Next Article in Journal
Male and Female Fertility: Prevention and Monitoring Hodgkin’ Lymphoma and Diffuse Large B-Cell Lymphoma Adult Survivors. A Systematic Review by the Fondazione Italiana Linfomi
Next Article in Special Issue
Targeting Bone Metastasis in Cancers
Previous Article in Journal
Could Ovarian Cancer Prediction Models Improve the Triage of Symptomatic Women in Primary Care? A Modelling Study Using Routinely Collected Data
Previous Article in Special Issue
Bone Marrow Adipocytes: A Link between Obesity and Bone Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanisms Supporting the Use of Beta-Blockers for the Management of Breast Cancer Bone Metastasis

by
Maria-Bernadette Madel
1 and
Florent Elefteriou
1,2,*
1
Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, USA
2
Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(12), 2887; https://doi.org/10.3390/cancers13122887
Submission received: 24 April 2021 / Revised: 6 June 2021 / Accepted: 8 June 2021 / Published: 9 June 2021
(This article belongs to the Special Issue Targeting Bone Metastasis in Cancers)

Abstract

:

Simple Summary

Bone represents the most common site of metastasis for breast cancer and the establishment and growth of metastatic cancer cells within the skeleton significantly reduces the quality of life of patients and their survival. The interplay between sympathetic nerves and bone cells, and its influence on the process of breast cancer bone metastasis is increasingly being recognized. Several mechanisms, all dependent on β-adrenergic receptor signaling in stromal bone cells, were shown to promote the establishment of disseminated cancer cells into the skeleton. This review provides a summary of these mechanisms in support of the therapeutic potential of β-blockers for the early management of breast cancer metastasis.

Abstract

The skeleton is heavily innervated by sympathetic nerves and represents a common site for breast cancer metastases, the latter being the main cause of morbidity and mortality in breast cancer patients. Progression and recurrence of breast cancer, as well as decreased overall survival in breast cancer patients, are associated with chronic stress, a condition known to stimulate sympathetic nerve outflow. Preclinical studies have demonstrated that sympathetic stimulation of β-adrenergic receptors in osteoblasts increases bone vascular density, adhesion of metastatic cancer cells to blood vessels, and their colonization of the bone microenvironment, whereas β-blockade prevented these events in mice with high endogenous sympathetic activity. These findings in preclinical models, along with clinical data from breast cancer patients receiving β-blockers, support the pathophysiological role of excess sympathetic nervous system activity in the formation of bone metastases, and the potential of commonly used, safe, and low-cost β-blockers as adjuvant therapy to improve the prognosis of bone metastases.

1. Introduction

Metastasis to distant organs represents a common and fatal complication in breast cancer patients [1,2]. About 70% of these patients experience metastatic bone disease, which can occur years after completion of the treatment of the primary tumor. Associated with a 5-year overall survival rate of approximately 23%, bone metastases represent a particularly unfavorable prognosis and can only be treated palliatively [1,3,4,5].
Cancer cell metastasis is a multistep process characterized by five major interrelated pathological events. As a consequence of their uncontrolled growth and low oxygen tension, tumor cells secrete proangiogenic factors that contribute to tumor vascularization. The high vascularity of the primary tumors provides a chance for metastatic cells to disseminate to other organs following detachment from the primary tumor, infiltration into the surrounding stroma, and migration through the basement membrane supporting the endothelium of local blood and/or lymphatic vessels. Intravasation into the blood/lymphatic circulation is followed by hematogenous or lymphagenous dissemination of a minimal number of anoikis-resistant tumor cells to distant organs. In these tissues, cancer cells extravasate into the tissue stroma in response to endothelial and stroma factors to form microfoci that can remain dormant or grow into full-blown metastatic macrofoci. Those macrofoci formed in skeletal tissues will impact skeletal homeostasis, structure and endocrine function [6,7,8,9,10]. These steps are driven in the primary and metastatic tumor microenvironment by direct cell-to-cell and paracrine interactions between cancer cells and different stromal cells [8,11,12,13,14,15,16].
The skeleton represents a preferred organ for metastasis of breast, prostate, and lung cancers [17,18,19,20,21,22]. It is a large, richly vascularized organ, abundantly innervated by sensory nerves found in the cancellous and mainly periosteal bone compartment, as well as sympathetic nerves, mostly associated with the bone marrow vasculature, sprouting toward bone surfaces [23,24,25,26,27,28,29,30,31,32]. Recent studies in mice have shown that sensory nerves, in addition to mediating pain postnatally, play an important role in the early formation of the skeleton and in the response of the skeleton to mechanical loading and fracture [33,34]. In contrast, sympathetic nerves were shown to be involved in the process of bone remodeling in adult mice, under the control of hypothalamic and brainstem centers in the central nervous system, with likely relevance to age-related bone loss, a condition concurrent with the time of most skeletal metastatic occurrences.
Over the past two decades, the relevance of skeletal innervation by sympathetic nerves extended to the process of bone metastasis, starting with the observation that chronic stress was associated with enhanced recurrence rates in breast cancer patients, and with metastasis formation, reduced overall survival, and poor patient prognosis [35,36,37,38,39,40,41,42,43,44], whereas β-blocker use was associated with increased relapse-free survival [40,42,44,45,46,47,48,49]. Chronic stress stimulates activity of the hypothalamic–pituitary axis (HPA) and the sympathetic nervous system (SNS), resulting in the release of norepinephrine (NE) from sympathetic nerve terminals and the stimulation of post-synaptic β-adrenergic receptors (βARs) on target cells, including bone [50] and cancer cells [51,52,53,54,55,56,57] (reviewed in [58]). These clinical observations were the first to support a putative role of excess sympathetic outflow in the mechanisms leading to the successful establishment of disseminated cancer cells into the skeleton. Since then, studies in vitro and in preclinical mouse models have refined our understanding of the mechanisms whereby sympathetic nerves can impact the metastatic process, and have revealed the complexity of these mechanisms. The effect of sympathetic nerves on the early stages of metastasis has recently been reviewed [58]. In the present review, we focus more specifically on mechanisms supporting the role of the SNS in the establishment of breast cancer cell metastases in the skeleton, and on how this knowledge supports the potential of βAR blockade as a new therapeutic strategy for a better management of patients with skeletal breast cancer metastases.

2. Sympathetic Innervation of the Skeleton and Evidence for an Interplay with the Process of Bone Metastasis

Stress has well-defined repercussions on body homeostasis. In the short-term, it stimulates the HPA and the SNS, resulting in the release of glucocorticoids and epinephrine from the adrenal glands and of NE from sympathetic nerve terminals to support the typical fight-or-flight response and survival. Chronic stress, however, has an overall negative impact on the body and has been associated with multiple pathological conditions, including coronary heart disease [59,60], obesity [61,62], and gastrointestinal diseases [63,64,65,66,67]. In the context of bone health, chronic psychological stress and severe depression have been associated with higher fracture risk and low bone mineral density (BMD) [68,69,70,71,72,73,74,75,76,77,78,79]. Although this association is likely multifactorial, one of its components is thought to be the abundant innervation of the skeleton by sympathetic nerves and the action of these nerves on bone cells and bone remodeling. Sympathetic nerve fibers are indeed located in close vicinity to bone cells, including osteoblasts, osteoclasts, and bone marrow cells, which all express post-synaptic βARs [51,52,53,54,55,56,57,80,81,82]. Studies in mice, using isoproterenol (ISO) as a surrogate for endogenous nerve-released NE, showed that βAR agonists cause bone loss by inhibiting osteoblast proliferation and by promoting their production of pro-osteoclastogenic factors and bone resorption [83,84]. On the other hand, mice lacking β2AR globally or specifically in osteoblasts were characterized by a high bone mass, and similar outcomes were observed in mice administered with the β-blocker propranolol [83,84,85]. These studies thus supported the critical role of βAR signaling in bone remodeling and pointed to osteoblasts as one of the critical targets of sympathetic nerves for their action on bone homeostasis.
β-blockers are commonly used to treat cardiovascular conditions, and retrospective studies have provided additional clinical insights to support the role of sympathetic nerves in the process of bone remodeling, while confirming the clinical relevance of the aforementioned preclinical findings [86,87,88,89,90,91,92,93,94]. These studies also revealed that β-blocker use at time of diagnosis in patients with triple negative breast cancer (but also non-small cell lung cancer, hemangiomas, and ovarian cancer) was associated with prolonged disease-free survival and reduced metastasis development and tumor recurrence [45,46,47,48,49,95,96,97,98,99,100,101,102]. Since no correlation was found between the post-diagnostic use of β-blockers and breast cancer progression [103], these observations implied that the βAR-dependent mechanisms leading to recurrence might mainly affect early stages of metastatic dissemination. As 70% of breast cancer patients succumb with skeletal metastases, these observations also suggested that sympathetic nerves, besides regulating bone homeostasis, participate in the molecular events that stimulate skeletal metastasis.
There are multiple mechanisms by which stress and sympathetic nerve activation can contribute to tumor metastasis. These include a direct effect on tumor cells, an indirect effect on the stroma of the primary tumor or distant metastatic tumor, and/or an effect on immune surveillance. Additional mechanisms might be involved, and they are not mutually exclusive. For instance, βAR stimulation in tumor cells and surrounding stromal elements in the primary tumor, such as tumor-associated macrophages and vascular endothelial cells, promotes molecular processes involved in tumor progression. These include DNA damage repair mechanisms by activation of the ataxia–telangiectasia and Rad3-related/p21 pathway [104,105,106], oncogene activation including Src [107], HER2 signaling [108,109], or resistance to chemotherapy-mediated cell death [110,111,112,113]. In line with the clinical findings associating stress and enhanced recurrence rate in cancer patients [42,43,44,45,46,47,48,49], in vitro studies demonstrated that NE decreases the efficacy of chemotherapeutics [104], and pharmacological βAR activation in breast cancer cells was shown to promote tumor cell growth and migration [114], as well as the expression of inflammatory and chemotactic cytokines required for metastasis and carcinogenesis [53,115,116,117,118]. These effects were abolished when using the β-blocker propranolol [53,115,116,117]. On the other hand, other studies showed an anti-proliferative effect of βAR agonists [118,119,120]. Results regarding a direct effect of catecholamines or βAR agonists on cancer cells are thus conflicting, but this is to be expected based on the in vitro nature of these assays, the focus on unique cell lines with specific characteristics, and the different doses of drugs used. The expression of αARs and βARs also differs between cancer cells lines and across molecular subtype (ER, PR, and HER2 status, reviewed in [58]). β2AR is the most widely expressed βAR in basal and luminal breast cancer cell lines and in tumor samples from patients with breast cancer [58,121,122,123]. β2AR expression was found to correlate with poor prognosis of ER breast cancer patients [123]. In another study, β2AR expression was associated with lower disease-free survival and higher lymph node metastasis rates in a small cohort of HER2+ breast cancer patients [121], but opposing results were obtained in another small cohort of HER2+ patients where it was associated with improved disease-free survival [124]. Inconsistencies regarding the type or level of expression of adrenergic receptors and the effect of their agonists in breast cancer cells thus limit confidence that modulation of βAR signaling in cancer cells may be of any solid predictive and therapeutic value, although this needs to be further examined.
β2AR is also widely expressed in host stromal tissues and βAR agonists can have distinct cellular targets and effects in different tissues and at specific stages of the metastatic process. The effect of βAR agonists and antagonists on this process requires all findings to be interpreted with this in mind. Further adding complexity to the mechanisms whereby sympathetic nerves influence breast cancer tumor progression is the difference between pharmacological βAR agonists and endogenous NE released by nerves in terms of adrenergic signaling and action. Pharmacological βAR agonists indeed acutely stimulate post-synaptic receptors on cancer cells or host cells, in contrast to endogenous nerve-released NE following SNS activation via chronic stress or other means, which is buffered by homeostatic mechanisms aimed at controlling excess SNS activation. Among the multiple components of this homeostatic system, the norepinephrine transporter (NET), located in the plasma membrane of noradrenergic neurons, is responsible for NE re-uptake and recycling. It serves as the primary mechanism for inactivating noradrenergic signaling and terminating the short-term biological effects of NE in the synaptic cleft [125,126], regulating adrenergic neurotransmission in the brain and peripheral organs [127,128]. NET is not only expressed in presynaptic neurons but also in bone cells, including mature osteoblasts and osteocytes [129,130] and its expression decreases during aging in mice [130], pointing to a potential role of this transporter in the regulation of bone remodeling, and possibly bone metastasis, and potentially explaining conflicting results between the effect of pharmacological βAR agonists and endogenous SNS activation on this process.
Although the assessment of an immune component is excluded in most studies related to cancer bone metastasis because of the common use of immunocompromised mice that do not reject human cancer cells, it is known that most immune cells express βARs and that SNS activation plays an integral role in the regulation of the anti-tumor immune response. For instance, sympathetic activation can promote the development of myeloid-derived suppressor cells (MDSCs) to favor an immunosuppressive environment [131] as well as macrophage tumor infiltration [132,133]. βAR signaling stimulates the expression of TGFβ, VEGF, IL-6, MMP9, and PTGS2 by macrophages, thereby promoting tumor progression and inhibiting the transcription of type I and II interferons [132,134,135], which are important in cell-mediated immune responses against cancer. Furthermore, βAR signaling can suppress the cytotoxic function of T cells and NK cells and contribute to the dissemination of cancer cells [136,137,138,139]. However, in vivo preclinical studies have demonstrated that stimulation of the SNS favors tumor metastasis in the absence of NK and cytotoxic T cells [53,110,132,140,141], thus suggesting the existence of T- and NK-cell-independent mechanisms contributing to the engraftment of metastatic tumor cells into the skeleton.

