Seminars in Cancer Biology 18 (2008) 150–163

Review

Neurobiology of cancer: Interactions between nervous, endocrine and immune systems as a base for monitoring and modulating the tumorigenesis by the brain Boris Mravec a,b,∗ , Yori Gidron c , Ivan Hulin a a

Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic b Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlarska 3, 833 06 Bratislava, Slovak Republic c Department of Psychology and Health, University of Tilburg, 5000 LE Tilburg, The Netherlands

Abstract The interactions between the nervous, endocrine and immune systems are studied intensively. The communication between immune and cancer cells, and multilevel and bi-directional interactions between the nervous and immune systems constitute the basis for a hypothesis assuming that the brain might monitor and modulate the processes associated with the genesis and progression of cancer. The aim of this article is to describe the data supporting this hypothesis. © 2007 Elsevier Ltd. All rights reserved. Keywords: Cholinergic anti-inflammatory pathway; Neurobiology of cancer; Neurotransmitters; Innervation of the tumors; Vagus nerve

Contents 1. 2.

3.

Neuro-endocrine–immune interactions as a base for neurobiology of peripheral diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Nervous system and tumorigenesis (tumor progression): facts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Clinical and experimental data supporting the assumption that the brain monitors and modulates tumorigenesis . . . . . . . . . . . . . 2.2. The impact of psychosocial factors on cancer incidence and progression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Nervous system and tumorigenesis: questions, assumptions, and hypotheses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. The tight interconnection between the immune and nervous systems elicits a question as to whether the brain might monitor and modulate the process of tumorigenesis, and if yes, at which level of the nervous system is it involved. . . . . . . . . . 3.2. If we assume that the brain can modulate the tumorigenesis then the brain must be informed about cancer. How is this information transmitted to the brain? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2.1. Indirect transmission of information about cancer to the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2.2. Direct transmission of information about cancer to the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3. How are brain functions influenced by cancer? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4. The monitoring of tumorigenesis by the brain as a new diagnostic approach. Could any of the functional imaging techniques (e.g. fMRI, PET) be able to detect an altered response in certain brain areas in cancer patients, especially after exposing them to experimental stimuli? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.5. Mechanisms potentially enabling the brain to modulate tumorigenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.5.1. Indirect modulation of cancer by the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.5.2. Direct modulation of cancer by the brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.6. Could the cholinergic anti-inflammatory pathway take part in the modulation of tumor growth? . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.7. Is there a role for axon reflexes in modulation of tumorigenesis? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .



Corresponding author at: Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovak Republic. Tel.: +421 2 59357389; fax: +421 2 59357601. E-mail address: [email protected] (B. Mravec). 1044-579X/$ – see front matter © 2007 Elsevier Ltd. All rights reserved. doi:10.1016/j.semcancer.2007.12.002

151 151 151 152 154 154 154 154 155 155

155 155 155 157 158 158

4.

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

151

3.8. Could a disrupted neural mechanism mean an increased risk for accelerated tumorigenesis? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.9. Modulation of tumorigenesis by the brain as a new therapeutic approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.10. What is the role of the CNS and tumor interactions in alternative therapeutic approaches? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

158 159 159 159 160 160

1. Neuro-endocrine–immune interactions as a base for neurobiology of peripheral diseases

might constitute the basis for the formation of a new branch in oncology—the neurobiology of cancer.

Homeostasis within the body of every higher animal is regulated by three interwoven systems—the nervous, endocrine and immune systems. It is increasingly clear that the exchange of information between these systems plays important roles in various physiological as well as pathological processes [1]. The data accumulated in the past decades show that the interactions between nervous, endocrine and immune systems constitute the basis for the involvement of the central nervous system (CNS) in etiopathogenesis of some pathological states and diseases, in which the role of CNS was previously either not recognized or neglected (e.g. arthritis, atherosclerosis, diabetes mellitus, hemorrhagic shock, ischemia-reperfusion, ileus, pancreatitis, sepsis; [2–4]). However, it is necessary to note that even when knowledge about the nervous system functions are increasing exponentially, our understanding of the brain’s role in the pathogenesis of various diseases of peripheral tissues is probably still at its beginnings and not precise. The development of cancer is a highly complex process, in which many known and unknown factors are involved, which we cannot now precisely quantify [5]. Close interactions between the nervous and immune systems, and the important role of the immune system in the development and progression of cancer [6,7] evoke the question whether the brain might monitor and modulate the tumorigenesis (tumor growth; [8–12]). The role of the immune system in tumorigenesis is of relevance to the CNS since the nervous and immune systems can bi-directionally communicate by using a common chemical language employing peptide and non-peptide neurotransmitters, hormones, cytokines and common receptors [13–15]. Through the sharing of ligands and receptors, the immune system could serve as the “sixth sense” to detect signals that the body cannot otherwise hear, see, smell, taste or touch. Pathogens, allergens as well as tumors may be detected with great sensitivity and specificity by the immune system. As the sixth sense, the immune system may have the capacity to signal information about changes of these types of mutative challenges to the brain [16,17]. In recent years, it has become certain that also neuroimmune mechanisms play a role in the defense against cancer as well as in its progression [18–20]. However, these interactions are highly complex, and many variations are possible according to the nature of the neoplasm involved [21]. This article depicts the findings that strongly indicate the involvement of the brain in cancer monitoring and modulating. We anticipate that focusing on the study of interactions between the brain and cancer

2. Nervous system and tumorigenesis (tumor progression): facts 2.1. Clinical and experimental data supporting the assumption that the brain monitors and modulates tumorigenesis Involvement of the nervous system in modulating cancer development and its progression is indicated by various clinical and experimental data. The interruption of interconnection between the brain and organs, namely that of vagal pathways, might cause aggravation or acceleration of tumor growth [22]. Several retrospective studies of patients who have undergone vagotomy suggest an increased risk of cancer development [23–26]. However, some controversial results were obtained from human and experimental studies in animals [27–31]. Conflicting results on the risk of cancer following vagotomy may be due to other factors such as hypochlorhydria, Helicobacter pylori, bile reflux and smoking, which might also play a role in the increased incidence of tumorigenesis in these patients [25,26,32,33]. Involvement of the sensory neurons of the vagus nerve in modulating the tumorigenesis is indicated by experiments in which mice were chemically vagotomized by capsaicin. Erin et al. [22] found out that chemical vagotomy increases metastasis of breast-cancer cells. Furthermore, when the tumor cells were administered long after capsaicin challenge, hence allowing nerve regeneration, animals given capsaicin had no more metastases when compared with placebo-treated mice. It is suggested that inactivation of sensory neurons with high dose of capsaicin enhanced metastases by promoting the growth of more aggressive cells. Moreover, it is hypothesized that loss of sensory nerve mediators (e.g. substance P (SP), calcitonin gene-related peptide (CGRP)) might have led to the loss of activators for certain genes involved in inhibition of cancer growth [34]. In a series of studies, Hodgson and coworkers [35,36] found that administration of IL-1␤ in the cerebral ventricles led to greater tumor retention in the lung in an animal model of adenocarcinoma. Their data suggests that observed exaggeration of tumorigenesis was mediated by possible immunosuppressive effects of increased activity of the sympathetic nervous system (SNS) and hypothalamic–pituitary–adrenal (HPA) axis [36]. It appears that effects of intracerebroventricularly administered IL-1␤ on peripheral cancer is mediated in the brain via central prostaglandins and in the periphery via ␤-adrenergic receptors [37].

152

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

It has been found that sympathectomy might influence tumorigenesis. The published data suggest that sympathectomy might suppress immune functions. Chemical sympathectomy in mice is followed by an increase in both hypothalamic Fos expression and levels of circulating corticosterone [38]. Sympathectomy might influence the tumorigenesis by modulating the activity of the immune system in two ways—by reducing the modulatory influences of catecholamines on immune cells as well as by increasing the secretion of glucocorticoids. However, it was found that chemical sympathectomy induced by 6-hydroxydopamine was connected with protective effects against colon carcinogenesis [39]. The role of the sympathetic system in tumorigenesis is further complicated by studies demonstrating that its neuroendocrine substances promote the tumor progression. Norepinephrine can promote certain tumors’ invasion angiogenesis via ␤-adrenergic receptors on various types of tumor cells (ovarian, nasopharyngeal), by increasing levels of vascular endothelial growth factor and matrix-metalloproteins [40,41]. Experiments in which were used stimulatory and lesion methods showed that specific immune functions are modulated by discrete brain areas [42]. Interestingly, lesion methods showed also links between tumorigenesis and the brain. Lesions of the median hypothalamus were found to result in a significant rise in the proliferation rate of Yoshida ascites tumor in rats and Erlich’s tumors and L1210 ascites tumor in mice, and a significant increase in cell multiplication in inoculated ascitic and solid tumors in mice and rats. Pinealectomy is associated with an increased incidence of induced breast cancer in rats, and this can by reversed by melatonin administration [43]. These studies suggest the tumor-modulatory role of certain CNS regions, particularly those involved in important homeostatic and neuroendocrine functions (e.g. hypothalamus, pineal gland). Surprisingly, there is only scattered data describing the changes in neuronal activity in CNS in animals with tumors. Immunohistochemical investigation of CNS in tumor-bearing rats showed an increased activity of spinal cord and forebrain neurons [44,45]. Recently we have found that the advanced stage of tumorigenesis is accompanied by an increased activity of brainstem and hypothalamic neurons (unpublished results). It is well known that these neurons are also activated by various immune challenges [46]. Therefore, our data might support the assumption that CNS receives signals related to tumorigenesis. Finally, some studies in humans have found interesting differences between cancer patients and control patients in the activity of various brain regions. Tashiro et al. [47,48] found a reduced prefrontal activation in cancer patients versus controls, and suggested that the brain’s response to a tumor resembles that of depressive states. This is important considering the prognostic role of depression in some cancers [49]. However, these studies need to be viewed with caution since it is difficult to distinguish the effects of the cancer per-se from the effects of cancer treatments (radiation, chemotherapy), that the patients underwent. In addition, the patients knew they had cancer, thus it is difficult to conclude whether these findings reflect any relationship between

