Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jul 2.
Published in final edited form as: Immunol Cell Biol. 2021 Sep 12;99(9):949–961. doi: 10.1111/imcb.12496

Neuroimmunology of cancer and associated symptomology

Nicole N Scheff 1, Jami L Saloman 1
PMCID: PMC9250294  NIHMSID: NIHMS1820096  PMID: 34355434

Abstract

Evolutionarily, the nervous system and immune cells have evolved to communicate with each other to control inflammation and host responses against injury. Recent findings in neuroimmune communication demonstrate that these mechanisms extend to cancer initiation and progression. Lymphoid structures and tumors, which are often associated with inflammatory infiltrate, are highly innervated by multiple nerve types (e.g. sympathetic, parasympathetic, sensory). Recent preclinical and clinical studies demonstrate that targeting the nervous system could be a therapeutic strategy to promote antitumor immunity while simultaneously reducing cancer-associated neurological symptoms, such as chronic pain, fatigue and cognitive impairment. Sympathetic nerve activity is associated with physiological or psychological stress, which can be induced by tumor development and cancer diagnosis. Targeting the stress response through suppression of sympathetic activity or activation of parasympathetic activity has been shown to drive activation of effector T cells and inhibition of myeloid-derived suppressor cells within the tumor. In addition, there is emerging evidence that sensory nerves may regulate tumor growth and metastasis by promoting or inhibiting immunosuppression in a tumor-type specific manner. Because neural effects are often tumor-type specific, further study is required to optimize clinical therapeutic strategies. This review examines the emerging evidence that neuroimmune communication can regulate antitumor immunity as well as contribute to development of cancer-related neurological symptoms.

Keywords: metastasis, peripheral nerves, proliferation, tumor immunity

INTRODUCTION

Local and systemic neuroimmune interactions serve as key regulators of homeostasis. Humoral signaling enables the central nervous system to coordinate systemic immune responses while local innervation of lymphoid organs13 and associated vasculature support more refined regulation.4 Given that neuroimmune crosstalk plays an important role in maintaining homeostasis, researchers have begun to examine how this communication impacts cancer. Many solid tumors are highly innervated (more so than the healthy organ)5 and are associated with a large inflammatory infiltrate.6 Anatomical studies indicate that within some solid tumors, immune cells aggregate in close proximity to nerves.7 Recently, the fields of neuroscience and immunology/oncology have made a concerted effort to elucidate the role of neuroimmune crosstalk in tumor development. Historically, investigations largely focused on the mechanistic underpinnings of neurological symptoms. Behavioral comorbidities including pain, sensorimotor deficits, depression, anxiety, fatigue and cognitive disturbances are prevalent in cancer patients.817 This review summarizes findings about the role of neuroimmune communication in both tumorigenesis and the development of cancer-related changes in neurological processes (i.e. cancer symptoms).

NEUROIMMUNOLOGY OF CANCER

Models of tumor stroma often exclude nerve endings and little attention has been paid to the role of nerves in the development of tumors that originate outside of the nervous system. However, many solid tumors are highly innervated by multiple types of nerves (e.g. sympathetic, parasympathetic, sensory).5 The prevailing hypothesis underlying tumorigenesis is that cancer cells exploit immunological processes to induce an immunosuppressive environment.18 Specifically, tumors evade immune surveillance by subduing effector cells and promoting myeloid-derived suppressor cells (MDSCs). It has now been shown that nerve activity plays an important role in regulating tumor growth in a nerve-type specific manner,19 often through control of antitumor immunity (Figure 1).

Figure 1.

Figure 1.

The cancer–neuroimmune network. The tumor microenvironment contains malignant cells, subsets of effector and regulatory immune cells and the peripheral nervous system composed of three different populations of peripheral nerves (e.g. sympathetic, parasympathetic and sensory). The representative schematic of communication between the peripheral nervous system and tumor-associated immune cells including myeloid-derived cells [i.e. myeloid-derived suppressor cell (MDSC), macrophage, monocyte, neutrophil] and lymphocytes [i.e. natural killer (NK) cell, CD8+ T cell] is based on recent findings. Selective manipulation of neurotransmission alters the number and/or activity of the indicated immune target in the direction indicated by the arrow. Cell communication can be either direct or mediated by cytokines, chemokines, growth factors and checkpoint proteins [i.e. tumor necrosis factor alpha (TNF-α), programmed cell death-1 (PD1)]. Intratumoral neurotransmitters and neuropeptides are known to regulate cancer progression by their direct effects on malignant cells and the modulation of tumor-infiltrating immune cell function. Activation/blockade of neurotransmission elicits protumor and antitumor immune responses in a nerve-type and tumor-type-dependent manner.

Sympathetic nervous system

The sympathetic nervous system and hypothalamic–pituitary–adrenal axis are responsible for regulating the body’s physical response to stressful stimuli. The primary sympathetic neurotransmitters epinephrine and norepinephrine bind to neuronal adrenergic receptors driving the “flight or fight” response. Adrenergic receptors are also expressed by immune cells enabling direct as well as indirect regulation of both neurons and immune cells.20 Adrenergic inhibitors can be used for the treatment of psychological (e.g. anxiety) and physiological (e.g. hypertension) stress.2123 The utility of adrenergic inhibitors as adjuvant cancer therapy has been a hot topic because epidemiological studies suggest that beta-adrenergic receptor blocker use is associated with prolonged survival; the greatest increases in cancer-specific survival rates have been observed with melanoma, pancreatic, ovarian and breast cancers.2426 The degree of improvement varies with not only tumor type, but also the receptor subtype that is targeted. For instance, type 2, but not type 1, beta-adrenergic receptor antagonists enhance responsiveness to immunotherapy.26

Sympathetic nerve ablation or pharmacological (e.g. beta-blockers) inhibition of adrenergic signaling reduces stress signaling and tumor growth. In some cases, this loss of sympathetic nerve activity has been linked directly to changes in immune responses. Systemic administration of 6-hydroxydopamine was used to ablate sympathetic nerves in Wistar rats prior to inoculation with hepatoma cells; while chemical sympathectomy did not alter tumor incidence or survival, it was associated with a systemic (i.e. blood) shift in the neutrophil-to-lymphocyte ratio as well as increased tumor necrosis factor-alpha (TNF-α), suggesting increased extratumoral mobilization of innate immune cells.27 By contrast, treatment with a beta-blocker, propranolol, was sufficient to slow tumor growth and metastasis in multiple models of melanoma, breast and pancreatic cancers.2830 Here, the authors analyzed tumor-infiltrating leukocytes (TILs) and found that blockade of beta adrenergic signaling was associated with a significant decrease in infiltration of myeloid cells (e.g. MDSCs), and a concomitant increase in the cytotoxic lymphocytes.2830 Given the shift in TIL profile, it may not be surprising that adrenergic signaling blockade exhibits synergy with immunotherapy. Propranolol reduces the expression of programmed cell death-1 (PD1) on CD8+ T cells, which results in improved responses to anti-PD1 immunotherapy.29 The monoclonal antibody trastuzumab is used to treat HER2-positive breast cancers, but treatment resistance is common.31 Liu et al.31 found that propranolol increases antitumor activity by resensitizing tumor cells to trastuzumab. Beta-blockade has also been tested in models for hemopoietic malignancies. The stress neurohormones epinephrine, prostaglandin and corticosterone are thought to promote tumor growth via their suppression of natural killer (NK) cell activity.32 In the CRNK-16 rat model of leukemia, individual or combination therapy with nadolol, a nonspecific beta-blocker, and indomethacin, a COX inhibitor, significantly prolonged survival.32 Thus, suppression of sympathetic nerve activity promotes local antitumor immunity in multiple tumor models and exhibits potential for synergistic combination therapy regimens. Given that both adrenergic receptors are expressed on both neurons and immune cells,20 future studies will be required to determine the exact cell types governing adrenergic-dependent changes in tumor immunity.