3. SNS-Induced Bone Stromal-Dependent Mechanisms Promote Skeletal Colonization by Cancer Cells

Multiple observations support the notion that sympathetic nerves act on cells of the osteoblast lineage to promote the formation of skeletal metastases. An important initial suggestive evidence of such mechanisms was the observation that hematopoietic stem cells from the bone marrow environment were mobilized into the circulatory system by sympathetic nerve signals and the induction of stromal-cell-derived factor 1 (SDF1/CXCL12), a chemokine also involved in the dissemination of metastatic cells into the skeleton [142,143]. Osteoblasts, osteocytes, and osteoclasts predominantly express β2AR and can thus respond to NE released by sympathetic nerves [51,52,83,84,144,145,146]. Angiogenesis represents a necessary component of both invasive tumor growth and distant metastasis. One of the main inducers of angiogenesis is vascular endothelial growth factor (VEGF) [147,148,149,150], whose expression correlates with increased primary tumor microvasculature and malignancy and metastasis in breast cancer patients [151]. In vitro studies revealed that VEGF synthesis is increased upon βAR stimulation of tumor cells [81,152] and animal studies showed that high endogenous sympathetic outflow increases VEGF and vascular density in primary tumors [53,153,154]. Besides its direct action on the primary tumor, the SNS also influences the vascular density of the bone marrow microenvironment. The skeleton is a highly vascularized tissue, and chondrocytes and osteoblasts are crucial regulators of angiogenesis during skeletal development and bone regeneration [155,156]. However, in adult mice, vascular density can be further increased upon stimulation of βARs in osteoblasts via β2AR-dependent induction of VEGF synthesis in osteoblasts [157]. This increase in VEGF and bone vascular density induced by βAR stimulation was shown to promote breast cancer cell bone metastasis, as VEGF blockade or β2AR deficiency in osteoblasts specifically reduced both vascularity and the number of metastatic bone lesions formed after intracardiac injection of triple negative MDA-MB-231 breast cancer cells and ISO administration [157]. Activation of βARs in breast cancer cell lines and osteoblasts also induces the production of IL-6, which has the potential to stimulate the βAR-driven pro-angiogenic pathway by promoting the expression of VEGF [81,158,159,160,161] and the proliferation of breast cancer cells [162,163]. Therefore, a first mechanism of action whereby sympathetic nerves can promote skeletal breast cancer metastasis is through a VEGF-dependent neo-angiogenic switch upon β2AR signaling in osteoblasts and stimulation of bone marrow vascular density, which increases the likelihood of circulating breast cancer cells colonizing the skeleton.
The arrest of circulating cancer cells into distant organs plays a central role in the process of tumor metastasis. In this context, the adhesive interaction between tumor cells and the vascular endothelium is important. Extravasation, which brings circulating tumor cells from the blood stream into the distant tissue stroma, is initialized by attachment and rolling of tumor cells along the endothelium via selectins, such as E- and P-selectin [164,165,166,167]. This process is followed by cancer cell arrest and tight adhesion mediated by integrins, before cancer cells transmigrate through the vascular endothelium [164,167]. This transient interaction between cancer cells and endothelial cells is promoted by inflammatory cytokines such as IL-1 and TNFα, whose action is to increase the expression of adhesion proteins on the endothelium [168,169,170,171,172]. IL-1β was shown to promote the adhesion of cancer cells to endothelial cells in vitro, and administration of IL-1β to mice increased the number of lung and skeletal metastases, while reducing IL-1β activity in melanoma mouse models reduced tumor burden and metastases [173,174,175,176]. In vitro experiments demonstrated that βAR stimulation of osteoblasts induced their expression of IL-1β [158]. Moreover, the conditioned medium from ISO-treated osteoblasts increased the expression of E- and P-selectin in endothelial cells and promoted the adhesion of human MDA-MB-231 breast cancer cells to these endothelial cultures, while preincubation with an IL-1β neutralizing antibody significantly reduced this effect [158]. Although the in vivo relevance of these data has not yet been experimentally addressed, a second level of SNS action on the bone microenvironment in the context of skeletal metastasis could thus be a stimulatory effect on osteoblast-derived IL1β, leading to enhanced adhesive properties of the bone vascular network for metastatic cancer cells and promotion of the arrest of these cells into the skeleton, in a E/P-selectin-dependent manner.
Whether metastatic cancer cells survive and grow in their new environment after extravasation depends on multiple cell-intrinsic and stromal-derived factors. Subjecting mice to chronic immobilization stress (CIS, known to transiently activate sympathetic nerves and the HPA axis) prior to injection of cancer cells increased the efficiency of skeletal metastasis compared to non-stressed control mice [141]. These findings indicated that sympathetic activation creates a pro-metastatic microenvironment that promotes the establishment of circulating cancer cells into the skeleton. This is in line with the aforementioned pro-angiogenic and pro-adhesive effect of β2AR signaling. The administration of the β-blocker propranolol reduced this effect, confirming the contribution of endogenous sympathetic activation and βAR signaling to this mechanism [141]. Further experiments demonstrated that the homing/migration properties of receptor activator of nuclear factor kappa-Β ligand (RANKL) were involved in this context. RANKL is expressed by osteoblasts and osteocytes as well as T cells and chondrocytes [177,178,179,180,181], and its expression in osteoblasts and osteocytes is readily increased upon βAR stimulation [141,145]. Besides its osteoclastogenic properties, RANKL is known to have a pro-migratory activity for RANK-expressing cancer cells [84,114,182,183,184,185]. High RANK expression (the receptor for RANKL) in the tumors of breast cancer patients was associated with poor prognosis and the combined expression of RANK and CXCR4 in breast cancer patients predicted recurrence of bone metastases [186,187]. In vitro, the observation that the conditioned medium of ISO-treated osteoblasts stimulated the migration of breast cancer cells, and that the RANKL decoy receptor osteoprotegerin (OPG) could block it, supported a stimulatory effect of βAR activation on breast cancer cell migration, via the upregulation of RANKL expression by the host stromal compartment [141]. These results were reinforced by the reduction in MDA-MD-231 cancer cell migration in vitro and bone metastasis in vivo following βAR stimulation and knockdown of RANK in breast cancer cells [141]. In conclusion and collectively, these results suggest that stimulation of βAR signaling in osteoblasts following activation of sympathetic nerves favors the colonization of the bone marrow environment by circulating breast cancer cells at multiple steps of the metastatic process, via (a) the promotion of a VEGF-dependent neoangiogenic switch leading to higher bone vascular density and (b) an IL-1β-dependent increase in the adhesive properties of this newly created vasculature, both of which potentially increase the likelihood of circulating breast cancer cells to arrest in bone tissues, and (c) a RANKL-dependent increase in cancer cell homing or retention into bone tissues, due to higher migratory activity toward RANKL-secreting niches (Figure 1). It is worth noting that osteocytes represent an essential source of RANKL during bone remodeling [5,188], and that β-adrenergic signals not only promote RANKL expression in osteoblasts but also in osteocytes [145]. Whether osteoblasts or osteocytes, or both, are targets of sympathetic-nerve-derived NE for its action on bone metastasis remains unknown.
Although pro-angiogenic and cancer cell bone-homing properties of βAR stimulation in the osteoblast lineage are mainly relevant to the early establishment of circulating breast cancer cells in the bone marrow environment, RANKL and IL-6 are, before all, two well-known potent osteoclastogenic cytokines [158,189,190,191]. Their high expression following βAR2 stimulation is likely to contribute to the development of osteolytic lesions and tumor progression through enhanced osteoclastogenesis once tumor cells are established in the skeleton [83,192]. Direct βAR-mediated stimulation of osteoclast differentiation has also been observed [193] and was reported to enhance the bone resorbing activity of βAR-expressing osteoclasts in vitro [194]. In addition, bone-derived growth factors, such as TGFβ, are released from the bone matrix during osteoclast-mediated resorption to promote the proliferation and survival of tumor cells [195,196,197,198,199,200,201,202]. Evidence that TGFβ plays a decisive role in tumor growth was substantiated by the demonstration that bone metastases can be effectively reduced by the TGFβ signaling blockade [200,202,203,204,205,206,207]. Therefore, stimulation of βAR in the osteoblast lineage and its effect on osteoclasts may not only favor the establishment of bone metastases at early disease stage but also fuel the feed-forward cycle of bone destruction at later stages of the bone metastatic process, by promoting osteoclastogenesis and the release of growth factors from the bone matrix [208]. The more recent discovery of the deleterious impact of bone-metastasis-derived TGFβ on the muscular system [209,210,211] also implies that high sympathetic nervous system activity would be associated with muscle weakness in the context of skeletal metastasis. In addition to their bone resorption function, osteoclasts are, as their progenitors, antigen-presenting cells [212]. Depending on their origin and environment, these cells induce regulatory CD4+ and CD8+ T cells [213,214,215] or inflammatory TNFα-producing CD4+ T cells [213]. Recently, an immunosuppressive subset of Cx3cr1+ osteoclasts was identified as promoting a pro-metastatic bone marrow microenvironment through its strong immunosuppressive capacity and upregulation of immunosuppressive checkpoint molecules including programmed death ligand 1 (PD-1L), herpes virus entry mediator (HVEM), and galectin 9 [216]. The immunosuppressive capacity of osteoclasts has also been described in the context of multiple myeloma, where the production of immunosuppressive molecules such as indoleamine 2,3-dioxygenase (IDO) and PD-L1 by osteoclasts protects myeloma cells against T-cell-mediated cytotoxicity [217]. Whether direct βAR activation in osteoclasts affects their immune function and whether this could entail an immunosuppressive and pro-metastatic microenvironment in the bone marrow remains to be investigated. Lastly, osteoclasts were shown to secrete miRNAs that trigger cancer cell proliferation, tumor cell survival, and angiogenesis [218,219,220]. Whether stimulation of adrenergic signals enhances this release and thus promotes a pro-tumorigenic microenvironment has yet to be addressed.

4. Treatment Strategies to Limit Metastatic Cancer Cell Engraftment into the Skeleton

Despite the successful use of anti-resorptive molecules such as bisphosphonates to limit bone pain and fracture as standard-of-care for patients with established bone metastases [221,222,223], the development of drug resistance and the persistence of minimal residual disease hampers cancer remission. An important implication of the findings summarized above is that βAR blockade should reduce the levels of VEGF, IL-1β, and RANKL produced by osteoblasts (and presumably osteocytes) in response to sympathetic activation, and thus offer some level of protection against bone metastatic events. It is important to emphasize that, in addition to a number of social stressors, the diagnosis of cancer and its treatment have a significant impact on the stress level of patients [44,224,225,226,227,228]. β-blockers are used for the treatment of multiple conditions, including congestive heart failure, hypertension, migraines, infantile hemangioma, and thyrotoxicosis [229]. The safety profile of these drugs is well-described, it is not associated with an increased incidence of breast cancer [230,231], and several clinical and preclinical studies support their use as adjuvant therapy in the treatment of breast cancer [45,46,47,48,49,95,96,97,98,99,100,101,102,232,233,234]. This beneficial effect of β-blockers on breast cancer patient survival, reduction of tumor recurrence, and reduced incidence of metastasis (summarized by Conceição and colleges [58]), remains, however, controversial because most of these studies present limitations, including retrospective design, small population size, difficulty in assessing duration of β-blocker treatment, or lack of data about comorbidities and intake of other medications. Nevertheless, improved relapse-free survival [45] and reduced metastasis and tumor recurrence were observed in breast cancer patients receiving β-blockers [46,48], and consistent with what was observed in some preclinical studies [141], a positive correlation was observed in patients taking propranolol the year before breast cancer diagnosis, specifically regarding tumor invasion and metastatic involvement at diagnosis, as well as reduced cancer-specific mortality [47]. This observation and the fact that there was no association between post-diagnostic use of β-blockers and breast cancer-specific mortality and progression [103] support the notion that metastatic dissemination, and not metastatic tumor growth, is the most relevant target of sympathetic nerves and that β-blocker therapy could improve prognosis. Of note, synergy was detected between β-blockers and chemotherapy in mice in terms of anti-tumor effect at the primary site and improved survival [235], thus further supporting the use of β-blockers concurrently with chemotherapy in the management of breast cancer. Although blockade of βAR signaling represents an attractive strategy to complement current therapeutic options, preclinical and adequately powered clinical trials focusing on overall survival and cancer recurrence are still needed to assess the therapeutic potential of β-blockers to restrain breast cancer bone metastasis. Whether the protective effect of non-selective β-blockers on skeletal metastasis is mediated by action on β1AR or β2AR also remains uncertain. It should be noted that β1AR blockade but not non-selective βAR blockade reduced bone resorption and had a favorable effect on cancellous BMD in clinical studies [94]. This action of β-blockers on BMD is another incentive to further evaluate this class of drugs for the treatment of cancer patients at risk of bone loss and fracture, especially since there are concerns regarding rare side-effects of current treatments targeting osteoclasts, such as osteonecrosis of the jaw or atypical femoral fractures [236]. One should also be aware of a recent study showing that the effect of β-blockers on overall survival may vary depending on the different subtypes of breast cancer—in contrast to improved relapse-free survival in patients with triple negative breast cancer [45,48,233,237], the pre-existing use of β-blockers in patients with advanced-stage HER2-positive breast cancer prior to anti-HER2 therapy initiation was associated with reduced overall survival compared to patients without β-blocker intake [238].