brain activity and cancer or reflect an anxiety accompanying the diagnosis of cancer. 2.2. The impact of psychosocial factors on cancer incidence and progression Several lines of evidence suggest that psychological or behavioral factors can influence the progression of cancer [50,51], though some reviews challenge these conclusions [52]. Cancer is associated with many circumstances, e.g. fear of death, side effects of treatment, cancer pain, disruption of social activities and social isolation. Approximately half of all cancer patients suffer from psychiatric disorders usually associated with depression [53]. A hyperactive HPA axis and sympathoadrenal system, due to stress and possible depression, might influence cancer progression by stimulating the tumor growth or immunosuppression (mainly via modulating activity of natural-killer cells) [53–56]. Stimulation of tumor growth by hypercortisolemia can be explained by a possible stimulation of angiogenesis, direct stimulation of tumor growth in hormone-sensitive tumors, and altered gluconeogenesis. The latter involves different responses of tumor cells to glucocorticoid signals compared to normal cells leading to a selective deprivation of normal cells of metabolic resources and facilitation of tumor cell growth instead [54]. Stress is associated with enhanced secretion of norepinephrine that may alter the NK cells availability and their function [50]. Ben-Eliyahu et al. [57] showed that the effects of stress on tumor growth were mediated by suppression of NK cell activity caused by catecholamines. Furthermore, as mentioned above, the activation of ␤-adrenergic receptors on tumor cells may promote the tumor growth [40,56]. Another possible mechanism of linking the stress with cancer progression is that intermediated by pro-inflammatory cytokines. Cerebral IL-1 may mediate the effects of helplessness [58] and cerebral administration of IL-1 can enhance peripheral tumor progression [35]. Since the peripheral IL-1 plays pivotal roles in tumor angiogenesis and metastasis [59], IL-1 may mediate the effects of helplessness on tumor progression [60]. Psychosocial factors may also influence the disease by disruption of neuroendocrine and immune circadian rhythms. The disruption of circadian systems was found in advanced cancer, and this is explained by the possible disruption of immune cell trafficking and cell proliferation cycles, as well as by altered hormone levels affecting the tumor versus host metabolism [61]. The damage of cellular DNA and consequent production of abnormal cells is the major trigger of tumors. Stress was found to reduce levels of methyltransferase, an important DNA repair enzyme induced in response to carcinogens [62]. A recent review also points at the quite consistent effects of stress on DNA-integrity in animal studies and points at significant associations between psychological factors and DNA-damage in humans [63]. Given the central role of DNA-damage in the onset of cancer and in the alterations of established tumor antigens, psychological factors may influence the tumorigenesis and progression via affecting DNA-integrity.

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

153

Fig. 1. Pathways, which might provide base for monitoring of tumorigenesis by the brain. Direct effect on the nervous system: molecules released actively from tumor cells (e.g. growth factors, CEA, PSA, CA125 [186]) or molecules released during necrosis of tumor cells (e.g. HMGB1, DNA fragments) might reach the brain via humoral pathway (A). Tumor cells might also influence vagal paraganglia (B), somatic afferents (C) or spinal visceral afferent nerves (D). Indirect effect mediated by immune system: Circulating cytokines (e.g. IL-1, IL-6, TNF) produced by tumor-activated immune cells might influence brain activity via circumventricular organs (e.g. subfornical organ, SFO, organum vasculosum laminae terminalis, OVLT, area postrema, AP) or via interaction with brain endothelial cells (A). Binding of cytokines (e.g. IL-1) to receptors on vagal paraganglion dendritic cells (grayish cell with protrusions) or directly to receptors of the vagus nerve activate the vagus nerve afferents that transmit information to the nucleus of the solitary tract (NTS) (B). Endorphins (␤-END) might bind to the endings of somatic afferents and produce an analgesic effect (C). Whether spinal visceral nerve afferents are influenced by some compound (?) released from immune cells remains to be investigated (D). As the vagus nerve innervates only limited visceral areas, it is possible that the immune signals are also carried via the spinal visceral afferents. The scheme omits sentinel cells (e.g. tissue fibroblasts) which may also play an important role in modulating the inflammatory processes and tumorigenesis [187,188] and might process and transmit signals from the immune system and tumor cells to the nervous system.

154

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

3. Nervous system and tumorigenesis: questions, assumptions, and hypotheses 3.1. The tight interconnection between the immune and nervous systems elicits a question as to whether the brain might monitor and modulate the process of tumorigenesis, and if yes, at which level of the nervous system is it involved. It has been suggested that the immune system might realize sensory functions that can in addition to infectious agents monitor tumor cells [14]. While Blalock has only indirectly approached the problem of interconnections between tumor cells, the immune system, and the brain, others have delineated this relationship more clearly [9,11,12]. 3.2. If we assume that the brain can modulate the tumorigenesis then the brain must be informed about cancer. How is this information transmitted to the brain? Information about cancer might reach brain indirectly, using the immune system as a transducer. Moreover, we hypothesize that some molecules released from tumor cells might represent messengers that might directly “inform” the brain about tumorigenesis (Fig. 1). 3.2.1. Indirect transmission of information about cancer to the brain The immune system might inform the brain about tumorigenesis via two pathways: humoral and neural [64–66]. The humoral pathways are relatively slow and less informative regarding the location or source of the immune signals. The neural pathways are fast and location-specific (Fig. 1). 3.2.1.1. Humoral pathways. Cytokines transmit signals from the immune to the nervous system, utilizing different routes [67,68]. Receptors for cytokines are present in peripheral nervous structures as well as in CNS [69]. The brain is informed about cytokines that circulate in the blood at least by three different pathways: (a) cytokines may be actively transported by the endothelium across the blood–brain barrier (BBB); (b) cytokines pass to the brain tissue at the level of circumventricular organs (CVOs) and activates CNS targets in the vicinity of CVOs; (c) cytokines induce the production of cytokines from cells of BBB, which then secrete cytokines into the brain parenchyma [70,71]. It is important to note that cytokines binding to receptors on macrophages, endothelial cells, or astrocytes induce the production of soluble molecules (prostaglandins, nitric oxide) that convey the signal from the circulation to CNS [71–75]. Therefore, prostaglandins may represent crucial messengers that constitute the links between circulatory cytokines and CNS [70,76,77]. 3.2.1.2. Neuronal pathways. Information from the immune system may reach the CNS also via peripheral nerves. Cytokines play a pivotal role in the transmission of signals from the immune system to peripheral nerves. However, other signaling molecules are also involved in the interaction between the immune and

peripheral nervous systems. Immune cells are capable of synthesizing many peptide hormones and neurotransmitters, e.g. corticotrophin releasing hormone, adrenocorticotropic hormone (ACTH), endorphins, thyroid stimulating hormone, growth hormone, prolactin, substance P, vasopressin, oxytocin, somatostatin, and neuropeptide Y [78–80]. These compounds do not act only in a paracrine manner. For example, immune cell-derived ␤-endorphins might act on opioid receptors on the peripheral terminals of sensory neurons [81]. Peripheral nerves could receive information directly from specialized immune cells or from sentinel cells, e.g. dendritic cells and subpopulations of tissue fibroblasts. Sentinel cells process information about the immune status of surrounding tissue and may consequently transmit these signals to the peripheral nervous system via production of cytokines [82–84]. It is suggested that sentinel cells might represent an analogy to taste cells. Both, the sentinel and taste cells are in the first line of contact with the chemical stimulus, and respond by generating a second signal capable of activating neural elements [65]. One of the most important visceral sensors is represented by the vagus nerve. It innervates the thorax and abdomen with fibers containing a variety of sensory receptors [85]. The role of the vagus nerve in the transmission of information about peripheral inflammatory processes is well recognized. The data indicates that capsaicin-sensitive afferent fibers of the hepatic vagus nerve constitute necessary components of the afferent mechanism of the first febrile phase [86]. This is supported by data showing that vagal sensory neurons themselves express mRNA for IL-1 receptors, suggesting a direct reaction of afferent vagal fibers to peripheral IL-1 [87]. Therefore, cytokines might activate the vagus nerve sensory afferents that transmit signals from the immune system to CNS, particularly to the nucleus of the solitary tract [88,89]. While the role of the vagus in immune-to-brain communication is quite established [87,90], this may be important especially in a situation when concentrations of peripheral pro-inflammatory cytokines are low [91]. This role may be pertinent to low concentrations of inflammation that can promote tumorigenesis, as described below. Another group of important visceral sensors are paraganglia, which represent structures supporting the transmission of information from the immune system to the brain via the vagus nerve [72]. Paraganglia, innervated by the vagus nerve, contain cells that express IL-1 receptors. IL-1 receptors appear to be located on dendritic-like cells as well as on cells interdigitating the vagus nerve parenchyma [92]. This arrangement constitutes an important link between the immune and nervous systems [93,94]. The vagus nerve does not innervate all visceral organs. Therefore, it can be hypothesized that spinal visceral afferent fibers and cutaneous sensory fibers might also transmit certain immune-related information from the vagus innervationfree visceral regions of the body. Experiments using bacterial lipopolysaccharide-induced inflammation and local anesthesia indicate that cutaneous sensory nerves can modestly participate in the transmission of inflammatory information to CNS [95]. Tactile hypersensitivity during inflammatory diseases and observations in patients with leprosies also suggest a possible role of cutaneous sensory afferent fibers in the transmission of

