scholar] [CrossRef] [Green Version]
  35. Kfoury, Y.; Baryawno, N.; Severe, N.; Mei, S.; Gustafsson, K.; Hirz, T.; Brouse, T.; Scadden, E.W.; Igolkina, A.A.; Kokkaliaris, K.; et al. Human Prostate Cancer Bone Metastases Have an Actionable Immunosuppressive Microenvironment. Cancer Cell 2021, 39, 1464–1478.e8. [Google Scholar] [CrossRef]
  36. Shibuya, T.; Kamiyama, A.; Sawada, H.; Kikuchi, K.; Maruyama, M.; Sawado, R.; Ikeda, N.; Asano, K.; Kurotaki, D.; Tamura, T.; et al. Immunoregulatory Monocyte Subset Promotes Metastasis Associated with Therapeutic Intervention for Primary Tumor. Front. Immunol. 2021, 12, 663115. [Google Scholar] [CrossRef]
  37. Condamine, T.; Ramachandran, I.; Youn, J.-I.; Gabrilovich, D.I. Regulation of Tumor Metastasis by Myeloid-Derived Suppressor Cells. Annu. Rev. Med. 2015, 66, 97–110. [Google Scholar] [CrossRef] [Green Version]
  38. Dong, Q.; Liu, X.; Cheng, K.; Sheng, J.; Kong, J.; Liu, T. Pre-Metastatic Niche Formation in Different Organs Induced by Tumor Extracellular Vesicles. Front. Cell Dev. Biol. 2021, 9, 733627. [Google Scholar] [CrossRef]
  39. Tămaș, F.; Bălașa, R.; Manu, D.; Gyorki, G.; Chinezu, R.; Tămaș, C.; Bălașa, A. The Importance of Small Extracellular Vesicles in the Cerebral Metastatic Process. Int. J. Mol. Sci. 2022, 23, 1449. [Google Scholar] [CrossRef]
  40. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour Exosome Integrins Determine Organotropic Metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
  41. Webber, J.P.; Spary, L.K.; Sanders, A.J.; Chowdhury, R.; Jiang, W.G.; Steadman, R.; Wymant, J.; Jones, A.T.; Kynaston, H.; Mason, M.D.; et al. Differentiation of Tumour-Promoting Stromal Myofibroblasts by Cancer Exosomes. Oncogene 2015, 34, 290–302. [Google Scholar] [CrossRef] [PubMed]
  42. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-Metastatic Phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Blanc, L.; Vidal, M. New Insights into the Function of Rab GTPases in the Context of Exosomal Secretion. Small GTPases 2017, 9, 95–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Ostenfeld, M.S.; Jeppesen, D.K.; Laurberg, J.R.; Boysen, A.T.; Bramsen, J.B.; Primdal-Bengtson, B.; Hendrix, A.; Lamy, P.; Dagnaes-Hansen, F.; Rasmussen, M.H.; et al. Cellular Disposal of MiR23b by RAB27-Dependent Exosome Release Is Linked to Acquisition of Metastatic Properties. Cancer Res. 2014, 74, 5758–5771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal MiR-135b Shed from Hypoxic Multiple Myeloma Cells Enhances Angiogenesis by Targeting Factor-Inhibiting HIF-1. Blood 2014, 124, 3748–3757. [Google Scholar] [CrossRef] [Green Version]
  46. Liu, Y.; Luo, F.; Wang, B.; Li, H.; Xu, Y.; Liu, X.; Shi, L.; Lu, X.; Xu, W.; Lu, L.; et al. STAT3-Regulated Exosomal MiR-21 Promotes Angiogenesis and Is Involved in Neoplastic Processes of Transformed Human Bronchial Epithelial Cells. Cancer Lett. 2016, 370, 125–135. [Google Scholar] [CrossRef]
  47. Gong, M.; Yu, B.; Wang, J.; Wang, Y.; Liu, M.; Paul, C.; Millard, R.W.; Xiao, D.-S.; Ashraf, M.; Xu, M. Mesenchymal Stem Cells Release Exosomes That Transfer MiRNAs to Endothelial Cells and Promote Angiogenesis. Oncotarget 2017, 8, 45200–45212. [Google Scholar] [CrossRef] [Green Version]
  48. Liu, S.J.; Dang, H.X.; Lim, D.A.; Feng, F.Y.; Maher, C.A. Long Noncoding RNAs in Cancer Metastasis. Nat. Rev. Cancer 2021, 21, 446–460. [Google Scholar] [CrossRef]
  49. Xie, Y.; Dang, W.; Zhang, S.; Yue, W.; Yang, L.; Zhai, X.; Yan, Q.; Lu, J. The Role of Exosomal Noncoding RNAs in Cancer. Mol. Cancer 2019, 18, 37. [Google Scholar] [CrossRef] [Green Version]
  50. Diederichs, S.; Bartsch, L.; Berkmann, J.C.; Fröse, K.; Heitmann, J.; Hoppe, C.; Iggena, D.; Jazmati, D.; Karschnia, P.; Linsenmeier, M.; et al. The Dark Matter of the Cancer Genome: Aberrations in Regulatory Elements, Untranslated Regions, Splice Sites, Non-Coding RNA and Synonymous Mutations. EMBO Mol. Med. 2016, 8, 442–457. [Google Scholar] [CrossRef]
  51. Sun, Z.; Yang, S.; Zhou, Q.; Wang, G.; Song, J.; Li, Z.; Zhang, Z.; Xu, J.; Xia, K.; Chang, Y.; et al. Emerging Role of Exosome-Derived Long Non-Coding RNAs in Tumor Microenvironment. Mol. Cancer 2018, 17, 82. [Google Scholar] [CrossRef] [PubMed]