5. Conclusions

Preclinical studies in mice demonstrated that stimulation of β2AR in osteoblasts promotes bone vascular density and the colonization of the bone microenvironment by metastatic breast cancer cells, and that pharmacological inhibition of βAR signaling by β-blockers inhibits skeletal colonization by metastatic cancer cells. These observations support the contribution of sympathetic nerves to the process of breast cancer bone metastasis, but there are still many unanswered questions to fully understand how these nerves change the behavior of metastatic cancer cells and the properties of their stroma along the multiple steps of the metastatic process. Variables in this equation include the variety of adrenergic receptors expressed by tumor cells and stromal cells, differential signaling upon various ligands, and the multiple homeostatic mechanisms regulating endogenous sympathetic NE release and AR signaling in both presynaptic and post-synaptic cells, as well as the contribution of various immune cells. Much remains to be done to broaden findings in different animal models of bone metastasis, in immunocompetent animals, and with different types of cancer cells. Clinical studies are also still needed to confirm the beneficial effect of low cost, safe β-blockers on cancer cell bone metastasis and relapse-free survival.

Author Contributions

Conceptualization, writing, review and editing, M.-B.M. and F.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NCI R01 CA168717 and NIH R01 AG055394.

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.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Manders, K.; van de Poll-Franse, L.V.; Creemers, G.-J.; Vreugdenhil, G.; van der Sangen, M.J.; Nieuwenhuijzen, G.A.; Roumen, R.M.; Voogd, A.C. Clinical management of women with metastatic breast cancer: A descriptive study according to age group. BMC Cancer 2006, 6, 179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Coleman, R.E. Clinical features of metastatic bone disease and risk of skeletal morbidity. Clin. Cancer Res. 2006, 12, 6243s–6249s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kennecke, H.; Yerushalmi, R.; Woods, R.; Cheang, M.C.U.; Voduc, D.; Speers, C.H.; Nielsen, T.O.; Gelmon, K. Metastatic Behavior of Breast Cancer Subtypes. J. Clin. Oncol. 2010, 28, 3271–3277. [Google Scholar] [CrossRef]
  4. Coleman, R.; Smith, P.; Rubens, R. Clinical course and prognostic factors following bone recurrence from breast cancer. Br. J. Cancer 1998, 77, 336–340. [Google Scholar] [CrossRef] [Green Version]
  5. Xiong, J.; Onal, M.; Jilka, R.L.; Weinstein, R.S.; Manolagas, S.C.; O’Brien, C.A. Matrix-embedded cells control osteoclast formation. Nat. Med. 2011, 17, 1235–1241. [Google Scholar] [CrossRef] [Green Version]
  6. Käkönen, S.-M.; Mundy, G.R. Mechanisms of osteolytic bone metastases in breast carcinoma. Cancer 2003, 97, 834–839. [Google Scholar] [CrossRef] [PubMed]
  7. van Zijl, F.; Krupitza, G.; Mikulits, W. Initial steps of metastasis: Cell invasion and endothelial transmigration. Mutat. Res. 2011, 728, 23–34. [Google Scholar] [CrossRef]
  8. Nguyen, D.X.; Bos, P.D.; Massagué, J. Metastasis: From dissemination to organ-specific colonization. Nat. Rev. Cancer 2009, 9, 274–284. [Google Scholar] [CrossRef]
  9. Feller, L.; Kramer, B.; Lemmer, J. Pathobiology of cancer metastasis: A short account. Cancer Cell Int. 2012, 12, 24. [Google Scholar] [CrossRef] [Green Version]
  10. Alizadeh, A.M.; Shiri, S.; Farsinejad, S. Metastasis review: From bench to bedside. Tumour Biol. 2014, 35, 8483–8523. [Google Scholar] [CrossRef]
  11. Nguyen, D.X.; Massagué, J. Genetic determinants of cancer metastasis. Nat. Rev. Genet. 2007, 8, 341–352. [Google Scholar] [CrossRef]
  12. Wels, J.; Kaplan, R.N.; Rafii, S.; Lyden, D. Migratory neighbors and distant invaders: Tumor-associated niche cells. Genes Dev. 2008, 22, 559–574. [Google Scholar] [CrossRef] [Green Version]
  13. Chiang, A.C.; Massagué, J. Molecular basis of metastasis. N. Engl. J. Med. 2008, 359, 2814–2823. [Google Scholar] [CrossRef] [Green Version]
  14. Sikes, R.A.; Nicholson, B.E.; Koeneman, K.S.; Edlund, N.M.; Bissonette, E.A.; Bradley, M.J.; Thalmann, G.N.; Cecchini, M.G.; Pienta, K.J.; Chung, L.W.K. Cellular interactions in the tropism of prostate cancer to bone. Int. J. Cancer 2004, 110, 497–503. [Google Scholar] [CrossRef]
  15. Kang, Y.; Siegel, P.M.; Shu, W.; Drobnjak, M.; Kakonen, S.M.; Cordón-Cardo, C.; Guise, T.A.; Massagué, J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003, 3, 537–549. [Google Scholar] [CrossRef] [Green Version]
  16. Minn, A.J.; Kang, Y.; Serganova, I.; Gupta, G.P.; Giri, D.D.; Doubrovin, M.; Ponomarev, V.; Gerald, W.L.; Blasberg, R.; Massagué, J. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J. Clin. Investig. 2005, 115, 44–55. [Google Scholar] [CrossRef] [Green Version]
  17. Hill, E.L.; Elde, R. Distribution of CGRP-, VIP-, D beta H-, SP-, and NPY-immunoreactive nerves in the periosteum of the rat. Cell Tissue Res. 1991, 264, 469–480. [Google Scholar] [CrossRef] [PubMed]
  18. Elefteriou, F. Role of sympathetic nerves in the establishment of metastatic breast cancer cells in bone. J. Bone Oncol. 2016, 5, 132–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Patel, M.S.; Elefteriou, F. The new field of neuroskeletal biology. Calcif. Tissue Int. 2007, 80, 337–347. [Google Scholar] [CrossRef]
  20. Artico, M.; Bosco, S.; Cavallotti, C.; Agostinelli, E.; Giuliani-Piccari, G.; Sciorio, S.; Cocco, L.; Vitale, M. Noradrenergic and cholinergic innervation of the bone marrow. Int. J. Mol. Med. 2002, 10, 77–80. [Google Scholar] [CrossRef] [PubMed]
  21. Mach, D.B.; Rogers, S.D.; Sabino, M.C.; Luger, N.M.; Schwei, M.J.; Pomonis, J.D.; Keyser, C.P.; Clohisy, D.R.; Adams, D.J.; O’Leary, P.; et al. Origins of skeletal pain: Sensory and sympathetic innervation of the mouse femur. Neuroscience 2002, 113, 155–166. [Google Scholar] [CrossRef]
  22. Goblirsch, M.J.; Zwolak, P.P.; Clohisy, D.R. Biology of bone cancer pain. Clin. Cancer Res. 2006, 12, 6231s–6235s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lorenz, M.R.; Brazill, J.M.; Beeve, A.T.; Shen, I.; Scheller, E.L. A neuroskeletal atlas: Spatial mapping and contextualization of axon subtypes innervating the long bones of C3H and B6 mice. J. Bone Miner. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  24. Sayilekshmy, M.; Hansen, R.B.; Delaissé, J.-M.; Rolighed, L.; Andersen, T.L.; Heegaard, A.-M. Innervation is higher above Bone Remodeling Surfaces and in Cortical Pores in Human Bone: Lessons from patients with primary hyperparathyroidism. Sci. Rep. 2019, 9, 5361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ahmed, M.; Bjurholm, A.; Kreicbergs, A.; Schultzberg, M. Neuropeptide Y, tyrosine hydroxylase and vasoactive intestinal polypeptide-immunoreactive nerve fibers in the vertebral bodies, discs, dura mater, and spinal ligaments of the rat lumbar spine. Spine 1993, 18, 268–273. [Google Scholar] [CrossRef] [PubMed]
  26. Fan, W.; Bouwense, S.A.W.; Crawford, R.; Xiao, Y. Structural and cellular features in metaphyseal and diaphyseal periosteum of osteoporotic rats. J. Mol. Histol. 2010, 41, 51–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hukkanen, M.; Konttinen, Y.T.; Rees, R.G.; Santavirta, S.; Terenghi, G.; Polak, J.M. Distribution of nerve endings and sensory neuropeptides in rat synovium, meniscus and bone. Int. J. Tissue React. 1992, 14, 1–10. [Google Scholar]
  28. Imai, S.; Tokunaga, Y.; Maeda, T.; Kikkawa, M.; Hukuda, S. Calcitonin gene-related peptide, substance P, and tyrosine hydroxylase-immunoreactive innervation of rat bone marrows: An immunohistochemical and ultrastructural investigation on possible efferent and afferent mechanisms. J. Orthop. Res. 1997, 15, 133–140. [Google Scholar] [CrossRef]
  29. Chartier, S.R.; Mitchell, S.A.T.; Majuta, L.A.; Mantyh, P.W. The Changing Sensory and Sympathetic Innervation of the Young, Adult and Aging Mouse Femur. Neuroscience 2018, 387, 178–190. [Google Scholar] [CrossRef]
  30. Castañeda-Corral, G.; Jimenez-Andrade, J.M.; Bloom, A.P.; Taylor, R.N.; Mantyh, W.G.; Kaczmarska, M.J.; Ghilardi, J.R.; Mantyh, P.W. The majority of myelinated and unmyelinated sensory nerve fibers that innervate bone express the tropomyosin receptor kinase A. Neuroscience 2011, 178, 196–207. [Google Scholar] [CrossRef] [Green Version]
  31. Martin, C.D.; Jimenez-Andrade, J.M.; Ghilardi, J.R.; Mantyh, P.W. Organization of a unique net-like meshwork of CGRP+ sensory fibers in the mouse periosteum: Implications for the generation and maintenance of bone fracture pain. Neurosci. Lett. 2007, 427, 148–152. [Google Scholar] [CrossRef] [Green Version]
  32. Jimenez-Andrade, J.M.; Mantyh, W.G.; Bloom, A.P.; Xu, H.; Ferng, A.S.; Dussor, G.; Vanderah, T.W.; Mantyh, P.W. A phenotypically restricted set of primary afferent nerve fibers innervate the bone versus skin: Therapeutic opportunity for treating skeletal pain. Bone 2010, 46, 306–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tomlinson, R.E.; Li, Z.; Zhang, Q.; Goh, B.C.; Li, Z.; Thorek, D.L.J.; Rajbhandari, L.; Brushart, T.M.; Minichiello, L.; Zhou, F.; et al. NGF-TrkA Signaling by Sensory Nerves Coordinates the Vascularization and Ossification of Developing Endochondral Bone. Cell Rep. 2016, 16, 2723–2735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Tomlinson, R.E.; Li, Z.; Li, Z.; Minichiello, L.; Riddle, R.C.; Venkatesan, A.; Clemens, T.L. NGF-TrkA signaling in sensory nerves is required for skeletal adaptation to mechanical loads in mice. Proc. Natl. Acad. Sci. USA 2017, 114, E3632–E3641. [Google Scholar] [CrossRef] [Green Version]
  35. Moreno-Smith, M.; Lutgendorf, S.K.; Sood, A.K. Impact of stress on cancer metastasis. Future Oncol. 2010, 6, 1863–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Heffner, K.L.; Loving, T.J.; Robles, T.F.; Kiecolt-Glaser, J.K. Examining psychosocial factors related to cancer incidence and progression: In search of the silver lining. Brain. Behav. Immun. 2003, 17 (Suppl. 1), S109–S111. [Google Scholar] [CrossRef]
  37. Clouston, S.A.P.; Kuan, P.; Kotov, R.; Mukherjee, S.; Thompson-Carino, P.; Bromet, E.J.; Luft, B.J. Risk factors for incident prostate cancer in a cohort of world trade center responders. BMC Psychiatry 2019, 19, 389. [Google Scholar] [CrossRef] [Green Version]
  38. Kikuchi, N.; Nishiyama, T.; Sawada, T.; Wang, C.; Lin, Y.; Watanabe, Y.; Tamakoshi, A.; Kikuchi, S. Perceived Stress and Colorectal Cancer Incidence: The Japan Collaborative Cohort Study. Sci. Rep. 2017, 7, 40363. [Google Scholar] [CrossRef]
  39. Vahdaninia, M.; Omidvari, S.; Montazeri, A. What do predict anxiety and depression in breast cancer patients? A follow-up study. Soc. Psychiatry Psychiatr. Epidemiol. 2010, 45, 355–361. [Google Scholar] [CrossRef]