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

signals from the immune system to CNS [96]. It is presumed, that disruption of sensory C-fibers and sympathetic innervation in leprosies are responsible for the loss of anti-inflammatory immune–nervous system communicative and modulatory circuits [97]. 3.2.2. Direct transmission of information about cancer to the brain As it is hypothesized that interactions between the nervous and immune systems might constitute the basis for monitoring the tumorigenesis, a question arises as to whether the brain is able to distinguish between inflammation and tumorigenesis? Presumably, the spectrum of cytokines and other chemical compounds emerging during tumorigenesis might provide a sufficient source of information necessary for the brain to “detect” the presence of tumor cells in organisms. Molecules released from tumor cells (CEA, PSA, CA125) or during their necrosis (HMGB1, DNA fragments; [98]) might represent another kind of messengers that might inform the brain about peripheral tumorigenesis (Fig. 1). Other, but unspecific signals might represent stimulation of tissue mechanoreceptors by tumor growth. Thus, although we cannot answer this question adequately, we propose that a pattern of signals, specific to tumorigenesis, may signal this process to the brain. 3.3. How are brain functions influenced by cancer? Anorexia–cachexia syndrome is observed in 80% of patients in advanced stages of cancer. The findings suggest that cancer anorexia–cachexia syndrome results from multifactorial processes involving various mediators, including cytokines, hormones and neuropeptides. It is likely that the hypothalamus can provide the basis for close interactions among these mediators given its role in feeding behavior [99]. Tumors might induce anorexia by modifying the brain function via two pathways, humoral and nervous. It was found, that the vagus nerve represents an important structure involved in processes associated with tumor-induced anorexia. Animals given a peripheral carcinogen, and subsequently undergoing chemical or surgical vagotomy, did not develop reduced food intake [100]. Depression represents another example of altered brain function common in patients with cancer. Even minimal peripheral inflammation can lead to negative moods in healthy humans [101]. It is suggested that depression (and also anorexia) in patients with cancer is caused by upregulation of production of inflammatory cytokines as a consequence of the immunological response to tumors [102]. As the brain activity may be altered by tumorigenesis, it can by hypothesized, that a modification of transmission of information from cancer cells to the brain, which is responsible for the induction of anorexia and depression, might represent a new potential method of restricting the negative consequence of cancer on the organism. On the other hand it can be hypothesized that the information related to tumorigenesis may induce the negative-feedback anti-inflammatory response by HPA axis or directly by the descending vagus, which may slow down the tumorigenesis. If this assumption is correct then the benefi-

155

cial effect can by induced by the activation of selected brain structures. 3.4. The monitoring of tumorigenesis by the brain as a new diagnostic approach. Could any of the functional imaging techniques (e.g. fMRI, PET) be able to detect an altered response in certain brain areas in cancer patients, especially after exposing them to experimental stimuli? In general, the tumorigenesis is a long-lasting process, and it may induce changes in the activity of some brain regions. Can these changes modulate the tumorigenesis and thus affect the prognosis of cancer disease? The nucleus of the solitary tract (NTS), which relays visceral information, might be modulated from peripheral tumors by receiving tumor-related inflammatory signals via the vagus nerve [9,12]. Other regions may represent the hypothalamic paraventricular (PVN) and suprachiasmatic (SCN) nuclei. The PVN is a coordinating center of autonomic, endocrine, and immune systems. Given the major roles of these systems in tumor development mentioned above, the PVN could potentially influence the tumorigenesis as well. The SCN is one of the key regulators of the circadian rhythm. The activation of SCN neurons by light induces complex neuroendocrine changes which can modulate the immune activity [103]. Therefore, it is not surprising that it is suggested that the disruption of the circadian rhythm might also participate in tumorigenesis [61,104]. For example, melatonin, a hormone of importance in circadian rhythms, influences the growth of spontaneous and induced tumors in animals. While the data in humans are conflicting, the majority of reports point toward protective actions of melatonin [43,105]. Whether the potential alteration of NTS neurons activity influences also the processing of gustatory information, and thus the change in quality or quantity of food intake in patients with cancer, is unclear. Similarly, a possible interference between cancer therapy and processing of the information in the abovementioned and other brain regions needs further investigation. Future studies need to examine whether the sensitivity and responses of such brain regions to tumor-related inflammatory signals may play a role in the early and later stages of cancer progression. To the best of our knowledge, only Tashiro et al. [47,48] have examined brain activity in cancer patients and found reduced pro-frontal activity, resembling the depressive symptoms (see above). 3.5. Mechanisms potentially enabling the brain to modulate tumorigenesis We suggest that the brain might modulate the course of tumorigenesis indirectly by modulating the immune functions as well as directly by released neurotransmitters that might locally influence the activity of cancer cells (Fig. 2). 3.5.1. Indirect modulation of cancer by the brain The nervous system can stimulate or inhibit activities of the innate and adaptive immune systems via neural and humoral pathways (Fig. 2) [106,107]. Due to the role of immune fac-

156

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

Fig. 2. Pathways, which might provide base for modulation of tumorigenesis by the brain. Tumorigenesis might be modulated directly by compounds released by the brain (e.g. melatonin; (A)) and by neurotransmitters released by vagal (acetylcholine; (B)) or sympathetic (norepinephrine, neuropeptide Y; (C)) postganglionic neurons. Moreover, the activation of sensory endings via axonal reflex (F) might induce the release of neuropeptides (e.g. substance P, calcitonin gene-related peptide) that might potentially modulate the tumorigenesis. The progression of cancer might be influenced by the brain also indirectly via modifying the immune cells activity. Hormones released from the pituitary gland (e.g. ACTH, prolactin, GH) might modulate the immune function (A). Acetylcholine released from postganglionic vagal neurons (VNpo) binds to the nicotine receptors of immune cells and produces an anti-inflammatory effect (B). Norepinephrine and neuropeptide Y released from postganglionic sympathetic neurons (SNpo) and epinephrine/norepinephrine released from adrenal medulla might influence immune functions after binding to adrenergic receptors on the immune cells (C and D). Glucocorticoids released from adrenal cortex have complex effects on the immune system (E). The scheme omits the modulation of immune cells by somatic afferent sensory fibers that after being activated by inflammatory processes release neuropeptides via axonal reflex manner. Similarly, the release of norepinephrine from sympathetic nerve endings might be modulated by cytokines released from neighboring immune cells [189]. However, these mechanisms are the primary consequence of local peripheral processes that are not initiated by the activity of the central nervous system. Abbreviations: CGRP, calcitonin gene-related peptide; SNpr, preganglionic sympathetic neurons; SP, substance P; VNpr, preganglionic vagal neurons.

tors in enhancing and inhibiting the tumorigenesis and given effects of the brain on immunity, CNS could potentially influence the tumorigenesis. In the regulation of immune functions, the innervation of the bone marrow by autonomic nerves plays important roles as well [108,109], possibly influencing the estab-

lishment of new immune cells and influencing the bone marrow microenvironment. 3.5.1.1. Humoral pathways. The main messengers of humoral communication between the brain and the immune system are

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

hormones released from the adenohypophysis [106]. It was shown that after parturition, the function of the bone marrow, the thymus and the maintenance of immunocompetence became all dependent on pituitary prolactin (PRL) and growth hormone (GH). Thyroid stimulating hormone modulates immune functions both by stimulation of thyroid hormones and by its action on the lymphoid cells [110–112]. The pro-opiomelanocortin derived peptides (ACTH, ␣melanocyte-stimulating hormone (␣-MSH) and ␤-endorphin (␤-END)) act antagonistically with GH and PRL and suppress the adaptive immune responses by acting on the nervous, endocrine and immune systems [106,113]. It has been shown that ␣-MSH suppresses the nuclear factor-␬B (NF-␬B) activated by various inflammatory agents and that this mechanism probably contributes to ␣-MSH-induced anti-inflammatory effects [114,115]. The influence of ACTH on immune status is mediated mainly via glucocorticoids, released from the adrenal gland, which affect the immune responses via glucocorticoid receptors expressed by immune cells. Whereas it was initially thought that glucocorticoids mediate immunosuppression, more recent studies indicate that they suppress Th1 and activate Th2 lymphocytes [116]. Thus, ACTH-induced changes of the immune system activity are not always immunosuppressive, but rather immunomodulatory [67]. It is necessary to take into consideration that immune cells also possess a capacity to produce some hormones, e.g. PRL, GH [78]. Another humoral “effector” of immunity is oxytocin, a hormone synthesized in the hypothalamus and secreted from the pituitary gland. Oxytocin has immunomodulatory roles [117] and is relevant to tumorigenesis since it may have a role in suppressing the tumor cell proliferation [118]. As immune cells might synthesize some neurotransmitters, the interplay between hormones released from CNS and immune cells might participate in the modulation of immune functions. Neurohormones may suppress the activity of natural-killer cells or cytotoxic T-cells, thus facilitating the tumors’ escape from immune surveillance [57,119]. Furthermore, the effects of adrenal hormones on eliciting the Th2 immune profile may influence the tumorigenesis since this profile has a prognostic value in certain cancers [120]. 3.5.1.2. Neuronal pathways. Both the sympathetic and parasympathetic parts of the autonomic nervous system may modulate immune processes in the organism. All lymphoid organs receive autonomic innervation, and cells located in the lymphoid tissues possess receptors for transmitters released from autonomic nerves [121–124]. The immune system is regulated, to a great extent, by the SNS, which innervates the majority of lymphoid organs [125–128]. It is well documented that catecholamines released from sympathetic nerve endings modulate the function of many components of the immune system via adrenergic and purinergic receptors on immune cells [122,129–132]. Recent findings also show that SNS plays an important role in the regulation of the egress of hematopoietic cells from bone marrow [133]. Moreover, SNS may modulate immune functions also by direct regulation of blood flow via immune tissues [134]. Experimental data shows

