Next Article in Journal
Role of RUNX3 in Restriction Point Regulation
Next Article in Special Issue
HTR1A, TPH2, and 5-HTTLPR Polymorphisms and Their Impact on the Severity of Depressive Symptoms and on the Concentration of Tryptophan Catabolites during Hepatitis C Treatment with Pegylated Interferon-α2a and Oral Ribavirin (PEG-IFN-α2a/RBV)
Previous Article in Journal
What Is the Significance of Lysosomal-Mediated Resistance to Imatinib?
Previous Article in Special Issue
Increased Lipid Peroxidation and Lowered Antioxidant Defenses Predict Methamphetamine Induced Psychosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Underlying Inflammation in Carcinoma Is Essential for the Resolution of Depressiveness

by
Milica M. Borovcanin
1,*,
Katarina Vesić
2,
Dragana Arsenijević
3,
Maja Milojević-Rakić
4,
Nataša R. Mijailović
5 and
Ivan P. Jovanovic
3
1
Department of Psychiatry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
2
Department of Neurology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
3
Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
4
Faculty of Physical Chemistry, University of Belgrade, 11000 Belgrade, Serbia
5
Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
*
Author to whom correspondence should be addressed.
Cells 2023, 12(5), 710; https://doi.org/10.3390/cells12050710
Submission received: 26 December 2022 / Revised: 3 February 2023 / Accepted: 22 February 2023 / Published: 23 February 2023

Abstract

:
In modern clinical practice and research on behavioral changes in patients with oncological problems, there are several one-sided approaches to these problems. Strategies for early detection of behavioral changes are considered, but they must take into account the specifics of the localization and phase in the course and treatment of somatic oncological disease. Behavioral changes, in particular, may correlate with systemic proinflammatory changes. In the up-to-date literature, there are a lot of useful pointers on the relationship between carcinoma and inflammation and between depression and inflammation. This review is intended to provide an overview of these similar underlying inflammatory disturbances in both oncological disease and depression. The specificities of acute and chronic inflammation are considered as a basis for causal current and future therapies. Modern therapeutic oncology protocols may also cause transient behavioral changes, so assessment of the quality, quantity, and duration of behavioral symptoms is necessary to prescribe adequate therapy. Conversely, antidepressant properties could be used to ameliorate inflammation. We will attempt to provide some impetus and present some unconventional potential treatment targets related to inflammation. It is certain that only an integrative oncology approach is justifiable in modern patient treatment.

1. Introduction

Treating patients with cancer is challenging. In modern clinical practice and research on behavioral changes in patients with oncologic problems, there are several one-sided approaches to this problem. Oncologists are concerned in great detail with localization of the primary oncologic process, pre- and post-operative care, protocols for chemotherapy and radiation therapy, and monitoring for recurrence. However, it is quite common for psychiatrists to be involved in some of the phases of integrative treatment. Mental predisposition could be discussed in the etiology of various carcinomas [1]. Mental disturbances could be a consequence of the patient’s awareness of the illness onset and its possible impact on the patient’s overall quality of life, or they may follow somatic perturbations and be an impact of the various therapies applied [2,3]. Mental disorders could also induce cancer recurrence [4].
In the new therapeutic strategies for the treatment of neuropathic pain as an oncological complication, it was very interesting to draw a parallel between the changes in the acute and chronic phases of pain and mental disorders management [5]. Inflammatory processes, both acute and chronic, are a hallmark of both oncological and mental disorders [6,7]. The exacerbation of somatic disorders and mental illnesses could reflect acute inflammation, whereas prolonged processes are related to chronic inflammation [8,9,10]. The question is if these landmarks could induce depressive symptomatology, and if so, to what extent and whether these phenomena should be treated as simple non-comorbid depression. In the up-to-date literature, there are a lot of useful pointers on the relationship between carcinoma and inflammation and between depression and inflammation. This review article aimed to compare and integrate these complex interactions in the same context of carcinoma and depression comorbidity. Further, we will try to use this information to potentially improve the clinical approach and discuss the importance of the resolution of inflammation as a new treatment strategy in the cooccurrence of carcinoma and depression.

2. Acute and Chronic Inflammation in Carcinoma

Inflammation represents the systemic host response to tissue damage. It is usually caused by injury, ischemia, infection, or chemical exposure [11,12]. Additionally, inflammation plays an important role in tissue repair, regeneration, and remodeling [13]. The inflammatory response involves the recruitment and action of the immune response [14].
Inflammation occurs in two stages, acute and chronic inflammation [13]. Acute inflammation is a part of innate immunity initiated by immune cells and lasts for a short time [15]. It serves as a defense against infection, tissue damage, and allergens. Receptors of innate immunity recognize the structures of microorganisms (pathogen-associated molecular patterns—PAMPs), but also molecules that are released from damaged host cells [16,17]. These molecules are called danger-associated molecular patterns (DAMPs) and represent proteins or nucleic acids that are not normally found outside the cell. The most important DAMPs include chromatin-associated protein high-mobility group box 1 (HMGB1), adenosine triphosphate (ATP), uric acid (UA), deoxyribonucleic acid (DNA), and degraded extracellular matrix (ECM)-like heparan sulfate and hyaluronan. PAMPs and DAMPs are recognized via pattern recognition receptors (PRRs) [16,17]. The term “alarmin” is today used as a synonym for DAMP [18].
In acute inflammation, pro-inflammatory mediators such as acute-phase proteins, prostaglandins, leukotrienes, oxygen- and nitrogen-derived free radicals, chemokines, growth factors, and cytokines that are released by immune defense cells locally at the site of inflammation cause neutrophil infiltration [19,20,21,22]. C-reactive protein (CRP), fibrinogen, and procalcitonin (PCT) are part of an innate immune response that is detectable in serum within a few hours of the initiation of inflammation [19]. They facilitate the inflammatory process and represent hallmarks of acute inflammation [19]. Subsequently, other cells of innate and adaptive immunity (e.g., macrophages and lymphocytes) are recruited to the inflammatory environment [14]. In response to DAMPs, innate immune cells secrete cytokines that mediate normal cellular processes and communication between leukocytes and other cells, but also regulate the host’s response to damage [21,22].
Cytokines can exert proinflammatory and anti-inflammatory effects both locally and systemically [23]. Activated cells of innate immunity produce the most important proinflammatory cytokines: interleukin (IL)-1, tumor necrosis factor-alpha (TNF-α), IL-6, IL-12, and IL-23 [23]. Conversely, the cells of adaptive immunity—activated T lymphocytes—produce interferon-gamma (IFN-γ) and IL-17 [21,22]. Some cytokines, such as IL-1α and IL-33, act as alarmins [18]. They are released from host cells as a result of injury or death and subsequently mobilize and activate immune cells [18].
The resolution of acute inflammation begins when PAMPs and DAMPs are no longer present [14]. However, if the pathogen cannot be completely eradicated or there is a constant source of self-antigens or a growing tumor that continuously disrupts tissue structure and induces the production of inflammatory cytokines, the second stage of inflammation, chronic inflammation, occurs [15]. Long-lasting chronic inflammation can lead to many chronic diseases including cardiovascular, respiratory, neurodegenerative diseases, and cancer via dysregulation of various signaling pathways [13,14,24].
When Rudolf Virchow described leukocytes within primary tumor tissue, a possible link between inflammation and cancer was established in the 19th century [25,26]. Today, it is obvious that inflammation plays an important role in the biology of tumors. Inflammation may play an anti- or pro-tumorigenic role. Acute inflammation in neoplastic tissues is indicative of an anti-tumor immune response [25,26]. In chronic inflammation, the inflammatory microenvironment facilitates cell mutations and proliferation leading to tumor development [27]. Alteration of several signaling pathways may contribute to the development of genetic and epigenetic changes in local tissue cells [28,29]. Additionally, chronic inflammation attenuates anti-tumor immunity and affects cell proliferation, death, senescence, DNA mutation, and angiogenesis [12,25,30,31,32]. The question remains whether the inflammation is a consequence of the anti-tumor immune response or whether the tumor arose in the setting of chronic inflammation.

3. Acute and Chronic Inflammation in Depression

Pro-inflammatory peripheral biomarkers elevation, a higher risk of depression in inflammatory and autoimmune diseases, the ability of immune mediators to induce depressive symptoms, and the fact that activated microglial cells reduce levels of serotonin and generate oxidative stress (OS) molecules all point to immune system involvement in the pathogenesis of depression [33]. Blood-brain barrier (BBB) permeability, the brain-gut axis, and the brain-fat axis bring systemic, particularly inflammatory, changes into the spotlight, not just central nervous system (CNS) disturbances in depression [34].
Specific depressive symptomatology was explored in correlation with inflammatory changes in the periphery. Majd et al. (2020) conducted a narrative review and indicated that there is an association between neurovegetative symptoms of depression, such as sleep problems, fatigue or loss of energy, appetite changes, and inflammation [35]. Increased inflammatory markers were measured in patients with major depressive disorder: IL-1β, IL-6, TNF-α, and CRP. Peripheral inflammation could signal the brain by leaky regions in the BBB, the cytokine transport system, and the vagus nerve. They based their conclusions on Capuron et al. (2002) [36], who demonstrated that IFN administration causes neurovegetative symptoms in the first two weeks, which are less responsive to antidepressant therapy, and depressed mood and cognitive symptoms later, which are responsive to antidepressants. Among other prominent theories of depression, the cytokine theory has played an important role in clinical practice [37]. Cytokines and peripheral immune cell counts could serve as biomarkers for distinct subgroups of inflamed depression and direct further treatment [38].
As recently noted in coronavirus disease (COVID-19), acute inflammation could be followed by behavioral changes termed “sickness behavior”, the resolution of which follows eradication of the infection, although in some cases psychotropic medications are required to resolve mental symptoms, particularly agitation [39]. It seems that some individuals have a predisposition to an exaggerated immune response to an infectious agent that could be harmful, not protective, and also lead to a later onset of depression [40]. The peripheral immune response is particularly exacerbated in depressive patients that are resistant to antidepressants [41]. Resilient animals do not display exacerbated immune responses following acute and chronic stress, suggesting that positive affectivity could buffer the negative impact of stress on immunity [42].
This hypersensitivity could be linked to the role of IL-6 as an important marker. A recently published first meta-analysis with a robust sample reported an adjusted association between IL-6 and future depression [43]. In addition, a small prospective association between depression and IL-6 was observed in both directions. If inflammation is prolonged and chronic, it is important to consider whether symptoms meet the threshold for a diagnosis of a depressive episode and require treatment. However, the elevation of IL-6 may be associated not only with chronic inflammation but also with other pathological processes that may also be observed in depression [43,44].