  40. Giese-Davis, J.; Wilhelm, F.H.; Conrad, A.; Abercrombie, H.C.; Sephton, S.; Yutsis, M.; Neri, E.; Taylor, C.B.; Kraemer, H.C.; Spiegel, D. Depression and stress reactivity in metastatic breast cancer. Psychosom. Med. 2006, 68, 675–683. [Google Scholar] [CrossRef] [Green Version]
  41. Ross, K. Mapping pathways from stress to cancer progression. J. Natl. Cancer Inst. 2008, 100, 914–915. [Google Scholar] [CrossRef] [Green Version]
  42. Burgess, C.; Cornelius, V.; Love, S.; Graham, J.; Richards, M.; Ramirez, A. Depression and anxiety in women with early breast cancer: Five year observational cohort study. BMJ 2005, 330, 702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Spiegel, D.; Giese-Davis, J. Depression and cancer: Mechanisms and disease progression. Biol. Psychiatry 2003, 54, 269–282. [Google Scholar] [CrossRef]
  44. Chida, Y.; Hamer, M.; Wardle, J.; Steptoe, A. Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat. Clin. Pract. Oncol. 2008, 5, 466–475. [Google Scholar] [CrossRef]
  45. Melhem-Bertrandt, A.; Chavez-Macgregor, M.; Lei, X.; Brown, E.N.; Lee, R.T.; Meric-Bernstam, F.; Sood, A.K.; Conzen, S.D.; Hortobagyi, G.N.; Gonzalez-Angulo, A.-M. Beta-blocker use is associated with improved relapse-free survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2011, 29, 2645–2652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Powe, D.G.; Voss, M.J.; Zänker, K.S.; Habashy, H.O.; Green, A.R.; Ellis, I.O.; Entschladen, F. Beta-blocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival. Oncotarget 2010, 1, 628–638. [Google Scholar] [CrossRef] [Green Version]
  47. Barron, T.I.; Connolly, R.M.; Sharp, L.; Bennett, K.; Visvanathan, K. Beta blockers and breast cancer mortality: A population- based study. J. Clin. Oncol. 2011, 29, 2635–2644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Botteri, E.; Munzone, E.; Rotmensz, N.; Cipolla, C.; De Giorgi, V.; Santillo, B.; Zanelotti, A.; Adamoli, L.; Colleoni, M.; Viale, G.; et al. Therapeutic effect of β-blockers in triple-negative breast cancer postmenopausal women. Breast Cancer Res. Treat. 2013, 140, 567–575. [Google Scholar] [CrossRef]
  49. Choi, C.H.; Song, T.; Kim, T.H.; Choi, J.K.; Park, J.-Y.; Yoon, A.; Lee, Y.-Y.; Kim, T.-J.; Bae, D.-S.; Lee, J.-W.; et al. Meta-analysis of the effects of beta blocker on survival time in cancer patients. J. Cancer Res. Clin. Oncol. 2014, 140, 1179–1188. [Google Scholar] [CrossRef]
  50. Elefteriou, F. Impact of the Autonomic Nervous System on the Skeleton. Physiol. Rev. 2018, 98, 1083–1112. [Google Scholar] [CrossRef] [Green Version]
  51. Ma, Y.; Nyman, J.S.; Tao, H.; Moss, H.H.; Yang, X.; Elefteriou, F. β2-Adrenergic receptor signaling in osteoblasts contributes to the catabolic effect of glucocorticoids on bone. Endocrinology 2011, 152, 1412–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Elefteriou, F.; Campbell, P.; Ma, Y. Control of bone remodeling by the peripheral sympathetic nervous system. Calcif. Tissue Int. 2014, 94, 140–151. [Google Scholar] [CrossRef] [Green Version]
  53. Thaker, P.H.; Han, L.Y.; Kamat, A.A.; Arevalo, J.M.; Takahashi, R.; Lu, C.; Jennings, N.B.; Armaiz-Pena, G.; Bankson, J.A.; Ravoori, M.; et al. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat. Med. 2006, 12, 939–944. [Google Scholar] [CrossRef]
  54. Shimizu, N.; Hori, T.; Nakane, H. An interleukin-1 beta-induced noradrenaline release in the spleen is mediated by brain corticotropin-releasing factor: An in vivo microdialysis study in conscious rats. Brain Behav. Immun. 1994, 8, 14–23. [Google Scholar] [CrossRef] [Green Version]
  55. Madden, K.S.; Sanders, V.M.; Felten, D.L. Catecholamine influences and sympathetic neural modulation of immune responsiveness. Annu. Rev. Pharmacol. Toxicol. 1995, 35, 417–448. [Google Scholar] [CrossRef]
  56. Badino, G.R.; Novelli, A.; Girardi, C.; Di Carlo, F. Evidence for functional beta-adrenoceptor subtypes in CG-5 breast cancer cell. Pharmacol. Res. 1996, 33, 255–260. [Google Scholar] [CrossRef] [PubMed]
  57. Schuller, H.M.; Cole, B. Regulation of cell proliferation by beta-adrenergic receptors in a human lung adenocarcinoma cell line. Carcinogenesis 1989, 10, 1753–1755. [Google Scholar] [CrossRef]
  58. Conceição, F.; Sousa, D.M.; Paredes, J.; Lamghari, M. Sympathetic activity in breast cancer and metastasis: Partners in crime. Bone Res. 2021, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  59. Kubzansky, L.D.; Koenen, K.C.; Spiro, A.; Vokonas, P.S.; Sparrow, D. Prospective study of posttraumatic stress disorder symptoms and coronary heart disease in the Normative Aging Study. Arch. Gen. Psychiatry 2007, 64, 109–116. [Google Scholar] [CrossRef] [Green Version]
  60. Cohen, B.E.; Marmar, C.R.; Neylan, T.C.; Schiller, N.B.; Ali, S.; Whooley, M.A. Posttraumatic stress disorder and health-related quality of life in patients with coronary heart disease: Findings from the Heart and Soul Study. Arch. Gen. Psychiatry 2009, 66, 1214–1220. [Google Scholar] [CrossRef] [PubMed]
  61. Block, J.P.; He, Y.; Zaslavsky, A.M.; Ding, L.; Ayanian, J.Z. Psychosocial stress and change in weight among US adults. Am. J. Epidemiol. 2009, 170, 181–192. [Google Scholar] [CrossRef] [Green Version]
  62. Gluck, M.E.; Geliebter, A.; Hung, J.; Yahav, E. Cortisol, hunger, and desire to binge eat following a cold stress test in obese women with binge eating disorder. Psychosom. Med. 2004, 66, 876–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Jansson, C.; Wallander, M.-A.; Johansson, S.; Johnsen, R.; Hveem, K. Stressful psychosocial factors and symptoms of gastroesophageal reflux disease: A population-based study in Norway. Scand. J. Gastroenterol. 2010, 45, 21–29. [Google Scholar] [CrossRef] [PubMed]
  64. Fass, R.; Naliboff, B.D.; Fass, S.S.; Peleg, N.; Wendel, C.; Malagon, I.B.; Mayer, E.A. The effect of auditory stress on perception of intraesophageal acid in patients with gastroesophageal reflux disease. Gastroenterology 2008, 134, 696–705. [Google Scholar] [CrossRef] [PubMed]
  65. Levenstein, S.; Prantera, C.; Varvo, V.; Scribano, M.L.; Andreoli, A.; Luzi, C.; Arcà, M.; Berto, E.; Milite, G.; Marcheggiano, A. Stress and exacerbation in ulcerative colitis: A prospective study of patients enrolled in remission. Am. J. Gastroenterol. 2000, 95, 1213–1220. [Google Scholar] [CrossRef]
  66. Mittermaier, C.; Dejaco, C.; Waldhoer, T.; Oefferlbauer-Ernst, A.; Miehsler, W.; Beier, M.; Tillinger, W.; Gangl, A.; Moser, G. Impact of depressive mood on relapse in patients with inflammatory bowel disease: A prospective 18-month follow-up study. Psychosom. Med. 2004, 66, 79–84. [Google Scholar] [CrossRef]
  67. Maunder, R.G.; Greenberg, G.R.; Hunter, J.J.; Lancee, W.J.; Steinhart, A.H.; Silverberg, M.S. Psychobiological subtypes of ulcerative colitis: pANCA status moderates the relationship between disease activity and psychological distress. Am. J. Gastroenterol. 2006, 101, 2546–2551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Erez, H.B.; Weller, A.; Vaisman, N.; Kreitler, S. The relationship of depression, anxiety and stress with low bone mineral density in post-menopausal women. Arch. Osteoporos. 2012, 7, 247–255. [Google Scholar] [CrossRef] [PubMed]
  69. Cizza, G.; Primma, S.; Coyle, M.; Gourgiotis, L.; Csako, G. Depression and osteoporosis: A research synthesis with meta-analysis. Horm. Metab. Res. 2010, 42, 467–482. [Google Scholar] [CrossRef] [Green Version]
  70. Huang, W.-S.; Hsu, J.-W.; Huang, K.-L.; Bai, Y.-M.; Su, T.-P.; Li, C.-T.; Lin, W.-C.; Chen, T.-J.; Tsai, S.-J.; Liou, Y.-J.; et al. Post-traumatic stress disorder and risk of osteoporosis: A nationwide longitudinal study. Stress Health 2018, 34, 440–445. [Google Scholar] [CrossRef] [PubMed]
  71. El-Gabalawy, R.; Blaney, C.; Tsai, J.; Sumner, J.A.; Pietrzak, R.H. Physical health conditions associated with full and subthreshold PTSD in U.S. military veterans: Results from the National Health and Resilience in Veterans Study. J. Affect. Disord. 2018, 227, 849–853. [Google Scholar] [CrossRef]
  72. Williams, L.J.; Pasco, J.A.; Jackson, H.; Kiropoulos, L.; Stuart, A.L.; Jacka, F.N.; Berk, M. Depression as a risk factor for fracture in women: A 10 year longitudinal study. J. Affect. Disord. 2016, 192, 34–40. [Google Scholar] [CrossRef] [PubMed]
  73. Yirmiya, R.; Bab, I. Major depression is a risk factor for low bone mineral density: A meta-analysis. Biol. Psychiatry 2009, 66, 423–432. [Google Scholar] [CrossRef]
  74. Wong, S.Y.S.; Lau, E.M.C.; Lynn, H.; Leung, P.C.; Woo, J.; Cummings, S.R.; Orwoll, E. Depression and bone mineral density: Is there a relationship in elderly Asian men? Results from Mr. Os (Hong Kong). Osteoporos. Int. 2005, 16, 610–615. [Google Scholar] [CrossRef]
  75. Schweiger, U.; Deuschle, M.; Körner, A.; Lammers, C.H.; Schmider, J.; Gotthardt, U.; Holsboer, F.; Heuser, I. Low lumbar bone mineral density in patients with major depression. Am. J. Psychiatry 1994, 151, 1691–1693. [Google Scholar] [CrossRef]
  76. Esel, E.; Ozsoy, S.; Tutus, A.; Sofuoglu, S.; Kartalci, S.; Bayram, F.; Kokbudak, Z.; Kula, M. Effects of antidepressant treatment and of gender on serum leptin levels in patients with major depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2005, 29, 565–570. [Google Scholar] [CrossRef] [PubMed]
  77. Michelson, D.; Stratakis, C.; Hill, L.; Reynolds, J.; Galliven, E.; Chrousos, G.; Gold, P. Bone mineral density in women with depression. N. Engl. J. Med. 1996, 335, 1176–1181. [Google Scholar] [CrossRef]
  78. Altindag, O.; Altindag, A.; Asoglu, M.; Gunes, M.; Soran, N.; Deveci, Z. Relation of cortisol levels and bone mineral density among premenopausal women with major depression. Int. J. Clin. Pract. 2007, 61, 416–420. [Google Scholar] [CrossRef] [PubMed]
  79. Cosman, F.; Ruffing, J.; Zion, M.; Uhorchak, J.; Ralston, S.; Tendy, S.; McGuigan, F.E.A.; Lindsay, R.; Nieves, J. Determinants of stress fracture risk in United States Military Academy cadets. Bone 2013, 55, 359–366. [Google Scholar] [CrossRef] [Green Version]
  80. Vandewalle, B.; Revillion, F.; Lefebvre, J. Functional beta-adrenergic receptors in breast cancer cells. J. Cancer Res. Clin. Oncol. 1990, 116, 303–306. [Google Scholar] [CrossRef]
  81. Madden, K.S.; Szpunar, M.J.; Brown, E.B. β-Adrenergic receptors (β-AR) regulate VEGF and IL-6 production by divergent pathways in high β-AR-expressing breast cancer cell lines. Breast Cancer Res. Treat. 2011, 130, 747–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Powe, D.G.; Voss, M.J.; Habashy, H.O.; Zänker, K.S.; Green, A.R.; Ellis, I.O.; Entschladen, F. Alpha- and beta-adrenergic receptor (AR) protein expression is associated with poor clinical outcome in breast cancer: An immunohistochemical study. Breast Cancer Res. Treat. 2011, 130, 457–463. [Google Scholar] [CrossRef]
  83. Takeda, S.; Elefteriou, F.; Levasseur, R.; Liu, X.; Zhao, L.; Parker, K.L.; Armstrong, D.; Ducy, P.; Karsenty, G. Leptin regulates bone formation via the sympathetic nervous system. Cell 2002, 111, 305–317. [Google Scholar] [CrossRef] [Green Version]