Several clinical trials have utilized pharmacological approaches to establish the effect of beta blockers on response to conventional therapies in breast, ovarian and colorectal cancers. In addition to psychological stress associated with a cancer diagnosis, primary interventions such as surgery or chemotherapy can drive physiological stress that induces secretion of prostaglandins and catecholamines such as epinephrine/norepinephrine, all of which promote immunosuppression and metastases as well as reduce survival.3336 In a phase II randomized clinical trial, women with early stage breast cancer undergoing surgical resection were given 11 days of perioperative placebo or propranolol combined with the COX-2 inhibitor etodolac.37,38 Transcriptomic analyses of the resected tumor indicated that the drug treatment was associated with reduced circulating levels of proinflammatory mediators [e.g. interleukin (IL)-6, C-reactive protein, interferon-γ] and increased anti-inflammatory mediators (e.g. IL-10).37,38 This dual drug treatment was associated with shifts in the TIL profile, including decreased monocytes, increased NK cell activity and an increase in B cells.38 Interestingly, similar shifts in monocytes and NK cells were observed in colorectal cancer patients given perioperative propranolol plus etodolac, but there was a decrease in B-cell infiltration.39 The effect of propranolol alone has been tested in the adjuvant setting for patients with epithelial ovarian cancer undergoing either chemotherapy or surgery.40 Propranolol was associated with improved mood as well as significant reductions in proinflammatory IL-6 and IL-8 which correlated with response to therapy.40 Propranolol has also been assessed for effects on biomarkers in the treatment of hemopoietic malignancies. In a phase II trial of patients receiving hematopoietic cell transplantation, transcriptomic analyses suggest that adjuvant/neoadjuvant propranolol results in greater downregulation of transcripts associated with monocyte activation and a shift toward a CD34 stem/progenitor cell profile.41 While these small clinical trials show promising results, additional studies are needed to parse out the impact of sympathetic nerve inhibition at various disease stages and across different tumor types.

The addition of behavioral stress paradigms to preclinical models has been used to mimic the psychological stress associated with the diagnosis of cancer. Psychological stress is commonly induced through behavioral stressors (e.g. chronic restraint stress) which elevate circulating levels of epinephrine, norepinephrine and glucocorticoids as well as promote anxiety- and depression-like behaviors.32,42,43,44,45,46,47,48,49 Multiple studies have shown that these behavioral stressors are sufficient to accelerate tumor progression, dissemination and metastasis of a diverse range of cancers, including melanoma,43 glioma,48 leukemia,32,46 pancreatic,45,47 breast42 and prostate cancer.44 In many cases, treatment with beta blockers was sufficient to reverse the effects of behavioral stressors by preventing immunosuppression. Using a chronic unpredictable stress model, Chen et al.50 showed that chronic unpredictable stress or isoproterenol (beta adrenergic agonist) promotes more severe colonization of the lung by breast cancer cells. Stress-induced lung metastasis caused upregulation of the monocyte chemoattractant CCL2, resulting in an accumulation of monocytes and macrophages; depletion of monocytes/macrophages or propranolol prevented stress-induced metastasis.50 Isolation or overcrowding can also cause stress in rodents. Both stressors are associated with significant decreases in the size of lymphoid organs such as thymus in melanoma-bearing mice; the shift in lymphoid organ size was prevented by administration of propanolol.43 Initially, propranolol also retarded tumor growth in the overcrowded mice.43 Social or group housing partially abrogates tumor-induced depressive behaviors in tumor-bearing mice.49 Furthermore, when a social hierarchy is established in the group housing setting, submissive mice exhibit elevated anxiety and stress. Submissive melanoma-bearing mice have increased metastases to lung, decreased NK cell cytotoxicity and decreased basal oxidative burst in neutrophils and monocytes compared with their dominant cagemates51; this is correlative and a direct link between the elevated stress response, sympathetic nervous system (SNS) activation and the immune system has yet to be established in this context.

Physiological environment has also been implicated in regulation of the stress response. For example, a deviation from thermoneutral ambient temperature is sufficient to drive stress as measured by elevated plasma norepinephrine.29,30 Thermally induced stress accelerates melanoma, pancreatic and mammary tumor growth.29,30,52 Maintaining mice at thermoneutral temperatures reduces plasma epinephrine as well as lowers stress and anxiety behaviors. Housing mice at thermoneutral temperatures was sufficient to increase intratumoral effector T cells (e.g. CD8+) and reduce the number of immunosuppressive MDSCs and regulatory T cells.29,52 Furthermore, intratumoral effector cells exhibited increased markers of activation and reduced expression of exhaustion markers (e.g. PD1).29,52 The effects of temperature were occluded in beta-2-adrenergic receptor knockout mice, implicating the sympathetic signaling pathway in the regulation of adaptive tumor immunity. Thermal treatments (e.g. weekly hyperthermia, localized heat source) for mice housed at subthermoneutral temperatures were sufficient to reduce the accumulation of MDSCs to a similar extent as propranolol.53 Given the important role of physiological stress and sympathetic nerve activity in antitumor immunity, it may be possible to leverage this system to improve therapeutic responsiveness in patients.

Stress reduces cancer patients’ quality of life and may independently influence prognosis.54 While the studies discussed earlier suggest that pharmacological interventions may be useful in the adjuvant setting, it is important to note that nonmedical interventions such as mindfulness, meditation, yoga and other psychosocial strategies also reportedly improve quality of life and prognosis.55 The effects of such lifestyle changes on immune responses in cancer, however, have not been characterized. It is known that exercise can mobilize cytotoxic T and NK cells.56 Thus, in addition to pharmacological manipulations, future studies should consider nonmedical interventions that reduce sympathetic nerve activity as a potential means to promote antitumor immunity.

Parasympathetic nervous system

The primary neurotransmitter of parasympathetic neurons is acetylcholine, which activates muscles, glands and other organs. The most common approach to study parasympathetic activity is through manipulation of the vagus nerve (e.g. transection, electrical stimulation) or by targeting cholinergic signaling pharmacologically. Vagotomy has been shown to inhibit prostate57 and stomach58 tumor growth and metastasis. Conversely, vagotomy has been shown to promote pancreatic ductal adenocarcinoma59,60 as well as colon61,62 and breast cancer.63 While the effects of manipulating the vagus nerve have been attributed to changes in parasympathetic activity, it is well known that parasympathetic axons make up only 15–20% of the axons traveling in the vagus nerve; the majority are sensory fibers.6466 In transplant, chemical and transgenic models of breast cancer, viral expression of a bacterial voltage-gated sodium channel or diphtheria toxin subunit A was used to drive activation or ablate parasympathetic neurons, respectively.63 Bacterial voltage-gated sodium channel-dependent increases in parasympathetic activity reduced metastasis while diphtheria toxin subunit A-mediated ablation accelerated tumor growth and metastasis.63 Given that parasympathetic activity appears to exert opposing effects depending on the tumor type, it is important to determine whether the effects of vagotomy and/or vagus nerve stimulation on tumor responses are due purely to the small percentage of parasympathetic axons or whether there is also a sensory nerve component.

In addition to modulating primary tumor growth, parasympathetic activity has been implicated in the process of tumor immunity. One study demonstrated that vagal nerve stimulation upregulated splenic Tff2, a memory T-cell derived anti-inflammatory peptide, in a CXCR4-dependent manner; Tff2-dependent blockade of immunosuppression has been shown to slow colorectal tumorigenesis.61 It should be noted that it is unclear whether the stimulation parameters were selectively activating only parasympathetic fibers. In mammary tumors, genetically induced parasympathetic activation reduced the expression of the immunosuppressive markers PD1 and Foxp3 on CD4+ T cells and PD1 on CD8+ T cells.63 Furthermore, activation of the vagal parasympathetic neurons via electroacupuncture in mammary tumors reduced proinflammatory cytokines and infiltration of MDSCs as well as increased proportions of CD8+ T cells and NK cells.67 In pancreatic cancer models, vagotomy was associated with significantly elevated levels of TNF-α and increased numbers of tumor-associated macrophages.60 Treatment with bethanechol, a selective muscarinic acetylcholine receptor agonist, suppressed infiltration of myeloid cells and TNF-α levels, promoting antitumor immunity and reducing liver metastases.59 However, another recent study on pancreatic cancer showed that the nonselective agonist acetylcholine inhibited CD8+ T-cell recruitment, interferon-γ production and promoted Th2 responses, thereby fostering immunosuppression; in this case vagotomy promoted recruitment of CD8+ T cells and improved survival.68 Despite these conflicting findings, bethanechol is currently in clinical trials as adjuvant therapy for pancreatic cancer (NCT03572283). Currently, cholinergic agonists are Food and Drug Administration approved for treatment of elevated ocular pressure and urinary retention.