  52. Garzon, R.; Marcucci, G.; Croce, C.M. Targeting MicroRNAs in Cancer: Rationale, Strategies and Challenges. Nat. Rev. Drug Discov. 2010, 9, 775–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Budakoti, M.; Panwar, A.S.; Molpa, D.; Singh, R.K.; Büsselberg, D.; Mishra, A.P.; Coutinho, H.D.M.; Nigam, M. Micro-RNA: The Darkhorse of Cancer. Cell. Signal. 2021, 83, 109995. [Google Scholar] [CrossRef] [PubMed]
  54. Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and Functional Features of Central Nervous System Lymphatic Vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Winkler, F. The Brain Metastatic Niche. J. Mol. Med. 2015, 93, 1213–1220. [Google Scholar] [CrossRef]
  56. Nicolson, G.L.; Menter, D.G.; Herrmann, J.; Cavanaugh, P.; Jia, L.; Hamada, J.; Yun, Z.; Nakajima, M.; Marchetti, D. Tumor Metastasis to Brain: Role of Endothelial Cells, Neurotrophins, and Paracrine Growth Factors. Crit. Rev. Oncog. 1994, 5, 451–471. [Google Scholar] [CrossRef]
  57. Robador, J.R.; Feinauer, M.J.; Schneider, S.W.; Mayer, F.T.; Gorzelanny, C.; Sacharow, A.; Liu, X.; Berghoff, A.; Brehm, M.A.; Hirsch, D.; et al. Involvement of Platelet-Derived VWF in Metastatic Growth of Melanoma in the Brain. Neurooncol. Adv. 2021, 3, vdab175. [Google Scholar] [CrossRef]
  58. Schwartz, H.; Blacher, E.; Amer, M.; Livneh, N.; Abramovitz, L.; Klein, A.; Ben-Shushan, D.; Soffer, S.; Blazquez, R.; Barrantes-Freer, A.; et al. Incipient Melanoma Brain Metastases Instigate Astrogliosis and Neuroinflammation. Cancer Res. 2016, 76, 4359–4371. [Google Scholar] [CrossRef] [Green Version]
  59. Klein, A.; Schwartz, H.; Sagi-Assif, O.; Meshel, T.; Izraely, S.; Ben Menachem, S.; Bengaiev, R.; Ben-Shmuel, A.; Nahmias, C.; Couraud, P.-O.; et al. Astrocytes Facilitate Melanoma Brain Metastasis via Secretion of IL-23. J. Pathol. 2015, 236, 116–127. [Google Scholar] [CrossRef]
  60. Xing, F.; Liu, Y.; Sharma, S.; Wu, K.; Chan, M.D.; Lo, H.-W.; Carpenter, R.L.; Metheny-Barlow, L.J.; Zhou, X.; Qasem, S.A.; et al. Activation of the C-Met Pathway Mobilizes an Inflammatory Network in the Brain Microenvironment to Promote Brain Metastasis of Breast Cancer. Cancer Res. 2016, 76, 4970–4980. [Google Scholar] [CrossRef] [Green Version]
  61. Doron, H.; Pukrop, T.; Erez, N. A Blazing Landscape: Neuroinflammation Shapes Brain Metastasis. Cancer Res. 2019, 79, 423–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Burda, J.E.; Sofroniew, M.V. Reactive Gliosis and the Multicellular Response to CNS Damage and Disease. Neuron 2014, 81, 229–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Rodrigues, G.; Hoshino, A.; Kenific, C.M.; Matei, I.R.; Steiner, L.; Freitas, D.; Kim, H.S.; Oxley, P.R.; Scandariato, I.; Casanova-Salas, I.; et al. Tumour Exosomal CEMIP Protein Promotes Cancer Cell Colonization in Brain Metastasis. Nat. Cell Biol. 2019, 21, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  64. Gril, B.; Paranjape, A.N.; Woditschka, S.; Hua, E.; Dolan, E.L.; Hanson, J.; Wu, X.; Kloc, W.; Izycka-Swieszewska, E.; Duchnowska, R.; et al. Reactive Astrocytic S1P3 Signaling Modulates the Blood–Tumor Barrier in Brain Metastases. Nat. Commun. 2018, 9, 2705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sevenich, L.; Bowman, R.L.; Mason, S.D.; Quail, D.F.; Rapaport, F.; Elie, B.T.; Brogi, E.; Brastianos, P.K.; Hahn, W.C.; Holsinger, L.J.; et al. Analysis of Tumour- and Stroma-Supplied Proteolytic Networks Reveals a Brain-Metastasis-Promoting Role for Cathepsin S. Nat. Cell Biol. 2014, 16, 876–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Klemm, F.; Möckl, A.; Salamero-Boix, A.; Alekseeva, T.; Schäffer, A.; Schulz, M.; Niesel, K.; Maas, R.R.; Groth, M.; Elie, B.T.; et al. Compensatory CSF2-Driven Macrophage Activation Promotes Adaptive Resistance to CSF1R Inhibition in Breast-to-Brain Metastasis. Nat. Cancer 2021, 2, 1086–1101. [Google Scholar] [CrossRef]
  67. Chuang, H.-N.; van Rossum, D.; Sieger, D.; Siam, L.; Klemm, F.; Bleckmann, A.; Bayerlová, M.; Farhat, K.; Scheffel, J.; Schulz, M.; et al. Carcinoma Cells Misuse the Host Tissue Damage Response to Invade the Brain. Glia 2013, 61, 1331–1346. [Google Scholar] [CrossRef] [Green Version]