  84. Elefteriou, F.; Ahn, J.D.; Takeda, S.; Starbuck, M.; Yang, X.; Liu, X.; Kondo, H.; Richards, W.G.; Bannon, T.W.; Noda, M.; et al. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature 2005, 434, 514–520. [Google Scholar] [CrossRef] [PubMed]
  85. Kajimura, D.; Hinoi, E.; Ferron, M.; Kode, A.; Riley, K.J.; Zhou, B.; Guo, X.E.; Karsenty, G. Genetic determination of the cellular basis of the sympathetic regulation of bone mass accrual. J. Exp. Med. 2011, 208, 841–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Schlienger, R.G.; Kraenzlin, M.E.; Jick, S.S.; Meier, C.R. Use of beta-blockers and risk of fractures. JAMA 2004, 292, 1326–1332. [Google Scholar] [CrossRef] [Green Version]
  87. Meisinger, C.; Heier, M.; Lang, O.; Döring, A. Beta-blocker use and risk of fractures in men and women from the general population: The MONICA/KORA Augsburg cohort study. Osteoporos. Int. 2007, 18, 1189–1195. [Google Scholar] [CrossRef]
  88. Toulis, K.A.; Hemming, K.; Stergianos, S.; Nirantharakumar, K.; Bilezikian, J.P. β-Adrenergic receptor antagonists and fracture risk: A meta-analysis of selectivity, gender, and site-specific effects. Osteoporos. Int. 2014, 25, 121–129. [Google Scholar] [CrossRef] [PubMed]
  89. Lary, C.W.; Hinton, A.C.; Nevola, K.T.; Shireman, T.I.; Motyl, K.J.; Houseknecht, K.L.; Lucas, F.L.; Hallen, S.; Zullo, A.R.; Berry, S.D.; et al. Association of Beta Blocker Use With Bone Mineral Density in the Framingham Osteoporosis Study: A Cross-Sectional Study. JBMR Plus 2020, 4, e10388. [Google Scholar] [CrossRef]
  90. Turker, S.; Karatosun, V.; Gunal, I. Beta-blockers increase bone mineral density. Clin. Orthop. Relat. Res. 2006, 443, 73–74. [Google Scholar] [CrossRef]
  91. Pasco, J.A.; Henry, M.J.; Sanders, K.M.; Kotowicz, M.A.; Seeman, E.; Nicholson, G.C. Geelong Osteoporosis Study Beta-adrenergic blockers reduce the risk of fracture partly by increasing bone mineral density: Geelong Osteoporosis Study. J. Bone Miner. Res. 2004, 19, 19–24. [Google Scholar] [CrossRef]
  92. Rejnmark, L.; Vestergaard, P.; Mosekilde, L. Treatment with beta-blockers, ACE inhibitors, and calcium-channel blockers is associated with a reduced fracture risk: A nationwide case-control study. J. Hypertens. 2006, 24, 581–589. [Google Scholar] [CrossRef] [PubMed]
  93. Bonnet, N.; Gadois, C.; McCloskey, E.; Lemineur, G.; Lespessailles, E.; Courteix, D.; Benhamou, C.L. Protective effect of beta blockers in postmenopausal women: Influence on fractures, bone density, micro and macroarchitecture. Bone 2007, 40, 1209–1216. [Google Scholar] [CrossRef]
  94. Khosla, S.; Drake, M.T.; Volkman, T.L.; Thicke, B.S.; Achenbach, S.J.; Atkinson, E.J.; Joyner, M.J.; Rosen, C.J.; Monroe, D.G.; Farr, J.N. Sympathetic β1-adrenergic signaling contributes to regulation of human bone metabolism. J. Clin. Investig. 2018, 128, 4832–4842. [Google Scholar] [CrossRef]
  95. Grytli, H.H.; Fagerland, M.W.; Fosså, S.D.; Taskén, K.A.; Håheim, L.L. Use of β-blockers is associated with prostate cancer-specific survival in prostate cancer patients on androgen deprivation therapy. Prostate 2013, 73, 250–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Grytli, H.H.; Fagerland, M.W.; Fosså, S.D.; Taskén, K.A. Association between use of β-blockers and prostate cancer-specific survival: A cohort study of 3561 prostate cancer patients with high-risk or metastatic disease. Eur. Urol. 2014, 65, 635–641. [Google Scholar] [CrossRef]
  97. Watkins, J.L.; Thaker, P.H.; Nick, A.M.; Ramondetta, L.M.; Kumar, S.; Urbauer, D.L.; Matsuo, K.; Squires, K.C.; Coleman, R.L.; Lutgendorf, S.K.; et al. Clinical impact of selective and nonselective beta-blockers on survival in patients with ovarian cancer. Cancer 2015, 121, 3444–3451. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, H.M.; Liao, Z.X.; Komaki, R.; Welsh, J.W.; O’Reilly, M.S.; Chang, J.Y.; Zhuang, Y.; Levy, L.B.; Lu, C.; Gomez, D.R. Improved survival outcomes with the incidental use of beta-blockers among patients with non-small-cell lung cancer treated with definitive radiation therapy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013, 24, 1312–1319. [Google Scholar] [CrossRef] [PubMed]
  99. Perron, L.; Bairati, I.; Harel, F.; Meyer, F. Antihypertensive drug use and the risk of prostate cancer (Canada). Cancer Causes Control 2004, 15, 535–541. [Google Scholar] [CrossRef] [PubMed]
  100. Lemeshow, S.; Sørensen, H.T.; Phillips, G.; Yang, E.V.; Antonsen, S.; Riis, A.H.; Lesinski, G.B.; Jackson, R.; Glaser, R. β-Blockers and survival among Danish patients with malignant melanoma: A population-based cohort study. Cancer Epidemiol. Biomarkers Prev. 2011, 20, 2273–2279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Aydiner, A.; Ciftci, R.; Karabulut, S.; Kilic, L. Does beta-blocker therapy improve the survival of patients with metastatic non-small cell lung cancer? Asian Pac. J. Cancer Prev. 2013, 14, 6109–6114. [Google Scholar] [CrossRef] [Green Version]
  102. De Giorgi, V.; Gandini, S.; Grazzini, M.; Benemei, S.; Marchionni, N.; Geppetti, P. Effect of β-blockers and other antihypertensive drugs on the risk of melanoma recurrence and death. Mayo Clin. Proc. 2013, 88, 1196–1203. [Google Scholar] [CrossRef]
  103. Cardwell, C.R.; Coleman, H.G.; Murray, L.J.; Entschladen, F.; Powe, D.G. Beta-blocker usage and breast cancer survival: A nested case-control study within a UK clinical practice research datalink cohort. Int. J. Epidemiol. 2013, 42, 1852–1861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Reeder, A.; Attar, M.; Nazario, L.; Bathula, C.; Zhang, A.; Hochbaum, D.; Roy, E.; Cooper, K.L.; Oesterreich, S.; Davidson, N.E.; et al. Stress hormones reduce the efficacy of paclitaxel in triple negative breast cancer through induction of DNA damage. Br. J. Cancer 2015, 112, 1461–1470. [Google Scholar] [CrossRef] [Green Version]
  105. Hara, M.R.; Sachs, B.D.; Caron, M.G.; Lefkowitz, R.J. Pharmacological blockade of a β(2)AR-β-arrestin-1 signaling cascade prevents the accumulation of DNA damage in a behavioral stress model. Cell Cycle 2013, 12, 219–224. [Google Scholar] [CrossRef] [Green Version]
  106. Hara, M.R.; Kovacs, J.J.; Whalen, E.J.; Rajagopal, S.; Strachan, R.T.; Grant, W.; Towers, A.J.; Williams, B.; Lam, C.M.; Xiao, K.; et al. A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1. Nature 2011, 477, 349–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Armaiz-Pena, G.N.; Allen, J.K.; Cruz, A.; Stone, R.L.; Nick, A.M.; Lin, Y.G.; Han, L.Y.; Mangala, L.S.; Villares, G.J.; Vivas-Mejia, P.; et al. Src activation by β-adrenoreceptors is a key switch for tumour metastasis. Nat. Commun. 2013, 4, 1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Shi, M.; Liu, D.; Duan, H.; Qian, L.; Wang, L.; Niu, L.; Zhang, H.; Yong, Z.; Gong, Z.; Song, L.; et al. The β2-adrenergic receptor and Her2 comprise a positive feedback loop in human breast cancer cells. Breast Cancer Res. Treat. 2011, 125, 351–362. [Google Scholar] [CrossRef]
  109. Gu, L.; Lau, S.K.; Loera, S.; Somlo, G.; Kane, S.E. Protein kinase A activation confers resistance to trastuzumab in human breast cancer cell lines. Clin. Cancer Res. 2009, 15, 7196–7206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Hassan, S.; Karpova, Y.; Baiz, D.; Yancey, D.; Pullikuth, A.; Flores, A.; Register, T.; Cline, J.M.; D’Agostino, R.; Danial, N.; et al. Behavioral stress accelerates prostate cancer development in mice. J. Clin. Investig. 2013, 123, 874–886. [Google Scholar] [CrossRef] [Green Version]
  111. Sastry, K.S.R.; Karpova, Y.; Prokopovich, S.; Smith, A.J.; Essau, B.; Gersappe, A.; Carson, J.P.; Weber, M.J.; Register, T.C.; Chen, Y.Q.; et al. Epinephrine protects cancer cells from apoptosis via activation of cAMP-dependent protein kinase and BAD phosphorylation. J. Biol. Chem. 2007, 282, 14094–14100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Pasquier, E.; Street, J.; Pouchy, C.; Carre, M.; Gifford, A.J.; Murray, J.; Norris, M.D.; Trahair, T.; Andre, N.; Kavallaris, M. β-blockers increase response to chemotherapy via direct antitumour and anti-angiogenic mechanisms in neuroblastoma. Br. J. Cancer 2013, 108, 2485–2494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wolter, J.K.; Wolter, N.E.; Blanch, A.; Partridge, T.; Cheng, L.; Morgenstern, D.A.; Podkowa, M.; Kaplan, D.R.; Irwin, M.S. Anti-tumor activity of the beta-adrenergic receptor antagonist propranolol in neuroblastoma. Oncotarget 2014, 5, 161–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Drell, T.L.; Joseph, J.; Lang, K.; Niggemann, B.; Zaenker, K.S.; Entschladen, F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA-MB-468 human breast carcinoma cells. Breast Cancer Res. Treat. 2003, 80, 63–70. [Google Scholar] [CrossRef]
  115. Lutgendorf, S.K.; Cole, S.; Costanzo, E.; Bradley, S.; Coffin, J.; Jabbari, S.; Rainwater, K.; Ritchie, J.M.; Yang, M.; Sood, A.K. Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines. Clin. Cancer Res. 2003, 9, 4514–4521. [Google Scholar] [PubMed]
  116. Nilsson, M.B.; Armaiz-Pena, G.; Takahashi, R.; Lin, Y.G.; Trevino, J.; Li, Y.; Jennings, N.; Arevalo, J.; Lutgendorf, S.K.; Gallick, G.E.; et al. Stress hormones regulate interleukin-6 expression by human ovarian carcinoma cells through a Src-dependent mechanism. J. Biol. Chem. 2007, 282, 29919–29926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Lang, K.; Drell, T.L.; Lindecke, A.; Niggemann, B.; Kaltschmidt, C.; Zaenker, K.S.; Entschladen, F. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int. J. Cancer 2004, 112, 231–238. [Google Scholar] [CrossRef]
  118. Pérez Piñero, C.; Bruzzone, A.; Sarappa, M.G.; Castillo, L.F.; Lüthy, I.A. Involvement of α2- and β2-adrenoceptors on breast cancer cell proliferation and tumour growth regulation. Br. J. Pharmacol. 2012, 166, 721–736. [Google Scholar] [CrossRef] [Green Version]
  119. Slotkin, T.A.; Zhang, J.; Dancel, R.; Garcia, S.J.; Willis, C.; Seidler, F.J. Beta-adrenoceptor signaling and its control of cell replication in MDA-MB-231 human breast cancer cells. Breast Cancer Res. Treat. 2000, 60, 153–166. [Google Scholar] [CrossRef]
  120. Gargiulo, L.; Copsel, S.; Rivero, E.M.; Galés, C.; Sénard, J.-M.; Lüthy, I.A.; Davio, C.; Bruzzone, A. Differential β₂-adrenergic receptor expression defines the phenotype of non-tumorigenic and malignant human breast cell lines. Oncotarget 2014, 5, 10058–10069. [Google Scholar] [CrossRef]
  121. Liu, D.; Deng, Q.; Sun, L.; Wang, T.; Yang, Z.; Chen, H.; Guo, L.; Liu, Y.; Ma, Y.; Guo, N.; et al. A Her2-let-7-β2-AR circuit affects prognosis in patients with Her2-positive breast cancer. BMC Cancer 2015, 15, 832. [Google Scholar] [CrossRef] [Green Version]