157

that an interruption of SNS in animals produces either enhancement or suppression of inflammation, depending on the stage of development, at which the system is ablated, and whether the system is interrupted at a local or systemic level [67]. It is well established that afferent neural pathways in the vagus nerve participate in the brain-mediated responses to inflammation [67]. In addition to this sensory function of the vagus nerve, an efferent or motor vagus nerve mechanism has also been described, by which acetylcholine, the principal vagus nerve neurotransmitter, inhibits cytokine release from resident tissue macrophages [135]. The findings show that both pharmacological and electrical stimulations of the vagus nerve can attenuate the systemic inflammatory response via cholinergic anti-inflammatory pathways [136]. Given the role of inflammatory signals in early [137] and late [59] stages of tumorigenesis, this anti-inflammatory actions of the vagus nerve may have implications for tumorigenesis as well [9]. It is necessary to point out that lymphocytes of various immunological compartments were found to be equipped with the key enzymes for the synthesis of both acetylcholine and catecholamines [138–140]. Therefore, the effects of acetylcholine and catecholamines released by immune cells in a paracrine manner might co-operate/interfere with effect of neurotransmitters released by autonomic nerves within immunological compartments. 3.5.2. Direct modulation of cancer by the brain In analogy with the potential antimicrobial activity of neuropeptides (substance P, neuropeptide Y, adrenomedullin, ␣-MSH, proenkephalin A) [141,142], arises a question as to whether the nervous system might produce substances with potential tumor-modulating activity. Receptors for neurotransmitters are often expressed in many primary human cancers [143]. Therefore, it is likely that tumor cells are susceptible to the same signal substances of the nervous system, just like the normal cells of the tissue they descent from [20]. This assumption is supported by accumulating the evidence suggesting the involvement of specific neuropeptides with defined physiological action such as neurotransmitters, in the modulation of progression of cancer of various organs [144,145]. The data suggests that neurotransmitters might influence apoptosis, mitogenesis, angiogenesis, and migration of cells as well as the genesis of metastasis [19,20,146,147]. Therefore, it is not surprising that some researchers propose the use of compounds affecting of the neurotransmitters receptors as a novel and promising approach for treating patients with cancer [10]. The migration of breast, prostate and colon cancer cells is enhanced by the stress-related neurotransmitter norepinephrine in vitro, and this effect can be inhibited by ␤-blocker propranolol [148]. Serotonin is able to significantly increase the apoptosis of cells of Burkitt lymphoma. It is speculated that serotonergic innervation might modulate the dynamics of this disease [149]. Another compound released from nerve endings that might regulate the apoptosis is the gaseous transmitter NO [150]. Also substance P is associated with various processes connected with tumorigenesis. SP might promote mitogenesis, angiogenesis, and genesis of metastasis [10]. It is suggested that SP released in

158

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

the skin might participate in photocarcinogenesis [151]. While the majority of neurotransmitters have a stimulatory effect on cell migration, an endogenous substance amantadine and GABA exhibit an inhibitory effect [152,153]. The pineal gland and its principal hormone melatonin are known to influence the initiation and progression of cancer [154]. It is suggested that melatonin may have an anti-tumor activity [155]. Callaghan [43] hypothesized that melatonin is involved in the mechanism of psychological effects in the promotion of tumorigenesis. Data indicates that brain-derived oxytocin might participate in modulation of the tumor progression [118]. This hormone has recently been related to social support and synergistically interacts with its effects in reducing the stress-response [156]. Given that social support predicts a better prognosis in several cancers [157], the role of oxytocin in these effects also needs to be investigated. However, it is necessary to distinguish whether neurotransmitters that might influence tumorigenesis are released from the brain or nerve endings in tumor tissues, or whether they are synthesized by local non-neuronal (immune) cells and act in autocrine or paracrine manners [158]. Whereas the lack of innervation in tumors was a generally accepted fact [159] recent experimental data suggests that nerve cells infiltrate and innervate tumors [160,161]. Based on these facts, the concept of neuro-neoplastic synapse has emerged [162,163]. How is the process of tumor innervation regulated? Tumor cells are able to release neurotrophic factors. These factors may stimulate adjacent nerve cells to develop nerve axons into the tumor. These nerve cells might in turn release neurotransmitters, for which the tumor cells are susceptible. It is suggested that innervations of the tumor might provide additional support for a nerve-driven induction of metastasis development [20,164,165]. According to the concept of neuroneoplastic synapse a question arises as to whether this structure might represent a new target for cancer therapy [166]. 3.6. Could the cholinergic anti-inflammatory pathway take part in the modulation of tumor growth? The progression of several types of cancer is determined primarily by the severity of the inflammatory response, which may be regulated by NF-␬B [167]. Therefore, the NF-␬B pathway plays an important role in linking the chronic inflammation with cancer [168], since disruption of the NF-␬B pathway results in a strong reduction of cancer in models of colorectal and hepatocellular cancers [169]. Only recently it was discovered that there is a strong antiinflammatory effect induced by the stimulation of efferent vagus nerve axons (the cholinergic anti-inflammatory pathway) [132]. Even if some controversies regarding the anatomical and functional aspects of the cholinergic anti-inflammatory pathway remains [124], it was repeatedly confirmed that acetylcholine released from postganglionic neurons of the vagus nerve induces a profound inhibition of synthesis of pro-inflammatory cytokines in macrophages [4]. This anti-inflammatory effect is mediated by ␣7-nicotinic receptors. The occupation of this subtype of nicotinic receptors inhibits nuclear activity of NF-␬B [170]. Thus, the activation of cholinergic anti-inflammatory pathways might

inhibit chronic inflammation and perhaps modulate tumorigenesis. It was observed that guanylhydrazone CNI-1493 has antiinflammatory effects that may take place through the cholinergic anti-inflammatory pathway [171]. CNI-1493 was already studied in the phase I trial in melanoma and renal cancer patients, showing the evidence of pharmacological activity as an inhibitor of TNF production [172]. In the case of melanoma, the interpretation of these findings in relation to the vagus nerve need to be taken with caution since this nerve does not innervate the skin. Though CNI-1493 activates the efferent vagus fibers, it is possible that by the effects of the vagus on the HPA axis, a systemic suppression of circulating cytokines may aid in melanoma treatment. Furthermore, anti-inflammatory pathways of the vagus nerve might also be activated by the occupation of central melanocortin receptors (e.g. by ACTH, ␣-melanocyte-stimulating hormone) [115,173]. Thus it is possible that therapeutic modulation of cancer progression via vagus efferent pathway-activating drugs might act at the level of CNS, in addition to the tumor microenvironmental level, and “stimulate” a defensive reaction against tumor cells. Accumulated data suggests that non-steroidal antiinflammatory drugs (NSAIDs), especially aspirin, prevent cancer development [174]. Interestingly, it was shown that NSAIDs modulate peripheral inflammation not only in regions of inflammation, but also by affecting the CNS [175]. Therefore, the preventive effects of NSAIDs on cancer development might be potentially mediated also by their actions via the CNS. 3.7. Is there a role for axon reflexes in modulation of tumorigenesis? The main role of peripheral sensory nerve fibers is the transmission of information to the CNS allowing the host to sense and respond to peripheral stimuli. However, peripheral sensory nerve fibers are also capable of transmitting the signals antidromically via branches of the peripheral nerves to transmit signals in the reverse direction back to the peripheral innervated tissues. Via this so-called axon reflex, neuropeptides (e.g. SP, CGRP) are released from peripheral nerve endings into the tissues where they might modulate the immune and inflammatory reactions. Legat and Wolf [151] suggest that these interactions might constitute a link between cutaneous sensory nerves, photoaging and tumorigenesis. 3.8. Could a disrupted neural mechanism mean an increased risk for accelerated tumorigenesis? Czura and Tracey [123] suggest that autonomic dysfunction of cholinergic anti-inflammatory pathways may predispose some individuals to excessive inflammatory responses. Pro-inflammatory processes are clearly implicated in hypermetabolism and weight loss associated with cancer-associated cachexia. Moreover, the presence of systemic inflammation is now clearly linked with adverse prognosis in patients with cancer, the fact of which cannot be fully explained by the association

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

with weight loss alone. Therefore, it is suggested, that the systemic inflammation remains an important therapeutic target in combating the cachexia [176]. Whether the dysfunction of neuroendocrine and immune interactions (e.g. cholinergic antiinflammatory pathway) might predispose to cancer diseases and influence the progression of tumorigenesis needs to be investigated. Shanks and Lightman [177] focused on the importance of the maternal–neonatal neuroimmune interactions. Some environmental stimuli might alter the development of these interactions during the intrauterine period. Shanks and Lightman [177] suggest that an altered neuroimmune developmental course might contribute to individual vulnerability to stress-related diseases as well as inflammation in adulthood. Whether intrauterine alterations of neuroimmune system interactions might potentially increase the vulnerability to cancer remains to be investigated. A rather simple manner for measuring such interactions is to test relations between pro-inflammatory cytokines and heart-rate variability, the latter (primarily its high-frequency component) reflecting descending vagal activity. Normally, an inverse relation exists between such parameters [178]. Future studies may wish to test whether the magnitude of (inverse) relations between such parameters predicts a risk of cancer or cancer prognosis, reflecting poor neuroimmune modulation. 3.9. Modulation of tumorigenesis by the brain as a new therapeutic approach Various immunomodulatory methods for cancer therapy have been developed in recent years [179]. It is hypothesized that immunomodulation by the autonomic nervous system might represent a new therapeutic approach for cancer [180]. Tracey [4] proposed a hypothetical “immunological homunculus” which regulates various immune functions by selected brain areas. For example, elevated right-hemisphere activity is associated with reduced natural-killer activity [181], which could be relevant to eradicating tumor cells. We suggest that modifying the activity of specific brain structures involved in regulation of selected immune functions might represent one possible way of cancer therapy by immunomodulation. Finally, Gidron et al. [9] proposes to test the effects of vagal nerve stimulation on tumorigenesis. Such efforts are currently underway by our research team. 3.10. What is the role of the CNS and tumor interactions in alternative therapeutic approaches? Pavlov et al. [66] suggest a role of alternative therapeutic approaches (e.g. hypnosis, biofeedback, acupuncture, and even Pavlovian conditioning) in modulating the inflammatory diseases. On the basis of the data reviewed in this article, it can be suggested that all of these methods can potentially modulate the inflammatory processes connected with the progression of cancer via modulating the CNS–tumor interactions since these therapies influence the sympathoadrenal system and vagal activ-

159

ity [182,183] as well as immune functioning in cancer patients [184,185]. 4. Conclusion The data accumulated in the past decades indicate that the brain is involved in etiopathogenesis of a much wider spectrum of diseases than previously expected. Various experimental and clinical data indicate that the brain might also be involved in etiopathogenesis of cancer. However, detection and modulation of tumorigenesis by the brain might represent a highly complex process with many unknown features. Known facts: • The transmission of information from the immune system to the central nervous system indicates that the brain might be involved in the monitoring of tumorigenesis or at least in monitoring of the tumor-associated inflammatory and other signals. • The transmission of signals from the brain to the immune system constitutes the basis for modulating the cancer growth by the brain. • The brain might also modulate tumorigenesis directly via neurotransmitters and hormones secreted from nerve endings and brain-associated endocrine organs. Unknown facts: • The hierarchical organization of neuroimmune processes involved in the detection of tumorigenesis. • The mode of operation of complex systems possibly responsible for regulating the cancer progression. • Nodal points “deciding” whether cancer will progress or regress. Action to be taken: • Define the manner of transmission of signals from cancer cells and the manner by which the brain might receive these signals. • Investigate the details of the role of the central nervous system in modulating the tumorigenesis. • Answer the question as to whether the differences in modulating the tumorigenesis by the brain (e.g. the density of vagal innervation; changes in cholinergic anti-inflammatory pathway activity) might predispose to the development of cancer. • Develop the methods of modulating the complex processes associated with tumorigenesis, from the level of cancer cells to the level of the brain. We expect that cancer research that focuses on the role of the brain in tumorigenesis might markedly extend our knowledge on the biology of cancer. We suggest that solely interdisciplinary and integrative oncological and neuroscientific approaches might effectively illuminate the possibility that the brain might “know” about tumorigenesis in the body and might modulate its progression. We suggest that creation of new scientific discipline, neurobiology of cancer, might open new avenues in cancer research with a possible impact on prevention, diagnosis and therapy of tumor diseases.