  68. Izraely, S.; Ben-Menachem, S.; Sagi-Assif, O.; Telerman, A.; Zubrilov, I.; Ashkenazi, O.; Meshel, T.; Maman, S.; Orozco, J.I.J.; Salomon, M.P.; et al. The Metastatic Microenvironment: Melanoma-Microglia Cross-Talk Promotes the Malignant Phenotype of Melanoma Cells. Int. J. Cancer 2019, 144, 802–817. [Google Scholar] [CrossRef] [Green Version]
  69. Gong, X.; Hou, Z.; Endsley, M.P.; Gronseth, E.I.; Rarick, K.R.; Jorns, J.M.; Yang, Q.; Du, Z.; Yan, K.; Bordas, M.L.; et al. Interaction of Tumor Cells and Astrocytes Promotes Breast Cancer Brain Metastases through TGF-Β2/ANGPTL4 Axes. NPJ Precis. Oncol. 2019, 3, 24. [Google Scholar] [CrossRef] [Green Version]
  70. Doron, H.; Amer, M.; Ershaid, N.; Blazquez, R.; Shani, O.; Lahav, T.G.; Cohen, N.; Adler, O.; Hakim, Z.; Pozzi, S.; et al. Inflammatory Activation of Astrocytes Facilitates Melanoma Brain Tropism via the CXCL10-CXCR3 Signaling Axis. Cell Rep. 2019, 28, 1785–1798.e6. [Google Scholar] [CrossRef] [Green Version]
  71. Valiente, M.; Obenauf, A.C.; Jin, X.; Chen, Q.; Zhang, X.H.-F.; Lee, D.J.; Chaft, J.E.; Kris, M.G.; Huse, J.T.; Brogi, E.; et al. Serpins Promote Cancer Cell Survival and Vascular Co-Option in Brain Metastasis. Cell 2014, 156, 1002–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Berghoff, A.S.; Lassmann, H.; Preusser, M.; Höftberger, R. Characterization of the Inflammatory Response to Solid Cancer Metastases in the Human Brain. Clin. Exp. Metastasis 2013, 30, 69–81. [Google Scholar] [CrossRef] [PubMed]
  73. Berghoff, A.S.; Fuchs, E.; Ricken, G.; Mlecnik, B.; Bindea, G.; Spanberger, T.; Hackl, M.; Widhalm, G.; Dieckmann, K.; Prayer, D.; et al. Density of Tumor-Infiltrating Lymphocytes Correlates with Extent of Brain Edema and Overall Survival Time in Patients with Brain Metastases. OncoImmunology 2016, 5, e1057388. [Google Scholar] [CrossRef] [PubMed]
  74. Neman, J.; Choy, C.; Kowolik, C.M.; Anderson, A.; Duenas, V.J.; Waliany, S.; Chen, B.T.; Chen, M.Y.; Jandial, R. Co-Evolution of Breast-to-Brain Metastasis and Neural Progenitor Cells. Clin. Exp. Metastasis 2013, 30, 753–768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Li, B.; Wang, C.; Zhang, Y.; Zhao, X.Y.; Huang, B.; Wu, P.F.; Li, Q.; Li, H.; Liu, Y.S.; Cao, L.Y.; et al. Elevated PLGF Contributes to Small-Cell Lung Cancer Brain Metastasis. Oncogene 2013, 32, 2952–2962. [Google Scholar] [CrossRef] [Green Version]
  76. Yang, Y.; Cao, Y. The Impact of VEGF on Cancer Metastasis and Systemic Disease. Semin. Cancer Biol. 2022, 86, 251–261. [Google Scholar] [CrossRef]
  77. Lorger, M.; Krueger, J.S.; O’Neal, M.; Staflin, K.; Felding-Habermann, B. Activation of Tumor Cell Integrin Avβ3 Controls Angiogenesis and Metastatic Growth in the Brain. Proc. Natl. Acad. Sci. USA 2009, 106, 10666–10671. [Google Scholar] [CrossRef] [Green Version]
  78. Fan, J.; Cai, B.; Zeng, M.; Hao, Y.; Giancotti, F.G.; Fu, B.M. Integrin Β4 Signaling Promotes Mammary Tumor Cell Adhesion to Brain Microvascular Endothelium by Inducing ErbB2-Mediated Secretion of VEGF. Ann. Biomed. Eng. 2011, 39, 2223–2241. [Google Scholar] [CrossRef] [Green Version]
  79. Treps, L.; Perret, R.; Edmond, S.; Ricard, D.; Gavard, J. Glioblastoma Stem-like Cells Secrete the pro-Angiogenic VEGF-A Factor in Extracellular Vesicles. J. Extracell. Vesicles 2017, 6, 1359479. [Google Scholar] [CrossRef] [Green Version]
  80. Contreras-Zárate, M.J.; Cittelly, D.M. Sex Steroid Hormone Function in the Brain Niche: Implications for Brain Metastatic Colonization and Progression. Cancer Rep. 2022, 5, e1241. [Google Scholar] [CrossRef]
  81. Wu, D.; Deng, S.; Li, L.; Liu, T.; Zhang, T.; Li, J.; Yu, Y.; Xu, Y. TGF-Β1-Mediated Exosomal Lnc-MMP2-2 Increases Blood–Brain Barrier Permeability via the MiRNA-1207-5p/EPB41L5 Axis to Promote Non-Small Cell Lung Cancer Brain Metastasis. Cell Death Dis. 2021, 12, 721. [Google Scholar] [CrossRef] [PubMed]
  82. Lu, Y.; Chen, L.; Li, L.; Cao, Y. Exosomes Derived from Brain Metastatic Breast Cancer Cells Destroy the Blood-Brain Barrier by Carrying LncRNA GS1-600G8.5. BioMed Res. Int. 2020, 2020, e7461727. [Google Scholar] [CrossRef] [PubMed]