4. Animal Models of Inflammatory-Induced Depression in Carcinoma

The estimated high prevalence of depression in cancer patients and the insufficient data on the mechanisms by which tumors per se may alter brain functions, including mood and cognition, have engaged the preclinical research community to search for novel cancer-induced models. The main advantage of using animal models in research is the control of confounding variables that are difficult to control in the clinical setting and the ability to unravel mechanistic interactions between neural, immune, and inflammatory processes through which tumors alter brain function. Animal models provide a better explanation for the independent impact of tumor-associated biological processes on affective and cognitive symptoms, independent of cancer-associated stress and treatments.
Significant behavioral changes were found in mice with implanted tumors, characterized primarily by an increase in avoidance behavior and a decrease in immobility, defensive-submissive behavior, and non-social exploration [45]. Changes in brain plasticity as a result of disturbed neural redox homeostasis were detected in the brains of tumor-bearing mice with depressive-like behavior [46,47,48]. Structural evidence for a depressive-like state induced in a model of mammary cell carcinoma was also observed through decreased dendritic branching of pyramidal neurons in the medial prefrontal cortex [49].
Lipopolysaccharide is a component of gram-negative bacteria commonly used to induce a potent inflammatory response and behavioral changes that rapidly resolve within 24 h, followed by hyperalgesia [42]. Cytokine production in the tumor microenvironment can be detectable in the general circulation of experimental models of various tumor types, as well as in brain areas responsible for mood regulation. These studies reported increased plasma levels of IL-6, IL-12, TNF-α, IL-10, and IL-1β, but also increased IL-1β, IL-10 expression of IL-1β mitochondrial ribonucleic acid in the cortex and hippocampus, and increased levels of IL-6 and TNF-α in the hippocampus [50,51,52,53,54]. Hippocampal inflammation was related to depressive-like behavior in breast cancer mice, and also gastric-cancer-bearing mice with a significant increase in IL-6, IL-1β, reactive oxygen species (ROS), and cyclooxygenase-2 (COX2) [55,56]. The model of chronic stress and smoke exposure induced depression-like behavior and lung cancer, respectively, in mice, with the synergistic effect in a combined model manifested through a more prominent inflammatory response [57]. However, the impact of antidepressant fluoxetine was significantly attenuated under the conditions of chronic stress and LPS-induced inflammation, suggesting the role of chronic inflammation in the development of treatment-resistant depression [58]. We could identify several important underlying cascades in the development of depressiveness induced by inflammation.
Activation of inflammasomes, particularly nod-like receptor family pyrin domain containing 3 (NLRP3), may occur through DAMPs or PAMPs mediated by toll-like receptors (TLRs) and subsequently activate important intracellular pathways such as IFN I and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) [34]. At the cellular level, repercussions of these processes could be the production of IL-1α, IL-1β, TNF-α, and IL-6, as well as the activation of microglia and the impairment of astrocytes in depression [34].

5. Underlying Inflammatory Disturbances in Carcinoma and Depression Co-Occurrence

Somatic illnesses could be followed by mental disturbances, or mental disorders could be a typical response to illness that vanished in reconvalescence with the illness resolution or could persist after somatic illness recovery [5,59]. Hart was the first to propose the concept that “sickness behavior” occurs as a short-term reaction in an acute inflammatory state and is crucial for the survival of the individual [60]. However, when inflammation becomes chronic, as in autoimmune diseases, neurodegenerative diseases, cardiovascular diseases, diabetes and obesity, and cancer, mood symptoms predominate and can worsen the disease.
Nearly 30% of cancer patients meet the criteria for a psychiatric diagnosis of depression, neurotic and stress-related disorders, adjustment disorders, sleep disorders, or delirium [61]. The problem of insomnia is very pronounced in patients in the active and stable phase of cancer, especially when associated with a pain syndrome and distress [62]. With regard to the onset and persistence of depressive symptomatology, it was very interesting for us to consider the overlap with pain and fatigue as symptoms of the cluster, presented as two or three concurrent and interrelated symptoms that may or may not have a common etiology and pathophysiological pathways [63,64]. Dodd et al. (2001) defined pain, fatigue, and insomnia in cancer patients as a cluster [65]. Recently, Charalambous et al. (2019) provided preliminary evidence that targeting fatigue, anxiety, and depression in patients with breast and prostate cancer may have a meaningful effect on pain as a related symptom [66]. A proposed underlying mechanism in the pathogenesis of these symptoms includes systemic inflammation with high pro-inflammatory cytokine levels, oxidative stress, and neuroendocrine-immune alterations [67,68,69,70,71,72]. Inflammation-mediated tryptophan catabolism along the kynurenine pathway might contribute significantly to the development of fatigue and depression in cancer patients [73].
Consideration of the common neuroimmune mechanisms of chronic pain and depression and the possible corrective anti-inflammatory effect of antidepressants seem to be of greater importance in this case [74]. Therefore, researchers have developed a model of inflammatory cytokine activity in cancer to explain the co-occurrence of pain, fatigue, and sleep disturbances [75] (summarized in Figure 1). Sometimes it is necessary to remember that the primary goal is to eliminate pain sensations to prevent the onset of depressive symptoms. Functioning could be especially compromised with pain sensations that are also correlated with ongoing inflammation [76]. Acute pain was also associated with acute inflammation, and chronic inflammation reflected chronic pain [59,77]. Chronic pain and depression in humans are associated with persistent low-grade inflammation rather than severe systemic inflammation, with only a partially common underlying mechanism [77]. Neuropathic pain has been shown to be associated with increases in the tryptophan-metabolizing enzyme indolamine 1,3 deoxygenase (IDO1) in the liver but not in the brain, and antagonism of the N-methyl-D-aspartate (NMDA) receptor by kynurenic acid [77]. On the contrary, co-morbid depression was mediated downstream of spinal cord IL-1β signaling and the formation of kynurenine and its metabolites in the brain [77,78].
Along with anxiety and depression, cancer-related fatigue is one of the most common symptoms in cancer patients [79]. Fatigue and depression have similar clinical presentations (Figure 1). Fatigue can occur independently, be a prodromal symptom of depressive disorders, or be part of a developed depression [80]. Fatigue is defined as a loss of energy that can affect physical, mental, or cognitive functioning and is manifested by loss of motivation, apathy, and reduced concentration and attention [81]. The above symptoms are important characteristics of depressive mood disorder. For these reasons, it is sometimes very difficult in clinical practice to distinguish whether it is just fatigue or depression. Recently, our research group has pointed out that acute and chronic inflammation have a significant impact on fatigue and depression in patients with the inflammatory and neurodegenerative disease multiple sclerosis. We observed that peripheral inflammation was related to fatigue and postulated that brain inflammation in acute episodes could further lead to neurodegeneration and mood and cognitive changes [70].
The new important clinical entity of paraneoplastic disorder should be considered in the context of the clinical field of autoimmune-mediated depression [82]. Paraneoplastic neurologic syndromes (PNSs) are rare cancer-related diseases that can affect any level of the central and peripheral nervous systems [83]. These disorders do not result from tissue invasion by the tumor, metastases, or metabolic or toxic effects of cancer therapy [84]. PNSs are caused by an immune response directed toward neural self-antigens aberrantly expressed by neoplastic cells and marked by specific autoantibodies [83,85]. Although PNSs can occur in any type of tumor, the most frequently associated malignancies include ovarian and breast cancer, small-cell lung cancer, thymoma, Hodgkin’s lymphoma, and neuroendocrine tumors [86]. The exact immunopathogenic mechanisms for most paraneoplastic syndromes are still unclear. The autoimmune theory postulates an immune cross-reaction between antigens expressed by tumor cells (“onconeural” antigens) and neurons [87]. The autoimmune response, initially directed against tumor cells, results in further damage to neurons that physiologically express the same antigen [86]. The target of the immune attack can be intracellular antigens (anti-Hu, anti-Yo, anti-Ma2, anti-Ri, GAD), antigens on synaptic receptors (NMDA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, γ-aminobutyric acid receptor) or ion channels, and other cell-surface proteins (LGI 1, GQ1b) [88,89,90,91,92,93,94,95]. The main effector of the immune response in PNSs associated with antibodies directed against intracellular antigens is the CD8+ cytotoxic T cell, whose action results in rapid and extensive neuronal death by cytotoxic activity [96]. Mild signs of inflammation are commonly detected in the cerebrospinal fluid in the early phases of these disorders [97]. Antibodies against plasma membrane antigens, such as ion channels and surface receptors, may play a pathogenic role as direct effectors in neural tissue injury. Mechanisms by which these antibodies affect the targeted cells include antigen internalization and degradation, activation of complement cascades, antibody-dependent cell-mediated cytotoxicity, and blockade of receptor function [96]. Paraneoplastic syndromes of the CNS can be present with neuropsychiatric and cognitive symptoms, abnormal movements, new-onset epilepsy, and sleep disorders [98].
Over the past decade, evidence has accumulated of an intriguing relationship between cancer and neurodegenerative diseases. Progression of both conditions is primarily defined by a set of molecular determinants that are complementarily dysregulated or share important underlying biological mechanisms that promote cell proliferation and apoptosis, including alarmins (discussed in detail in [99]). DNA, cell cycle aberrations, redox imbalance, inflammation, and immunity are closely associated with shared characteristics of cancer and neurodegenerative diseases. The question arises whether each depressive episode and these kinds of repeated excessive immune and autonomic dysregulation could also contribute to neurodegeneration.