160

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

Acknowledgments This work was supported by the Slovak Research and Development Agency under the contract no. APVV-0045-06 and VEGA grant (1/4312/07). References [1] Besedovsky HO, Rey AD. Physiology of psychoneuroimmunology: a personal view. Brain Behav Immun 2007;21:34–44. [2] Razavi R, Chan Y, Afifiyan FN, Liu XJ, Wan X, Yantha J, et al. TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes. Cell 2006;127:1123–35. [3] Gidron Y, Kupper N, Kwaijtaal M, Winter J, Denollet J. Vagus-brain communication in atherosclerosis-related inflammation: a neuroimmunomodulation perspective of CAD. Atherosclerosis 2007;195: 1–9. [4] Tracey KJ. Physiology and immunology of the cholinergic antiinflammatory pathway. J Clin Invest 2007;117:289–96. [5] Mareel M, Leroy A. Clinical, cellular, and molecular aspects of cancer invasion. Physiol Rev 2003;83:337–76. [6] Balkwill F, Coussens LM. Cancer: an inflammatory link. Nature 2004;431:405–6. [7] de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 2006;6:24–37. [8] Prolo P, Chiappelli F, Bernard G, Fiala M, Ibarra A, Sartori ML, et al. Neuroendocrine-immune surveillance of osteosarcoma: emerging hypothesis. J Dent Res 2003;82:417–21. [9] Gidron Y, Perry H, Glennie M. Does the vagus nerve inform the brain about preclinical tumours and modulate them? Lancet Oncol 2005;6:245–8. [10] Esteban F, Munoz M, Gonzalez-Moles MA, Rosso M. A role for substance P in cancer promotion and progression: a mechanism to counteract intracellular death signals following oncogene activation or DNA damage. Cancer Metastasis Rev 2006;25:137–45. [11] Mravec B, Hulin I. Does vagus nerve constitute a self-organization complexity or a “hidden network”? Bratisl Lek Listy 2006;107:3–8. [12] Mravec B, Gidron Y, Kukanova B, Bizik J, Kiss A, Hulin I. Neural–endocrine–immune complex in the central modulation of tumorigenesis: facts, assumptions, and hypotheses. J Neuroimmunol 2006;180:104–16. [13] Serafeim A, Gordon J. The immune system gets nervous. Curr Opin Pharmacol 2001;1:398–403. [14] Blalock JE. The immune system as the sixth sense. J Intern Med 2005;257:126–38. [15] Meredith EJ, Chamba A, Holder MJ, Barnes NM, Gordon J. Close encounters of the monoamine kind: immune cells betray their nervous disposition. Immunology 2005;115:289–95. [16] Ferencik M, Stvrtinova V. Is the immune system our sixth sense? Relation between the immune and neuroendocrine systems. Bratisl Lek Listy 1997;98:187–98. [17] Blalock JE, Smith EM. Conceptual development of the immune system as a sixth sense. Brain Behav Immun 2007;21:23–33. [18] Berczi I, Chow DA, Baral E, Nagy E. Neuroimmunoregulation and cancer (review). Int J Oncol 1998;13:1049–60. [19] Entschladen F, Drell 4th TL, Lang K, Joseph J, Zaenker KS. Neurotransmitters and chemokines regulate tumor cell migration: potential for a new pharmacological approach to inhibit invasion and metastasis development. Curr Pharm Des 2005;11:403–11. [20] Lang K, Bastian P. Neurotransmitter effects on tumor cells and leukocytes. Prog Exp Tumor Res 2007;39:99–121. [21] Conti A. Oncology in neuroimmunomodulation. What progress has been made? Ann NY Acad Sci 2000;917:68–83. [22] Erin N, Boyer PJ, Bonneau RH, Clawson GA, Welch DR. Capsaicinmediated denervation of sensory neurons promotes mammary tumor metastasis to lung and heart. Anticancer Res 2004;24:1003–9.

[23] Watt PC, Patterson CC, Kennedy TL. Late mortality after vagotomy and drainage for duodenal ulcer. Br Med J (Clin Res Ed) 1984;288:1335–8. [24] Caygill CP, Hill MJ, Kirkham JS, Northfield TC. Mortality from colorectal and breast cancer in gastric-surgery patients. Int J Colorectal Dis 1988;3:144–8. [25] Caygill CP, Knowles RL, Hall R. Increased risk of cancer mortality after vagotomy for peptic ulcer: a preliminary analysis. Eur J Cancer Prev 1991;1:35–7. [26] Ekbom A, Lundegardh G, McLaughlin JK, Nyren O. Relation of vagotomy to subsequent risk of lung cancer: population based cohort study. BMJ 1998;316:518–9. [27] Nelson RL, Briley S, Vaz OP, Abcarian H. The effect of vagotomy and pyloroplasty on colorectal tumor induction in the rat. J Surg Oncol 1992;51:281–6. [28] Caygill CP, Hill MJ, Kirkham JS, Northfield TC. Oesophageal cancer in gastric surgery patients. Ital J Gastroenterol 1993;25:168–70. [29] Fisher SG, Davis F, Nelson R, Weber L, Haenszel W. Large bowel cancer following gastric surgery for benign disease: a cohort study. Am J Epidemiol 1994;139:684–92. [30] Lundegardh G, Ekbom A, McLaughlin JK, Nyren O. Gastric cancer risk after vagotomy. Gut 1994;35:946–9. [31] Bayon LAM, Landa GI, Alcalde EJ, Rodriguez DS, Ortega ML, Balibrea CJL. Colonic carcinogenesis in vagotomyzed rats. Rev Esp Enferm Dig 2001;93:576–86. [32] Bahmanyar S, Ye W, Dickman PW, Nyren O. Long-term risk of gastric cancer by subsite in operated and unoperated patients hospitalized for peptic ulcer. Am J Gastroenterol 2007;102:1185–91. [33] Jenkins JT, Duncan JR, Hole D, O’dwyer PJ, McGregor JR. Malignant disease in peptic ulcer surgery patients after long term follow-up: a cohort study of 1992 patients. Eur J Surg Oncol 2007;33:706–12. [34] Erin N, Zhao W, Bylander J, Chase G, Clawson G. Capsaicin-induced inactivation of sensory neurons promotes a more aggressive gene expression phenotype in breast cancer cells. Breast Cancer Res Treat 2006;99:351–64. [35] Hodgson DM, Yirmiya R, Chiappelli F, Taylor AN. Intracerebral HIV glycoprotein (gp120) enhances tumor metastasis via centrally released interleukin-1. Brain Res 1998;781:244–51. [36] Hodgson DM, Yirmiya R, Chiappelli F, Taylor AN. Intracerebral interleukin-1beta impairs response to tumor invasion: involvement of adrenal catecholamines. Brain Res 1999;816:200–8. [37] Hodgson DM, Yirmiya R, Taylor AN. Intracerebroventricular interleukin1beta impairs clearance of tumor cells from the lungs: role of brain prostaglandins. J Neuroimmunol 2001;119:57–63. [38] Leo NA, Bonneau RH. Chemical sympathectomy alters cytotoxic T lymphocyte responses to herpes simplex virus infection. Ann N Y Acad Sci 2000;917:923–34. [39] Tatsuta M, Iishi H, Baba M, Taniguchi H. Inhibition of azoxymethaneinduced experimental colon carcinogenesis in Wistar rats by 6hydroxydopamine. Int J Cancer 1992;50:298–301. [40] Sood AK, Bhatty R, Kamat AA, Landen CN, Han L, Thaker PH, et al. Stress hormone-mediated invasion of ovarian cancer cells. Clin Cancer Res 2006;12:369–75. [41] Yang EV, Sood AK, Chen M, Li Y, Eubank TD, Marsh CB, et al. Norepinephrine up-regulates the expression of vascular endothelial growth factor, matrix metalloproteinase (MMP)-2, and MMP-9 in nasopharyngeal carcinoma tumor cells. Cancer Res 2006;66:10357–64. [42] Wrona D. Neural–immune interactions: an integrative view of the bidirectional relationship between the brain and immune systems. J Neuroimmunol 2006;172:38–58. [43] Callaghan BD. Does the pineal gland have a role in the psychological mechanisms involved in the progression of cancer? Med Hypotheses 2002;59:302–11. [44] Kergozien S, Delcros JG, Jouan H, Moulinoux JP. Induction of Fos protein expression in spinal cord neurons of tumour-bearing rats. Br J Cancer 1999;80:1512–7. [45] Konsman JP, Blomqvist A. Forebrain patterns of c-Fos and FosB induction during cancer-associated anorexia–cachexia in rat. Eur J Neurosci 2005;21:2752–66.