  83. Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.F.; Li, S.; Chin, A.R.; et al. Breast-Cancer-Secreted MiR-122 Reprograms Glucose Metabolism in Premetastatic Niche to Promote Metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kanchan, R.K.; Siddiqui, J.A.; Mahapatra, S.; Batra, S.K.; Nasser, M.W. MicroRNAs Orchestrate Pathophysiology of Breast Cancer Brain Metastasis: Advances in Therapy. Mol. Cancer 2020, 19, 29. [Google Scholar] [CrossRef] [Green Version]
  85. Alsidawi, S.; Malek, E.; Driscoll, J.J. MicroRNAs in Brain Metastases: Potential Role as Diagnostics and Therapeutics. Int. J. Mol. Sci. 2014, 15, 10508–10526. [Google Scholar] [CrossRef] [Green Version]
  86. Oliveira, F.D.; Castanho, M.A.R.B.; Neves, V. Exosomes and Brain Metastases: A Review on Their Role and Potential Applications. Int. J. Mol. Sci. 2021, 22, 10899. [Google Scholar] [CrossRef]
  87. Zhang, L.; Zhang, S.; Yao, J.; Lowery, F.J.; Zhang, Q.; Huang, W.-C.; Li, P.; Li, M.; Wang, X.; Zhang, C.; et al. Microenvironment-Induced PTEN Loss by Exosomal MicroRNA Primes Brain Metastasis Outgrowth. Nature 2015, 527, 100–104. [Google Scholar] [CrossRef] [Green Version]
  88. Connolly, M.K.; Mallen-St. Clair, J.; Bedrosian, A.S.; Malhotra, A.; Vera, V.; Ibrahim, J.; Henning, J.; Pachter, H.L.; Bar-Sagi, D.; Frey, A.B.; et al. Distinct Populations of Metastases-Enabling Myeloid Cells Expand in the Liver of Mice Harboring Invasive and Preinvasive Intra-Abdominal Tumor. J. Leukoc. Biol. 2010, 87, 713–725. [Google Scholar] [CrossRef] [Green Version]
  89. Toh, B.; Wang, X.; Keeble, J.; Sim, W.J.; Khoo, K.; Wong, W.-C.; Kato, M.; Prevost-Blondel, A.; Thiery, J.-P.; Abastado, J.-P. Mesenchymal Transition and Dissemination of Cancer Cells Is Driven by Myeloid-Derived Suppressor Cells Infiltrating the Primary Tumor. PLoS Biol. 2011, 9, e1001162. [Google Scholar] [CrossRef] [Green Version]
  90. Hiratsuka, S.; Watanabe, A.; Sakurai, Y.; Akashi-Takamura, S.; Ishibashi, S.; Miyake, K.; Shibuya, M.; Akira, S.; Aburatani, H.; Maru, Y. The S100A8–Serum Amyloid A3–TLR4 Paracrine Cascade Establishes a Pre-Metastatic Phase. Nat. Cell Biol. 2008, 10, 1349–1355. [Google Scholar] [CrossRef]
  91. Gil-Bernabé, A.M.; Ferjancic, S.; Tlalka, M.; Zhao, L.; Allen, P.D.; Im, J.H.; Watson, K.; Hill, S.A.; Amirkhosravi, A.; Francis, J.L.; et al. Recruitment of Monocytes/Macrophages by Tissue Factor-Mediated Coagulation Is Essential for Metastatic Cell Survival and Premetastatic Niche Establishment in Mice. Blood 2012, 119, 3164–3175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Seike, T.; Fujita, K.; Yamakawa, Y.; Kido, M.A.; Takiguchi, S.; Teramoto, N.; Iguchi, H.; Noda, M. Interaction between Lung Cancer Cells and Astrocytes via Specific Inflammatory Cytokines in the Microenvironment of Brain Metastasis. Clin. Exp. Metastasis 2011, 28, 13–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Qian, B.-Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 Recruits Inflammatory Monocytes to Facilitate Breast-Tumour Metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Qian, B.; Deng, Y.; Im, J.H.; Muschel, R.J.; Zou, Y.; Li, J.; Lang, R.A.; Pollard, J.W. A Distinct Macrophage Population Mediates Metastatic Breast Cancer Cell Extravasation, Establishment and Growth. PLoS ONE 2009, 4, e6562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Camerer, E.; Qazi, A.A.; Duong, D.N.; Cornelissen, I.; Advincula, R.; Coughlin, S.R. Platelets, Protease-Activated Receptors, and Fibrinogen in Hematogenous Metastasis. Blood 2004, 104, 397–401. [Google Scholar] [CrossRef]
  96. Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Jirousková, M.; Degen, J.L. Platelets and Fibrin(Ogen) Increase Metastatic Potential by Impeding Natural Killer Cell-Mediated Elimination of Tumor Cells. Blood 2005, 105, 178–185. [Google Scholar] [CrossRef] [Green Version]
  97. Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Hu, Z.; Barney, K.A.; Degen, J.L. Tumor Cell-Associated Tissue Factor and Circulating Hemostatic Factors Cooperate to Increase Metastatic Potential through Natural Killer Cell-Dependent and-Independent Mechanisms. Blood 2007, 110, 133–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Strilic, B.; Yang, L.; Albarrán-Juárez, J.; Wachsmuth, L.; Han, K.; Müller, U.C.; Pasparakis, M.; Offermanns, S. Tumour-Cell-Induced Endothelial Cell Necroptosis via Death Receptor 6 Promotes Metastasis. Nature 2016, 536, 215–218. [Google Scholar] [CrossRef]