  122. Rivero, E.M.; Martinez, L.M.; Bruque, C.D.; Gargiulo, L.; Bruzzone, A.; Lüthy, I.A. Prognostic significance of α- and β2-adrenoceptor gene expression in breast cancer patients. Br. J. Clin. Pharmacol. 2019, 85, 2143–2154. [Google Scholar] [CrossRef] [PubMed]
  123. Kurozumi, S.; Kaira, K.; Matsumoto, H.; Hirakata, T.; Yokobori, T.; Inoue, K.; Horiguchi, J.; Katayama, A.; Koshi, H.; Shimizu, A.; et al. β2-Adrenergic receptor expression is associated with biomarkers of tumor immunity and predicts poor prognosis in estrogen receptor-negative breast cancer. Breast Cancer Res. Treat. 2019, 177, 603–610. [Google Scholar] [CrossRef] [PubMed]
  124. Caparica, R.; Richard, F.; Brandão, M.; Awada, A.; Sotiriou, C.; de Azambuja, E. Prognostic and Predictive Impact of Beta-2 Adrenergic Receptor Expression in HER2-Positive Breast Cancer. Clin. Breast Cancer 2020, 20, 262–273.e7. [Google Scholar] [CrossRef] [PubMed]
  125. Pacholczyk, T.; Blakely, R.D.; Amara, S.G. Expression cloning of a cocaine- and antidepressant-sensitive human noradrenaline transporter. Nature 1991, 350, 350–354. [Google Scholar] [CrossRef]
  126. Zavosh, A.; Schaefer, J.; Ferrel, A.; Figlewicz, D.P. Desipramine treatment decreases 3H-nisoxetine binding and norepinephrine transporter mRNA in SK-N-SHSY5Y cells. Brain Res. Bull. 1999, 49, 291–295. [Google Scholar] [CrossRef]
  127. Tellioglu, T.; Robertson, D. Genetic or acquired deficits in the norepinephrine transporter: Current understanding of clinical implications. Expert Rev. Mol. Med. 2001, 2001, 1–10. [Google Scholar] [CrossRef]
  128. Blakely, R.D.; Bauman, A.L. Biogenic amine transporters: Regulation in flux. Curr. Opin. Neurobiol. 2000, 10, 328–336. [Google Scholar] [CrossRef]
  129. Ma, Y.; Krueger, J.J.; Redmon, S.N.; Uppuganti, S.; Nyman, J.S.; Hahn, M.K.; Elefteriou, F. Extracellular norepinephrine clearance by the norepinephrine transporter is required for skeletal homeostasis. J. Biol. Chem. 2013, 288, 30105–30113. [Google Scholar] [CrossRef] [Green Version]
  130. Zhu, Y.; Ma, Y.; Elefteriou, F. Cortical bone is an extraneuronal site of norepinephrine uptake in adult mice. Bone Rep. 2018, 9, 188–198. [Google Scholar] [CrossRef]
  131. Mohammadpour, H.; MacDonald, C.R.; Qiao, G.; Chen, M.; Dong, B.; Hylander, B.L.; McCarthy, P.L.; Abrams, S.I.; Repasky, E.A. β2 adrenergic receptor-mediated signaling regulates the immunosuppressive potential of myeloid-derived suppressor cells. J. Clin. Investig. 2019, 129, 5537–5552. [Google Scholar] [CrossRef]
  132. Sloan, E.K.; Priceman, S.J.; Cox, B.F.; Yu, S.; Pimentel, M.A.; Tangkanangnukul, V.; Arevalo, J.M.G.; Morizono, K.; Karanikolas, B.D.W.; Wu, L.; et al. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 2010, 70, 7042–7052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Armaiz-Pena, G.N.; Gonzalez-Villasana, V.; Nagaraja, A.S.; Rodriguez-Aguayo, C.; Sadaoui, N.C.; Stone, R.L.; Matsuo, K.; Dalton, H.J.; Previs, R.A.; Jennings, N.B.; et al. Adrenergic regulation of monocyte chemotactic protein 1 leads to enhanced macrophage recruitment and ovarian carcinoma growth. Oncotarget 2015, 6, 4266–4273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cole, S.W.; Korin, Y.D.; Fahey, J.L.; Zack, J.A. Norepinephrine accelerates HIV replication via protein kinase A-dependent effects on cytokine production. J. Immunol. 1998, 161, 610–616. [Google Scholar] [PubMed]
  135. Collado-Hidalgo, A.; Sung, C.; Cole, S. Adrenergic inhibition of innate anti-viral response: PKA blockade of Type I interferon gene transcription mediates catecholamine support for HIV-1 replication. Brain Behav. Immun. 2006, 20, 552–563. [Google Scholar] [CrossRef] [PubMed]
  136. Inbar, S.; Neeman, E.; Avraham, R.; Benish, M.; Rosenne, E.; Ben-Eliyahu, S. Do stress responses promote leukemia progression? An animal study suggesting a role for epinephrine and prostaglandin-E2 through reduced NK activity. PLoS ONE 2011, 6, e19246. [Google Scholar] [CrossRef]
  137. Goldfarb, Y.; Sorski, L.; Benish, M.; Levi, B.; Melamed, R.; Ben-Eliyahu, S. Improving postoperative immune status and resistance to cancer metastasis: A combined perioperative approach of immunostimulation and prevention of excessive surgical stress responses. Ann. Surg. 2011, 253, 798–810. [Google Scholar] [CrossRef]
  138. Lee, J.-W.; Shahzad, M.M.K.; Lin, Y.G.; Armaiz-Pena, G.; Mangala, L.S.; Han, H.-D.; Kim, H.-S.; Nam, E.J.; Jennings, N.B.; Halder, J.; et al. Surgical stress promotes tumor growth in ovarian carcinoma. Clin. Cancer Res. 2009, 15, 2695–2702. [Google Scholar] [CrossRef] [Green Version]
  139. Glasner, A.; Avraham, R.; Rosenne, E.; Benish, M.; Zmora, O.; Shemer, S.; Meiboom, H.; Ben-Eliyahu, S. Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a beta-adrenergic antagonist and a cyclooxygenase-2 inhibitor. J. Immunol. 2010, 184, 2449–2457. [Google Scholar] [CrossRef] [Green Version]
  140. Magnon, C.; Hall, S.J.; Lin, J.; Xue, X.; Gerber, L.; Freedland, S.J.; Frenette, P.S. Autonomic nerve development contributes to prostate cancer progression. Science 2013, 341, 1236361. [Google Scholar] [CrossRef] [Green Version]
  141. Campbell, J.P.; Karolak, M.R.; Ma, Y.; Perrien, D.S.; Masood-Campbell, S.K.; Penner, N.L.; Munoz, S.A.; Zijlstra, A.; Yang, X.; Sterling, J.A.; et al. Stimulation of host bone marrow stromal cells by sympathetic nerves promotes breast cancer bone metastasis in mice. PLoS Biol. 2012, 10, e1001363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Katayama, Y.; Battista, M.; Kao, W.-M.; Hidalgo, A.; Peired, A.J.; Thomas, S.A.; Frenette, P.S. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 2006, 124, 407–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zheng, H.; Fu, G.; Dai, T.; Huang, H. Migration of endothelial progenitor cells mediated by stromal cell-derived factor-1alpha/CXCR4 via PI3K/Akt/eNOS signal transduction pathway. J. Cardiovasc. Pharmacol. 2007, 50, 274–280. [Google Scholar] [CrossRef]
  144. Elefteriou, F. Regulation of bone remodeling by the central and peripheral nervous system. Arch. Biochem. Biophys. 2008, 473, 231–236. [Google Scholar] [CrossRef] [Green Version]
  145. Yao, Q.; Liang, H.; Huang, B.; Xiang, L.; Wang, T.; Xiong, Y.; Yang, B.; Guo, Y.; Gong, P. Beta-adrenergic signaling affect osteoclastogenesis via osteocytic MLO-Y4 cells’ RANKL production. Biochem. Biophys. Res. Commun. 2017, 488, 634–640. [Google Scholar] [CrossRef]
  146. Jiao, K.; Niu, L.-N.; Li, Q.; Ren, G.; Zhao, C.; Liu, Y.; Tay, F.R.; Wang, M. β2-Adrenergic signal transduction plays a detrimental role in subchondral bone loss of temporomandibular joint in osteoarthritis. Sci. Rep. 2015, 5, 12593. [Google Scholar] [CrossRef] [Green Version]
  147. Ferrara, N.; Davis-Smyth, T. The biology of vascular endothelial growth factor. Endocr. Rev. 1997, 18, 4–25. [Google Scholar] [CrossRef]
  148. Achen, M.G.; Jeltsch, M.; Kukk, E.; Mäkinen, T.; Vitali, A.; Wilks, A.F.; Alitalo, K.; Stacker, S.A. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc. Natl. Acad. Sci. USA 1998, 95, 548–553. [Google Scholar] [CrossRef] [Green Version]
  149. Ogawa, S.; Oku, A.; Sawano, A.; Yamaguchi, S.; Yazaki, Y.; Shibuya, M. A novel type of vascular endothelial growth factor, VEGF-E (NZ-7 VEGF), preferentially utilizes KDR/Flk-1 receptor and carries a potent mitotic activity without heparin-binding domain. J. Biol. Chem. 1998, 273, 31273–31282. [Google Scholar] [CrossRef] [Green Version]
  150. Meyer, M.; Clauss, M.; Lepple-Wienhues, A.; Waltenberger, J.; Augustin, H.G.; Ziche, M.; Lanz, C.; Büttner, M.; Rziha, H.J.; Dehio, C. A novel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. EMBO J. 1999, 18, 363–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Weidner, N.; Semple, J.P.; Welch, W.R.; Folkman, J. Tumor angiogenesis and metastasis—Correlation in invasive breast carcinoma. N. Engl. J. Med. 1991, 324, 1–8. [Google Scholar] [CrossRef]
  152. Yang, E.V.; Kim, S.; Donovan, E.L.; Chen, M.; Gross, A.C.; Webster Marketon, J.I.; Barsky, S.H.; Glaser, R. Norepinephrine upregulates VEGF, IL-8, and IL-6 expression in human melanoma tumor cell lines: Implications for stress-related enhancement of tumor progression. Brain Behav. Immun. 2009, 23, 267–275. [Google Scholar] [CrossRef] [Green Version]
  153. Liu, J.; Deng, G.-H.; Zhang, J.; Wang, Y.; Xia, X.-Y.; Luo, X.-M.; Deng, Y.-T.; He, S.-S.; Mao, Y.-Y.; Peng, X.-C.; et al. The effect of chronic stress on anti-angiogenesis of sunitinib in colorectal cancer models. Psychoneuroendocrinology 2015, 52, 130–142. [Google Scholar] [CrossRef] [PubMed]
  154. Xie, H.; Li, C.; He, Y.; Griffin, R.; Ye, Q.; Li, L. Chronic stress promotes oral cancer growth and angiogenesis with increased circulating catecholamine and glucocorticoid levels in a mouse model. Oral Oncol. 2015, 51, 991–997. [Google Scholar] [CrossRef] [PubMed]
  155. Kusumbe, A.P.; Ramasamy, S.K.; Adams, R.H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014, 507, 323–328. [Google Scholar] [CrossRef] [PubMed]
  156. Kusumbe, A.P.; Ramasamy, S.K.; Itkin, T.; Mäe, M.A.; Langen, U.H.; Betsholtz, C.; Lapidot, T.; Adams, R.H. Age-dependent modulation of vascular niches for haematopoietic stem cells. Nature 2016, 532, 380–384. [Google Scholar] [CrossRef] [Green Version]
  157. Mulcrone, P.L.; Campbell, J.P.; Clément-Demange, L.; Anbinder, A.L.; Merkel, A.R.; Brekken, R.A.; Sterling, J.A.; Elefteriou, F. Skeletal Colonization by Breast Cancer Cells Is Stimulated by an Osteoblast and β2AR-Dependent Neo-Angiogenic Switch. J. Bone Miner. Res. 2017, 32, 1442–1454. [Google Scholar] [CrossRef]
  158. Clément-Demange, L.; Mulcrone, P.L.; Tabarestani, T.Q.; Sterling, J.A.; Elefteriou, F. β2ARs stimulation in osteoblasts promotes breast cancer cell adhesion to bone marrow endothelial cells in an IL-1β and selectin-dependent manner. J. Bone Oncol. 2018, 13, 1–10. [Google Scholar] [CrossRef]
  159. Gopinathan, G.; Milagre, C.; Pearce, O.M.T.; Reynolds, L.E.; Hodivala-Dilke, K.; Leinster, D.A.; Zhong, H.; Hollingsworth, R.E.; Thompson, R.; Whiteford, J.R.; et al. Interleukin-6 Stimulates Defective Angiogenesis. Cancer Res. 2015, 75, 3098–3107. [Google Scholar] [CrossRef] [Green Version]
  160. Catar, R.; Witowski, J.; Zhu, N.; Lücht, C.; Derrac Soria, A.; Uceda Fernandez, J.; Chen, L.; Jones, S.A.; Fielding, C.A.; Rudolf, A.; et al. IL-6 Trans-Signaling Links Inflammation with Angiogenesis in the Peritoneal Membrane. J. Am. Soc. Nephrol. 2017, 28, 1188–1199. [Google Scholar] [CrossRef] [Green Version]
  161. Shinriki, S.; Jono, H.; Ueda, M.; Ota, K.; Ota, T.; Sueyoshi, T.; Oike, Y.; Ibusuki, M.; Hiraki, A.; Nakayama, H.; et al. Interleukin-6 signalling regulates vascular endothelial growth factor-C synthesis and lymphangiogenesis in human oral squamous cell carcinoma. J. Pathol. 2011, 225, 142–150. [Google Scholar] [CrossRef]
  162. Jiang, X.-P.; Yang, D.C.; Elliott, R.L.; Head, J.F. Down-regulation of expression of interleukin-6 and its receptor results in growth inhibition of MCF-7 breast cancer cells. Anticancer Res. 2011, 31, 2899–2906. [Google Scholar]
  163. Studebaker, A.W.; Storci, G.; Werbeck, J.L.; Sansone, P.; Sasser, A.K.; Tavolari, S.; Huang, T.; Chan, M.W.Y.; Marini, F.C.; Rosol, T.J.; et al. Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008, 68, 9087–9095. [Google Scholar] [CrossRef] [Green Version]
  164. Strilic, B.; Offermanns, S. Intravascular Survival and Extravasation of Tumor Cells. Cancer Cell 2017, 32, 282–293. [Google Scholar] [CrossRef] [Green Version]
  165. Biancone, L.; Araki, M.; Araki, K.; Vassalli, P.; Stamenkovic, I. Redirection of tumor metastasis by expression of E-selectin in vivo. J. Exp. Med. 1996, 183, 581–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Bendas, G.; Borsig, L. Cancer cell adhesion and metastasis: Selectins, integrins, and the inhibitory potential of heparins. Int. J. Cell Biol. 2012, 2012, 676731. [Google Scholar] [CrossRef]
  167. Sökeland, G.; Schumacher, U. The functional role of integrins during intra- and extravasation within the metastatic cascade. Mol. Cancer 2019, 18, 12. [Google Scholar] [CrossRef] [PubMed]
  168. Makó, V.; Czúcz, J.; Weiszhár, Z.; Herczenik, E.; Matkó, J.; Prohászka, Z.; Cervenak, L. Proinflammatory activation pattern of human umbilical vein endothelial cells induced by IL-1β, TNF-α, and LPS. Cytometry A 2010, 77, 962–970. [Google Scholar] [CrossRef] [PubMed]
  169. Bokhari, S.M.; Zhou, L.; Karasek, M.A.; Paturi, S.G.; Chaudhuri, V. Regulation of skin microvasculature angiogenesis, cell migration, and permeability by a specific inhibitor of PKCalpha. J. Investig. Dermatol. 2006, 126, 460–467. [Google Scholar] [CrossRef] [PubMed]
  170. Voronov, E.; Dotan, S.; Krelin, Y.; Song, X.; Elkabets, M.; Carmi, Y.; Rider, P.; Idan, C.; Romzova, M.; Kaplanov, I.; et al. Unique Versus Redundant Functions of IL-1α and IL-1β in the Tumor Microenvironment. Front. Immunol. 2013, 4, 177. [Google Scholar] [CrossRef]
  171. Khatib, A.-M.; Auguste, P.; Fallavollita, L.; Wang, N.; Samani, A.; Kontogiannea, M.; Meterissian, S.; Brodt, P. Characterization of the host proinflammatory response to tumor cells during the initial stages of liver metastasis. Am. J. Pathol. 2005, 167, 749–759. [Google Scholar] [CrossRef] [Green Version]
  172. Okahara, H.; Yagita, H.; Miyake, K.; Okumura, K. Involvement of very late activation antigen 4 (VLA-4) and vascular cell adhesion molecule 1 (VCAM-1) in tumor necrosis factor alpha enhancement of experimental metastasis. Cancer Res. 1994, 54, 3233–3236. [Google Scholar] [PubMed]
  173. Giavazzi, R.; Garofalo, A.; Bani, M.R.; Abbate, M.; Ghezzi, P.; Boraschi, D.; Mantovani, A.; Dejana, E. Interleukin 1-induced augmentation of experimental metastases from a human melanoma in nude mice. Cancer Res. 1990, 50, 4771–4775. [Google Scholar] [PubMed]
  174. Lauri, D.; Bertomeu, M.C.; Orr, F.W.; Bastida, E.; Sauder, D.; Buchanan, M.R. Interleukin-1 increases tumor cell adhesion to endothelial cells through an RGD dependent mechanism: In vitro and in vivo studies. Clin. Exp. Metastasis 1990, 8, 27–32. [Google Scholar] [CrossRef]
  175. Arguello, F.; Baggs, R.B.; Graves, B.T.; Harwell, S.E.; Cohen, H.J.; Frantz, C.N. Effect of IL-1 on experimental bone/bone-marrow metastases. Int. J. Cancer 1992, 52, 802–807. [Google Scholar] [CrossRef]
  176. Voronov, E.; Shouval, D.S.; Krelin, Y.; Cagnano, E.; Benharroch, D.; Iwakura, Y.; Dinarello, C.A.; Apte, R.N. IL-1 is required for tumor invasiveness and angiogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 2645–2650. [Google Scholar] [CrossRef] [Green Version]
  177. Udagawa, N.; Takahashi, N.; Jimi, E.; Matsuzaki, K.; Tsurukai, T.; Itoh, K.; Nakagawa, N.; Yasuda, H.; Goto, M.; Tsuda, E.; et al. Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colony-stimulating factor: Receptor activator of NF-kappa B ligand. Bone 1999, 25, 517–523. [Google Scholar] [CrossRef]
  178. Tatsumi, S.; Ishii, K.; Amizuka, N.; Li, M.; Kobayashi, T.; Kohno, K.; Ito, M.; Takeshita, S.; Ikeda, K. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 2007, 5, 464–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Horwood, N.J.; Kartsogiannis, V.; Quinn, J.M.; Romas, E.; Martin, T.J.; Gillespie, M.T. Activated T lymphocytes support osteoclast formation in vitro. Biochem. Biophys. Res. Commun. 1999, 265, 144–150. [Google Scholar] [CrossRef]
  180. Komuro, H.; Olee, T.; Kühn, K.; Quach, J.; Brinson, D.C.; Shikhman, A.; Valbracht, J.; Creighton-Achermann, L.; Lotz, M. The osteoprotegerin/receptor activator of nuclear factor kappaB/receptor activator of nuclear factor kappaB ligand system in cartilage. Arthritis Rheum. 2001, 44, 2768–2776. [Google Scholar] [CrossRef]
  181. Kwan Tat, S.; Amiable, N.; Pelletier, J.-P.; Boileau, C.; Lajeunesse, D.; Duval, N.; Martel-Pelletier, J. Modulation of OPG, RANK and RANKL by human chondrocytes and their implication during osteoarthritis. Rheumatology 2009, 48, 1482–1490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Feng, X.; Teitelbaum, S.L. Osteoclasts: New Insights. Bone Res. 2013, 1, 11–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Jones, D.H.; Nakashima, T.; Sanchez, O.H.; Kozieradzki, I.; Komarova, S.V.; Sarosi, I.; Morony, S.; Rubin, E.; Sarao, R.; Hojilla, C.V.; et al. Regulation of cancer cell migration and bone metastasis by RANKL. Nature 2006, 440, 692–696. [Google Scholar] [CrossRef] [Green Version]
  184. Chen, L.-M.; Kuo, C.-H.; Lai, T.-Y.; Lin, Y.-M.; Su, C.-C.; Hsu, H.-H.; Tsai, F.-J.; Tsai, C.-H.; Huang, C.-Y.; Tang, C.-H. RANKL increases migration of human lung cancer cells through intercellular adhesion molecule-1 up-regulation. J. Cell. Biochem. 2011, 112, 933–941. [Google Scholar] [CrossRef]
  185. Armstrong, A.P.; Miller, R.E.; Jones, J.C.; Zhang, J.; Keller, E.T.; Dougall, W.C. RANKL acts directly on RANK-expressing prostate tumor cells and mediates migration and expression of tumor metastasis genes. Prostate 2008, 68, 92–104. [Google Scholar] [CrossRef] [Green Version]
  186. Zhang, L.; Teng, Y.; Zhang, Y.; Liu, J.; Xu, L.; Qu, J.; Hou, K.; Yang, X.; Liu, Y.; Qu, X. Receptor activator for nuclear factor κ B expression predicts poor prognosis in breast cancer patients with bone metastasis but not in patients with visceral metastasis. J. Clin. Pathol. 2012, 65, 36–40. [Google Scholar] [CrossRef] [PubMed]
  187. Ibrahim, T.; Sacanna, E.; Gaudio, M.; Mercatali, L.; Scarpi, E.; Zoli, W.; Serra, P.; Ricci, R.; Serra, L.; Kang, Y.; et al. Role of RANK, RANKL, OPG, and CXCR4 tissue markers in predicting bone metastases in breast cancer patients. Clin. Breast Cancer 2011, 11, 369–375. [Google Scholar] [CrossRef]
  188. Nakashima, T.; Hayashi, M.; Fukunaga, T.; Kurata, K.; Oh-Hora, M.; Feng, J.Q.; Bonewald, L.F.; Kodama, T.; Wutz, A.; Wagner, E.F.; et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat. Med. 2011, 17, 1231–1234. [Google Scholar] [CrossRef]
  189. Axmann, R.; Böhm, C.; Krönke, G.; Zwerina, J.; Smolen, J.; Schett, G. Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum. 2009, 60, 2747–2756. [Google Scholar] [CrossRef]
  190. Kudo, O.; Sabokbar, A.; Pocock, A.; Itonaga, I.; Fujikawa, Y.; Athanasou, N.A. Interleukin-6 and interleukin-11 support human osteoclast formation by a RANKL-independent mechanism. Bone 2003, 32, 1–7. [Google Scholar] [CrossRef]
  191. Devlin, R.D.; Reddy, S.V.; Savino, R.; Ciliberto, G.; Roodman, G.D. IL-6 mediates the effects of IL-1 or TNF, but not PTHrP or 1,25(OH)2D3, on osteoclast-like cell formation in normal human bone marrow cultures. J. Bone Miner. Res. 1998, 13, 393–399. [Google Scholar] [CrossRef] [PubMed]
  192. Elefteriou, F. Neuronal signaling and the regulation of bone remodeling. Cell. Mol. Life Sci. 2005, 62, 2339–2349. [Google Scholar] [CrossRef] [PubMed]
  193. Aitken, S.J.; Landao-Bassonga, E.; Ralston, S.H.; Idris, A.I. Beta2-adrenoreceptor ligands regulate osteoclast differentiation in vitro by direct and indirect mechanisms. Arch. Biochem. Biophys. 2009, 482, 96–103. [Google Scholar] [CrossRef] [PubMed]
  194. Arai, M.; Nagasawa, T.; Koshihara, Y.; Yamamoto, S.; Togari, A. Effects of beta-adrenergic agonists on bone-resorbing activity in human osteoclast-like cells. Biochim. Biophys. Acta 2003, 1640, 137–142. [Google Scholar] [CrossRef] [Green Version]
  195. Sanders, J.L.; Chattopadhyay, N.; Kifor, O.; Yamaguchi, T.; Butters, R.R.; Brown, E.M. Extracellular calcium-sensing receptor expression and its potential role in regulating parathyroid hormone-related peptide secretion in human breast cancer cell lines. Endocrinology 2000, 141, 4357–4364. [Google Scholar] [CrossRef]
  196. Mundy, G.R. Metastasis to bone: Causes, consequences and therapeutic opportunities. Nat. Rev. Cancer 2002, 2, 584–593. [Google Scholar] [CrossRef]
  197. Guise, T.A.; Yin, J.J.; Taylor, S.D.; Kumagai, Y.; Dallas, M.; Boyce, B.F.; Yoneda, T.; Mundy, G.R. Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J. Clin. Investig. 1996, 98, 1544–1549. [Google Scholar] [CrossRef]
  198. Weilbaecher, K.N.; Guise, T.A.; McCauley, L.K. Cancer to bone: A fatal attraction. Nat. Rev. Cancer 2011, 11, 411–425. [Google Scholar] [CrossRef]
  199. Kagiya, T. MicroRNAs and Osteolytic Bone Metastasis: The Roles of MicroRNAs in Tumor-Induced Osteoclast Differentiation. J. Clin. Med. 2015, 4, 1741–1752. [Google Scholar] [CrossRef] [Green Version]
  200. Chiechi, A.; Waning, D.L.; Stayrook, K.R.; Buijs, J.T.; Guise, T.A.; Mohammad, K.S. Role of TGF-β in breast cancer bone metastases. Adv. Biosci. Biotechnol. 2013, 4, 15–30. [Google Scholar] [CrossRef] [Green Version]
  201. Kakonen, S.-M.; Selander, K.S.; Chirgwin, J.M.; Yin, J.J.; Burns, S.; Rankin, W.A.; Grubbs, B.G.; Dallas, M.; Cui, Y.; Guise, T.A. Transforming growth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J. Biol. Chem. 2002, 277, 24571–24578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Buijs, J.T.; Stayrook, K.R.; Guise, T.A. TGF-β in the Bone Microenvironment: Role in Breast Cancer Metastases. Cancer Microenviron. 2011, 4, 261–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Yin, J.J.; Selander, K.; Chirgwin, J.M.; Dallas, M.; Grubbs, B.G.; Wieser, R.; Massagué, J.; Mundy, G.R.; Guise, T.A. TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J. Clin. Investig. 1999, 103, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Biswas, S.; Nyman, J.S.; Alvarez, J.; Chakrabarti, A.; Ayres, A.; Sterling, J.; Edwards, J.; Rana, T.; Johnson, R.; Perrien, D.S.; et al. Anti-transforming growth factor ß antibody treatment rescues bone loss and prevents breast cancer metastasis to bone. PLoS ONE 2011, 6, e27090. [Google Scholar] [CrossRef]
  205. Ganapathy, V.; Ge, R.; Grazioli, A.; Xie, W.; Banach-Petrosky, W.; Kang, Y.; Lonning, S.; McPherson, J.; Yingling, J.M.; Biswas, S.; et al. Targeting the Transforming Growth Factor-beta pathway inhibits human basal-like breast cancer metastasis. Mol. Cancer 2010, 9, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Korpal, M.; Yan, J.; Lu, X.; Xu, S.; Lerit, D.A.; Kang, Y. Imaging transforming growth factor-beta signaling dynamics and therapeutic response in breast cancer bone metastasis. Nat. Med. 2009, 15, 960–966. [Google Scholar] [CrossRef]
  207. Guise, T.A.; Mundy, G.R. Cancer and Bone*. Endocr. Rev. 1998, 19, 18–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Guise, T.A.; Chirgwin, J.M. Transforming growth factor-beta in osteolytic breast cancer bone metastases. Clin. Orthop. Relat. Res. 2003, S32–S38. [Google Scholar] [CrossRef]
  209. Waning, D.L.; Mohammad, K.S.; Reiken, S.; Xie, W.; Andersson, D.C.; John, S.; Chiechi, A.; Wright, L.E.; Umanskaya, A.; Niewolna, M.; et al. Excess TGF-β mediates muscle weakness associated with bone metastases in mice. Nat. Med. 2015, 21, 1262–1271. [Google Scholar] [CrossRef]
  210. Regan, J.N.; Mikesell, C.; Reiken, S.; Xu, H.; Marks, A.R.; Mohammad, K.S.; Guise, T.A.; Waning, D.L. Osteolytic Breast Cancer Causes Skeletal Muscle Weakness in an Immunocompetent Syngeneic Mouse Model. Front. Endocrinol. 2017, 8, 358. [Google Scholar] [CrossRef] [Green Version]
  211. Regan, J.N.; Trivedi, T.; Guise, T.A.; Waning, D.L. The Role of TGFβ in Bone-Muscle Crosstalk. Curr. Osteoporos. Rep. 2017, 15, 18–23. [Google Scholar] [CrossRef] [PubMed]
  212. Madel, M.-B.; Ibáñez, L.; Wakkach, A.; de Vries, T.J.; Teti, A.; Apparailly, F.; Blin-Wakkach, C. Immune Function and Diversity of Osteoclasts in Normal and Pathological Conditions. Front. Immunol. 2019, 10, 1408. [Google Scholar] [CrossRef]
  213. Ibáñez, L.; Abou-Ezzi, G.; Ciucci, T.; Amiot, V.; Belaïd, N.; Obino, D.; Mansour, A.; Rouleau, M.; Wakkach, A.; Blin-Wakkach, C. Inflammatory Osteoclasts Prime TNFα-Producing CD4+ T Cells and Express CX3 CR1. J. Bone Miner. Res. 2016, 31, 1899–1908. [Google Scholar] [CrossRef] [PubMed]
  214. Buchwald, Z.S.; Kiesel, J.R.; DiPaolo, R.; Pagadala, M.S.; Aurora, R. Osteoclast activated FoxP3+ CD8+ T-cells suppress bone resorption in vitro. PLoS ONE 2012, 7, e38199. [Google Scholar] [CrossRef] [Green Version]
  215. Kiesel, J.R.; Buchwald, Z.S.; Aurora, R. Cross-presentation by osteoclasts induces FoxP3 in CD8+ T cells. J. Immunol. 2009, 182, 5477–5487. [Google Scholar] [CrossRef] [Green Version]
  216. Madel, M.-B.; Ibáñez, L.; Ciucci, T.; Halper, J.; Rouleau, M.; Boutin, A.; Hue, C.; Duroux-Richard, I.; Apparailly, F.; Garchon, H.-J.; et al. Dissecting the phenotypic and functional heterogeneity of mouse inflammatory osteoclasts by the expression of Cx3cr1. Elife 2020, 9. [Google Scholar] [CrossRef] [PubMed]
  217. An, G.; Acharya, C.; Feng, X.; Wen, K.; Zhong, M.; Zhang, L.; Munshi, N.C.; Qiu, L.; Tai, Y.-T.; Anderson, K.C. Osteoclasts promote immune suppressive microenvironment in multiple myeloma: Therapeutic implication. Blood 2016, 128, 1590–1603. [Google Scholar] [CrossRef] [Green Version]
  218. Ell, B.; Mercatali, L.; Ibrahim, T.; Campbell, N.; Schwarzenbach, H.; Pantel, K.; Amadori, D.; Kang, Y. Tumor-induced osteoclast miRNA changes as regulators and biomarkers of osteolytic bone metastasis. Cancer Cell 2013, 24, 542–556. [Google Scholar] [CrossRef] [Green Version]
  219. Lee, D.Y.; Deng, Z.; Wang, C.-H.; Yang, B.B. MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression. Proc. Natl. Acad. Sci. USA 2007, 104, 20350–20355. [Google Scholar] [CrossRef] [Green Version]
  220. O’Day, E.; Lal, A. MicroRNAs and their target gene networks in breast cancer. Breast Cancer Res. 2010, 12, 201. [Google Scholar] [CrossRef] [Green Version]
  221. Gainford, M.C.; Dranitsaris, G.; Clemons, M. Recent developments in bisphosphonates for patients with metastatic breast cancer. BMJ 2005, 330, 769–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Wong, M.; Pavlakis, N. Optimal management of bone metastases in breast cancer patients. Breast Cancer 2011, 3, 35–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Clines, G.A.; Guise, T.A. Molecular mechanisms and treatment of bone metastasis. Expert Rev. Mol. Med. 2008, 10, e7. [Google Scholar] [CrossRef]
  224. Barre, P.V.; Padmaja, G.; Rana, S. Tiamongla Stress and Quality of Life in Cancer Patients: Medical and Psychological Intervention. Indian J. Psychol. Med. 2018, 40, 232–238. [Google Scholar] [CrossRef]
  225. Weber, D.; O’Brien, K. Cancer and Cancer-Related Fatigue and the Interrelationships with Depression, Stress, and Inflammation. J. Evid. Based Complement. Altern. Med. 2017, 22, 502–512. [Google Scholar] [CrossRef]
  226. Porcerelli, J.H.; Bornstein, R.F.; Porcerelli, D.; Arterbery, V.E. The complex role of personality in cancer treatment: Impact of dependency-detachment on health status, distress, and physician-patient relationship. J. Nerv. Ment. Dis. 2015, 203, 264–268. [Google Scholar] [CrossRef] [Green Version]
  227. Thornton, L.M.; Andersen, B.L.; Carson, W.E. Immune, endocrine, and behavioral precursors to breast cancer recurrence: A case-control analysis. Cancer Immunol. Immunother. 2008, 57, 1471–1481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Baqutayan, S.M.S. The effect of anxiety on breast cancer patients. Indian J. Psychol. Med. 2012, 34, 119–123. [Google Scholar] [CrossRef] [Green Version]
  229. Park, C.S.; Lee, H.-Y. Clinical utility of sympathetic blockade in cardiovascular disease management. Expert Rev. Cardiovasc. Ther. 2017, 15, 277–288. [Google Scholar] [CrossRef]
  230. Fryzek, J.P.; Poulsen, A.H.; Lipworth, L.; Pedersen, L.; Nørgaard, M.; McLaughlin, J.K.; Friis, S. A cohort study of antihypertensive medication use and breast cancer among Danish women. Breast Cancer Res. Treat. 2006, 97, 231–236. [Google Scholar] [CrossRef]
  231. Li, C.I.; Malone, K.E.; Weiss, N.S.; Boudreau, D.M.; Cushing-Haugen, K.L.; Daling, J.R. Relation between use of antihypertensive medications and risk of breast carcinoma among women ages 65–79 years. Cancer 2003, 98, 1504–1513. [Google Scholar] [CrossRef] [PubMed]
  232. Le, C.P.; Nowell, C.J.; Kim-Fuchs, C.; Botteri, E.; Hiller, J.G.; Ismail, H.; Pimentel, M.A.; Chai, M.G.; Karnezis, T.; Rotmensz, N.; et al. Chronic stress in mice remodels lymph vasculature to promote tumour cell dissemination. Nat. Commun. 2016, 7, 10634. [Google Scholar] [CrossRef] [Green Version]
  233. Spera, G.; Fresco, R.; Fung, H.; Dyck, J.R.B.; Pituskin, E.; Paterson, I.; Mackey, J.R. Beta blockers and improved progression-free survival in patients with advanced HER2 negative breast cancer: A retrospective analysis of the ROSE/TRIO-012 study. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1836–1841. [Google Scholar] [CrossRef]
  234. Montoya, A.; Amaya, C.N.; Belmont, A.; Diab, N.; Trevino, R.; Villanueva, G.; Rains, S.; Sanchez, L.A.; Badri, N.; Otoukesh, S.; et al. Use of non-selective β-blockers is associated with decreased tumor proliferative indices in early stage breast cancer. Oncotarget 2017, 8, 6446–6460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Pasquier, E.; Ciccolini, J.; Carre, M.; Giacometti, S.; Fanciullino, R.; Pouchy, C.; Montero, M.-P.; Serdjebi, C.; Kavallaris, M.; Andre, N. Propranolol potentiates the anti-angiogenic effects and anti-tumor efficacy of chemotherapy agents: Implication in breast cancer treatment. Oncotarget 2011, 2. [Google Scholar] [CrossRef] [Green Version]
  236. Khosla, S.; Hofbauer, L.C. Osteoporosis treatment: Recent developments and ongoing challenges. Lancet Diabetes Endocrinol. 2017, 5, 898–907. [Google Scholar] [CrossRef] [Green Version]
  237. Munzone, E.; Botteri, E.; Rotmensz, N.; Cipolla, C.M.; Zanelotti, A.; Adamoli, L.; Viale, G.; Goldhirsch, A.; Gandini, S. Prognostic effect of beta blockers (BB) in triple-negative breast cancer (TNBC) patients. J. Clin. Oncol. 2013, 31, 1061. [Google Scholar] [CrossRef]
  238. Modi, N.D.; Tan, J.Q.E.; Rowland, A.; Koczwara, B.; Kichenadasse, G.; McKinnon, R.A.; Wiese, M.D.; Sorich, M.J.; Hopkins, A.M. The Influence of Pre-Existing Beta-Blockers Use on Survival Outcomes in HER2 Positive Advanced Breast Cancer: Pooled Analysis of Clinical Trial Data. Front. Oncol. 2020, 10, 1130. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the different mechanisms by which sympathetic nerve activation promotes the establishment of metastatic breast cancer cells in the skeleton. Secretion of NE from sympathetic nerve terminals in the bone microenvironment stimulates post-synaptic βARs on various target cells, including bone and cancer cells. βAR activation on osteoblasts results in increased IL-1β, VEGF-a, and RANKL secretion, leading to endothelial activation and neoangiogenesis as well as tumor stromal retention and increased osteoclastogenesis, respectively. It also promotes tumor growth through increased osteoclastogenesis, bone resorption, and subsequent release of bone-derived growth factors such as TGFβ. βAR, β-adrenergic receptor; OB, osteoblast; OCL, osteoclast; NE, norepinephrine; RANK, receptor activator of nuclear factor kappa-Β; RANKL, receptor activator of nuclear factor kappa-Β ligand; VEGF-a, vascular endothelial growth factor A; VEGFR, vascular endothelial growth factor receptor.
Figure 1. Overview of the different mechanisms by which sympathetic nerve activation promotes the establishment of metastatic breast cancer cells in the skeleton. Secretion of NE from sympathetic nerve terminals in the bone microenvironment stimulates post-synaptic βARs on various target cells, including bone and cancer cells. βAR activation on osteoblasts results in increased IL-1β, VEGF-a, and RANKL secretion, leading to endothelial activation and neoangiogenesis as well as tumor stromal retention and increased osteoclastogenesis, respectively. It also promotes tumor growth through increased osteoclastogenesis, bone resorption, and subsequent release of bone-derived growth factors such as TGFβ. βAR, β-adrenergic receptor; OB, osteoblast; OCL, osteoclast; NE, norepinephrine; RANK, receptor activator of nuclear factor kappa-Β; RANKL, receptor activator of nuclear factor kappa-Β ligand; VEGF-a, vascular endothelial growth factor A; VEGFR, vascular endothelial growth factor receptor.
Cancers 13 02887 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Madel, M.-B.; Elefteriou, F. Mechanisms Supporting the Use of Beta-Blockers for the Management of Breast Cancer Bone Metastasis. Cancers 2021, 13, 2887. https://doi.org/10.3390/cancers13122887

AMA Style

Madel M-B, Elefteriou F. Mechanisms Supporting the Use of Beta-Blockers for the Management of Breast Cancer Bone Metastasis. Cancers. 2021; 13(12):2887. https://doi.org/10.3390/cancers13122887

Chicago/Turabian Style

Madel, Maria-Bernadette, and Florent Elefteriou. 2021. "Mechanisms Supporting the Use of Beta-Blockers for the Management of Breast Cancer Bone Metastasis" Cancers 13, no. 12: 2887. https://doi.org/10.3390/cancers13122887

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