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163 [46] Hollis JH, Lightman SL, Lowry CA. Integration of systemic and visceral sensory information by medullary catecholaminergic systems during peripheral inflammation. Ann N Y Acad Sci 2004;1018:71–5. [47] Tashiro M, Kubota K, Itoh M, Yoshioka T, Yoshida M, Nakagawa Y, et al. Hypometabolism in the limbic system of cancer patients observed by positron emission tomography. Psychooncology 1999;8:283–6. [48] Tashiro M, Itoh M, Kubota K, Kumano H, Masud MM, Moser E, et al. Relationship between trait anxiety, brain activity and natural killer cell activity in cancer patients: a preliminary PET study. Psychooncology 2001;10:541–6. [49] Watson M, Haviland JS, Greer S, Davidson J, Bliss JM. Influence of psychological response on survival in breast cancer: a population-based cohort study. Lancet 1999;354:1331–6. [50] Spiegel D, Kato PM. Psychosocial influences on cancer incidence and progression. Harv Rev Psychiatry 1996;4:10–26. [51] Kiecolt-Glaser JK, Glaser R. Psychoneuroimmunology and cancer: fact or fiction? Eur J Cancer 1999;35:1603–7. [52] Petticrew M, Bell R, Hunter D. Influence of psychological coping on survival and recurrence in people with cancer: systematic review. BMJ 2002;325:1066. [53] Spiegel D. Cancer and depression. Br J Psychiatry Suppl 1996;109:16. [54] Spiegel D. Embodying the mind in psychooncology research. Adv Mind Body Med 1999;15:267–73. [55] Reiche EM, Morimoto HK, Nunes SM. Stress and depression-induced immune dysfunction: implications for the development and progression of cancer. Int Rev Psychiatry 2005;17:515–27. [56] Antoni MH, Lutgendorf SK, Cole SW, Dhabhar FS, Sephton SE, McDonald PG, et al. The influence of bio-behavioural factors on tumour biology: pathways and mechanisms. Nat Rev Cancer 2006;6:240–8. [57] Ben-Eliyahu S, Shakhar G, Page GG, Stefanski V, Shakhar K. Suppression of NK cell activity and of resistance to metastasis by stress: a role for adrenal catecholamines and beta-adrenoceptors. Neuroimmunomodulation 2000;8:154–64. [58] Maier SF, Watkins LR. Intracerebroventricular interleukin-1 receptor antagonist blocks the enhancement of fear conditioning and interference with escape produced by inescapable shock. Brain Res 1995;695:279–82. [59] Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, Iwakura Y, et al. IL-1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci USA 2003;100:2645–50. [60] Argaman M, Gidron Y, Ariad S. Interleukin-1 may link helplessness– hopelessness with cancer progression: a proposed model. Int J Behav Med 2005;12:161–70. [61] Sephton S, Spiegel D. Circadian disruption in cancer: a neuroendocrine– immune pathway from stress to disease? Brain Behav Immun 2003;17:321–8. [62] Heffner KL, Loving TJ, Robles TF, Kiecolt-Glaser JK. Examining psychosocial factors related to cancer incidence and progression: in search of the silver lining. Brain Behav Immun 2003;17:109–11. [63] Gidron Y, Russ K, Tissarchondou H, Warner J. The relation between psychological factors and DNA-damage: a critical review. Biol Psychol 2006;72:291–304. [64] Dantzer R, Konsman JP, Bluthe RM, Kelley KW. Neural and humoral pathways of communication from the immune system to the brain: parallel or convergent? Auton Neurosci 2000;85:60–5. [65] Goehler LE, Gaykema RP, Hansen MK, Anderson K, Maier SF, Watkins LR. Vagal immune-to-brain communication: a visceral chemosensory pathway. Auton Neurosci 2000;85:49–59. [66] Pavlov VA, Wang H, Czura CJ, Friedman SG, Tracey KJ. The cholinergic anti-inflammatory pathway: a missing link in neuroimmunomodulation. Mol Med 2003;9:125–34. [67] Sternberg EM. Neural–immune interactions in health and disease. J Clin Invest 1997;100:2641–7. [68] Mantovani A. The chemokine system: redundancy for robust outputs. Immunol Today 1999;20:254–7. [69] Rothwell NJ, Hopkins SJ. Cytokines and the nervous system II: actions and mechanisms of action. Trends Neurosci 1995;18:130–6. [70] Quan N, Herkenham M. Connecting cytokines and brain: a review of current issues. Histol Histopathol 2002;17:273–88.

161

[71] Turrin NP, Rivest S. Unraveling the molecular details involved in the intimate link between the immune and neuroendocrine systems. Exp Biol Med 2004;229:996–1006. [72] Watkins LR, Maier SF, Goehler LE. Cytokine-to-brain communication: a review and analysis of alternative mechanisms. Life Sci 1995;57:1011–26. [73] Nadeau S, Rivest S. Effects of circulating tumor necrosis factor on the neuronal activity and expression of the genes encoding the tumor necrosis factor receptors (p55 and p75) in the rat brain: a view from the blood–brain barrier. Neuroscience 1999;93:1449–64. [74] Szelenyi J. Cytokines and the central nervous system. Brain Res Bull 2001;54:329–38. [75] Konsman JP, Parnet P, Dantzer R. Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci 2002;25:154–9. [76] Turnbull AV, Rivier CL. Regulation of the hypothalamic–pituitary– adrenal axis by cytokines: actions and mechanisms of action. Physiol Rev 1999;79:1–71. [77] Rivest S. How circulating cytokines trigger the neural circuits that control the hypothalamic–pituitary–adrenal axis. Psychoneuroendocrinology 2001;26:761–88. [78] Savino W, Dardenne M. Immune–neuroendocrine interactions. Immunol Today 1995;16:318–22. [79] Petrovsky N. Towards a unified model of neuroendocrine–immune interaction. Immunol Cell Biol 2001;79:350–7. [80] Shepherd AJ, Downing JE, Miyan JA. Without nerves, immunology remains incomplete—in vivo veritas. Immunology 2005;116:145– 63. [81] Blalock JE. The syntax of immune–neuroendocrine communication. Immunol Today 1994;15:504–11. [82] Smith RS, Smith TJ, Blieden TM, Phipps RP. Fibroblasts as sentinel cells. Synthesis of chemokines and regulation of inflammation. Am J Pathol 1997;151:317–22. [83] Buckley CD, Pilling D, Lord JM, Akbar AN, Scheel-Toellner D, Salmon M. Fibroblasts regulate the switch from acute resolving to chronic persistent inflammation. Trends Immunol 2001;22:199–204. [84] Kaufman J, Graf BA, Leung EC, Pollock SJ, Koumas L, Reddy SY, et al. Fibroblasts as sentinel cells: role of the CDcd40–CDcd40 ligand system in fibroblast activation and lung inflammation and fibrosis. Chest 2001;120:S53–5. [85] Paintal AS. Vagal sensory receptors and their reflex effects. Physiol Rev 1973;53:159–227. [86] Romanovsky AA, Ivanov AI, Szekely M. Neural route of pyrogen signaling to the brain. Clin Infect Dis 2000;31:S162–7. [87] Ek M, Kurosawa M, Lundeberg T, Ericsson A. Activation of vagal afferents after intravenous injection of interleukin-1beta: role of endogenous prostaglandins. J Neurosci 1998;18:9471–9. [88] Maier SF, Goehler LE, Fleshner M, Watkins LR. The role of the vagus nerve in cytokine-to-brain communication. Ann N Y Acad Sci 1998;840:289–300. [89] Perry VH. The impact of systemic inflammation on brain inflammation. ACNR 2004;4:8–9. [90] Goehler LE, Gaykema RP, Hammack SE, Maier SF, Watkins LR. Interleukin-1 induces c-Fos immunoreactivity in primary afferent neurons of the vagus nerve. Brain Res 1998;804:306–10. [91] Hansen MK, Daniels S, Goehler LE, Gaykema RP, Maier SF, Watkins LR. Subdiaphragmatic vagotomy does not block intraperitoneal lipopolysaccharide-induced fever. Auton Neurosci 2000;85:83– 7. [92] Licinio J, Wong ML. Pathways and mechanisms for cytokine signaling of the central nervous system. J Clin Invest 1997;100:2941–7. [93] Goehler LE, Relton JK, Dripps D, Kiechle R, Tartaglia N, Maier SF, et al. Vagal paraganglia bind biotinylated interleukin-1 receptor antagonist: a possible mechanism for immune-to-brain communication. Brain Res Bull 1997;43:357–64. [94] Goehler LE, Gaykema RP, Nguyen KT, Lee JE, Tilders FJ, Maier SF, et al. Interleukin-1beta in immune cells of the abdominal vagus nerve: a link between the immune and nervous systems? J Neurosci 1999;19:2799–806.