  99. Kienast, Y.; von Baumgarten, L.; Fuhrmann, M.; Klinkert, W.E.F.; Goldbrunner, R.; Herms, J.; Winkler, F. Real-Time Imaging Reveals the Single Steps of Brain Metastasis Formation. Nat. Med. 2010, 16, 116–122. [Google Scholar] [CrossRef]
  100. Miarka, L.; Valiente, M. Animal Models of Brain Metastasis. Neuro-Oncol. Adv. 2021, 3, v144–v156. [Google Scholar] [CrossRef] [PubMed]
  101. Valiente, M.; Van Swearingen, A.E.D.; Anders, C.K.; Bairoch, A.; Boire, A.; Bos, P.D.; Cittelly, D.M.; Erez, N.; Ferraro, G.B.; Fukumura, D.; et al. Brain Metastasis Cell Lines Panel: A Public Resource of Organotropic Cell Lines. Cancer Res. 2020, 80, 4314–4323. [Google Scholar] [CrossRef] [PubMed]
  102. Broggini, T.; Piffko, A.; Hoffmann, C.J.; Harms, C.; Vajkoczy, P.; Czabanka, M. Passive Entrapment of Tumor Cells Determines Metastatic Dissemination to Spinal Bone and Other Osseous Tissues. PLoS ONE 2016, 11, e0162540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Entenberg, D.; Oktay, M.H.; Condeelis, J.S. Intravital Imaging to Study Cancer Progression and Metastasis. Nat. Rev. Cancer 2023, 23, 25–42. [Google Scholar] [CrossRef]
  104. Lorger, M.; Felding-Habermann, B. Capturing Changes in the Brain Microenvironment during Initial Steps of Breast Cancer Brain Metastasis. Am. J. Pathol. 2010, 176, 2958–2971. [Google Scholar] [CrossRef]
  105. Feinauer, M.J.; Schneider, S.W.; Berghoff, A.S.; Robador, J.R.; Tehranian, C.; Karreman, M.A.; Venkataramani, V.; Solecki, G.; Grosch, J.K.; Gunkel, K.; et al. Local Blood Coagulation Drives Cancer Cell Arrest and Brain Metastasis in a Mouse Model. Blood 2021, 137, 1219–1232. [Google Scholar] [CrossRef] [PubMed]
  106. Broggini, T.; Czabanka, M.; Piffko, A.; Harms, C.; Hoffmann, C.; Mrowka, R.; Wenke, F.; Deutsch, U.; Grötzinger, C.; Vajkoczy, P. ICAM1 Depletion Reduces Spinal Metastasis Formation in Vivo and Improves Neurological Outcome. Eur. Spine J. 2015, 24, 2173–2181. [Google Scholar] [CrossRef] [PubMed]
  107. Zheng, H.; Yuan, C.; Cai, J.; Pu, W.; Wu, P.; Li, C.; Li, G.; Zhang, Y.; Zhang, J.; Guo, J.; et al. Early Diagnosis of Breast Cancer Lung Metastasis by Nanoprobe-Based Luminescence Imaging of the Pre-Metastatic Niche. J. Nanobiotechnol. 2022, 20, 134. [Google Scholar] [CrossRef] [PubMed]
  108. Hayashi, M.; Takai, J.; Yu, L.; Motohashi, H.; Moriguchi, T.; Yamamoto, M. Whole-Body In Vivo Monitoring of Inflammatory Diseases Exploiting Human Interleukin 6-Luciferase Transgenic Mice. Mol. Cell. Biol. 2015, 35, 3590–3601. [Google Scholar] [CrossRef] [Green Version]
  109. Hoffman, R.M. Advantages of Multi-Color Fluorescent Proteins for Whole-Body and in Vivo Cellular Imaging. J. Biomed. Opt. 2005, 10, 41202. [Google Scholar] [CrossRef]
  110. Broggini, T.; Piffko, A.; Hoffmann, C.J.; Ghori, A.; Harms, C.; Adams, R.H.; Vajkoczy, P.; Czabanka, M. Ephrin-B2–EphB4 Communication Mediates Tumor–Endothelial Cell Interactions during Hematogenous Spread to Spinal Bone in a Melanoma Metastasis Model. Oncogene 2020, 39, 7063–7075. [Google Scholar] [CrossRef]
  111. Kokkaliaris, K.D.; Kunz, L.; Cabezas-Wallscheid, N.; Christodoulou, C.; Renders, S.; Camargo, F.; Trumpp, A.; Scadden, D.T.; Schroeder, T. Adult Blood Stem Cell Localization Reflects the Abundance of Reported Bone Marrow Niche Cell Types and Their Combinations. Blood 2020, 136, 2296–2307. [Google Scholar] [CrossRef] [PubMed]
  112. Dodt, H.-U.; Leischner, U.; Schierloh, A.; Jährling, N.; Mauch, C.P.; Deininger, K.; Deussing, J.M.; Eder, M.; Zieglgänsberger, W.; Becker, K. Ultramicroscopy: Three-Dimensional Visualization of Neuronal Networks in the Whole Mouse Brain. Nat. Methods 2007, 4, 331–336. [Google Scholar] [CrossRef] [PubMed]
  113. Ertürk, A.; Becker, K.; Jährling, N.; Mauch, C.P.; Hojer, C.D.; Egen, J.G.; Hellal, F.; Bradke, F.; Sheng, M.; Dodt, H.-U. Three-Dimensional Imaging of Solvent-Cleared Organs Using 3DISCO. Nat. Protoc. 2012, 7, 1983–1995. [Google Scholar] [CrossRef] [PubMed]