Sensory nervous system

The sensory nervous system has been considered largely in the context of cancer symptoms such as paraneoplastic syndromes and sensorimotor deficits.8,9 Sensory nerves innervating the body below the neck arise from spinal dorsal root ganglia and the nodose/jugular ganglia (vagal). Afferent signals transduce information to the central nervous system, but sensory neurons also have an important efferent component. For example, release of neurotransmitters and neuropeptides such as substance P (SP) and calcitonin gene related peptide (CGRP) significantly contribute to neurogenic inflammation and pain.69 Several denervation studies have provided evidence that sensory nerves promote pancreatic ductal adenocarcinoma and basal cell carcinoma growth (reviewed in Saloman et al.19). Sensory denervation prior to pancreatic tumor development significantly reduced the size of the inflammatory stromal compartment, which includes infiltrating leukocytes.70 Unfortunately, no direct analysis of immune cells was done in this study to examine the phenotype of TILs. However, it has been shown that SP increases cytotoxic immune responses to colorectal cancer cells in vitro.71 With only few studies done so far, it is difficult to conclude whether neuroimmune crosstalk is responsible for the effects of denervation on primary tumor growth and whether this is a targetable pathway for therapeutic interventions.

Perineural invasion (PNI) by tumor cells as well as hypertrophy and axonogenesis of intratumoral nerve fibers has been documented in peripheral nerves; both are associated with reduced quality of life, increased metastasis and poorer overall prognosis.72 Anatomical studies from resected pancreatic tumors show that these intratumoral nerves are prominently surrounded and infiltrated by immune cells.7,73 Furthermore, transcriptomic analyses of resected pancreatic tumors show that the degree of PNI is associated with reduced expression for cytotoxic T cells and increased expression of Th2 signatures.68 With respect to sensory neuron–immune interactions, in vitro studies have demonstrated that PNI is dependent on immune cells. While most of the clinical work on PNI does not identify what “type” of nerve is involved, many preclinical studies use sensory nerves as the model. These studies incorporated dorsal root ganglia or sciatic nerve into the classic tumor cell migration assay. Prior to the formation of pancreatic tumor cell–sensory neuron contacts, endoneurial macrophages are recruited and release glial cell derived neurotrophic factor (GDNF) that acts as a chemoattractant for the tumor cells to migrate to the sciatic nerve.73 Neonatal ablation of sensory afferents systemically reduces primary pancreatic tumor incidence (metastasis was not directly studied)19,70; however, systemic ablation and selective ablation of vagal sensory afferents result in increased breast cancer metastasis.74 This suggests that vagal versus spinal sensory afferents may have differing effects on tumor immunity. Alternatively, the role of sensory afferents could be tumor specific, as has been reported for the vagus nerve. For example, blocking SP promoted antitumor immunity in mice bearing tumors derived from a brain metastasis by upregulating CD8+ and CD4+ T cells.75 However, SP blockade in mice bearing tumors derived from a liver metastasis had a significant increase in MDSCs promoting an immunosuppressive environment.75 Regardless, sensory-derived SP clearly regulates metastasis through modulation of TILs; thus, sensory neuropeptides may be targets for adjuvant therapy.

NEUROIMMUNE REGULATION OF GLIAL TUMORS

Immune interactions with peripheral nerves, including neurons and glia (e.g. satellite, Schwann), are an important focus area for tumors of non-neural origin; however, there is also emerging evidence that neuroimmune crosstalk regulates glial-derived tumors. Neurons induced from human pluripotent stem cells harboring mutations in Nf1 were found to secrete the chemokine midkine, which directly activates CD8+ T cells.76 Canonically, activation of CD8+ T cells is associated with increased cytotoxic capacity, but here T-cell-derived CCL4 induced microglia to make growth factors that promote stem cell survival and glioma growth. Tumor cell expression of ligands that activate inhibitory immune checkpoint proteins is one of the major pathways thought to contribute to tumor-induced immunosuppression. However, cerebellar and cortical neuron expression of the canonical immune checkpoint protein PD-L1 is associated with increased survival in glioblastoma patients, suggesting that neuronal expression of canonical immune checkpoint proteins could contribute to regulation of antitumor immunity and tumor growth.77 More studies are needed to assess the impact of nonimmune cell expression of “immune” proteins to elucidate whether this is a potential new therapeutic target.

NEUROIMMUNOLOGY OF CANCER-RELATED PAIN

Almost half of all cancer patients struggle with moderate to severe cancer-related pain,78 which can arise throughout the disease process79; the pain incidence and intensity can increase with the progression of the cancer.78 While the etiology of cancer-related pain is debated, the common consensus is that cancer pain arises from processes involving crosstalk between tumor cells, the immune system and peripheral and central nervous system.8082

Primary tumor-related pain

An often-cited hypothesis regarding primary tumor-related pain is that tumor cells and immune cells in the cancer microenvironment produce mediators that activate and sensitize pain-sensing sensory neurons (i.e. nociceptors).82 TNF-α, a proinflammatory mediator released by both cancer and immune cells, has been implicated in nociceptive signaling in the presence of cancer. TNF-α antagonist, given systemically in a bone cancer pain mouse model, only partially reduced cancer-mediated mechanical hyperalgesia, indicating that TNF-α present in the tumor microenvironment, not circulating TNF-α, may contribute to tumor-related nociception. In an oral cancer mouse model, inhibition of TNF-α signaling reduced orofacial nociception induced by tongue cancer and decreased the number of CD3+ T cells infiltrating into the tongue83; clinical trials using a monoclonal antibody against TNF-α (i.e. infliximab) demonstrated a similar decrease in epidermal CD3+ T cells in patients with moderate to severe psoriasis vulgaris,84 but it is yet to be investigated clinically for cancer-related pain. Proteases, released from both primary tumor cells and immune cells (e.g. neutrophils and mast cells) in the tumor, bind and activate protease-activated receptor 2 (PAR2) on sensory neurons which also drives cancer pain.85,86 In a murine breast cancer model,87 proteases released after cell activation and degranulation evoked SP release in peripheral sensory nerves via PAR2 signaling, which subsequently contributed to sustained neuroinflammation and pain.88 Lastly, nerve growth factor (NGF), which promotes the local growth and survival of neurons, is secreted by both cancer and cancer-infiltrating immune cells.89,90 NGF can induce cancer pain via its association with PNI91; NGF has been demonstrated to promote PNI in mouse models of pancreatic92 and oral90 cancer. In addition, NGF can drive axonal sprouting in sensory and sympathetic nerves within the tumor microenvironment and intratumoral space; anti-NGF significantly decreased bone cancer-mediated nociceptive behavior, as well as sensory and sympathetic nerve sprouting in tumor-bearing mice.93

Inflammation within the peripheral nerve, called neuritis, arises from perineural immune cell infiltration into the nerve bundle. Most neuritis episodes in cancer-related studies have been studied in pancreatic cancer.94 Nerve presence within the tumor microenvironment positively correlates with the presence and severity of pancreatic neuritis as well as with the degree of pain in patients.94 Demir et al.7 have demonstrated that spatial interaction of peripheral nerves with mast cells may be at the root of the inflammatory response and hypersensitivity within the nerve. Furthermore, perineural mast cell numbers, but not macrophages or cytotoxic T cells, were increased in pancreatic patients with severe pain compared with those without pain.7