162

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

[95] Roth J, De Souza GE. Fever induction pathways: evidence from responses to systemic or local cytokine formation. Braz J Med Biol Res 2001;34:301–14. [96] Hermann GE, Holmes GM, Rogers RC. TNF(alpha) modulation of visceral and spinal sensory processing. Curr Pharm 2005;11:1391– 409. [97] Rook GA, Lightman SL, Heijnen CJ. Can nerve damage disrupt neuroendocrine immune homeostasis? Leprosy as a case in point. Trends Immunol 2002;23:18–22. [98] Ito N, DeMarco RA, Mailliard RB, Han J, Rabinowich H, Kalinski P, et al. Cytolytic cells induce HMGB1 release from melanoma cell lines. J Leukoc Biol 2007;81:75–83. [99] Ramos EJ, Suzuki S, Marks D, Inui A, Asakawa A, Meguid MM. Cancer anorexia–cachexia syndrome: cytokines and neuropeptides. Curr Opin Clin Nutr Metab Care 2004;7:427–34. [100] Bernstein IL. Neutral mediation of food aversions and anorexia induced by tumor necrosis factor and tumors. Neurosci Biobehav Rev 1996;20:177–81. [101] Reichenberg A, Yirmiya R, Schuld A, Kraus T, Haack M, Morag A, et al. Cytokine-associated emotional and cognitive disturbances in humans. Arch Gen Psychiatry 2001;58:445–52. [102] Illman J, Corringham R, Robinson Jr D, Davis HM, Rossi JF, Cella D, et al. Are inflammatory cytokines the common link between cancer-associated cachexia and depression? J Support Oncol 2005;3:37–50. [103] Roberts JE. Light and immunomodulation. Ann N Y Acad Sci 2000;917:435–45. [104] Filipski E, King VM, Li X, Granda TG, Mormont MC, Liu X, et al. Host circadian clock as a control point in tumor progression. J Natl Cancer Inst 2002;94:690–7. [105] Brzezinski A. Melatonin in humans. N Engl J Med 1997;336:186–95. [106] Berczi I. Neuroimmune biology—an introduction. In: Berczi I, Gorezynski RM, editors. New foundation of biology. Amsterdam: Elsevier Science; 2001. p. 3–45. [107] Brogden KA, Guthmiller JM, Salzet M, Zasloff M. The nervous system and innate immunity: the neuropeptide connection. Nat Immunol 2005;6:558–64. [108] Broome CS, Miyan JA. Neuropeptide control of bone marrow neutrophil production. A key axis for neuroimmunomodulation. Ann N Y Acad Sci 2000;917:424–34. [109] Maestroni GJ. Neurohormones and catecholamines as functional components of the bone marrow microenvironment. Ann N Y Acad Sci 2000;917:29–37. [110] Berczi I. The role of the growth and lactogenic hormone family in immune function. Neuroimmunomodulation 1994;1:201–16. [111] Berczi I. Pituitary hormones and immune function. Acta Paediatr 1997;423:70–5. [112] Fabris N, Mocchegiani E, Provinciali M. Pituitary–thyroid axis and immune system: a reciprocal neuroendocrine–immune interaction. Horm Res 1995;43:29–38. [113] Vamvakopoulos NC, Chrousos GP. Hormonal regulation of human corticotropin-releasing hormone gene expression: implications for the stress response and immune/inflammatory reaction. Endocr Rev 1994;15:409–20. [114] Manna SK, Aggarwal BB. Alpha-melanocyte-stimulating hormone inhibits the nuclear transcription factor NF-kappa B activation induced by various inflammatory agents. J Immunol 1998;161:2873–80. [115] Ichiyama T, Sato S, Okada K, Catania A, Lipton JM. The neuroimmunomodulatory peptide alpha-MSH. Ann N Y Acad Sci 2000;917:221–6. [116] Almawi WY, Melemedjian OK, Rieder MJ. An alternate mechanism of glucocorticoid anti-proliferative effect: promotion of a Th2 cytokinesecreting profile. Clin Transplant 1999;13:365–74. [117] Yang H, Wang L, Ju G. Evidence for hypothalamic paraventricular nucleus as an integrative center of neuroimmunomodulation. Neuroimmunomodulation 1997;4:120–7. [118] Cassoni P, Sapino A, Marrocco T, Chini B, Bussolati G. Oxytocin and oxytocin receptors in cancer cells and proliferation. J Neuroendocrinol 2004;16:362–4.

[119] Ben-Eliyahu S, Yirmiya R, Liebeskind JC, Taylor AN, Gale RP. Stress increases metastatic spread of a mammary tumor in rats: evidence for mediation by the immune system. Brain Behav Immun 1991;5:193– 205. [120] Inagaki A, Ishida T, Ishii T, Komatsu H, Iida S, Ding J, et al. Clinical significance of serum Th1-, Th2- and regulatory T cells-associated cytokines in adult T-cell leukemia/lymphoma: high interleukin-5 and -10 levels are significant unfavorable prognostic factors. Int J Cancer 2006;118:3054–61. [121] Dardenne M, Savino W. Control of thymus physiology by peptidic hormones and neuropeptides. Immunol Today 1994;15:518–23. [122] Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve-an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev 2000;52:595–638. [123] Czura CJ, Tracey KJ. Autonomic neural regulation of immunity. J Intern Med 2005;257:156–66. [124] Nance DM, Sanders VM. Autonomic innervation and regulation of the immune system (1987–2007). Brain Behav Immun 2007;21:736–45. [125] Weigent DA, Blalock JE. Interactions between the neuroendocrine and immune systems: common hormones and receptors. Immunol Rev 1987;100:79–108. [126] Stevens-Felten SY, Bellinger DL. Noradrenergic and peptidergic innervation of lymphoid organs. Chem Immunol 1997;69:99–131. [127] Basu S, Dasgupta PS. Dopamine, a neurotransmitter, influences the immune system. J Neuroimmunol 2000;102:113–24. [128] Denes A, Boldogkoi Z, Uhereczky G, Hornyak A, Rusvai M, Palkovits M, et al. Central autonomic control of the bone marrow: multisynaptic tract tracing by recombinant pseudorabies virus. Neuroscience 2005;134:947–63. [129] Elenkov IJ, Hasko G, Kovacs KJ, Vizi ES. Modulation of lipopolysaccharide-induced tumor necrosis factor-alpha production by selective alpha- and beta-adrenergic drugs in mice. J Neuroimmunol 1995;61:123–31. [130] Vizi ES, Orso E, Osipenko ON, Hasko G, Elenkov IJ. Neurochemical, electrophysiological and immunocytochemical evidence for a noradrenergic link between the sympathetic nervous system and thymocytes. Neuroscience 1995;68:1263–76. [131] Hasko G, Szabo C. Regulation of cytokine and chemokine production by transmitters and co-transmitters of the autonomic nervous system. Biochem Pharmacol 1998;56:1079–87. [132] Tracey KJ. The inflammatory reflex. Nature 2002;420:853–9. [133] Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 2006;124:407–21. [134] Vizi ES. Receptor-mediated local fine-tuning by noradrenergic innervation of neuroendocrine and immune systems. Ann N Y Acad Sci 1998;851:388–96. [135] Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 2000;405:458–62. [136] Bernik TR, Friedman SG, Ochani M, DiRaimo R, Ulloa L, Yang H, et al. Pharmacological stimulation of the cholinergic antiinflammatory pathway. J Exp Med 2002;195:781–8. [137] Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004;431:461–6. [138] Rinner I, Felsner P, Liebmann PM, Hofer D, Wolfler A, Globerson A, et al. Adrenergic/cholinergic immunomodulation in the rat model-in vivo veritas? Dev Immunol 1998;6:245–52. [139] Kawashima K, Fujii T. The lymphocytic cholinergic system and its biological function. Life Sci 2003;72:2101–9. [140] Qiu YH, Peng YP, Jiang JM, Wang JJ. Expression of tyrosine hydroxylase in lymphocytes and effect of endogenous catecholamines on lymphocyte function. Neuroimmunomodulation 2004;11:75–83. [141] Kowalska K, Carr DB, Lipkowski AW. Direct antimicrobial properties of substance P. Life Sci 2002;71:747–50. [142] Metz-Boutigue MH, Goumon Y, Strub JM, Lugardon K, Aunis D. Antimicrobial chromogranins and proenkephalin-A-derived pep-

B. Mravec et al. / Seminars in Cancer Biology 18 (2008) 150–163

[143] [144] [145]

[146]

[147]

[148]

[149]

[150] [151]

[152]

[153]

[154]

[155]

[156]

[157]

[158]

[159]

[160] [161] [162] [163] [164]

tides: Antibacterial and antifungal activities of chromogranins and proenkephalin-A-derived peptides. Ann N Y Acad Sci 2003;992:168– 78. Reubi JC. Peptide receptors as molecular targets for cancer diagnosis and therapy. Endocr Rev 2003;24:389–427. Heasley LE. Autocrine and paracrine signaling through neuropeptide receptors in human cancer. Oncogene 2001;20:1563–9. Schuller HM. Neurotransmitter receptor-mediated signaling pathways as modulators of carcinogenesis. Prog Exp Tumor Res 2007;39:45– 63. Drell 4th TL, Joseph J, Lang K, Niggemann B, Zaenker KS, 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. Zukowska Z, Grant DS, Lee EW. Neuropeptide Y: a novel mechanism for ischemic angiogenesis. Trends Cardiovasc Med 2003;13:86– 92. Lang K, Drell 4th TL, Lindecke A, Niggemann B, Kaltschmidt C, Zaenker KS, et al. Induction of a metastatogenic tumor cell type by neurotransmitters and its pharmacological inhibition by established drugs. Int J Cancer 2004;112:231–8. Serafeim A, Grafton G, Chamba A, Gregory CD, Blakely RD, Bowery NG, et al. 5-Hydroxytryptamine drives apoptosis in biopsylike Burkitt lymphoma cells: reversal by selective serotonin reuptake inhibitors. Blood 2002;99:2545–53. Tarr JM, Eggleton P, Winyard PG. Nitric oxide and the regulation of apoptosis in tumour cells. Curr Pharm Des 2006;12:4445–68. Legat FJ, Wolf P. Photodamage to the cutaneous sensory nerves: role in photoaging and carcinogenesis of the skin? Photochem Photobiol Sci 2006;5:170–6. Joseph J, Niggemann B, Zaenker KS, Entschladen F. The neurotransmitter gamma-aminobutyric acid is an inhibitory regulator for the migration of SW 480 colon carcinoma cells. Cancer Res 2002;62:6467– 9. Joseph J, Niggemann B, Zaenker KS, Entschladen F. Anandamide is an endogenous inhibitor for the migration of tumor cells and T lymphocytes. Cancer Immunol Immunother 2004;53:723–8. Giannoulia-Karantana A, Vlachou A, Polychronopoulou S, Papassotiriou I, Chrousos GP. Melatonin and immunomodulation: connections and potential clinical applications. Neuroimmunomodulation 2006;13:133–44. Kajdaniuk D, Marek B, Kos-Kudła B, Ciesielska-Kopacz N, Buntner B. Oncostatic effect of melatonin action—facts and hypotheses. Med Sci Monit 1999;5:RA350–6. Heinrichs M, Baumgartner T, Kirschbaum C, Ehlert U. Social support and oxytocin interact to suppress cortisol and subjective responses to psychosocial stress. Biol Psychiatry 2003;54:1389–98. Soler-Vila H, Kasl SV, Jones BA. Prognostic significance of psychosocial factors in African–American and white breast cancer patients: a population-based study. Cancer 2003;98:1299–308. Russo P, Catassi A, Cesario A, Servent D. Development of novel therapeutic strategies for lung cancer: targeting the cholinergic system. Curr Med Chem 2006;13:3493–512. Vachkov IH, Huang X, Yamada Y, Tonchev AB, Yamashima T, Kato S, et al. Inhibition of axonal outgrowth in the tumor environment: involvement of class 3 semaphorins. Cancer Sci 2007;98:1192–7. Seifert P, Spitznas M. Tumours may be innervated. Virchows Arch 2001;438:228–31. Seifert P, Benedic M, Effert P. Nerve fibers in tumors of the human urinary bladder. Virchows Arch 2002;440:291–7. Palm D, Entschladen F. Neoneurogenesis and the neuro-neoplastic synapse. Prog Exp Tumor Res 2007;39:91–8. Zanker KS. The neuro-neoplastic synapse: does it exist? Prog Exp Tumor Res 2007;39:154–61. Entschladen F, Palm D, Lang K, Drell 4th TL, Zaenker KS. Neoneurogenesis: tumors may initiate their own innervation by the release of neurotrophic factors in analogy to lymphangiogenesis and neoangiogenesis. Med Hypotheses 2006;67:33–5.

163

[165] Lang K, Entschladen F, Weidt C, Zaenker KS. Tumor immune escape mechanisms: impact of the neuroendocrine system. Cancer Immunol Immunother 2006;55:749–60. [166] Muller JM. Potential inhibition of the neuro-neoplastic interactions: the clue of a GPCR-targeted therapy. Prog Exp Tumor Res 2007;39:130–53. [167] Houghton J, Morozov A, Smirnova I, Wang TC. Stem cells and cancer. Semin Cancer Biol 2007;17:191–203. [168] Li Q, Withoff S, Verma IM. Inflammation-associated cancer: NF-kappaB is the lynchpin. Trends Immunol 2005;26:318–25. [169] Maeda A, Ebata T, Matsunaga K, Kanemoto H, Bando E, Yamaguchi S, et al. Primary liver cancer with bidirectional differentiation into hepatocytes and biliary epithelium. J Hepatobiliary Pancreat Surg 2005;12:484–7. [170] Gallowitsch-Puerta M, Pavlov VA. Neuro-immune interactions via the cholinergic anti-inflammatory pathway. Life Sci 2007;80:2325–9. [171] Borovikova LV, Ivanova S, Nardi D, Zhang M, Yang H, Ombrellino M, et al. Role of vagus nerve signaling in CNI-1493-mediated suppression of acute inflammation. Auton Neurosci 2000;85:141–7. [172] Atkins MB, Redman B, Mier J, Gollob J, Weber J, Sosman J, et al. A phase I study of CNI-1493, an inhibitor of cytokine release, in combination with high-dose interleukin-2 in patients with renal cancer and melanoma. Clin Cancer Res 2001;7:486–92. [173] Guarini S, Cainazzo MM, Giuliani D, Mioni C, Altavilla D, Marini H, et al. Adrenocorticotropin reverses hemorrhagic shock in anesthetized rats through the rapid activation of a vagal anti-inflammatory pathway. Cardiovasc Res 2004;63:357–65. [174] Shiff SJ, Shivaprasad P, Santini DL. Cyclooxygenase inhibitors: drugs for cancer prevention. Curr Opin Pharmacol 2003;3:352–61. [175] Catania A, Arnold J, Macaluso A, Hiltz ME, Lipton JM. Inhibition of acute inflammation in the periphery by central action of salicylates. Proc Natl Acad Sci USA 1991;88:8544–7. [176] Deans C, Wigmore SJ. Systemic inflammation, cachexia and prognosis in patients with cancer. Curr Opin Clin Nutr Metab Care 2005;8:265–9. [177] Shanks N, Lightman SL. The maternal–neonatal neuro-immune interface: are there long-term implications for inflammatory or stress-related disease? J Clin Invest 2001;108:1567–73. [178] Janszky I, Ericson M, Lekander M, Blom M, Buhlin K, Georgiades A, et al. Inflammatory markers and heart rate variability in women with coronary heart disease. J Intern Med 2004;256:421–8. [179] Waldmann TA. Effective cancer therapy through immunomodulation. Annu Rev Med 2006;57:65–81. [180] Abo T, Kawamura T. Immunomodulation by the autonomic nervous system: therapeutic approach for cancer, collagen diseases, and inflammatory bowel diseases. Ther Apher 2002;6:348–57. [181] Davidson RJ, Coe CC, Dolski I, Donzella B. Individual differences in prefrontal activation asymmetry predict natural killer cell activity at rest and in response to challenge. Brain Behav Immun 1999;2:93–108. [182] Bernardi L, Sleight P, Bandinelli G, Cencetti S, Fattorini L, WdowczycSzulc J, et al. Effect of rosary prayer and yoga mantras on autonomic cardiovascular rhythms: comparative study. BMJ 2001;323:1446–9. [183] Infante JR, Torres-Avisbal M, Pinel P, Vallejo JA, Peran F, Gonzalez F, et al. Catecholamine levels in practitioners of the transcendental meditation technique. Physiol Behav 2001;72:141–6. [184] Gruber BL, Hersh SP, Hall NR, Waletzky LR, Kunz JF, Carpenter JK, et al. Immunological responses of breast cancer patients to behavioral interventions. Biofeedback Self Regul 1993;18:1–22. [185] Bakke AC, Purtzer MZ, Newton P. The effect of hypnotic-guided imagery on psychological well-being and immune function in patients with prior breast cancer. J Psychosom Res 2002;53:1131–7. [186] Sidransky D. Emerging molecular markers of cancer. Nat Rev Cancer 2002;2:210–9. [187] Silzle T, Randolph GJ, Kreutz M, Kunz-Schughart LA. The fibroblast: sentinel cell and local immune modulator in tumor tissue. Int J Cancer 2004;108:173–80. [188] Tlsty TD, Hein PW. Know thy neighbor: stromal cells can contribute oncogenic signals. Curr Opin Genet Dev 2001;11:54–9. [189] Straub RH, Westermann J, Scholmerich J, Falk W. Dialogue between the CNS and the immune system in lymphoid organs. Immunol Today 1998;19:409–13.

Neurobiology of cancer Interactions between nervous, endocrine ...

Boris Mravec a,b,∗, Yori Gidron c, Ivan Hulin a .... B. Mravec et al. .... by increasing levels of vascular endothelial growth factor and .... of cancer Interactions between nervous, e ... ring and modulating the tumorigenesis by the brain.pdf.

782KB Sizes 0 Downloads 215 Views

Recommend Documents

Drug interactions in cancer therapy
Pharmacia & Upjohn Company. Emcyt prescribing information. Pfizer [online] .... Crewe, H. K., Ellis, S. W., Lennard, M. S. & Tucker, G. T.. Variable contribution of ...

Drug interactions in cancer therapy
Improvements in in vitro methods and early clinical testing have made the prediction of potentially clinically ... as part of their cancer treatment or for the management of .... limited data that pertains to the interactions between grapefruit juice

Interactions between iboga agents and ... - Springer Link
K.K. Szumlinski (✉) · I.M. Maisonneuve · S.D. Glick. Center for Neuropharmacology and Neuroscience (MC-136),. Albany Medical College, 47 New Scotland ...

Dynamic Interactions between Visual Experiences ... - IEEE Xplore
Abstract—The primary aim of this special session is to inform the conference's interdisciplinary audience about the state-of-the-art in developmental studies of ...

D ynamic interactions between crimes
time series data we find that an increase in minor crimes dynamically triggers more severe crimes without the ... empirical support of the economic model of crime, it seems that favourable labour market conditions. 1 .... O denotes the (per capita) n

Interactions between macromolecules and ions: the ...
Oct 10, 2006 - not alter the hydrogen bonding network outside the direct vicinity of the .... layers, it is the direct interactions of the ions with the film that are ...

Multiple Endocrine Neoplasias - DidacticsOnline
Pancreas/GI endocrine. Pituitary. MEN II. Pheochromocytoma. Medullary thyroid cancer. (Mutation of RET proto-oncogene). Mucosal neuromas. Parathyroid. IIb.

Dynamic Interactions between Large-Scale Brain ...
Apr 19, 2012 - A separate account of errors in perceptual tasks focuses on reduced .... nents analysis was computed using eeglab software (Delorme and.

Interactions between fire and permafrost on peatland hydrology and ...
The following positions are currently available at University of. Alberta, Edmonton, Canada, supervised by Dr. David Olefeldt at the department of Renewable ... energy in downstream aquatic ecosystems, where it may be mineralized and ...

Membrane-mediated interactions between rigid ... - APS Link Manager
Sep 7, 2012 - ... do not deform the membrane, proportional to kBT ; an elastic and an entropic .... theory, we move on to computing interaction (free) energies.

Interactions between the Agulhas Current and the ...
Mar 29, 2014 - Interactions between the Agulhas Current and the ecologically important region of the Agulhas Bank are studied through the analysis of high resolution along-track altimetry, merged mapped altimetry and in situ measurements of current s

Hydrogen-Bonding Interactions between Formic Acid ...
controversy. These carboxylic acid-pyridine systems may adopt two different ... both P4VPy and P2VPy were ionic, while their data for the corresponding PMAA ...

Small-scale spatial variation in the interactions between ...
dispersal services (Snow 1971; McKey 1975; Howe and Estabrook 1977). However, the ..... Chavez-Ramirez F. and Slack R.D. 1994. Effects of avian foraging.

Opponent interactions between serotonin and dopamine
Neural Networks 15 (2002) 603–616 ... 1988), it starts from the simple idea of using two systems to .... administration to show that such an inhibitory action of.

Interactions between fire and permafrost on peatland hydrology and ...
Field work will be carried out in the vicinity of Fort Simpson, in the Northwest Territories. Qualifications: BSc or MSc in physical geography or related field.

Interactions Between Number and Space in Parietal ...
Patients who neglect the left portion of representational space show a similar neglect of the left portion of the 'mental number line'9. When asked to bisect a ...

anatomy of nervous system pdf
Sign in. Loading… Whoops! There was a problem loading more pages. Retrying... Whoops! There was a problem previewing this document. Retrying.

nervous system
നോഡീോകോശം - ഘടന. Page 12. TYPICAL NERVE CELL. Page 13. Page 14. Page 15. Page 16. TRANSMISSION OF IMPULSES. Page 17. ആോവഗപസരണം ...

The Costs of Cancer - American Cancer Society Cancer Action Network
solutions designed to eliminate cancer as a major ...... suffering and maintain the best possible quality of life for patients and .... wordpress.com/2016/01/8806-the-burden-of-medical- ... February 22, 2017. https://www.acscan.org/sites/default/.