  114. Renier, N.; Wu, Z.; Simon, D.J.; Yang, J.; Ariel, P.; Tessier-Lavigne, M. IDISCO: A Simple, Rapid Method to Immunolabel Large Tissue Samples for Volume Imaging. Cell 2014, 159, 896–910. [Google Scholar] [CrossRef] [Green Version]
  115. Glaser, A.K.; Bishop, K.W.; Barner, L.A.; Susaki, E.A.; Kubota, S.I.; Gao, G.; Serafin, R.B.; Balaram, P.; Turschak, E.; Nicovich, P.R.; et al. A Hybrid Open-Top Light-Sheet Microscope for Versatile Multi-Scale Imaging of Cleared Tissues. Nat. Methods 2022, 19, 613–619. [Google Scholar] [CrossRef]
  116. Yang, B.; Treweek, J.B.; Kulkarni, R.P.; Deverman, B.E.; Chen, C.-K.; Lubeck, E.; Shah, S.; Cai, L.; Gradinaru, V. Single-Cell Phenotyping within Transparent Intact Tissue through Whole-Body Clearing. Cell 2014, 158, 945–958. [Google Scholar] [CrossRef] [Green Version]
  117. Cai, R.; Pan, C.; Ghasemigharagoz, A.; Todorov, M.I.; Förstera, B.; Zhao, S.; Bhatia, H.S.; Parra-Damas, A.; Mrowka, L.; Theodorou, D.; et al. Panoptic Imaging of Transparent Mice Reveals Whole-Body Neuronal Projections and Skull-Meninges Connections. Nat. Neurosci. 2019, 22, 317–327. [Google Scholar] [CrossRef] [PubMed]
  118. Mai, H.; Luo, J.; Hoeher, L.; Al-Maskari, R.; Horvath, I.; Paetzold, J.C.; Todorov, M.; Hellal, F.; Ertürk, A. Whole Mouse Body Histology Using Standard IgG Antibodies. bioRxiv 2023. [Google Scholar] [CrossRef]
  119. Wassie, A.T.; Zhao, Y.; Boyden, E.S. Expansion Microscopy: Principles and Uses in Biological Research. Nat. Methods 2019, 16, 33–41. [Google Scholar] [CrossRef]
  120. Chen, F.; Tillberg, P.W.; Boyden, E.S. Expansion Microscopy. Science 2015, 347, 543–548. [Google Scholar] [CrossRef] [Green Version]
  121. Gallagher, B.R.; Zhao, Y. Expansion Microscopy: A Powerful Nanoscale Imaging Tool for Neuroscientists. Neurobiol. Dis. 2021, 154, 105362. [Google Scholar] [CrossRef] [PubMed]
  122. Perrin, J.; Capitao, M.; Mougin-Degraef, M.; Guérard, F.; Faivre-Chauvet, A.; Rbah-Vidal, L.; Gaschet, J.; Guilloux, Y.; Kraeber-Bodéré, F.; Chérel, M.; et al. Cell Tracking in Cancer Immunotherapy. Front. Med. 2020, 7, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Eisenblaetter, M.; Flores-Borja, F.; Lee, J.J.; Wefers, C.; Smith, H.; Hueting, R.; Cooper, M.S.; Blower, P.J.; Patel, D.; Rodriguez-Justo, M.; et al. Visualization of Tumor-Immune Interaction-Target-Specific Imaging of S100A8/A9 Reveals Pre-Metastatic Niche Establishment. Theranostics 2017, 7, 2392–2401. [Google Scholar] [CrossRef] [Green Version]
  124. Lobaz, V.; Konefał, R.; Pánek, J.; Vlk, M.; Kozempel, J.; Petřík, M.; Novy, Z.; Gurská, S.; Znojek, P.; Štěpánek, P.; et al. In Situ In Vivo Radiolabeling of Polymer-Coated Hydroxyapatite Nanoparticles to Track Their Biodistribution in Mice. Colloids Surf. B Biointerfaces 2019, 179, 143–152. [Google Scholar] [CrossRef]
  125. Olmeda, D.; Cerezo-Wallis, D.; Riveiro-Falkenbach, E.; Pennacchi, P.C.; Contreras-Alcalde, M.; Ibarz, N.; Cifdaloz, M.; Catena, X.; Calvo, T.G.; Cañón, E.; et al. Whole-Body Imaging of Lymphovascular Niches Identifies Pre-Metastatic Roles of Midkine. Nature 2017, 546, 676–680. [Google Scholar] [CrossRef] [PubMed]
  126. Liu, T.; Li, Z.; Li, X.; Zhao, R.; Wei, X.; Wang, Z.; Xin, S.X. In Vivo Visualization of Murine Melanoma Cells B16-Derived Exosomes through Magnetic Resonance Imaging. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2022, 1866, 130062. [Google Scholar] [CrossRef]
  127. Hwang, B.; Lee, J.H.; Bang, D. Single-Cell RNA Sequencing Technologies and Bioinformatics Pipelines. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Nam, A.S.; Chaligne, R.; Landau, D.A. Integrating Genetic and Non-Genetic Determinants of Cancer Evolution by Single-Cell Multi-Omics. Nat. Rev. Genet. 2021, 22, 3–18. [Google Scholar] [CrossRef]
  129. Kim, N.; Kim, H.K.; Lee, K.; Hong, Y.; Cho, J.H.; Choi, J.W.; Lee, J.-I.; Suh, Y.-L.; Ku, B.M.; Eum, H.H.; et al. Single-Cell RNA Sequencing Demonstrates the Molecular and Cellular Reprogramming of Metastatic Lung Adenocarcinoma. Nat. Commun. 2020, 11, 2285. [Google Scholar] [CrossRef]
  130. Lei, Y.; Tang, R.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Yu, X.; Shi, S. Applications of Single-Cell Sequencing in Cancer Research: Progress and Perspectives. J. Hematol. Oncol. 2021, 14, 91. [Google Scholar] [CrossRef]