6. Potential for New Anti-Inflammatory Strategies in Cooccurrence of Carcinoma and Depression

The basic mechanism of action of conventional therapy for malignant diseases, such as radiotherapy and chemotherapy, is to induce the death of tumor cells [100]. However, the process of tumor cell necrosis is often triggered as an accompanying phenomenon in addition to the desired apoptosis. Necrosis is followed by the release of cellular contents outside the cell. Thus, endogenous alarmins reach the intercellular space and become inducers and facilitators of inflammation [100]. In this way, therapeutically induced tumor necrosis may be beneficial to the host [101,102]. Therefore, another no less important mechanism of action of the therapy is the induction of inflammation and the strengthening of the antitumor immune response. New therapeutically induced tumor necrosis may benefit the innate antitumor immune response, as necrotic cells facilitate the maturation of antigen-presenting cells [103,104]. Mature antigen-presenting cells, especially dendritic cells, induce a potent acquired antitumor response. Thus, the increase in systemic values of proinflammatory cytokines of innate immunity is accompanied by an increase in values of cytokines of acquired immunity. Chronic inflammation is present in and around most tumors, including those not causally related to an inflammatory process [105]. The percentage of patients with inflammatory components in the tumor microenvironment varied from 28% to 63% depending on tumor type [106]. Anti-tumor therapy is usually followed by a wave of acute inflammation that changes the intensity and course of the antitumor immune response [100].
Although radiotherapy and chemotherapy are options for the treatment of cancer, other treatments are increasingly being explored today, such as immunotherapy [107]. The use of monoclonal antibodies, immunomodulatory agents, modulated immunocompetent cells, or blocking antibodies for checkpoint molecules has shown significant results in cancer therapy and has fundamentally changed the approach to cancer therapy [108,109]. The discovery of checkpoint molecule inhibitors was awarded the Nobel Prize [110]. The blockade of cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death protein 1 (PD1) molecules with antibodies is now very topical and has also found its application in clinical practice [111]. Research on blocking other checkpoint molecules such as T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), the cluster of differentiation 96 (CD96), natural killer receptor NKG2A is in full swing [112,113,114,115]. A strong effect of the application of this type of therapy is the enhancement of both innate and acquired antitumor immune responses [110,111,112,113,114,115]. This phenomenon is almost always accompanied by increased production of pro-inflammatory cytokines and momentum of inflammation in the host. These effects could be unwanted in the propagation of inflammation and consequently trigger depressive symptomatology.
Since alterations of various cytokines have been established in both depression and cancer, cytokine inhibitors deserve more detailed discussion. Infliximab, a TNF antagonist, improves depressive symptomatology by decreasing CRP levels [116] but has also shown beneficial effects in treating cancer-related fatigue [117]. Adalimumab, another TNF-α-specific neutralizing monoclonal antibody similar to infliximab, has been shown to significantly improve depressive symptomatology in patients with various chronic diseases [118,119,120], but without studies in psychiatric patients. Etanercept, another TNF-α antagonist, reduced depressive-like behavior in preclinical models, but also clinical studies in patients with psoriasis and rheumatoid arthritis [121,122,123,124,125]. Pentoxifylline, a methylxanthine drug that acts as a strong non-selective TNF-α inhibitor, has improved depressive behavior in animal models but has also shown positive results as an add-on treatment for depression [126,127,128]. Ustekinumab, an inhibitor of IL-12 and IL-23, dupilumab, an antagonist of the receptor of IL-4, ixekizumab, an IL-17A inhibitor, and guselkumab, an IL-23 inhibitor, have all been for their antidepressant action [129,130,131,132]. Although cytokine inhibitors have a more targeted effect on depression-related inflammation, these results were limited to specific patient groups. Because cytokine inhibitors are large molecules, they cannot cross the BBB, suggesting that their anti-inflammatory action is limited to peripheral TNF-α. This does not preclude their efficacy, but further studies are needed to determine their potential for treating depression in the presence of concomitant carcinoma.
Conversely, re-establishing balance in the peripheral secretion of cytokines is observed after antidepressant use and the resolution of depression. The most recent pharmacological protocols for the treatment of depression in carcinoma target monoamine neurotransmitters, brain-derived neurotrophic and inflammatory factors, and glutamate and its receptors, using monoamine oxidase inhibitors, tricyclic drugs, selective serotonin reuptake inhibitors (SSRIs) and selective serotonin noradrenaline reuptake inhibitors (SNRIs), glutamatergic drugs, opioids, and benzodiazepines [74]. In vitro, SSRIs have been shown to inhibit the release of TNF-α and NO from activated microglia, impede calcium ion influx, decrease the activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway, and also reduce inflammatory changes [133]. SSRIs and SNRIs decrease blood and tissue cytokines and regulate complex inflammatory pathways of NF-κB, inflammasomes, TLR4, and peroxisome proliferator-activated receptor gamma (PPAR-γ) [134]. Liu et al. (2020) showed in their systematic review and meta-analysis that patients with depression who responded to treatment had lower baseline levels of the chemotactic factor for neutrophils IL-8 than non-responders [135]. In addition, treatment with antidepressants decreases TNF-α and IL-5 levels. However, long-term treatment with SSRIs has been postulated to increase Th1 and decreases Th2-derived cytokines [136].
Celecoxib, nonsteroidal anti-inflammatory drugs, minocycline, but also statins, polyunsaturated fatty acids, pioglitazone, modafinil, corticosteroids, the vitamin D2 analog i.e., paricalcitol, etc. have already been reported as classical anti-inflammatory drugs with consequent antidepressant effects [137,138,139,140] (Table 1).
Celecoxib, a selective COX-2 inhibitor, exerts anti-depressive action by decreasing IL-6 expression and/or levels [137]. Minocycline, the second-generation tetracycline antibiotic, can cross the BBB more efficiently than other tetracycline antibiotics. It has anti-inflammatory, antioxidant, and neuroprotective effects within the CNS by preventing the release of inflammatory cytokines such as IL-6 and TNF-α [141]. It also inhibits neutrophil migration, degranulation, oxygen-free radical production, and NO release. Statins, known as lipid-lowering agents, have shown anti-inflammatory potency by decreasing levels of CRP and low-density lipoprotein (LDL) cholesterol, TNF-α and IFN-γ production in stimulated T cells, but also by reducing immune activation of T-helper cells [142]. Pioglitazone, primarily used as an antidiabetic drug, acts as a PPAR-γ agonist and decreases the expression of IL-1β, IL-6, TNF-α, inducible nitric oxide synthase (iNOS), and chemoattractant protein-1 (MCP-1/CCL2) [143]. It ameliorates depression-like behaviors by inducing the neuroprotective phenotype of microglia [144]. The psychostimulant modafinil reduces brain inflammation by impacting monocyte recruitment and activation, T cell recruitment and differentiation, cytokine production, and glial activation [145]. Corticosteroids, known for their anti-inflammatory properties, have also been studied for their antidepressant properties [146]. Because of their various side effects, which depend on their dosage and duration of treatment, they should be used with caution [147]. Paricalcitol, a vitamin D2 analog, regulates microglia-mediated neuroinflammation via decreased production of IL-1-β, inhibition of NF-κB and NLRP3 signaling, and caspase-1 overexpression [140]. In examining the link between depression and cancer, numerous experimental studies have revealed that activation of the kynurenine pathway of tryptophan degradation due to inflammation plays an important role in the evolution and persistence of both diseases [148].
The Hamilton group study showed that a history of depression, anxiety, and fear of tumor recurrence was associated with greater use of complementary treatment approaches [149,150]. The supplements most commonly used by patients are selenium (Se), folic acid, and omega-3 fatty acids [151]. Cancer patients often turn to antioxidants; among them, Se is particularly interesting, either from an inorganic source (sodium selenate) or the amino acid (selenomethionine) [151]. However, it is questionable whether its action can be considered exclusively as an antioxidant because it can also act as an oxidant and exhibit an anticarcinogenic effect [152]. Due to its antioxidant effect, Se is suitable as a supplement in depressive states and is an essential trace element for thyroxine metabolism. Thus, Se deficiency lowers antioxidant protection of the brain and may lead to brain damage—the turnover of dopamine and serotonin increases, while it decreases for noradrenaline and 5-hydroxy-3-indoleacetic acid compared to controls [153].
The role of folic acid is reflected in the synthesis of serotonin, and its supplementation is advised for patients with depression [154]. However, at high doses of folic acid, an adverse action may be observed because its role in metabolism controls the potential proliferative action for cancer cells [151].
Other widely used supplements are omega-3 fatty acids, predominantly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). They influence optimal cell structure/function and affect synaptic neurotransmission [155]. Therefore, they are recommended for complementary therapy in depression [156]. Improvements are also expected from fatty acid supplementation in chemotherapy and radiotherapy, as they affect inflammation, apoptosis, eicosanoid synthesis, etc. [157].
New therapeutic approaches may include drug-supporting/delivery systems as well as assorted supplements. Among the various supplements, zeolites are in the spotlight. There are a number of zeolite-associated positive effects reported in the literature: antioxidant and anticancer performance, ion exchange, and adsorption/encapsulation features, to name a few [158,159]. These aluminosilicates can be of synthetic or natural origin, such as clinoptilolite, and are recognized for human application [160]. Interestingly, synthetic zeolites can be designed to meet the specific demands of drug carrier systems and seem to be a far better choice, but are still awaiting general pharmaceutical recognition. Some therapeutic approaches may benefit from zeolite use, i.e., sustained drug-delivery systems, which are considered to be improved therapeutic pathways compared to regular ones [161]. Over the past two decades, researchers have competed to find ideal carriers, exploring a possible synergistic effect between the selected support and the drug itself [162,163]. There are several reasons for this—firstly, a specially designed carrier of nanometric dimensions must be considered to sustain BBB pass [164]. To meet this requirement, animal testing is set forward with some interesting applications. For example, infrared-activated BBB permeability may be accomplished by utilizing zeolitic imidazolate-based nanocomposite for intracerebral quercetin delivery providing neuroprotective effects [165]. Furthermore, the zeolite platform must encompass enough functional centers to efficiently adsorb/encapsulate drugs. Thus, zeolitic composites were proposed for synergetic tumor thermo-chemotherapy using doxorubicin drug delivery that sustains tumor reduction [166].
In the field of mental disorders, zeolite testing is under-explored with the majority of studies employing only animal models. One way to treat induced bipolar disorder in rats with probiotic cultures, alone and in zeolite-supported formulations, was suggested by Alchujyan et al. [167]. Interestingly, probiotics expressed a positive effect on arginase/nitric oxide synthase activities without significant benefits of zeolite carrier, as both formulations led to beneficial histopathological brain alterations and subsequent behavioral progress in rats.
Several reports suggest that recovery of cancer patients can be promoted by zeolite supplementation [168]. This hypothesis is based on zeolites’ excellent adsorption capacity for histamine which may be regarded as beneficial for pain relief [169]. In vitro and in vivo experiments on zeolite frameworks safety are extensively studied, while others investigated double-blinded trials of oral clinoptilolite intake in cancer patients to treat peripheral neuropathy induced by chemotherapy [170]. As reported by Vitale et al., the neuropathy extent was quite similar, occurring in 70.6% and 64.3% of patients in the placebo and zeolite supplementation groups, respectively [171].
Bearing in mind the good adsorption properties of zeolites, their role in the removal of heavy metals is often mentioned in the context of the prevention of mental disorders. A prospective use of zeolite/ethylenediaminetetraacetic acid as a lead scavenger is reported [172], confirming the role of clinoptilolite in reducing neurotoxicity in mice. Another removal of lead addresses issues with autism spectrum disorder [173]. Injection of zeolite particles is proposed with the possibility of stool excretion after metal adsorption, with no analysis of the detrimental effect zeolite nanoparticles could have on the hematological and gastrointestinal region. As a multifunctional material, Y zeolite is applied as an electrode support for the ruthenium ammine complex in the electrochemical detection of dopamine/serotonin [174]. Extending this system toward zeolite’s possible interaction with L-dopa, as a dopamine precursor may be sound due to several hydrogen bonds that can be formed. However, this emerged as a premise for rising dopamine levels, which is challenging to test/confirm [175]. Expectedly, these propositions are left with only hypothetical opinions.

7. Conclusions

Immune system alteration is the common denominator of depression and cancer. Additionally, alterations in the immune response seem to overlap in both pathological conditions. The psychiatric correlates are followed by immune disturbances, and we still wonder to what extent the resolution of inflammation in carcinoma might simultaneously contribute to the resolution of the associated depressive symptomatology. Recognition of acute mediators of inflammation is very important, and it is even more important to prevent the transition from acute to chronic inflammation through early anti-inflammatory interventions. The alarmins induce local (central) inflammation by TLR signaling and facilitating NF-κB transcriptional activity and NLRP3 inflammasome in neuronal and nonneuronal cells. Thus, pro-inflammatory cytokines produced in the periphery could activate inflammation in the brain and subsequently modulate the release and function of neurotransmitters, leading to the onset of depression. Previous clinical investigations have shown that the cytokines IL-1, IL-6, IFN-γ, and TNF-α play key roles in these processes. These same cytokines are among the major mediators of the anti-tumor immune response and the chronic inflammation that usually accompanies it. Hypersensitivity and chronification of inflammation suggest an exhausted and insufficient immune response. In conclusion, peripheral inflammation could trigger central immune-inflammatory pathways that lead to pain, fatigue, and depressive symptomatology in patients with cancer. Cancer treatment strategies, as well as conventional psychotropic drugs, could help balance the inflammatory milieu. The new equilibrium in both conditions may be achieved by variously targeted anti-inflammatory strategies. Anti-inflammatory drugs are well known, but new possible pathways and challenging add-on therapies have yet to be found.