Metastasis-related pain

Many cancers, most commonly breast and prostate, will metastasize from their original site to the bone. Bone cancer is thought to be the most common cause of cancer-related pain, with the resulting pain thought to be a distinct type of persistent pain; the neurochemical changes documented in peripheral sensory neurons as well as the spinal cord are unique compared to the response to peripheral inflammation or nerve injury.95 Cancer-induced injury to sensory nerve fiber terminals, release of inflammatory mediators and increased bone degradation all impact the development of bone cancer pain.96 ATP, secreted from both cancer and immune cells, and P2X7 receptor upregulation in spinal microglia have been implicated in preclinical models of bone cancer pain; spinal P2X7 inhibition reduced cancer-related nociceptive behavior and blocked hyperactivity in the spinal cord.97 Sigma-1 receptor is a modulator of calcium signaling on the endoplasmic reticulum; in a rat model of bone cancer pain, administration of sigma-1 antagonist, BD-1047, resulted in a decrease in the number of activated microglia in the spinal cord dorsal horn.98 However, these studies were done prior to the establishment of pain and neuroinflammation and additional investigation is needed to determine the impact of sigma-1 antagonism after the onset of cancer-induce pain and neuroinflammation.

Osteoclasts, the bone’s innate immune cells, contribute to pain via inflammatory cytokine and chemokine production to create an acidic extracellular environment and increase bone resorption; this process ultimately leads to sensory and sympathetic nerve compression as well as bone fractures. Osteoclasts can be involved in several processes that lead to bone metastasis-related cancer pain, such as tumor growth and angiogenesis.99 Recent preclinical and clinical studies have demonstrated that inhibition of osteoclast signaling via src, a nonreceptor protein tyrosine kinase linked to spinal glutamate excitatory neurotransmission, may be used to treat cancer-related bone resorption and metastasis.100102 In bone cancer pain rat model, src inhibitors reduced nociceptive behavior via reduced N-methyl-d-aspartate activity as well as inhibition of bone resorption in the tumor.103 However, a recent randomized controlled trial of src inhibitor, saracatinib, did not show a significant effect on self-reported bone pain scores in patients with metastatic bone pain.104

Immune-mediated endogenous analgesia

Endogenous opioid peptides consist of beta-endorphin, met- and leu-enkephalin, dynorphin and nociceptin105; endogenous opioid peptides and their receptors are distributed in the central nervous system and peripheral nervous system and their contribution to antinociception has been extensively characterized.106 Outside of the nervous system, endogenous opioids have been identified in non-neuronal peripheral tissues (e.g. leukocytes, keratinocytes) and found to impact nociceptive signaling.107 For example, endothelin-1, an endogenous vasoconstrictor secreted by endothelin cells, contributes to endogenous analgesia by driving opioid secretion from keratinocytes.108

While the immune system is best known for its contribution to inflammatory pain, there are instances in which leukocytes provide an endogenous opioid-based mechanism of pain control.109 Lymphocytes, macrophages, neutrophils and mast cells have been found to contain beta-endorphin and met-enkephalin; minimal dynorphin has been detected in many immune subtypes.110,111 The current understanding is that when stressed or when activated by specific inflammatory mediators (e.g. IL-1β, corticotropic factor), immune cells secrete opioids, which activate opioid receptors on sensory neurons and promote analgesia.112 Cancer-recruited neutrophils make beta-endorphin and met-enkephalin, which are antinociceptive, while neutrophil depletion exacerbated nociception in a mouse oral cancer model.113 Endogenous opioid analgesia has a therapeutic benefit over exogenous opioids as it is restricted to the inflammatory site; thus, there are no side effects of opioid receptor activation in the central nervous system.114 These preclinical studies suggest that tumor-infiltrating immune cells can release opioids in the tumor microenvironment to inhibit cancer-related pain.113,115 Further research is needed to investigate whether cancer-fighting immune cells evoked by recently developed immunotherapies for cancer treatment116 increase the endogenous opioid signaling within the tumor as well. However, a recent clinical study involving multiple different solid tumor cases found that severe pain correlated with high amounts of circulating beta-endorphin, and that oxycontin treatment reduced pain as well as beta-endorphin levels.117 Therefore, additional preclinical and clinical studies are needed to determine whether immune-mediated analgesia can be leveraged for the treatment of cancer-related pain.

NEUROIMMUNE–CANCER COMMUNICATION IN CANCER-RELATED FATIGUE AND OTHER SYMPTOMS

Tumor-associated biological processes have a direct impact on affective and cognitive symptoms. Preclinical and clinical studies have provided evidence that cancer-induced immune-secreted cytokines can induce behavioral comorbidities, including depression, fatigue, impaired sleep and cognitive dysfunction.118 Furthermore, activation of the SNS as a result of the psychological stress from cancer diagnosis, treatment and survivorship can lead to alterations in the immune system such as decreased NK cell cytotoxicity and decreased basal oxidative burst in neutrophils and monocytes.51

Fatigue and sleep

Cancer patients often report cancer-related fatigue (CRF), a multifaceted condition coupled with decreased muscle mass, weakness and depression. CRF can present at any time throughout the disease process; it can decrease quality of life, limit functionality and decrease overall survival in patients. Recent research suggests that activation of proinflammatory cytokines in the periphery can impact the brain, leading to fatigue and other behavioral changes.119,120 Several clinical studies have shown an association between inflammatory markers and fatigue in cancer patients.119 For example, cytokine gene polymorphisms121 and human papilloma virus infection associated with head and neck cancer can influence inflammatory activity and fatigue.122 Immune cells recruited to the tumor microenvironment can produce proinflammatory cytokines. Shifts in circulating immune cell subtypes have also been noted in relation to CRF. There were more circulating T lymphocytes, specifically CD4+ and CD56+ effector T cells, in fatigued breast cancer survivors, compared with nonfatigued patients.123 However, because of the fact that most fatigue-related studies focus on cancer survivors after treatment, it is uncertain whether the neuroimmune communication associated with fatigue is driven by cancer treatment (i.e. chemotherapy) or the cancer itself. Preclinical models have begun to demonstrate that changes in peripheral immune cells can influence behavioral responses in mice,124 but not yet in the context of cancer.

Central nervous system-derived immune and inflammatory activity plays an important role in CRF. Tumors can influence neuroinflammatory processes, including alterations in brain proinflammatory cytokines.125 Norden et al.126 found that CRF and depression-like behaviors were correlated with increased IL-1β messenger RNA in the cortex and hippocampus. Furthermore, microgliosis has been implicated in the pathogenesis of CRF.125 A microglial inhibitor (minocycline) reduced tumor-induced expression of IL-1β in the brain as well as CRF and depression-like behavior.126 Although circulating inflammatory markers related to CRF have been extensively studied, additional data integrating in neural processes are needed to allow for a more thorough evaluation of biobehavioral contributors.