  131. Rao, A.; Barkley, D.; França, G.S.; Yanai, I. Exploring Tissue Architecture Using Spatial Transcriptomics. Nature 2021, 596, 211–220. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, K.H.; Boettiger, A.N.; Moffitt, J.R.; Wang, S.; Zhuang, X. RNA Imaging. Spatially Resolved, Highly Multiplexed RNA Profiling in Single Cells. Science 2015, 348, aaa6090. [Google Scholar] [CrossRef] [Green Version]
  133. Ståhl, P.L.; Salmén, F.; Vickovic, S.; Lundmark, A.; Navarro, J.F.; Magnusson, J.; Giacomello, S.; Asp, M.; Westholm, J.O.; Huss, M.; et al. Visualization and Analysis of Gene Expression in Tissue Sections by Spatial Transcriptomics. Science 2016, 353, 78–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Merritt, C.R.; Ong, G.T.; Church, S.E.; Barker, K.; Danaher, P.; Geiss, G.; Hoang, M.; Jung, J.; Liang, Y.; McKay-Fleisch, J.; et al. Multiplex Digital Spatial Profiling of Proteins and RNA in Fixed Tissue. Nat. Biotechnol. 2020, 38, 586–599. [Google Scholar] [CrossRef] [PubMed]
  135. Han, Y.; Wang, D.; Peng, L.; Huang, T.; He, X.; Wang, J.; Ou, C. Single-Cell Sequencing: A Promising Approach for Uncovering the Mechanisms of Tumor Metastasis. J. Hematol. Oncol. 2022, 15, 59. [Google Scholar] [CrossRef]
  136. Phan, H.V.; van Gent, M.; Drayman, N.; Basu, A.; Gack, M.U.; Tay, S. High-Throughput RNA Sequencing of Paraformaldehyde-Fixed Single Cells. Nat. Commun. 2021, 12, 5636. [Google Scholar] [CrossRef]
Figure 1. Concept drawing of the pre-metastatic niche. The primary tumor releases signals via exosomes and soluble factors to distant organs, preparing them for metastasis. Most of the distant modulation takes place in and around the microvasculature, with the endothelium being the first barrier for micrometastatic seeding. Among the most important aspects that define the pre-metastatic niche are the remodeling of the ECM, pioneering angiogenesis, the direct modulation of the endothelium and influences on the local immunological milieu.
Figure 1. Concept drawing of the pre-metastatic niche. The primary tumor releases signals via exosomes and soluble factors to distant organs, preparing them for metastasis. Most of the distant modulation takes place in and around the microvasculature, with the endothelium being the first barrier for micrometastatic seeding. Among the most important aspects that define the pre-metastatic niche are the remodeling of the ECM, pioneering angiogenesis, the direct modulation of the endothelium and influences on the local immunological milieu.
Ijms 24 10055 g001
Figure 2. The blood–brain barrier in the brain PMN. The blood–brain barrier (BBB) is a unique feature of the brain and is virtually the only entrance site for circulating tumor cells. As such, it has unique gatekeeping properties that are routinely modulated and hijacked by primary tumors. What little is known about the brain pre-metastatic niche overwhelmingly concerns the BBB. Dissolution of the tight junctions by immunological factors and growth factors has been shown in a pre-metastatic setting. Astrogliosis plays an important role in pro-metastatic vascular leakiness, and multiple routes lead to it; inflammation of surrounding astrocytes leads to scarring and increased BBB permeability, as does the differentiation of neural progenitor cells into astrocytes.
Figure 2. The blood–brain barrier in the brain PMN. The blood–brain barrier (BBB) is a unique feature of the brain and is virtually the only entrance site for circulating tumor cells. As such, it has unique gatekeeping properties that are routinely modulated and hijacked by primary tumors. What little is known about the brain pre-metastatic niche overwhelmingly concerns the BBB. Dissolution of the tight junctions by immunological factors and growth factors has been shown in a pre-metastatic setting. Astrogliosis plays an important role in pro-metastatic vascular leakiness, and multiple routes lead to it; inflammation of surrounding astrocytes leads to scarring and increased BBB permeability, as does the differentiation of neural progenitor cells into astrocytes.