Author Contributions

The first author M.M.B. presented the concept of the manuscript and discussed it with all contributors, K.V., D.A., M.M.-R., N.R.M. and I.P.J. Authors M.M.B., K.V., D.A., M.M.-R., N.R.M. and I.P.J. contributed equally to the writing subsections by their specific competencies. The first author M.M.B. wrote the preliminary version. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Faculty of Medical Sciences, University of Kragujevac (projects JP 03-17 and JP 03-16) and the Ministry of Education, Science and Technological Development of the Republic of Serbia (Grants Nos. 451-03-47/2023-01/200146 and ON175069).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Bojana Mircetic for language editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Uchikawa, H.; Fujii, K.; Shiohama, T.; Nakazato, M.; Shimizu, E.; Miyashita, T.; Shimojo, N. Specific temperament in patients with nevoid basal cell carcinoma syndrome. Pediatr. Int. 2021, 63, 177–182. [Google Scholar] [CrossRef]
  2. Verma, M.; Paik, J.M.; Younossi, I.; Tan, D.; Abdelaal, H.; Younossi, Z.M. The impact of hepatocellular carcinoma diagnosis on patients’ health-related quality of life. Cancer Med. 2021, 10, 6273–6281. [Google Scholar] [CrossRef]
  3. Pham, H.; Torres, H.; Sharma, P. Mental health implications in bladder cancer patients: A review. Urol. Oncol. 2019, 37, 97–107. [Google Scholar] [CrossRef]
  4. Oh, H.M.; Son, C.G. The Risk of Psychological Stress on Cancer Recurrence: A Systematic Review. Cancers 2021, 13, 5816. [Google Scholar] [CrossRef] [PubMed]
  5. Kelley, K.W.; Bluthé, R.M.; Dantzer, R.; Zhou, J.H.; Shen, W.H.; Johnson, R.W.; Broussard, S.R. Cytokine-induced sickness behavior. Brain Behav. Immun. 2003, 1, 12–118. [Google Scholar] [CrossRef]
  6. Takeuchi, Y.; Gotoh, N. Inflammatory cytokines-enriched microenvironment plays key roles for the development of breast cancers. Cancer Sci. 2023; epub ahead of print. [Google Scholar] [CrossRef]
  7. Bauer, M.E.; Teixeira, A.L. Inflammation in psychiatric disorders: What comes first? Ann. N. Y. Acad. Sci. 2019, 1437, 57–67. [Google Scholar] [CrossRef] [PubMed]
  8. Poole, L.; Dickens, C.; Steptoe, A. The puzzle of depression and acute coronary syndrome: Reviewing the role of acute inflammation. J. Psychosom. Res. 2011, 71, 61–68. [Google Scholar] [CrossRef] [Green Version]
  9. Beurel, E.; Toups, M.; Nemeroff, C.B. The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron. 2020, 107, 234–256. [Google Scholar] [CrossRef] [PubMed]
  10. Rohleder, N. Stress and inflammation—The need to address the gap in the transition between acute and chronic stress effects. Psychoneuroendocrinology 2019, 105, 164–171. [Google Scholar] [CrossRef]
  11. Lu, H.; Ouyang, W.; Huang, C. Inflammation, a key event in cancer development. Mol. Cancer Res. 2006, 4, 221–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Freyermuth-Trujillo, X.; Segura-Uribe, J.J.; Salgado-Ceballos, H.; Orozco-Barrios, C.E.; Coyoy-Salgado, A. Inflammation: A Target for Treatment in Spinal Cord Injury. Cells 2022, 11, 2692. [Google Scholar] [CrossRef] [PubMed]
  13. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
  14. Serhan, C.; Ward, P.; Gilroy, D. Fundamentals of Inflammation; Cambridge University Press: Cambridge, UK, 2010. [Google Scholar] [CrossRef] [Green Version]
  15. Munn, L. Cancer and Inflammation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2017, 9, e1370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 1994, 12, 991–1045. [Google Scholar] [CrossRef]
  17. Gallucci, S.; Matzinger, P. Danger signals: SOS to the immune system. Curr. Opin. Immunol. 2001, 13, 114–119. [Google Scholar] [CrossRef] [PubMed]
  18. Yang, D.; Han, Z.; Oppenheim, J.J. Alarmins and immunity. Immunol. Rev. 2017, 280, 41–56. [Google Scholar] [CrossRef]
  19. Chalmers, S.; Khawaja, A.; Wieruszewski, P.M.; Gajic, O.; Odeyemi, Y. Diagnosis and treatment of acute pulmonary inflammation in critically ill patients: The role of inflammatory biomarkers. World J. Crit. Care Medl. 2019, 8, 59–71. [Google Scholar] [CrossRef] [Green Version]
  20. Kumar, V.; Abbas, A.K.; Aster, J.C. Robbins Basic Pathology; Elsevier Health Sciences: Philadephia, PA, USA, 2013. [Google Scholar]
  21. Dinarello, C.A. Proinflammatory cytokines. Chest 2000, 118, 503–508. [Google Scholar] [CrossRef]
  22. Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef] [Green Version]
  23. Zhang, J.M.; An, J. Cytokines, inflammation and pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef] [Green Version]
  24. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
  26. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef] [PubMed]
  27. Philip, M.; Rowley, D.A.; Schreiber, H. Inflammation as a tumor promoter in cancer induction. Semin. Cancer Biol. 2004, 14, 433–439. [Google Scholar] [CrossRef] [PubMed]
  28. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  30. Jovanovic, I.P.; Pejnovic, N.N.; Radosavljevic, G.D.; Arsenijevic, N.N.; Lukic, M.L. IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology 2012, 1, 229–231. [Google Scholar] [CrossRef] [Green Version]
  31. Pollard, J.W. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer 2004, 4, 71–78. [Google Scholar] [CrossRef]
  32. Schetter, A.J.; Heegaard, N.H.; Harris, C.C. Inflammation and cancer: Interweaving microRNA, free radical, cytokine and p53 pathways. Carcinogenesis 2010, 31, 37–49. [Google Scholar] [CrossRef] [Green Version]
  33. Nettis, M.A.; Pariante, C.M. Is there neuroinflammation in depression? Understanding the link between the brain and the peripheral immune system in depression. Int. Rev. Neurobiol. 2020, 152, 23–40. [Google Scholar] [CrossRef]
  34. Carlessi, A.S.; Borba, L.A.; Zugno, A.I.; Quevedo, J.; Réus, G.Z. Gut microbiota-brain axis in depression: The role of neuroinflammation. Eur. J. Neurosci. 2021, 53, 222–235. [Google Scholar] [CrossRef] [PubMed]
  35. Majd, M.; Saunders, E.F.H.; Engeland, C.G. Inflammation and the dimensions of depression: A review. Front. Neuroendocrinol. 2020, 56, 100800. [Google Scholar] [CrossRef] [PubMed]
  36. Capuron, L.; Gumnick, J.F.; Musselman, D.L.; Lawson, D.H.; Reemsnyder, A.; Nemeroff, C.B.; Miller, A.H. Neurobehavioral effects of interferon-alpha in cancer patients: Phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology 2002, 26, 643–652. [Google Scholar] [CrossRef] [PubMed]
  37. Loftis, J.M.; Huckans, M.; Morasco, B.J. Neuroimmune mechanisms of cytokine-induced depression: Current theories and novel treatment strategies. Neurobiol. Dis. 2010, 37, 519–533. [Google Scholar] [CrossRef] [Green Version]
  38. Lynall, M.E.; Turner, L.; Bhatti, J.; Cavanagh, J.; de Boer, P.; Mondelli, V.; Jones, D.; Drevets, W.C.; Cowen, P.; Harrison, N.A.; et al. Neuroimmunology of Mood Disorders and Alzheimer’s Disease (NIMA) Consortium. Peripheral Blood Cell-Stratified Subgroups of Inflamed Depression. Biol. Psychiatry 2020, 88, 185–196. [Google Scholar] [CrossRef] [Green Version]
  39. Borovcanin, M.M.; Vesic, K.; Balcioglu, Y.H.; Mijailović, N.R. Prescription of selective serotonin reuptake inhibitors in COVID-19 infection needs caution. Front. Psychiatry 2022, 13, 1052710. [Google Scholar] [CrossRef]
  40. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From inflammation to sickness and depression: When the immune system subjugates the brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef] [Green Version]
  41. Kiraly, D.D.; Horn, S.R.; Van Dam, N.T.; Costi, S.; Schwartz, J.; Kim-Schulze, S.; Patel, M.; Hodes, G.E.; Russo, S.J.; Merad, M.; et al. Altered peripheral immune profiles in treatment-resistant depression: Response to ketamine and prediction of treatment outcome. Transl. Psychiatry 2017, 7, e1065. [Google Scholar] [CrossRef] [Green Version]
  42. Dudek, K.A.; Dion-Albert, L.; Kaufmann, F.N.; Tuck, E.; Lebel, M.; Menard, C. Neurobiology of resilience in depression: Immune and vascular insights from human and animal studies. Eur. J. Neurosci. 2021, 53, 183–221. [Google Scholar] [CrossRef] [Green Version]
  43. Mac Giollabhui, N.; Ng, T.H.; Ellman, L.M.; Alloy, L.B. The longitudinal associations of inflammatory biomarkers and depression revisited: Systematic review, meta-analysis, and meta-regression. Mol. Psychiatry 2021, 26, 3302–3314. [Google Scholar] [CrossRef]
  44. Del Giudice, M.; Gangestad, S.W. Rethinking IL-6 and CRP: Why they are more than inflammatory biomarkers, and why it matters. Brain Behav. Immun. 2018, 70, 61–75. [Google Scholar] [CrossRef] [PubMed]
  45. Vegas, O.; Beitia, G.; Sánchez-Martin, J.R.; Arregi, A.; Azpiroz, A. Behavioral and neurochemical responses in mice bearing tumors submitted to social stress. Behav. Brain Res. 2004, 155, 125–134. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, J.; Zhang, W.; Lu, T.; Li, J.; Zheng, Y.; Kong, L. Morphological and genetic characterization of a cultivated Cordyceps sinensis fungus and its polysaccharide component possessing antioxidant property in H22 tumor-bearing mice. Life Sci. 2006, 78, 2742–2748. [Google Scholar] [CrossRef]
  47. Papiez, M.A.; Dybala, M.; Sowa-Kucma, M.; Krzysciak, W.; Taha, H.; Jozkowicz, A.; Nowak, G. Evaluation of oxidative status and depression-like responses in Brown Norway rats with acute myeloid leukemia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2009, 33, 596–604. [Google Scholar] [CrossRef] [PubMed]