Sickness behavior, affective behaviors and cognitive impairment

Sickness behavior is characterized as lethargy, sleepiness, hyperalgesia and depressed mood, in response to an illness.127 The driving force behind sickness behavior is thought to be multiple proinflammatory incidences in response to cancer, resulting in a flood of proinflammatory cytokines such as TNF-α, IL-1β and IL-6 in circulation128; these peripheral cytokines then act on the brain through the vagus nerve or by crossing the blood–brain barrier, causing neuroinflammation.129 In a mouse model of human papilloma virus-positive head and neck cancer, increased IL-6, IL-1β and TNF-α expression in the liver and increased IL-1β expression in the brain were found in tumor-bearing animals as well as deficits in burrowing behaviors; minocycline was able to attenuate tumor-induced inflammation and depression-like behaviors, suggesting that microglial activation and associated neuroinflammation play a significant role.130

Dysregulated cytokine production associated with peripheral tumors can induce affective and somatic symptoms that can present months prior to and at diagnosis of cancer,131,132 which is often consistent with the tumor development timeframe.133,134 Prior to diagnosis, pancreatic cancer patients commonly report experiencing affective symptoms (e.g. crying, hopelessness) in addition to somatic symptoms (e.g. loss of appetite, insomnia).135 A study examining medical records of ovarian cancer patients found that, in the 6 months prior to diagnosis, these patients are significantly more likely to report affective and somatic symptoms compared with controls.136 Increased basal levels of brain proinflammatory cytokines were found mainly in the cortex and hippocampus in tumor-bearing rats with depression-like behavior and select cognitive impairments.126 In addition, inflammatory insult, such as lipopolysaccharide, in a mammary tumor-bearing rat exacerbated the neuroinflammatory IL-1β signaling and microglial activation across various brain regions.131 However, increased peripheral inflammatory markers linked to somatic and affective symptoms appear to be tumor-type specific. In prostate cancer patients with nonmetastatic disease, elevated serum TNF-α is associated with fatigue, depression and anxiety.137 By contrast, in a study of colorectal cancer patients, levels of TNF-α and C-reactive protein in plasma were not associated with baseline depressive symptoms.138 Additional studies are needed to understand changes in subtypes of circulating immune cells from cancer patients prior to treatment which will allow for a broader view of the neuroimmune interaction beyond systemic inflammatory markers.

CONCLUSIONS/FUTURE DIRECTIONS

Until recently, it was thought that the nervous system’s role in cancer was to react to tumor growth and immunity. It is now evident that neuroimmune communication can modulate cancer progression. Manipulation of the nervous system is associated with stark changes in TILs and circulating inflammatory cytokines, which directly affect tumor growth and neuroplasticity as well as contribute to the development of the neurological symptoms reported by many cancer patients. While the field of neuroimmunology in cancer is still in its infancy, there is sufficient evidence to suggest that the points of interaction between these systems could potentially reveal novel therapeutic targets and strategies. Furthermore, inflammatory signaling molecules currently being studied as targets for the treatment of cancer (e.g. checkpoint proteins, type I interferons) should be considered in the context of neurological signaling prior to translation to clinic. Additional interdisciplinary collaboration is needed to identify the cell type of origin and the cell type targeted by individual signaling pathways. We can then take advantage of the biology to refine therapeutic interventions and minimize undesired side effects.

ACKNOWLEDGMENTS

This work was supported by funding provided by the National Institutes of Health (R00DE28019, NNS) and the UPMC Hillman Cancer Center (JLS).

Footnotes

CONFLICT OF INTEREST

The authors have no conflicts of interest to report.