Ijms 24 10055 g002
Figure 3. Advanced and experimental techniques for characterizing the pre-metastatic niche in the brain. In order to adequately describe the brain pre-metastatic niche, new and emerging techniques should be adapted to gain more insights into this elusive concept. In a spontaneous metastasis model, micro- and later oligmetastases will form in the brain. The timepoint right before the formation of the first micrometastases will represent the pre-metastatic niche most closely. Techniques such as elaborate CSF analysis for tumor cells or in vivo imaging of the brain could distinguish this timeframe. In a second phase, advanced microscopy methods combined with single-cell sequencing and spatial transcriptomics can be used to define it in more detail.
Figure 3. Advanced and experimental techniques for characterizing the pre-metastatic niche in the brain. In order to adequately describe the brain pre-metastatic niche, new and emerging techniques should be adapted to gain more insights into this elusive concept. In a spontaneous metastasis model, micro- and later oligmetastases will form in the brain. The timepoint right before the formation of the first micrometastases will represent the pre-metastatic niche most closely. Techniques such as elaborate CSF analysis for tumor cells or in vivo imaging of the brain could distinguish this timeframe. In a second phase, advanced microscopy methods combined with single-cell sequencing and spatial transcriptomics can be used to define it in more detail.
Ijms 24 10055 g003
Table 1. Comparison of the pre-metastatic niche (PMN) and the metastatic niche (MN).
Table 1. Comparison of the pre-metastatic niche (PMN) and the metastatic niche (MN).
PMNMN
Function [7,8]Access, anchorage, and early survivalSurvival, protection, and proliferation
Effects [7,8]
  • Angiogenesis;
  • Lymphangiogenesis;
  • Vascular leakiness;
  • Remodeling of the ECM;
  • Inflammatory response.
  • Metabolic changes in the TME;
  • Pro-survival signaling in the ECM;
  • Induction of chemoresistance;
  • Growth factor production;
  • Early angiogenesis;
  • Epithelial–mesenchymal-/mesenchymal–epithelial-transitions.
Exosomes
  • Transport of ncRNA at a distance [39,49,51,86];
  • Transport of proteins such as CEMIP and MIF [25,63];
  • Organ specificity via integrin expression [40];
  • Exosomal disposal of tumor-supressor miRNA in metastatic cells [44];
  • Paracrine ncRNA transfer [45].
ncRNA
  • miR-21: Angiogenesis upregulation through STAT3 [46];
  • miR-105: destruction of tight junction protein ZO-1 [19];
  • miR-122: modulation of glucose metabolism in stromal cells to favor metastatic cells [83];
  • lnc-MMP2-2: miRNA sponge, mitigating tumor-suppressive RNAs to increase BBB permeability [81];
  • TLR3 upregulation and neutrophil infiltration in alveor epithelium by exosomal RNA [18].
  • miR-19a: PTEN downregulation in tumor cells, secreted by astrocytes [87];
  • miR-135b: secreted by hypoxic Multiple Myeloma cells to form endothelial tubes [45].
Inflammatory response
  • Decrease in IFN-γ and increase in pro-inflammatory cytokines in pre-metastatic lungs by immature myeloid cells [17];
  • CXCR2 binding of chemokines might direct immunosuppressive MDSCs to the pre-metastatic site [37,88,89];
  • S100A8/9 signal to accumulate macrophages in the pre-metastatic lung [90];
  • TAM recruitment by tissue-factor mediated coagulation has been discussed to take place even before metastatic cell arrival [91].
  • Direct cytokine regulation between tumor cells and astrocytes [92];
  • IL-23 upregulation in metastasis-associated astrocytes [59];
  • CCL2-recruited TAMs promote metastatic outgrowth and angiogenesis [93,94];
  • Immune escape through T-cell suppression at the early metastatic site [28];
  • Immune shielding through platelet activation [95,96,97];
  • TAM recruitment by tissue-factor mediated coagulation promotes metastatic survival [91];
  • In vitro data regarding Treg cells in the MN (discussed in [37]).
Vascular response
  • miR-105: modulates brain endothelial tight junctions [19];
  • Endothelial cell remodeling through STAT3-highjacking [20,46];
  • Remodeling through Notch1-receptor upregulation [22];
  • Remodeling through MMP9 [17].
  • Tumor-cell-mediated necroptosis of endothelial cells [98];
  • Vascular cooption in brain vessels [71,99];
  • Interplay between microvasculature and dormant tumor cells [7,27].
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

Geissler, M.; Jia, W.; Kiraz, E.N.; Kulacz, I.; Liu, X.; Rombach, A.; Prinz, V.; Jussen, D.; Kokkaliaris, K.D.; Medyouf, H.; et al. The Brain Pre-Metastatic Niche: Biological and Technical Advancements. Int. J. Mol. Sci. 2023, 24, 10055. https://doi.org/10.3390/ijms241210055

AMA Style

Geissler M, Jia W, Kiraz EN, Kulacz I, Liu X, Rombach A, Prinz V, Jussen D, Kokkaliaris KD, Medyouf H, et al. The Brain Pre-Metastatic Niche: Biological and Technical Advancements. International Journal of Molecular Sciences. 2023; 24(12):10055. https://doi.org/10.3390/ijms241210055

Chicago/Turabian Style

Geissler, Maximilian, Weiyi Jia, Emine Nisanur Kiraz, Ida Kulacz, Xiao Liu, Adrian Rombach, Vincent Prinz, Daniel Jussen, Konstantinos D. Kokkaliaris, Hind Medyouf, and et al. 2023. "The Brain Pre-Metastatic Niche: Biological and Technical Advancements" International Journal of Molecular Sciences 24, no. 12: 10055. https://doi.org/10.3390/ijms241210055

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