  48. Qi, H.; Ma, J.; Liu, Y.M.; Yang, L.; Peng, L.; Wang, H.; Chen, H.Z. Allostatic tumor-burden induces depression-associated changes in hepatoma-bearing mice. J. Neuro-Oncol. 2009, 94, 367–372. [Google Scholar] [CrossRef]
  49. Nashed, M.G.; Seidlitz, E.P.; Frey, B.N.; Singh, G. Depressive-like behaviours and decreased dendritic branching in the medial prefrontal cortex of mice with tumors: A novel validated model of cancer-induced depression. Behav. Brain Res. 2015, 294, 25–35. [Google Scholar] [CrossRef]
  50. Lamkin, D.M.; Lutgendorf, S.K.; Lubaroff, D.; Sood, A.K.; Beltz, T.G.; Johnson, A.K. Cancer induces inflammation and depressive-like behavior in the mouse: Modulation by social housing. Brain Behav. Immun. 2011, 25, 555–564. [Google Scholar] [CrossRef] [Green Version]
  51. Lebeña, A.; Vegas, O.; Gómez-Lázaro, E.; Arregi, A.; Garmendia, L.; Beitia, G.; Azpiroz, A. Melanoma tumors alter proinflammatory cytokine production and monoamine brain function, and induce depressive-like behavior in male mice. Behav. Brain Res. 2014, 272, 83–92. [Google Scholar] [CrossRef]
  52. Fang, C.K.; Chen, H.W.; Chiang, I.T.; Chen, C.C.; Liao, J.F.; Su, T.P.; Hwang, J.J. Mirtazapine inhibits tumor growth via immune response and serotonergic system. PLoS ONE 2012, 7, e38886. [Google Scholar] [CrossRef]
  53. Pyter, L.M.; Pineros, V.; Galang, J.A.; McClintock, M.K.; Prendergast, B.J. 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. [Google Scholar] [CrossRef] [Green Version]
  54. Norden, D.M.; Bicer, S.; Clark, Y.; Jing, R.; Henry, C.J.; Wold, L.E.; McCarthy, D.O. Tumor growth increases neuroinflammation, fatigue and depressive-like behavior prior to alterations in muscle function. Brain Behav. Immun. 2014, 43, 76–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yan, W.; Zhu, H.; Zhang, P.; Sun, T.; Gong, S.; Sun, G.; Ren, Z. Heterotopic 4T1 breast cancer transplantation induces hippocampal inflammation and depressive-like behaviors in mice. Metab. Brain Dis. 2022, 37, 2955–2963. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, T.; Zhou, F.; Yuan, X.; Yang, T.; Liang, X.; Wang, Y.; Tu, H.; Chang, J.; Nan, K.; Wei, Y. Reactive oxygen species are involved in the development of gastric cancer and gastric cancer-related depression through ABL1-mediated inflammation signaling pathway. Oxidative Med. Cell Longev. 2019, 2019, 5813985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Liu, B.P.; Zhang, C.; Zhang, Y.P.; Li, K.W.; Song, C. The combination of chronic stress and smoke exacerbated depression-like changes and lung cancer factor expression in A/J mice: Involve inflammation and BDNF dysfunction. PLoS ONE 2022, 17, e0277945. [Google Scholar] [CrossRef] [PubMed]
  58. Almutabagani, L.F.; Almanqour, R.A.; Alsabhan, J.F.; Alhossan, A.M.; Alamin, M.A.; Alrajeh, H.M.; Alonazi, A.S.; El-Malky, A.M.; Alrasheed, N.M. Inflammation and Treatment-Resistant Depression from Clinical to Animal Study: A Possible Link? Neurol. Int. 2023, 15, 100–120. [Google Scholar] [CrossRef] [PubMed]
  59. Walker, A.K.; Kavelaars, A.; Heijnen, C.J.; Dantzer, R. Neuroinflammation and comorbidity of pain and depression. Pharmacol. Rev. 2013, 66, 80–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hart, B.L. Biological basis of the behavior of sick animals. Neurosci. Biobehav. Rev. 1988, 12, 123–137. [Google Scholar] [CrossRef] [PubMed]
  61. Mitchell, A.J.; Chan, M.; Bhatti, H.; Halton, M.; Grassi, L.; Johansen, C.; Meader, N. Prevalence of depression, anxiety, and adjustment disorder in oncological, haematological, and palliative-care settings: A meta-analysis of 94 interview-based studies. Lancet Oncol. 2011, 12, 160–174. [Google Scholar] [CrossRef]
  62. Sharma, N.; Hansen, C.H.; O’Connor, M.; Thekkumpurath, P.; Walker, J.; Kleiboer, A.; Murray, G.; Espie, C.; Storey, D.; Sharpe, M.; et al. Sleep problems in cancer patients: Prevalence and association with distress and pain. Psychooncology 2012, 21, 1003–1009. [Google Scholar] [CrossRef]
  63. Miaskowski, C.; Dodd, M.; Lee, K. Symptom clusters: The new frontier in symptom management research. J. Natl. Cancer Inst. Monogr. 2004, 32, 17–21. [Google Scholar] [CrossRef]
  64. Dodd, M.J.; Miaskowski, C.; Lee, K.A. Occurrence of symptom clusters. J. Natl. Cancer Inst. Monogr. 2004, 32, 76–78. [Google Scholar] [CrossRef] [PubMed]
  65. Dodd, M.J.; Miaskowski, C.; Paul, S.M. Symptom clusters and their effect on the functional status of patients with cancer. Oncol. Nurs. Forum. 2001, 28, 465–470. [Google Scholar] [PubMed]
  66. Charalambous, A.; Giannakopoulou, M.; Bozas, E.; Paikousis, L. Parallel and serial mediation analysis between pain, anxiety, depression, fatigue and nausea, vomiting and retching within a randomised controlled trial in patients with breast and prostate cancer. BMJ Open 2019, 9, e026809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Holliday, E.B.; Dieckmann, N.F.; McDonald, T.L.; Hung, A.Y.; Thomas, C.R., Jr.; Wood, L.J. Relationship between fatigue, sleep quality and inflammatory cytokines during external beam radiation therapy for prostate cancer: A prospective study. Radiother Oncol. 2016, 118, 105–111. [Google Scholar] [CrossRef] [Green Version]
  68. Lee, C.H.; Giuliani, F. The Role of Inflammation in Depression and Fatigue. Front. Immunol. 2019, 10, 1696. [Google Scholar] [CrossRef] [Green Version]
  69. Matsuda, M.; Huh, Y.; Ji, R.R. Roles of inflammation, neurogenic inflammation, and neuroinflammation in pain. J. Anesth. 2019, 33, 131–139. [Google Scholar] [CrossRef]
  70. Katarina, V.; Gordana, T.; Svetlana, M.D.; Milica, B. Oxidative stress and neuroinflammation should be both considered in the occurrence of fatigue and depression in multiple sclerosis. Acta Neurol. Belg. 2020, 120, 853–861. [Google Scholar] [CrossRef]
  71. Lynn, M.; Maclachlan, L.; Finkelmeyer, A.; Clark, J.; Locke, J.; Todryk, S.; Ng, W.F.; Newton, J.L.; Watson, S. Reduction of Glucocorticoid Receptor Function in Chronic Fatigue Syndrome. Mediators Inflamm. 2018, 2018, 3972104. [Google Scholar] [CrossRef]
  72. Thornton, L.M.; Andersen, B.L.; Blakely, W.P. The pain, depression, and fatigue symptom cluster in advanced breast cancer: Covariation with the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system. Health Psychol. 2010, 29, 333–337. [Google Scholar] [CrossRef] [Green Version]
  73. Lanser, L.; Kink, P.; Egger, E.M.; Willenbacher, W.; Fuchs, D.; Weiss, G.; Kurz, K. Inflammation-Induced Tryptophan Breakdown is Related With Anemia, Fatigue, and Depression in Cancer. Front. Immunol. 2020, 11, 249. [Google Scholar] [CrossRef] [Green Version]
  74. Sheng, J.; Liu, S.; Wang, Y.; Cui, R.; Zhang, X. The Link between Depression and Chronic Pain: Neural Mechanisms in the Brain. Neural Plast. 2017, 2017, 9724371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kwekkeboom, K.L.; Tostrud, L.; Costanzo, E.; Coe, C.L.; Serlin, R.C.; Ward, S.E.; Zhang, Y. The Role of Inflammation in the Pain, Fatigue, and Sleep Disturbance Symptom Cluster in Advanced Cancer. J. Pain Symptom Manage. 2018, 55, 1286–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Bamonti, P.M.; Moye, J.; Naik, A.D. Pain is associated with continuing depression in cancer survivors. Psychol. Health Med. 2018, 23, 1182–1195. [Google Scholar] [CrossRef] [PubMed]
  77. Zhou, W.; Dantzer, R.; Budac, D.P.; Walker, A.K.; Mao-Ying, Q.L.; Lee, A.W.; Heijnen, C.J.; Kavelaars, A. Peripheral indoleamine 2,3-dioxygenase 1 is required for comorbid depression-like behavior but does not contribute to neuropathic pain in mice. Brain Behav. Immun. 2015, 46, 147–153. [Google Scholar] [CrossRef] [Green Version]
  78. Jovanovic, F.; Candido, K.D.; Knezevic, N.N. The Role of the Kynurenine Signaling Pathway in Different Chronic Pain Conditions and Potential Use of Therapeutic Agents. Int. J. Mol. Sci. 2020, 21, 6045. [Google Scholar] [CrossRef]
  79. Berger, A.M.; Mooney, K.; Alvarez-Perez, A.; Breitbart, W.S.; Carpenter, K.M.; Cella, D.; Cleeland, C.; Dotan, E.; Eisenberger, M.A.; Escalante, C.P.; et al. National comprehensive cancer network. Cancer-Related Fatigue, Version 2.2015. J. Natl. Compr. Canc. Netw. 2015, 13, 1012–1039. [Google Scholar] [CrossRef]
  80. Addington, A.M.; Gallo, J.J.; Ford, D.E.; Eaton, W.W. Epidemiology of unexplained fatigue and major depression in the community: The Baltimore ECA follow-up, 1981–1994. Psychol. Med. 2001, 31, 1037–1044. [Google Scholar] [CrossRef]
  81. Fava, M. Symptoms of fatigue and cognitive/executive dysfunction in major depressive disorder before and after antidepressant treatment. J. Clin. Psychiatry 2003, 64, 30–34. [Google Scholar]
  82. Endres, D.; Pollak, T.A.; Bechter, K.; Prüss, H.; Tebartz van Elst, L. Cerebrospinal Fluid Biomarkers for the Detection of Autoimmune Depression. Biol. Psychiatry 2022, 92, 530–532. [Google Scholar] [CrossRef]
  83. Höftberger, R.; Lassmann, H. Immune-mediated disorders. Handb. Clin. Neurol. 2018, 145, 285–299. [Google Scholar] [CrossRef]
  84. Grativvol, R.S.; Cavalcante, W.C.P.; Castro, L.H.M.; Nitrini, R.; Simabukuro, M.M. Updates in the Diagnosis and Treatment of Paraneoplastic Neurologic Syndromes. Curr. Oncol. Rep. 2018, 20, 92. [Google Scholar] [CrossRef] [PubMed]