REFERENCES

  • 1.Mignini F, Streccioni V, Amenta F. Autonomic innervation of immune organs and neuroimmune modulation. Auton Autacoid Pharmacol 2003; 23: 1–25. [DOI] [PubMed] [Google Scholar]
  • 2.Coutu DL, Kokkaliaris KD, Kunz L, Schroeder T. Three-dimensional map of nonhematopoietic bone and bone-marrow cells and molecules. Nat Biotechnol 2017; 35: 1202–1210. [DOI] [PubMed] [Google Scholar]
  • 3.Hu D, Al-Shalan HAM, Shi Z, et al. Distribution of nerve fibers and nerve-immune cell association in mouse spleen revealed by immunofluorescent staining. Sci Rep 2020; 10: 9850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dantzer R Neuroimmune interactions: from the brain to the immune system and vice versa. Physiol Rev 2018; 98: 477–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Reavis HD, Chen HI, Drapkin R. Tumor innervation: cancer has some nerve. Trends Cancer 2020; 6: 1059–1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674. [DOI] [PubMed] [Google Scholar]
  • 7.Demir IE, Schorn S, Schremmer-Danninger E, et al. Perineural mast cells are specifically enriched in pancreatic neuritis and neuropathic pain in pancreatic cancer and chronic pancreatitis. PLoS One 2013; 8: e60529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Clouston PD, DeAngelis LM, Posner JB. The spectrum of neurological disease in patients with systemic cancer. Ann Neurol 1992; 31: 268–273. [DOI] [PubMed] [Google Scholar]
  • 9.Giglio P, Gilbert MR. Neurologic complications of cancer and its treatment. Curr Oncol Rep 2010; 12: 50–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kosturakis AK, He Z, Li Y, et al. Subclinical peripheral neuropathy in patients with multiple myeloma before chemotherapy is correlated with decreased fingertip innervation density. J Clin Oncol 2014; 32: 3156–3162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Boyette-Davis JA, Eng C, Wang XS, et al. Subclinical peripheral neuropathy is a common finding in colorectal cancer patients prior to chemotherapy. Clin Cancer Res 2012; 18: 3180–3187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Meyers CA, Albitar M, Estey E. Cognitive impairment, fatigue, and cytokine levels in patients with acute myelogenous leukemia or myelodysplastic syndrome. Cancer 2005; 104: 788–793. [DOI] [PubMed] [Google Scholar]
  • 13.Ahles TA, Saykin AJ, McDonald BC, et al. Cognitive function in breast cancer patients prior to adjuvant treatment. Breast Cancer Res Treat 2008; 110: 143–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cimprich B, Reuter-Lorenz P, Nelson J, et al. Prechemotherapy alterations in brain function in women with breast cancer. J Clin Exp Neuropsychol 2010; 32: 324–331. [DOI] [PubMed] [Google Scholar]
  • 15.Vin-Raviv N, Hillyer GC, Hershman DL, et al. Racial disparities in posttraumatic stress after diagnosis of localized breast cancer: the BQUAL study. J Natl Cancer Inst 2013; 105: 563–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hermelink K, Voigt V, Kaste J, et al. Elucidating pretreatment cognitive impairment in breast cancer patients: the impact of cancer-related post-traumatic stress. J Natl Cancer Inst 2015; 107: djv009. [DOI] [PubMed] [Google Scholar]
  • 17.Wefel JS, Vidrine DJ, Veramonti TL, et al. Cognitive impairment in men with testicular cancer prior to adjuvant therapy. Cancer 2011; 117: 190–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ribatti D The concept of immune surveillance against tumors. The first theories. Oncotarget 2017; 8: 7175–7180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Saloman JL, Albers KM, Rhim AD, Davis BM. Can stopping nerves, stop cancer? Trends Neurosci 2016; 39: 880–889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sharma D, Farrar JD. Adrenergic regulation of immune cell function and inflammation. Semin Immunopathol 2020; 42: 709–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Garakani A, Murrough JW, Freire RC, et al. Pharmacotherapy of anxiety disorders: current and emerging treatment options. Front Psychiatry 2020; 11: 595584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mohseni S, Joseph B, Peden CJ. Mitigating the stress response to improve outcomes for older patients undergoing emergency surgery with the addition of beta-adrenergic blockade. Eur J Trauma Emerg Surg 2021. 10.1007/s00068-00021-01647-00067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pathak A, Mrabeti S. β-blockade for patients with hypertension, ischemic heart disease or heart failure: where are we now? Vasc Health Risk Manag 2021; 17: 337–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yap A, Lopez-Olivo MA, Dubowitz J, et al. Effect of beta-blockers on cancer recurrence and survival: a meta-analysis of epidemiological and perioperative studies. Br J Anaesth 2018; 121: 45–57. [DOI] [PubMed] [Google Scholar]
  • 25.Na Z, Qiao X, Hao X, et al. The effects of beta-blocker use on cancer prognosis: a meta-analysis based on 319,006 patients. Onco Targets Ther 2018; 11: 4913–4944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kokolus KM, Zhang Y, Sivik JM, et al. Beta blocker use correlates with better overall survival in metastatic melanoma patients and improves the efficacy of immunotherapies in mice. Oncoimmunology 2018; 7: e1405205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Horvathova L, Tillinger A, Sivakova I, Mikova L, Mravec B, Bucova M. Chemical sympathectomy increases neutrophil-to-lymphocyte ratio in tumor-bearing rats but does not influence cancer progression. J Neuroimmunol 2015; 278: 255–261. [DOI] [PubMed] [Google Scholar]
  • 28.Jean Wrobel L, Bod L, Lengagne R, Kato M, Prévost-Blondel A, Le Gal FA. Propranolol induces a favourable shift of anti-tumor immunity in a murine spontaneous model of melanoma. Oncotarget 2016; 7: 77825–77837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bucsek MJ, Qiao G, MacDonald CR, et al. β-adrenergic signaling in mice housed at standard temperatures suppresses an effector phenotype in CD8+ T cells and undermines checkpoint inhibitor therapy. Cancer Res 2017; 77: 5639–5651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Eng JW, Reed CB, Kokolus KM, et al. Housing temperature-induced stress drives therapeutic resistance in murine tumour models through β2-adrenergic receptor activation. Nat Commun 2015; 6: 6426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Liu D, Yang Z, Wang T, et al. β2-AR signaling controls trastuzumab resistance-dependent pathway. Oncogene 2016; 35: 47–58. [DOI] [PubMed] [Google Scholar]
  • 32.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Armaiz-Pena GN, Gonzalez-Villasana V, Nagaraja AS, et al. Adrenergic regulation of monocyte chemotactic protein 1 leads to enhanced macrophage recruitment and ovarian carcinoma growth. Oncotarget 2015; 6: 4266–4273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Horowitz M, Neeman E, Sharon E, Ben-Eliyahu S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat Rev Clin Oncol 2015; 12: 213–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Neeman E, Ben-Eliyahu S. Surgery and stress promote cancer metastasis: new outlooks on perioperative mediating mechanisms and immune involvement. Brain Behav Immun 2013; 30(Suppl): S32–S40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shakhar G, Ben-Eliyahu S. Potential prophylactic measures against postoperative immunosuppression: could they reduce recurrence rates in oncological patients? Ann Surg Oncol 2003; 10: 972–992. [DOI] [PubMed] [Google Scholar]
  • 37.Shaashua L, Shabat-Simon M, Haldar R, et al. Perioperative COX-2 and β-adrenergic blockade improves metastatic biomarkers in breast cancer patients in a phase-II randomized trial. Clin Cancer Res 2017; 23: 4651–4661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Haldar R, Shaashua L, Lavon H, et al. Perioperative inhibition of β-adrenergic and COX2 signaling in a clinical trial in breast cancer patients improves tumor Ki-67 expression, serum cytokine levels, and PBMCs transcriptome. Brain Behav Immun 2018; 73: 294–309. [DOI] [PubMed] [Google Scholar]
  • 39.Haldar R, Ricon-Becker I, Radin A, et al. Perioperative COX2 and beta-adrenergic blockade improves biomarkers of tumor metastasis, immunity, and inflammation in colorectal cancer: a randomized controlled trial. Cancer 2020; 126: 3991–4001. [DOI] [PubMed] [Google Scholar]
  • 40.Ramondetta LM, Hu W, Thaker PH, et al. Prospective pilot trial with combination of propranolol with chemotherapy in patients with epithelial ovarian cancer and evaluation on circulating immune cell gene expression. Gynecol Oncol 2019; 154: 524–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Knight JM, Rizzo JD, Hari P, et al. Propranolol inhibits molecular risk markers in HCT recipients: a phase 2 randomized controlled biomarker trial. Blood Adv 2020; 4: 467–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Flaherty RL, Intabli H, Falcinelli M, et al. Stress hormone-mediated acceleration of breast cancer metastasis is halted by inhibition of nitric oxide synthase. Cancer Lett. 2019; 459: 59–71. [DOI] [PubMed] [Google Scholar]
  • 43.Hasegawa H, Saiki I. Psychosocial stress augments tumor development through β-adrenergic activation in mice. Jpn J Cancer Res 2002; 93: 729–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hassan S, Karpova Y, Baiz D, et al. Behavioral stress accelerates prostate cancer development in mice. J Clin Invest 2013; 123: 874–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kim-Fuchs C, Le CP, Pimentel MA, et al. Chronic stress accelerates pancreatic cancer growth and invasion: a critical role for beta-adrenergic signaling in the pancreatic microenvironment. Brain Behav Immun 2014; 40: 40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lamkin DM, Sloan EK, Patel AJ, et al. Chronic stress enhances progression of acute lymphoblastic leukemia via β-adrenergic signaling. Brain Behav Immun 2012; 26: 635–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Renz BW, Takahashi R, Tanaka T, et al. beta2 adrenergic-neurotrophin feedforward loop promotes pancreatic cancer. Cancer Cell 2018; 33: e77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang P, Wang Z, Yan Y, et al. Psychological stress up-regulates CD147 expression through beta-arrestin1/ERK to promote proliferation and invasiveness of glioma cells. Front Oncol 2020; 10: 571181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lamkin DM, Lutgendorf SK, Lubaroff D, Sood AK, Beltz TG, Johnson AK. Cancer induces inflammation and depressive-like behavior in the mouse: modulation by social housing. Brain Behav Immun 2011; 25: 555–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chen H, Liu D, Guo L, Cheng X, Guo N, Shi MA-O. Chronic psychological stress promotes lung metastatic colonization of circulating breast cancer cells by decorating a pre-metastatic niche through activating β-adrenergic signaling. J Pathol 244: 49–60. [DOI] [PubMed] [Google Scholar]
  • 51.Sá-Rocha VM, Sá-Rocha LC, Palermo-Neto J. Variations in behavior, innate immunity and host resistance to B16F10 melanoma growth in mice that present social stable hierarchical ranks. Physiol Behav 2006; 88: 108–115. [DOI] [PubMed] [Google Scholar]
  • 52.Kokolus KM, Capitano ML, Lee CT, et al. Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature. Proc Natl Acad Sci USA 2013; 110: 20176–20181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.MacDonald C, Ministero S, Pandey M, et al. Comparing thermal stress reduction strategies that influence MDSC accumulation in tumor bearing mice. Cell Immunol 2021; 361: 104285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Butler LD, Koopman C, Cordova MJ, Garlan RW, DiMiceli S, Spiegel D. Psychological distress and pain significantly increase before death in metastatic breast cancer patients. Psychosom Med 2003; 65: 416–426. [DOI] [PubMed] [Google Scholar]
  • 55.Alimujiang A, Khoja L, Wiensch A, et al. “I am not a statistic” ovarian cancer survivors’ views of factors that influenced their long-term survival. Gynecol Oncol 2019; 155: 461–467. [DOI] [PubMed] [Google Scholar]
  • 56.Graff RM, Kunz HE, Agha NH, et al. β2-Adrenergic receptor signaling mediates the preferential mobilization of differentiated subsets of CD8+ T-cells, NK-cells and non-classical monocytes in response to acute exercise in humans. Brain Behav Immun 2018; 74: 143–153. [DOI] [PubMed] [Google Scholar]
  • 57.Magnon C, Hall SJ, Lin J, et al. Autonomic nerve development contributes to prostate cancer progression. Science 2013; 341: 1236361. [DOI] [PubMed] [Google Scholar]
  • 58.Zhao CM, Hayakawa Y, Kodama Y, et al. Denervation suppresses gastric tumorigenesis. Sci. Transl. Med 2014; 6: 250ra115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Renz BW, Tanaka T, Sunagawa M, et al. Cholinergic signaling via muscarinic receptors directly and indirectly suppresses pancreatic tumorigenesis and cancer stemness. Cancer Discov 2018; 8: 1458–1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Partecke LI, Kading A, Trung DN, et al. Subdiaphragmatic vagotomy promotes tumor growth and reduces survival via TNFα in a murine pancreatic cancer model. Oncotarget 2017; 8: 22501–22512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Dubeykovskaya Z, Si Y, Chen X, et al. Neural innervation stimulates splenic TFF2 to arrest myeloid cell expansion and cancer. Nat Commun 2016; 7: 10517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.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: 281–286. [DOI] [PubMed] [Google Scholar]
  • 63.Kamiya A, Hayama Y, Kato S, et al. Genetic manipulation of autonomic nerve fiber innervation and activity and its effect on breast cancer progression. Nat Neurosci 2019; 22: 1289–1305. [DOI] [PubMed] [Google Scholar]
  • 64.Berthoud HR, Neuhuber WL. Functional and chemical anatomy of the afferent vagal system. Auton Neurosci 2000; 85: 1–17. [DOI] [PubMed] [Google Scholar]
  • 65.Asala SA, Bower AJ. An electron microscope study of vagus nerve composition in the ferret. Anat Embryol (Berl) 1986; 175: 247–253. [DOI] [PubMed] [Google Scholar]
  • 66.Atsumi K, Yajima T, Tachiya D, et al. Sensory neurons in the human jugular ganglion. Tissue Cell 2020; 64: 101344. [DOI] [PubMed] [Google Scholar]
  • 67.Zhang Z, Yu Q, Zhang X, et al. Electroacupuncture regulates inflammatory cytokines by activating the vagus nerve to enhance antitumor immunity in mice with breast tumors. Life Sci 2021; 272: 119259. [DOI] [PubMed] [Google Scholar]
  • 68.Yang MW, Tao LY, Jiang YS, et al. Perineural invasion reprograms the immune microenvironment through cholinergic signaling in pancreatic ductal adenocarcinoma. Cancer Res 2020; 80: 1991–2003. [DOI] [PubMed] [Google Scholar]
  • 69.Sousa-Valente J, Brain SD. A historical perspective on the role of sensory nerves in neurogenic inflammation. Semin Immunopathol 2018; 40: 229–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sinha S, Fu YY, Grimont A, et al. PanIN neuroendocrine cells promote tumorigenesis via neuronal cross-talk. Cancer Res 2017; 77: 1868–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Flageole H, Senterman M, Trudel JL. Substance P increases in vitro lymphokine-activated-killer (LAK) cell cytotoxicity against fresh colorectal cancer cells. J Surg Res 1992; 53: 445–449. [DOI] [PubMed] [Google Scholar]
  • 72.Dwivedi S, Krishnan A. Neural invasion: a scenic trail for the nervous tumor and hidden therapeutic opportunity. Am J Cancer Res 2020; 10: 2258–2270. [PMC free article] [PubMed] [Google Scholar]
  • 73.Cavel O, Shomron O, Shabtay A, et al. Endoneurial macrophages induce perineural invasion of pancreatic cancer cells by secretion of GDNF and activation of RET tyrosine kinase receptor. Cancer Res 2012; 72: 5733–5743. [DOI] [PubMed] [Google Scholar]
  • 74.Erin N Role of sensory neurons, neuroimmune pathways, and transient receptor potential vanilloid 1 (TRPV1) channels in a murine model of breast cancer metastasis. Cancer Immunol Immunother 2020; 69: 307–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Nizam E, Koksoy S, Erin N. NK1R antagonist decreases inflammation and metastasis of breast carcinoma cells metastasized to liver but not to brain; phenotype-dependent therapeutic and toxic consequences. Cancer Immunol Immunother 2020; 69: 1639–1650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Guo X, Pan Y, Xiong M, et al. Midkine activation of CD8+ T cells establishes a neuron-immune-cancer axis responsible for low-grade glioma growth. Nat Commun 2020; 11: 2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu Y, Carlsson R, Ambjorn M, et al. PD-L1 expression by neurons nearby tumors indicates better prognosis in glioblastoma patients. J Neurosci 2013; 33: 14231–14245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mercadante S, Arcuri E. Breakthrough pain in cancer patients: pathophysiology and treatment. Cancer Treat Rev 1998; 24: 425–432. [DOI] [PubMed] [Google Scholar]
  • 79.Caraceni A, Shkodra M. Cancer pain assessment and classification. Cancers (Basel) 2019; 11: 510–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jimenez Andrade JM, Mantyh P. Cancer pain, Chapter 4. In: Kruger L, Light A, eds. Translational Pain Research: From Mouse to Man. Boca Raton, FL: CRC Press/Taylor & Francis, 2009: 10.1201/9781439812105-c4 [DOI] [Google Scholar]
  • 81.Lozano-Ondoua AN, Symons-Liguori AM, Vanderah TW. Cancer-induced bone pain: Mechanisms and models. Neurosci Lett 2013; 557(Pt A): 52–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Schmidt BL. The neurobiology of cancer pain. Neuroscientist 2014; 20: 546–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Scheff NN, Ye Y, Bhattacharya A, et al. Tumor necrosis factor alpha secreted from oral squamous cell carcinoma contributes to cancer pain and associated inflammation. Pain 2017; 158: 2396–2409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gottlieb AB, Masud S, Ramamurthi R, et al. Pharmacodynamic and pharmacokinetic response to anti-tumor necrosis factor-α monoclonal antibody (infliximab) treatment of moderate to severe psoriasis vulgaris. J Am Acad Dermatol 2003; 48: 68–75. [DOI] [PubMed] [Google Scholar]
  • 85.Lam DK, Dang D, Zhang J, Dolan JC, Schmidt BL. Novel animal models of acute and chronic cancer pain: a pivotal role for PAR2. J Neurosci 2012; 32: 14178–14183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Viet CT, Schmidt BL. Biologic mechanisms of oral cancer pain and implications for clinical therapy. J Dent Res 2012; 91: 447–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Aich A, Afrin LB, Gupta K. Mast cell-mediated mechanisms of nociception. Int J Mol Sci 2015; 16: 29069–29092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Nguyen J, Luk K, Vang D, et al. Morphine stimulates cancer progression and mast cell activation and impairs survival in transgenic mice with breast cancer. Br J Anaesth 2014; 113(Suppl 1): i4–i13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Aloe L, Rocco ML, Balzamino BO, Micera A. Nerve growth factor: role in growth, differentiation and controlling cancer cell development. J Exp Clin Cancer Res 2016; 35: 116–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ye Y, Dang D, Zhang J, et al. Nerve growth factor links oral cancer progression, pain, and cachexia. Mol Cancer Ther 2011; 10: 1667–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liebig C, Ayala G, Wilks JA, Berger DH, Albo D. Perineural invasion in cancer: a review of the literature. Cancer 2009; 115: 3379–3391. [DOI] [PubMed] [Google Scholar]
  • 92.Saloman JL, Singhi AD, Hartman DJ, Normolle DP, Albers KM, Davis BM. Systemic depletion of nerve growth factor inhibits disease progression in a genetically engineered model of pancreatic ductal adenocarcinoma. Pancreas 2018; 47: 856–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jimenez-Andrade JM, Ghilardi JR, Castaneda-Corral G, Kuskowski MA, Mantyh PW. Preventive or late administration of anti-NGF therapy attenuates tumor-induced nerve sprouting, neuroma formation, and cancer pain. Pain 2011; 152: 2564–2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ceyhan GO, Bergmann F, Kadihasanoglu M, et al. The neurotrophic factor artemin influences the extent of neural damage and growth in chronic pancreatitis. Gut 2007; 56: 534–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Honore P, Rogers SD, Schwei MJ, et al. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience 2000; 98: 585–598. [DOI] [PubMed] [Google Scholar]
  • 96.Appel CK, Gallego-Pedersen S, Andersen L, et al. The Src family kinase inhibitor dasatinib delays pain-related behaviour and conserves bone in a rat model of cancer-induced bone pain. Sci Rep 2017; 7: 4792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Yang Y, Li H, Li TT, et al. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18. J Neurosci 2015; 35: 7950–7963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ruiz-Cantero MC, Gonzalez-Cano R, Tejada MA, et al. Sigma-1 receptor: A drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacol Res 2021; 163: 105339. [DOI] [PubMed] [Google Scholar]
  • 99.Andriessen AS, Donnelly CR, Ji RR. Reciprocal interactions between osteoclasts and nociceptive sensory neurons in bone cancer pain. Pain Rep 2021; 6: e867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Gnoni A, Marech I, Silvestris N, Vacca A, Lorusso V. Dasatinib: an anti-tumour agent via Src inhibition. Curr Drug Targets 2011; 12: 563–578. [DOI] [PubMed] [Google Scholar]
  • 101.Kim LC, Song L, Haura EB. Src kinases as therapeutic targets for cancer. Nat Rev Clin Oncol 2009; 6: 587–595. [DOI] [PubMed] [Google Scholar]
  • 102.Zhang S, Huang WC, Zhang L, et al. SRC family kinases as novel therapeutic targets to treat breast cancer brain metastases. Cancer Res 2013; 73: 5764–5774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.De Felice M, Lambert D, Holen I, Escott KJ, Andrew D. Effects of Src-kinase inhibition in cancer-induced bone pain. Mol Pain 2016; 12: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Danson S, Mulvey MR, Turner L, et al. An exploratory randomized-controlled trial of the efficacy of the Src-kinase inhibitor saracatinib as a novel analgesic for cancer-induced bone pain. J Bone Oncol 2019; 19: 100261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Przewlocki R, Przewlocka B. Opioids in chronic pain. Eur J Pharmacol 2001; 429: 79–91. [DOI] [PubMed] [Google Scholar]
  • 106.Niikura K, Narita M, Butelman ER, Kreek MJ, Suzuki T. Neuropathic and chronic pain stimuli downregulate central μ-opioid and dopaminergic transmission. Trends Pharmacol Sci 2010; 31: 299–305. [DOI] [PubMed] [Google Scholar]
  • 107.Khodorova A, Navarro B, Jouaville LS, et al. Endothelin-B receptor activation triggers an endogenous analgesic cascade at sites of peripheral injury. Nat Med 2003; 9: 1055–1061. [DOI] [PubMed] [Google Scholar]
  • 108.Viet CT, Ye Y, Dang D, et al. Re-expression of the methylated EDNRB gene in oral squamous cell carcinoma attenuates cancer-induced pain. Pain 2011; 152: 2323–2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hua S Neuroimmune interaction in the regulation of peripheral opioid-mediated analgesia in inflammation. Front Immunol 2016; 7: 293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Stein C, Pfluger M, Yassouridis A, et al. No tolerance to peripheral morphine analgesia in presence of opioid expression in inflamed synovia. J Clin Invest 1996; 98: 793–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chuang YC, Yang LC, Chiang PH, et al. Gene gun particle encoding preproenkephalin cDNA produces analgesia against capsaicin-induced bladder pain in rats. Urology 2005; 65: 804–810. [DOI] [PubMed] [Google Scholar]
  • 112.Hua S, Cabot PJ. Mechanisms of peripheral immune-cell-mediated analgesia in inflammation: clinical and therapeutic implications. Trends Pharmacol Sci 2010; 31: 427–433. [DOI] [PubMed] [Google Scholar]
  • 113.Scheff NN, Bhattacharya A, Dowse E, et al. Neutrophil-mediated endogenous analgesia contributes to sex differences in oral cancer pain. Front Integr Neurosci 2018; 12: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Kapitzke D, Vetter I, Cabot PJ. Endogenous opioid analgesia in peripheral tissues and the clinical implications for pain control. Ther Clin Risk Manag 2005; 1: 279–297. [PMC free article] [PubMed] [Google Scholar]
  • 115.Scheff NN, Alemu RG, Klares R 3rd, et al. Granulocyte-colony stimulating factor-induced neutrophil recruitment provides opioid-mediated endogenous anti-nociception in female mice with oral squamous cell carcinoma. Front Mol Neurosci 2019; 12: 217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Cramer JD, Burtness B, Ferris RL. Immunotherapy for head and neck cancer: Recent advances and future directions. Oral Oncol 2019; 99: 104460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Chong D, Shao L, Yang Y, Wang R, Yang C, Zhang B. Correlations of cancer pain degree with levels of β-EP, CGRP and PGE2 and the effects of oxycontin on them. J BUON 2018; 23: 1552–1557. [PubMed] [Google Scholar]
  • 118.Dantzer R Cytokine-induced sickness behavior: where do we stand? Brain Behav Immun 2001; 15: 7–24. [DOI] [PubMed] [Google Scholar]
  • 119.Bower JE. The role of neuro-immune interactions in cancer-related fatigue: Biobehavioral risk factors and mechanisms. Cancer 2019; 125: 353–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci 2008; 9: 46–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wang T, Yin J, Miller AH, Xiao C. A systematic review of the association between fatigue and genetic polymorphisms. Brain Behav Immun 2017; 62: 230–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Xiao C, Beitler JJ, Higgins KA, et al. Associations among human papillomavirus, inflammation, and fatigue in patients with head and neck cancer. Cancer 2018; 124: 3163–3170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bower JE, Ganz PA, Aziz N, Fahey JL, Cole SW. T-cell homeostasis in breast cancer survivors with persistent fatigue. J Natl Cancer Inst 2003; 95: 1165–1168. [DOI] [PubMed] [Google Scholar]
  • 124.Hodes GE, Pfau ML, Leboeuf M, et al. Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress. Proc Natl Acad Sci USA 2014; 111: 16136–16141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Santos JC, Pyter LM. Neuroimmunology of behavioral comorbidities associated with cancer and cancer treatments. Front Immunol 2018; 9: 1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Norden DM, Bicer S, Clark Y, et al. Tumor growth increases neuroinflammation, fatigue and depressive-like behavior prior to alterations in muscle function. Brain Behav Immun 2015; 43: 76–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hart BL. Biological basis of the behavior of sick animals. Neurosci Biobehav Rev 1988; 12: 123–137. [DOI] [PubMed] [Google Scholar]
  • 128.Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun 2007; 21: 153–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Maier SF. Bi-directional immune-brain communication: Implications for understanding stress, pain, and cognition. Brain Behav Immun 2003; 17: 69–85. [DOI] [PubMed] [Google Scholar]
  • 130.Vichaya EG, Vermeer DW, Christian DL, et al. Neuroimmune mechanisms of behavioral alterations in a syngeneic murine model of human papilloma virus-related head and neck cancer. Psychoneuroendocrinology 2017; 79: 59–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pyter LM, El Mouatassim BS, Sattar H, Prendergast BJ. Peripheral tumors alter neuroinflammatory responses to lipopolysaccharide in female rats. Brain Res. 2014; 1552: 55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Pyter LM, Pineros V, Galang JA, McClintock MK, Prendergast BJ. Peripheral tumors induce depressive-like behaviors and cytokine production and alter hypothalamic-pituitary-adrenal axis regulation. Proc Natl Acad Sci USA 2009; 106: 9069–9074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Aldea M, Craciun L, Tomuleasa C, Crivii C. The role of depression and neuroimmune axis in the prognosis of cancer patients. J BUON 2014; 19: 5–14. [PubMed] [Google Scholar]
  • 134.Schrepf A, Lutgendorf SK, Pyter LM. Pre-treatment effects of peripheral tumors on brain and behavior: neuroinflammatory mechanisms in humans and rodents. Brain Behav Immun 2015; 49: 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Green AI, Austin CP. Psychopathology of pancreatic cancer. A psychobiologic probe. Psychosomatics 1993; 34: 208–221. [DOI] [PubMed] [Google Scholar]
  • 136.Friedman GD, Skilling JS, Udaltsova NV, Smith LH. Early symptoms of ovarian cancer: a case-control study without recall bias. Fam Pract 2005; 22: 548–553. [DOI] [PubMed] [Google Scholar]
  • 137.Dirksen SR, Kirschner KF, Belyea MJ. Association of symptoms and cytokines in prostate cancer patients receiving radiation treatment. Biol Res Nurs 2014; 16: 250–257. [DOI] [PubMed] [Google Scholar]
  • 138.Archer JA, Hutchison IL, Dorudi S, Stansfeld SA, Korszun A. Interrelationship of depression, stress and inflammation in cancer patients: a preliminary study. J Affect Disord 2012; 143: 39–46. [DOI] [PubMed] [Google Scholar]

RESOURCES