  85. Iorio, R.; Spagni, G.; Masi, G. Paraneoplastic neurological syndromes. Semin. Diagn. Pathol. 2019, 36, 279–292. [Google Scholar] [CrossRef] [PubMed]
  86. Dropcho, E.J. Neurologic paraneoplastic syndromes. Curr. Oncol. Rep. 2004, 6, 26–31. [Google Scholar] [CrossRef] [PubMed]
  87. Lancaster, E. Paraneoplastic disorders. Continuum (Minneap Minn). Neuro-Oncol. 2015, 21, 452–475. [Google Scholar] [CrossRef] [Green Version]
  88. Senties-Madrid, H.; Vega-Boada, F. Paraneoplastic syndromes associated with anti-Hu antibodies. Isr. Med. Assoc. J. 2001, 3, 94–103. [Google Scholar] [PubMed]
  89. Venkatraman, A.; Opal, P. Paraneoplastic cerebellar degeneration with anti-Yo antibodies—A review. Ann. Clin. Transl. Neurol. 2016, 3, 655–663. [Google Scholar] [CrossRef]
  90. Ortega Suero, G.; Sola-Valls, N.; Escudero, D.; Saiz, A.; Graus, F. Anti-Ma and anti-Ma2-associated paraneoplastic neurological syndromes. Neurologia 2018, 33, 18–27. [Google Scholar] [CrossRef]
  91. Olmez, O.F.; Kinikoglu, O.; Yilmaz, N.H.; Bilici, A.; Cubukcu, E.; Seker, M.; Cakir, T.; Yildiz, O.; Hamdard, J. Anti-Ri-associated paraneoplastic neurological syndrome: Initial symptom of breast cancer with HER2 overexpression and treatment by dual HER2 blockade. J. Oncol. Pharm. Pract. 2019, 25, 1526–1530. [Google Scholar] [CrossRef]
  92. Dalmau, J.; Tüzün, E.; Wu, H.Y.; Masjuan, J.; Rossi, J.E.; Voloschin, A.; Baehring, J.M.; Shimazaki, H.; Koide, R.; King, D.; et al. Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann. Neurol. 2007, 61, 25–36. [Google Scholar] [CrossRef] [Green Version]
  93. Laurido-Soto, O.; Brier, M.R.; Simon, L.E.; McCullough, A.; Bucelli, R.C.; Day, G.S. Patient characteristics and outcome associations in AMPA receptor encephalitis. J. Neurol. 2019, 266, 450–460. [Google Scholar] [CrossRef]
  94. Lancaster, E.; Lai, M.; Peng, X.; Hughes, E.; Constantinescu, R.; Raizer, J.; Friedman, D.; Skeen, M.B.; Grisold, W.; Kimura, A.; et al. Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: Case series and characterisation of the antigen. Lancet Neurol. 2010, 9, 67–76. [Google Scholar] [CrossRef] [Green Version]
  95. Lai, M.; Huijbers, M.G.; Lancaster, E.; Graus, F.; Bataller, L.; Balice-Gordon, R.; Cowel, J.K.; Dalmau, J. Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: A case series. Lancet Neurol. 2010, 9, 776–785. [Google Scholar] [CrossRef] [Green Version]
  96. Bien, C.G.; Vincent, A.; Barnett, M.H.; Becker, A.J.; Blümcke, I.; Graus, F.; Jellinger, K.A.; Reuss, D.E.; Ribalta, T.; Schlegel, J.; et al. Immunopathology of autoantibody-associated encephalitides: Clues for pathogenesis. Brain 2012, 135, 1622–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Psimaras, D.; Carpentier, A.F.; Rossi, C. PNS Euronetwork. Cerebrospinal fluid study in paraneoplastic syndromes. J. Neurol. Neurosurg. Psychiatry 2010, 81, 42–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Binks, S.; Uy, C.; Honnorat, J.; Irani, S.R. Paraneoplastic neurological syndromes: A practical approach to diagnosis and management. Pract. Neurol. 2022, 22, 19–31. [Google Scholar] [CrossRef] [PubMed]
  99. Borovcanin, M.M.; Vesic, K. Breast cancer in schizophrenia could be interleukin-33-mediated. World J. Psychiatry 2021, 11, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  100. Vyas, D.; Laput, G.; Vyas, A.K. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. OncoTargets Ther. 2014, 7, 1015–1023. [Google Scholar] [CrossRef] [Green Version]
  101. Proskuryakov, S.Y.; Gabai, V.L. Mechanisms of Tumor Cell Necrosis. Curr. Pharm. Des. 2010, 16, 56–68. [Google Scholar] [CrossRef]
  102. Jin, S.; DiPaola, R.S.; Mathew, R.; White, E. Metabolic catastrophe as a means to cancer cell death. J. Cell Sci. 2007, 120, 379–383. [Google Scholar] [CrossRef] [Green Version]
  103. Leek, R.D.; Landers, R.J.; Harris, A.L.; Lewis, C.E. Necrosis correlates with high vascular density and focal macrophage infltration in invasive carcinoma of the breast. Br. J. Cancer 1999, 79, 991–995. [Google Scholar] [CrossRef] [Green Version]
  104. Sauter, B.; Albert, M.L.; Francisco, L.; Larsson, M.; Somersan, S.; Bhardwaj, N. Consequences of cell death: Exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J. Exp. Med. 2000, 191, 423–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Francesco Colotta, F.; Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis 2009, 30, 1073–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Dolan, R.D.; McMillan, D.C. The prevalence of cancer associated systemic inflammation: Implications of prognostic studies using the Glasgow Prognostic Score. Crit. Rev. Oncol. Hematol. 2020, 150, 102962. [Google Scholar] [CrossRef] [PubMed]
  107. Ernst, B.; Anderson, K.S. Immunotherapy for the treatment of breast cancer. Curr. Oncol. Rep. 2015, 17, 5. [Google Scholar] [CrossRef] [PubMed]
  108. Dhupkar, P.; Gordon, N.; Stewart, J.; Kleinerman, E.S. Anti-PD-1 therapy redirects macrophages from an M2 to an M1 phenotype inducing regression of OS lung metastases. Cancer Med. 2018, 7, 2654–2664. [Google Scholar] [CrossRef]
  109. Parekh, V.V.; Lalani, S.; Kim, S.; Halder, R.; Azuma, M.; Yagita, H.; Kumar, V.; Wu, L.; Kaer, L.V. PD-1/PD-L blockade prevents anergy induction and enhances the anti-tumor activities of glycolipid-activated invariant NKT cells. J. Immunol. 2009, 182, 2816–2826. [Google Scholar] [CrossRef] [Green Version]
  110. Rotte, A.; D’Orazi, G.; Bhandaru, M. Nobel committee honors tumor immunologists. J. Exp. Clin. Cancer Res. 2018, 37, 262. [Google Scholar] [CrossRef] [Green Version]
  111. Helmy, K.Y.; Patel, S.A.; Nahas, G.R.; Rameshwar, P. Cancer immunotherapy: Accomplishments to date and future promise. Ther. Deliv. 2013, 4, 1307–1320. [Google Scholar] [CrossRef]
  112. Solomon, B.L.; Garrido-Laguna, I. TIGIT: A novel immunotherapy target moving from bench to bedside. Cancer Immunol. Immunother. 2018, 67, 1659–1667. [Google Scholar] [CrossRef]
  113. Zhang, Q.; Zhong, H.; Fan, Y.; Liu, Q.; Song, J.; Yao, S.; Cao, F. Immune and Clinical Features of CD96 Expression in Glioma by in silico Analysis. Front. Bioeng. Biotechnol. 2020, 8, 592. [Google Scholar] [CrossRef]
  114. van Hall, T.; André, P.; Horowitz, A.; Ruan, D.F.; Borst, L.; Zerbib, R.; Narni-Mancinelli, E.; van der Burg, S.H.; Vivier, E. Monalizumab: Inhibiting the novel immune checkpoint NKG2A. J. Immunother. Cancer 2019, 7, 263. [Google Scholar] [CrossRef] [PubMed]
  115. Burugu, S.; Dancsok, A.R.; Nielsen, T.O. Emerging targets in cancer immunotherapy. Semin. Cancer Biol. 2018, 52, 39–52. [Google Scholar] [CrossRef] [PubMed]
  116. Raison, C.L.; Rutherford, R.E.; Woolwine, B.J.; Shuo, C.; Schettler, P.; Drake, D.F.; Haroon, E.; Miller, A.H. A Randomized Controlled Trial of the Tumor Necrosis Factor Antagonist Infliximab for Treatment-Resistant Depression: The Role of Baseline Inflammatory Biomarkers. JAMA Psychiatry. 2013, 70, 31–41. [Google Scholar] [CrossRef] [PubMed]
  117. Tookman, A.J.; Jones, C.L.; DeWitte, M.; Fatigu Lodge, P.J. Fatigue in patients with advanced cancer: A pilot study of an intervention with infliximab. Support Care Cancer 2008, 16, 1131–1140. [Google Scholar] [CrossRef]
  118. Loftus, E.V.; Feagan, B.G.; Colombel, J.F.; Rubin, D.T.; Wu, E.Q.; Yu, A.P.; Pollack, P.F.; Chao, J.; Mulani, P. Effects of adalimumab maintenance therapy on health-related quality of life of patients with Crohn’s disease: Patient-reported outcomes of the CHARM trial. Am. J. Gastroenterol. 2008, 103, 3132–3141. [Google Scholar] [CrossRef]
  119. Leman, J.; Walton, S.; Layton, A.M.; Ward, K.A.; McBride, S.; Cliff, S.; Downs, A.; Landeira, M.; Bewley, A. The real world impact of adalimumab on quality of life and the physical and psychological effects of moderate-to-severe psoriasis: A UK prospective, multicenter, observational study. J Dermatolog Treat. 2020, 31, 213–221. [Google Scholar] [CrossRef]
  120. Scheinfeld, N.; Sundaram, M.; Teixeira, H.; Gu, Y.; Okun, M. Reduction in pain scores and improvement in depressive symptoms in patients with hidradenitis suppurativa treated with adalimumab in a phase 2, randomized, placebo-controlled trial. Dermatol. Online J. 2016, 22, 13030/qt38x5922j. [Google Scholar] [CrossRef]
  121. Bayramgürler, D.; Karson, A.; Özer, C.; Utkan, T. Effects of long-term etanercept treatment on anxiety- and depression-like neurobehaviors in rats. Physiol Behav. 2013, 119, 145–148. [Google Scholar] [CrossRef] [PubMed]
  122. Brymer, K.J.; Fenton, E.Y.; Kalynchuk, L.E.; Caruncho, H.J. Peripheral etanercept administration normalizes behavior, hippocampal neurogenesis, and hippocampal reelin and GABAA receptor expression in a preclinical model of depression. Front. Pharmacol. 2018, 9, 121. [Google Scholar] [CrossRef]
  123. Alshammari, M.A.; Khan, M.R.; Majid Mahmood, H.; Alshehri, A.O.; Alasmari, F.F.; Alqahtani, F.M.; Alasmari, A.F.; Alsharari, S.D.; Alhossan, A.; Ahmad, S.F. Systemic TNF-α blockade attenuates anxiety and depressive-like behaviors in db/db mice through downregulation of inflammatory signaling in peripheral immune cells. Saudi Pharm. J. 2020, 28, 621–629. [Google Scholar] [CrossRef]
  124. Tyring, S.; Gottlieb, A.; Papp, K.; Gordon, K.; Leonardi, C.; Wang, A.; Lalla, D.; Woolley, M.; Jahreis, A.; Zitnik, R.; et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: Double-blind placebo-controlled randomised phase III trial. Lancet 2006, 367, 29–35. [Google Scholar] [CrossRef] [PubMed]
  125. Bae, S.C.; Gun, S.C.; Mok, C.C.; Khandker, R.; Nab, H.W.; Koenig, A.S.; Vlahos, B.; Pedersen, R.; Singh, A. Improved health outcomes with Etanercept versus usual DMARD therapy in an Asian population with established rheumatoid arthritis. BMC Musculoskelet. Disord. 2013, 14, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Mohamed, B.; Aboul-Fotouh, S.; Ibrahim, E.A.; Shehata, H.; Mansour, A.A.; Az Yassin, N.; El-Eraky, W.; Abdel-Twab, A.M. Effects of pentoxifylline, 7-nitroindazole, and imipramine on tumor necrosis factor-α and indoleamine 2,3-dioxygenase enzyme activity in the hippocampus and frontal cortex of chronic mild-stress-exposed rats. Neuropsychiatr. Dis. Treat. 2013, 9, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Elgarf, A.S.A.; Aboul-Fotouh, S.; Abd-Alkhalek, H.A.; El Tabbal, M.; Hassan, A.N.; Kassim, S.K.; Hammouda, G.A.; Farrag, K.A.; Abdel-Tawab, A.M. Lipopolysaccharide repeated challenge followed by chronic mild stress protocol introduces a combined model of depression in rats: Reversibility by imipramine and pentoxifylline. Pharmacol. Biochem. Behav. 2014, 126, 152–162. [Google Scholar] [CrossRef]
  128. El-Haggar, S.M.; Eissa, M.A.; Mostafa, T.M.; El-Attar, K.S.; Abdallah, M.S. The phosphodiesterase inhibitor pentoxifylline as a novel adjunct to antidepressants in major depressive disorder patients: A proof-of-concept, randomized, double-blind, placebo-controlled trial. Psychother. Psychosom. 2018, 87, 331–339. [Google Scholar] [CrossRef]
  129. Langley, R.G.; Feldman, S.R.; Han, C.; Schenkel, B.; Szapary, P.; Hsu, M.C.; Ortonne, J.P.; Gordon, K.B.; Kimball, A.B. Ustekinumab significantly improves symptoms of anxiety, depression, and skin-related quality of life in patients with moderate-to-severe psoriasis: Results from a randomized, double-blind, placebo-controlled phase III trial. J. Am Acad Dermatol. 2010, 63, 457–465. [Google Scholar] [CrossRef]
  130. Cork, M.J.; Eckert, L.; Simpson, E.L.; Armstrong, A.; Barbarot, S.; Puig, L.; Girolomoni, G.; de Bruin-Weller, M.; Wollenberg, A.; Kataoka, Y.; et al. Dupilumab Improves Patient-Reported Symptoms of Atopic Dermatitis, Symptoms of Anxiety and Depression, and Health-Related Quality of Life in Moderate-to-Severe Atopic Dermatitis: Analysis of Pooled Data from the Randomized Trials SOLO 1 and SOLO 2. J. Dermatolog. Treat. 2020, 31, 606–614. [Google Scholar] [CrossRef] [Green Version]
  131. Griffiths, C.E.M.; Fava, M.; Miller, A.H.; Russell, J.; Ball, S.G.; Xu, W.; Acharya, N.; Rapaport, M.H. Impact of Ixekizumab Treatment on Depressive Symptoms and Systemic Inflammation in Patients with Moderate-to-Severe Psoriasis: An Integrated Analysis of Three Phase 3 Clinical Studies. Psychother. Psychosom. 2017, 86, 260–267. [Google Scholar] [CrossRef]
  132. Gordon, K.B.; Armstrong, A.W.; Han, C.; Foley, P.; Song, M.; Wasfi, Y.; You, Y.; Shen, Y.K.; Reich, K. Anxiety and Depression in Patients with Moderate-to-Severe Psoriasis and Comparison of Change from Baseline after Treatment with Guselkumab vs. Adalimumab: Results from the Phase 3 VOYAGE 2 Study. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1940–1949. [Google Scholar] [CrossRef]
  133. Leonard, B.E. Inflammation and depression: A causal or coincidental link to the pathophysiology? Acta Neuropsychiatr. 2018, 30, 1–16. [Google Scholar] [CrossRef] [Green Version]
  134. Dionisie, V.; Filip, G.A.; Manea, M.C.; Manea, M.; Riga, S. The anti-inflammatory role of SSRI and SNRI in the treatment of depression: A review of human and rodent research studies. Inflammopharmacology 2021, 29, 75–90. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, J.J.; Wei, Y.B.; Strawbridge, R.; Bao, Y.; Chang, S.; Shi, L.; Que, J.; Gadad, B.S.; Trivedi, M.H.; Kelsoe, J.R.; et al. Peripheral cytokine levels and response to antidepressant treatment in depression: A systematic review and meta-analysis. Mol. Psychiatry 2020, 25, 339–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Levite, M. Neurotransmitters activate T-cells and elicit crucial functions via neurotransmitter receptors. Curr. Opin. Pharmacol. 2008, 8, 460–471. [Google Scholar] [CrossRef] [PubMed]
  137. Abbasi, S.H.; Hosseini, F.; Modabbernia, A.; Ashrafi, M.; Akhondzadeh, S. Effect of celecoxib add-on treatment on symptoms and serum IL-6 concentrations in patients with major depressive disorder: Randomized double-blind placebo-controlled study. J. Affect Disord. 2012, 141, 308–314. [Google Scholar] [CrossRef] [PubMed]
  138. Zheng, L.S.; Kaneko, N.; Sawamoto, K. Minocycline treatment ameliorates interferon-alpha- induced neurogenic defects and depression-like behaviors in mice. Front. Cell Neurosci. 2015, 9, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Kohler, O.; Krogh, J.; Mors, O.; Benros, M.E. Inflammation in Depression and the Potential for Anti-Inflammatory Treatment. Curr. Neuropharmacol 2016, 14, 732–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. He, M.; Shi, Z.; Sha, N.N.; Chen, N.; Peng, S.Y.; Liao, D.F.; Wong, M.S.; Dong, X.L.; Wang, Y.J.; Yuan, T.F.; et al. Paricalcitol alleviates lipopolysaccharide-induced depressive-like behavior by suppressing hypothalamic microglia activation and neuroinflammation. Biochem. Pharmacol. 2019, 163, 1–8. [Google Scholar] [CrossRef] [PubMed]
  141. Elewa, H.F.; Hilali, H.; Hess, D.C.; Machado, L.S.; Fagan, S.C. Minocycline for short-term neuroprotection. Pharmacotherapy 2006, 26, 515–521. [Google Scholar] [CrossRef] [Green Version]
  142. Avan, R.; Sahebnasagh, A.; Hashemi, J.; Monajati, M.; Faramarzi, F.; Henney, N.C.; Montecucco, F.; Jamialahmadi, T.; Sahebkar, A. Update on Statin Treatment in Patients with Neuropsychiatric Disorders. Life 2021, 11, 1365. [Google Scholar] [CrossRef]
  143. Zhao, Q.; Wu, X.; Yan, S.; Xie, X.; Fan, Y.; Zhang, J.; Peng, C.; You, Z. The antidepressant-like effects of pioglitazone in a chronic mild stress mouse model are associated with PPARγ-mediated alteration of microglial activation phenotypes. J. Neuroinflammation. 2016, 13, 259. [Google Scholar] [CrossRef] [Green Version]
  144. Kemp, D.E.; Schinagle, M.; Gao, K.; Conroy, C.; Ganocy, S.J.; Ismail-Beigi, F.; Calabrese, J.R. PPAR-gamma agonism as a modulator of mood: Proof-of-concept for pioglitazone in bipolar depression. CNS Drugs. 2014, 28, 571–581. [Google Scholar] [CrossRef] [Green Version]
  145. Zager, A. Modulating the immune response with the wake-promoting drug modafinil: A potential therapeutic approach for inflammatory disorders. Brain Behav. Immunity. 2020, 88, 878–886. [Google Scholar] [CrossRef]
  146. DeBattista, C.; Posener, J.A.; Kalehzan, B.M.; Schatzberg, A.F. Acute antidepressant effects of intravenous hydrocortisone and CRH in depressed patients: A double-blind, placebo-controlled study. Am. J. Psychiatry. 2000, 157, 1334–1337. [Google Scholar] [CrossRef]
  147. Rice, J.B.; White, A.G.; Scarpati, L.M.; Wan, G.; Nelson, W.W. Long-term Systemic Corticosteroid Exposure: A Systematic Literature Review. Clin. Ther. 2017, 39, 2216–2229. [Google Scholar] [CrossRef] [Green Version]
  148. Sforzini, L.; Nettis, M.A.; Mondelli, V.; Pariante, C.M. Inflammation in cancer and depression: A starring role for the kynurenine pathway. Psychopharmacology 2019, 236, 2997–3011. [Google Scholar] [CrossRef] [Green Version]
  149. Sohl, S.J.; Weaver, K.E.; Birdee, G.; Kent, E.E.; Danhauer, S.C.; Hamilton, A.S. Characteristics Associated with the Use of Complementary Health Approaches among Long-Term Cancer Survivors. Support. Care Cancer 2014, 22, 927–936. [Google Scholar] [CrossRef] [Green Version]
  150. Gansler, T.; Strollo, S.; Fallon, E.; Leach, C. Use of Complementary/Integrative Methods: Cancer Survivors’ Misconceptions about Recurrence Prevention. J. Cancer Surviv. 2019, 13, 418–428. [Google Scholar] [CrossRef] [PubMed]
  151. Werneke, U. Risk Management of Nutritional Supplements in Chronic Illness: The Implications for the Care of Cancer and Depression. Proc. Nutr. Soc. 2007, 66, 483–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Drake, E.N. Cancer Chemoprevention: Selenium as a Prooxidant, Not an Antioxidant. Med. Hypotheses 2006, 67, 318–322. [Google Scholar] [CrossRef] [PubMed]
  153. Castaño, A. Low Selenium Diet Increases the Dopamine Turnover in Prefrontal Cortex of the Rat. Neurochem. Int. 1997, 30, 549–555. [Google Scholar] [CrossRef] [PubMed]
  154. Young, S.N. Folate and Depression—A Neglected Problem. J. Psychiatry Neurosci. 2007, 32, 80–82. [Google Scholar] [PubMed]
  155. Grosso, G.; Galvano, F.; Marventano, S.; Malaguarnera, M.; Bucolo, C.; Drago, F.; Caraci, F. Omega-3 Fatty Acids and Depression: Scientific Evidence and Biological Mechanisms. Oxid. Med. Cell Longev. 2014, 2014, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Freeman, M.P.; Hibbeln, J.R.; Wisner, K.L.; Davis, J.M.; Mischoulon, D.; Peet, M.; Keck, P.E.; Marangell, L.B.; Richardson, A.J.; Lake, J. Omega-3 Fatty Acids: Evidence Basis for Treatment and Future Research in Psychiatry. J. Clin. Psychiatry 2006, 67, 1954–1967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Hardman, W.E. (N-3) Fatty Acids and Cancer Therapy. J. Nutr. 2004, 134, S3427–S3430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Jevremović, A.; Božinović, N.; Arsenijević, D.; Marmakov, S.; Nedić Vasiljević, B.; Uskoković-Marković, S.; Bajuk-Bogdanović, D.; Milojević-Rakić, M. Modulation of cytotoxicity by consecutive adsorption of tannic acid and pesticides on surfactant functionalized zeolites, Environ Sci Process. Impacts 2020, 22, 2199–2211. [Google Scholar] [CrossRef]
  159. Janićijević, A.; Jevremović, A.; Janošević Ležaić, B.; Nedić Vasiljević, S.; Uskoković-Marković, D.; Bajuk-Bogdanović, M.; Milojević-Rakić, M. Comparative assessment of pesticide adsorption capacity and antioxidant activity of Silver Dodecatungstophosphate/HΒEA zeolite composites. J. Environ. Chem. Eng. 2021, 9, 106341. [Google Scholar] [CrossRef]
  160. Pavelić, S.K.; Medica, J.S.; Gumbarević, D.; Filošević, A.; Pržulj, N.; Pavelić, K. Critical review on zeolite clinoptilolite safety and medical applications in vivo. Front. Pharmacol. 2018, 9, 1350. [Google Scholar] [CrossRef]
  161. Mijailović, N.R.; Nedić Vasiljević, B.; Ranković, M.; Milanović, V.; Uskoković-Marković, S. Environmental and Pharmacokinetic Aspects of Zeolite/Pharmaceuticals Systems—Two Facets of Adsorption Ability. Catalysts 2022, 12, 837. [Google Scholar] [CrossRef]
  162. Krajišnik, D.; Daković, A.; Milojević, M.; Malenović, A.; Kragović, M.; Bajuk-Bogdanović, D.; Dondur, V.; Milić, J. Properties of diclofenac sodium sorption onto natural zeolite modified with cetylpyridinium chloride. Colloids Surf. B Biointerfaces 2011, 83, 165–172. [Google Scholar] [CrossRef]
  163. Krajišnik, D.; Daković, A.; Malenović, M.; Milojević-Rakić, M.; Dondur, V.; Radulović, Ž.; Milić, J.; Radulović, Z.; Milić, J. Investigation of adsorption and release of diclofenac sodium by modified zeolites composites. Appl. Clay Sci. 2013, 83–84, 322–326. [Google Scholar] [CrossRef]
  164. Hassanvand, A.; Gharibzadeh, S. Is It Possible to Treat Some Brain Diseases by Drug-Substituted Zeolites? J. Neuropsychiatry Clin. Neurosci 2013, 25, E04. [Google Scholar] [CrossRef] [PubMed]
  165. Liu, Z.; Hong, Z.; Xue, J.; Luo, J.; Liu, Q.; Chen, X.; Pan, Z.; Zhou, J.; Liu, Z.; Chen, T. Near-Infrared Radiation-Assisted Drug Delivery Nanoplatform to Realize Blood–Brain Barrier Crossing and Protection for Parkinsonian Therapy. ACS Appl. Mater. Interfaces 2021, 13, 37746–37760. [Google Scholar] [CrossRef] [PubMed]
  166. Wu, Q.; Niu, M.; Chen, X.; Tan, L.; Fu, C.; Ren, X.; Ren, J.; Li, L.; Xu, K.; Zhong, H.; et al. Biocompatible and biodegradable zeolitic imidazolate framework/polydopamine nanocarriers for dual stimulus triggered tumor thermo-chemotherapy. Biomaterials 2018, 162, 132–143. [Google Scholar] [CrossRef] [PubMed]
  167. Alchujyan, N.K.; Hovhannisyan, M.R.; Movsesyan, N.H.; Madoyan, R.A.; Sargsyan, H.H.; Aghababova, A.A.; Minasyan, G.H.; Hairapetyan, H.L.; Kevorkian, R.G.; Chailyan, S.G.; et al. Free and zeolite-immobilized mixture versus sodium valproate in prevention of oxidative stress and modulation of the larginine intracellular metabolic pathways in the rat brain and blood following dexamphetamine-induced bipolar disorder. Eur. Chem. Bull 2018, 7, 42–51. [Google Scholar] [CrossRef] [Green Version]
  168. Eisenwagen, S.; Pavelic, K. Potential Role of Zeolites in Rehabilitation of Cancer Patients. Arch. Physiother. Rehabil 2022, 3, 29–40. Available online: http://www.fotunejournals.com/potential-role-of-zeolites-in-rehabilitation-of-cancer-patients.html (accessed on 21 August 2022).
  169. Johnson, H.H.; Microchemical, A. Method for the Determination of Histamine. AMA Arch. Derm. 1955, 72, 307. [Google Scholar] [CrossRef]
  170. Mück-Šeler, D.; Pivac, N. The effect of natural clinoptilolite on the serotonergic receptors in the brain of mice with mammary carcinoma. Life Sci. 2003, 73, 2059–2069. [Google Scholar] [CrossRef]
  171. Vitale, M.G.; Barbato, C.; Crispo, A.; Habetswallner, F.; De Martino, B.M.; Riccardi, F.; Maione, A.; Eisenwagen, S.; Vitale, G.; Cartenì, G.; et al. Multi Trial: A Randomized, Double-Blinded, Placebo-Controlled Trial of Oral PMA-Zeolite to Prevent Chemotherapy-Induced Side Effects, in Particular, Peripheral Neuropathy. Molecules 2020, 25, 2297. [Google Scholar] [CrossRef]
  172. Basha, M.P.; Begum, S.; Mir, B.A. Neuroprotective Actions of Clinoptilolite and Ethylenediaminetetraacetic Acid Against Lead-induced Toxicity in Mice Mus musculus. Toxicol. Int. 2013, 20, 201–207. [Google Scholar] [CrossRef] [Green Version]
  173. Delavarian, M.; Hassanvand, A.; Gharibzadeh, S. Increasing performance in children with ADHD by trapping lead with a nano-zeolite. J. Neuropsychiatry Clin. Neurosci. 2013, 25, E23. [Google Scholar] [CrossRef]
  174. Teixeira, M.F.S.; Bergamini, M.F.; Marques, C.M.P.; Bocchi, N. Voltammetric determination of L-dopa using an electrode modified with trinuclear ruthenium ammine complex (Ru-red) supported on Y-type zeolite. Talanta 2004, 63, 1083–1088. [Google Scholar] [CrossRef] [PubMed]
  175. Delavarian, M.; Hassanvand, A.; Gharibzadeh, S. Using natural zeolite as a transporter of dopamine. J. Neuropsychiatry Clin. Neurosci. 2013, 25, E21. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Inflammation as a substrate of depression and carcinoma co-occurrence. Peripheral inflammation is initiated with DAMPs through TLR, NLRP3, and NF-κB pathways. DAMPS are molecules released from damaged host cells that can be used as “alarmins”. Pro-inflammatory mediators such as acute-phase proteins, prostaglandins, leukotrienes, oxygen- and nitrogen-derived free radicals, chemokines, growth factors, and cytokines released by immune defense cells are particularly important for the early detection of acute inflammation. CRP, fibrinogen, and PCT as part of an innate immune response are detectable in serum within a few hours of inflammation initiation. The pro-inflammatory cytokines IL-1, IL-6, IFN, and TNF-α are elevated in carcinoma and consequently in the brain of patients with pain, fatigue, and depression. Acute and transitory inflammation must be distinguished from chronic and persistent inflammation. Anti-inflammatory strategies could lead to the resolution of inflammation and at the same time stabilization of depression and carcinoma. DAMP—danger-associated molecular patterns; TLR—toll-like receptors; NLRP3—nod-like receptor family pyrin domain containing 3; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; CRP—C-reactive protein; PCT—procalcitonin; IL—interleukin; TNF-α—tumor necrosis factor-alpha.
Figure 1. Inflammation as a substrate of depression and carcinoma co-occurrence. Peripheral inflammation is initiated with DAMPs through TLR, NLRP3, and NF-κB pathways. DAMPS are molecules released from damaged host cells that can be used as “alarmins”. Pro-inflammatory mediators such as acute-phase proteins, prostaglandins, leukotrienes, oxygen- and nitrogen-derived free radicals, chemokines, growth factors, and cytokines released by immune defense cells are particularly important for the early detection of acute inflammation. CRP, fibrinogen, and PCT as part of an innate immune response are detectable in serum within a few hours of inflammation initiation. The pro-inflammatory cytokines IL-1, IL-6, IFN, and TNF-α are elevated in carcinoma and consequently in the brain of patients with pain, fatigue, and depression. Acute and transitory inflammation must be distinguished from chronic and persistent inflammation. Anti-inflammatory strategies could lead to the resolution of inflammation and at the same time stabilization of depression and carcinoma. DAMP—danger-associated molecular patterns; TLR—toll-like receptors; NLRP3—nod-like receptor family pyrin domain containing 3; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; CRP—C-reactive protein; PCT—procalcitonin; IL—interleukin; TNF-α—tumor necrosis factor-alpha.
Cells 12 00710 g001
Table 1. Drugs targeting inflammation with potential for joint antidepressant effects.
Table 1. Drugs targeting inflammation with potential for joint antidepressant effects.
DrugClassReference
CelecoxibNSAIDs[137]
Minocyclinetetracycline antibiotics[138]
StatinsHMG-CoA reductase inhibitor[139]
PioglitazoneAntihyperglycemic[139]
Modafinilwakefulness promoting agents[139]
Steroidscorticosteroids[139]
Paricalcitolvitamin D analog[140]
Abbreviation: MAO—monoamine neurotransmitters; SSRIs—selective serotonin reuptake inhibitors; SNRIs—selective serotonin noradrenaline reuptake inhibitors; NSAIDs—non-steroidal anti-inflammatory drugs; HMG-CoA—β-hydroxy β-methylglutaryl-CoA.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Borovcanin, M.M.; Vesić, K.; Arsenijević, D.; Milojević-Rakić, M.; Mijailović, N.R.; Jovanovic, I.P. Targeting Underlying Inflammation in Carcinoma Is Essential for the Resolution of Depressiveness. Cells 2023, 12, 710. https://doi.org/10.3390/cells12050710

AMA Style

Borovcanin MM, Vesić K, Arsenijević D, Milojević-Rakić M, Mijailović NR, Jovanovic IP. Targeting Underlying Inflammation in Carcinoma Is Essential for the Resolution of Depressiveness. Cells. 2023; 12(5):710. https://doi.org/10.3390/cells12050710

Chicago/Turabian Style

Borovcanin, Milica M., Katarina Vesić, Dragana Arsenijević, Maja Milojević-Rakić, Nataša R. Mijailović, and Ivan P. Jovanovic. 2023. "Targeting Underlying Inflammation in Carcinoma Is Essential for the Resolution of Depressiveness" Cells 12, no. 5: 710. https://doi.org/10.3390/cells12050710

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop