Published online Oct 24, 2025. doi: 10.5306/wjco.v16.i10.108954
Revised: June 2, 2025
Accepted: September 17, 2025
Published online: October 24, 2025
Processing time: 180 Days and 20.7 Hours
Neurotransmitter-mediated regulation plays a multi-dimensional role in the tumor microenvironment, profoundly influencing key processes such as tumor immune evasion, metabolic reprogramming, and metastasis. However, the upst
Core Tip: This article presents a novel perspective on the role of neural signaling in orchestrating tumor progression through the neuro-metabolic-immune axis within the tumor microenvironment. It illustrates that neurotransmitters can directly regulate immune evasion while also mediating metabolic reprogramming, thereby indirectly facilitating immunosuppression. This integrative framework unveils a dynamic bidirectional network wherein neural signals coordinate both metabolic and immune adaptations, offering new therapeutic targets aimed at disrupting this axis to enhance conventional cancer treatments.
- Citation: Li ZK, Liao JL, Luo MR, Fang SJ, Huang WZ, Zhang DY. Neuro-tumor interactions: Multi-dimensional mechanisms of neurotransmitter regulation in tumor immune evasion and metabolic reprogramming. World J Clin Oncol 2025; 16(10): 108954
- URL: https://www.wjgnet.com/2218-4333/full/v16/i10/108954.htm
- DOI: https://dx.doi.org/10.5306/wjco.v16.i10.108954
Cancer remains one of the leading causes of death worldwide. In 2020, there were an estimated 19.3 million new cancer cases and 10 million cancer-related deaths globally. This burden is projected to rise further, reaching approximately 20 million new cases and 10 million deaths by 2025, underscoring cancer’s persistent and escalating impact on global public health[1]. The biological heterogeneity and complexity of cancer significantly challenge conventional therapeutic approaches[2]. In recent years, advances in immunotherapy and metabolism-targeted treatments have emphasized the tumor microenvironment (TME) as a central regulatory nexus influencing cancer progression and response to therapy[3]. While the mechanisms of immune evasion and metabolic reprogramming have been widely studied, the upstream regulatory systems that orchestrate these processes remain incompletely understood. The nervous system is increasingly recognized as a cross-system integrator in cancer biology, capable of shaping the TME through modulation of immune activity, metabolic states, and neurochemical signaling[4].
In recent years, the crucial role of the nervous system in tumor progression has gradually been revealed. For instance, the impact of psychological stress on tumor progression has been confirmed in several studies, with the nervous system playing an important role in this process, particularly in regulating immune responses and the TME. Research has shown that excessive activation of the sympathetic nervous system (SNS) may lead to the disruption of body homeostasis, promoting tumor formation and progression[5]. The nervous system mediates extensive interactions with tumor cells and their microenvironment through neurotransmitters, neuropeptides, and extracellular vesicles like exosomes, regulating tumor initiation, proliferation, invasion, and metastasis[6-8]. The nervous system can not only directly drive tumor cell proliferation, but also additionally modulate immune cell functions to facilitate tumor immune evasion[9,10]. Furthermore, in certain cancers, such as prostate cancer, mechanisms like tumor axonogenesis and neurogenesis further highlight the complexity of neural components in the TME[11]. The density and distribution of nerve fibers may even serve as prognostic biomarkers, providing a basis for personalized treatment[12]. These studies reveal the multidimensional regulatory role of the nervous system in the TME, involving immune modulation, metabolic reprogramming, and tumor cell proliferation and invasion. A deeper understanding of the molecular mechanisms underlying neuro-tumor communication not only provides a new perspective for elucidating tumor biological behavior but also lays a theoretical foundation for developing novel cancer treatment strategies targeting neuro-regulation. This paper aims to summarize the progress in understanding the neuro-regulation of tumorigenesis, with a particular focus on the key roles of neurotransmitter signaling in tumor immune evasion and metabolic reprogramming, and explore the potential therapeutic implications of targeting neural signaling in cancer treatment, providing new perspectives and a theoretical foundation for future research.
The SNS plays a crucial role in tumor progression through the release of norepinephrine (NE), which influences immune cell polarization and function. NE activates β-adrenergic receptor (β-AR) on tumor-associated macrophages (TAMs), promoting their polarization toward the M2 phenotype. This M2 polarization supports tumor growth by enhancing immune suppression and promoting tissue remodeling, which creates a more favorable environment for tumor expansion. Additionally, SNS signaling via NE contributes to the exhaustion of CD8+ T-cells by upregulating inhibitory receptors such as programmed death 1 (PD-1) and T cell immunoglobulin and mucin domain-containing protein 3, which impairs the cytotoxic function of these immune cells. Chronic stress, which elevates NE levels, accelerates tumor progression by further enhancing vascularization within the tumor and promoting immune escape. These processes allow tumors to evade immune surveillance and metastasize more effectively[13-15].
In contrast, parasympathetic nervous system signaling, primarily through acetylcholine (ACh), generally exerts anti-inflammatory effects that help regulate immune responses in the TME. ACh activates the cholinergic anti-inflammatory pathway, binding to α7-nicotinic ACh receptors on immune cells such as macrophages, inhibiting the release of pro-inflammatory cytokines like interleukin-6 and tumor necrosis factor-α (TNF-α). This dampening of inflammation helps regulate excessive immune responses in the TME, potentially preventing inflammation-related tumor progression and supporting immune responses against the tumor. However, in some contexts, ACh can also facilitate immune evasion by promoting the expansion of regulatory T cells (Tregs), which suppresses anti-tumor immune responses, and by inhibiting the infiltration of cytotoxic T cells into tumors. This paradoxical effect highlights ACh’s dual role in cancer: While it can modulate inflammation, it can also contribute to immune escape, thereby supporting tumor survival and progression[16,17].
Neuropeptides are an important means through which the nervous system directly regulates tumor immunity. Among them, calcitonin gene-related peptide (CGRP) and substance P, primarily released from sensory neurons, have been shown to modulate immune responses in the TME. CGRP can suppress CD8+ T-cell infiltration and function in tumors, thereby facilitating immune escape and tumor progression. This effect is reversible, as blocking CGRP signaling can enhance T cell responses and improve therapeutic outcomes such as those of radiotherapy and checkpoint inhibition[18,19]. CGRP is also involved in shaping the innate immune landscape, including inhibition of group 2 innate lymphoid cells and promoting M2-like macrophage polarization via the phosphoinositide 3-kinases/protein kinase B pathway, which contributes to immunosuppressive conditions in tumors[20,21]. Substance P, through its receptor neurokinin-1 receptor, has pro-inflammatory effects in general immune contexts, but in tumors, it has been associated with enhanced immunosuppressive activity driven by both TAMs and myeloid-derived suppressor cells (MDSCs)[22]. Neuropeptide Y has also been implicated in immune modulation, with studies indicating that it may influence T cell activity and Tregs function in a receptor- and dose-dependent manner[23]. In addition, neuropeptides influence immune activity by interacting with inflammasome and cytokine signaling. CGRP, for example, can inhibit NLRP3 inflammasome activation in macrophages, reducing interleukin-1β release and limiting inflammatory responses under infection-related conditions[24].
5-hydroxytryptamine (5-HT), beyond its classical role as a neurotransmitter, is increasingly recognized as an immunomodulatory molecule that actively shapes both innate and adaptive immune responses. A variety of immune cells—including T cells, macrophages, dendritic cells, and monocytes—express 5-HT receptors (e.g., 5-HT1, 5-HT2, 5-HT3, and 5-HT7) and can also synthesize or uptake 5-HT from the environment. Through these receptors, 5-HT regulates immune cell proliferation, cytokine production, and differentiation. For instance, 5-HT promotes T cell activation and modulates the T helper 17 cell/Treg balance, while also driving dendritic cell maturation and functional polarization. In macrophages, 5-HT signaling influences the shift between pro-inflammatory M1 and anti-inflammatory M2 phenotypes, suggesting that 5-HT contributes broadly to immune homeostasis and the resolution of inflammation[25,26].
Recent studies further reveal that different 5-HT receptors mediate distinct immunological outcomes. The 5-HT7 receptor, for example, can enhance the secretion of interleukin-6 and TNF-α in monocytes and microglia, while activation of the 5-HT3 receptor has been associated with modulation of cytokine release via p38 mitogen-activated protein kinase signaling. These receptor-specific pathways highlight 5-HT’s nuanced role in immune signaling. In the context of tumor immunology, 5-HT can contribute to immune evasion by promoting immunosuppressive cell phenotypes—such as M2 macrophages and Tregs—within the TME. At the same time, antagonizing certain 5-HT receptors has been shown to restore anti-tumor immune activity, indicating that serotonergic pathways may be viable targets in cancer immunotherapy[27-29].
Warburg effect: The Warburg effect, a hallmark of cancer metabolism, refers to the preference of cancer cells for glycolysis over oxidative phosphorylation even in the presence of sufficient oxygen. This phenomenon, first observed by Otto Warburg in the early 1920s, has since been identified as a key feature of tumor cell metabolism. The driving factors behind the Warburg effect are multifaceted and involve both genetic mutations and alterations in signaling pathways. Key contributors include hypoxia-inducible factors, cellular myelocytomatosis oncogene, and mutations in tumor suppressors, such as p53, which collectively promote the reprogramming of cancer cell metabolism[30]. These factors regulate the expression of crucial enzymes involved in glycolysis, such as hexokinase, phosphofructokinase, and pyruvate kinase isoform M2, which facilitate the rapid consumption of glucose and the production of lactate, even under normoxic conditions[31]. Additionally, the altered TME, characterized by low oxygen levels, acidic conditions, and nutrient deprivation, further enhances the reliance on glycolysis as the primary energy source, ensuring continuous cell proliferation and survival[32]. This metabolic shift not only supports cancer cell growth but also promotes immune evasion and resistance to conventional therapies, making the Warburg effect a central driver of tumor progression[30].
Glutamine metabolism dependency: Tumor cells often exhibit a heightened dependency on glutamine metabolism, a key feature of their metabolic reprogramming. This dependency is driven by the need for glutamine to support rapid cell proliferation and maintain tumor survival. Glutamine, though a non-essential amino acid, is consumed at significantly higher rates by cancer cells due to the upregulation of specific transporters, such as solute carrier family 1, member 5, and enzymes like glutaminase, which facilitate enhanced glutamine uptake and its catabolism[33]. This reprogrammed metabolism supports the replenishment of tricarboxylic acid cycle intermediates through anaplerosis, which in turn fuels critical biosynthetic pathways for nucleotides, amino acids, and lipids—key building blocks for rapidly dividing cancer cells[34]. Furthermore, glutamine-derived α-ketoglutarate plays a crucial role in epigenetic regulation, influencing tumorigenesis by modulating gene expression and chromatin structure. In hypoxic environments, glutamine also supports oxidative phosphorylation, providing energy even in the absence of adequate glucose[35]. These adaptations, however, are not uniform across all cancer types, with specific TME and genetic mutations influencing the extent of glutamine dependency. This metabolic heterogeneity highlights the potential for targeting glutamine metabolism as a therapeutic strategy for cancer treatment[36].
Enhanced lipid metabolism: Lipid metabolic reprogramming has emerged as a crucial driver of cancer progression, enabling tumor cells to adapt to the hostile TME characterized by low oxygen and nutrient availability. The reprogramming of lipid metabolism pathways supports tumor growth by providing essential building blocks for cell membranes, signaling molecules, and energy storage. One of the key mechanisms is the increased lipid uptake, particularly through transporters like CD36, fatty acid transfer proteins, and fatty acid binding proteins, which are upregulated in various cancers, including breast and ovarian cancers[37,38]. Moreover, tumor cells upregulate de novo lipogenesis to sustain their rapid growth, utilizing acetyl-CoA for fatty acid synthesis, which is facilitated by enzymes such as fatty acid synthase[39]. These metabolic alterations also extend to the TME, where stromal and immune cells undergo lipid reprogramming, further promoting an immunosuppressive environment that favors tumor progression[40]. The enhanced lipid metabolism not only supports the structural and energy needs of tumor cells but also modulates intracellular signaling pathways that contribute to malignant phenotypes, including metastasis and resistance to therapy[41].
Lactate contributes to the immunosuppressive microenvironment: Lactate, a metabolic byproduct of glycolysis, plays a significant role in shaping the immune-suppressive microenvironment in cancer and chronic inflammatory diseases. In the TME, the rapid glycolysis of tumor cells leads to high lactate levels, which contribute to local hypoxia and acidification. These conditions impair the function of effector immune cells, such as cytotoxic T cells and natural killer cells, by reducing glucose uptake and forcing these cells into metabolic stress, thus inhibiting their proliferation and cytokine production. Additionally, lactate fosters the polarization of immunosuppressive cells, such as Tregs and MDSCs, both of which contribute to immune evasion and promote tumor growth[42,43]. Lactate also directly influences macrophage polarization, shifting them toward an M2-like phenotype, which further reinforces the immunosuppressive environment and aids in tumor progression[44]. This dual role of lactate in fueling both metabolic and immune suppression highlights its pivotal function in cancer immunology.
Beyond its metabolic impact, lactate also induces epigenetic changes that contribute to the suppression of immune responses. Lactate promotes histone acetylation in macrophages, which leads to the transcriptional repression of pro-inflammatory cytokines and genes crucial for immune activation. This epigenetic modification not only weakens the inflammatory response but also leads to a form of “trained immunosuppression”, where immune cells, particularly macrophages, exhibit a diminished capacity to respond to subsequent inflammatory signals. This effect of lactate extends beyond cancer to other inflammatory conditions, such as rheumatoid arthritis and multiple sclerosis, where it similarly contributes to immune dysregulation[39,45]. The accumulation of lactate in these various pathological conditions underscores its role as a key modulator of immune responses, influencing both chronic inflammation and cancer progression[42,43].
Glutamine metabolic competition shapes the immunosuppressive microenvironment: Glutamine metabolism plays a pivotal role in shaping the immunosuppressive TME by influencing the function and phenotype of both innate and adaptive immune cells. In the TME, glutamine depletion caused by tumor cells not only impairs T cell effector function but also drives the reprogramming of tumor-associated immune cells toward suppressive phenotypes. For example, G protein-coupled receptor 109A+ myeloid cells, induced under glutamine-starved conditions via endoplasmic reticulum stress and the inositol-requiring enzyme 1α/X-box binding protein 1 axis, promote the accumulation of M2 phenotype TAMs and MDSCs, which inhibit CD8+ T cell responses[46,47]. Concurrently, TAMs can undergo metabolic reprogramming under glutamine-deficient and hypoxic conditions, shifting toward increased glutaminolysis, which supports their M2 phenotype through production of metabolites such as α-ketoglutarate. This metabolite not only fuels oxidative phosphorylation and fatty acid oxidation but also modulates epigenetic programs via Jmjd3-dependent mechanisms to sustain M2 activation[48,49]. Moreover, in renal cancer, local glutamine deprivation triggers interleukin-23 secretion from TAMs via hypoxia-inducible factor 1α activation, which in turn promotes Tregs expansion and suppresses cytotoxic lymphocyte activity, further contributing to immune evasion[50]. In dendritic cells, particularly type 1 conventional dendritic cells, glutamine uptake through the solute carrier family 38, member 2 transporter is essential for maintaining their cross-priming capacity and promoting CD8+ T cell responses. However, competition with tumor cells for glutamine can suppress these critical functions[47]. Notably, systemic glutamine blockade through prodrug-based inhibitors can impair tumor metabolic activity while simultaneously reprogramming the TME to enhance T cell infiltration and improve responsiveness to immunotherapy, highlighting the therapeutic potential of exploiting the differential glutamine metabolic flexibility between tumor and immune cells[51]. In summary, glutamine metabolism serves as a central axis in the metabolic crosstalk between tumor and immune cells, orchestrating an immunosuppressive niche that facilitates tumor progression and immune evasion, and thereby represents a critical target for therapeutic intervention in cancer immunometabolism.
Lipid metabolism as a driver of immunosuppressive TME: Lipid metabolism plays a critical role in shaping the immunosuppressive TME by influencing immune cell function and promoting immune evasion by tumor cells. In response to the nutrient and oxygen scarcity in the TME, tumor cells upregulate lipid uptake, synthesis, and oxidation to sustain their growth. This metabolic reprogramming leads to an accumulation of lipids, particularly fatty acids, within immune cells such as CD8+ T cells and Tregs, impairing their anti-tumor functions. For instance, excess lipids in CD8+ T cells, due to increased lipid uptake via CD36 and other transporters, hinder their secretion of critical cytokines like interferon-gamma and TNF-α, diminishing their cytotoxic activity against tumor cells[38,52]. Moreover, TAMs and MDSCs also undergo lipid metabolic reprogramming, enhancing their immunosuppressive functions and promoting tumor progression[38]. Additionally, the activation of sterol regulatory element-binding proteins in Tregs drives fatty acid synthesis, contributing to their functional specialization and further suppressing anti-tumor immune responses[53]. This lipid-driven metabolic reprogramming not only affects immune cell function but also reshapes the interactions between tumor cells and stromal cells, further reinforcing a pro-tumor immune environment. Collectively, these findings also suggest that targeting lipid metabolism in the TME could offer a promising therapeutic strategy to enhance anti-tumor immunity[54].
Sympathetic adrenergic signaling mediates metabolic reprogramming: Sympathetic adrenergic signaling plays a critical role in regulating metabolic reprogramming, a key feature of cancer cell metabolism. The release of NE from the SNS binds to β-AR on immune cells and tumor cells, modulating their metabolic activities. In immune cells, NE–β-AR signaling suppresses glycolysis by downregulating the expression of glucose transporter 1, which is essential for glucose uptake and effective immune function[55]. In addition to immune cells, chronic stress and adrenergic signaling also modulate tumor metabolism by enhancing glycolysis. For example, chronic stress-induced release of epinephrine activates the β2-adrenergic receptor (β2-AR)/protein kinase A/cAMP responsive element binding protein 1 pathway, increasing the expression of glycolytic enzymes such as hexokinase 2 and phosphofructokinase, thus promoting colorectal cancer (CRC) progression[56]. This metabolic shift towards glycolysis under adrenergic stress enables tumor cells to meet their high energy demands, crucial for sustaining rapid cell growth and proliferation. By enhancing glucose availability and facilitating the production of metabolic intermediates, this shift supports tumor cells in adapting to their high-energy needs, promoting both tumor growth and metastasis[57]. Additionally, chronic stress-induced epinephrine stabilizes the ubiquitin-specific peptidase 22 protein, which promotes the expression of forkhead box protein O1, leading to the activation of adipose triglyceride lipase-mediated lipolysis and driving breast cancer metastasis. Inhibiting ubiquitin-specific peptidase 22 can block this signaling pathway, thus suppressing epinephrine-induced breast cancer metastasis[58]. In conclusion, metabolic reprogramming can bring about complex changes to the TME and profoundly influence tumor progression, while sympathetic adrenergic signaling acts as one of the ways in neural regulation to mediate metabolic reprogramming.
Regulation of metabolic reprogramming by 5-HT: Beyond its traditional role as a neurotransmitter, 5-HT not only directly regulates the immune microenvironment as mentioned earlier to influence immune function, but also has a role worth exploring in regulating metabolism. In peripheral tissues, 5-HT regulates metabolic processes by influencing insulin secretion in pancreatic β-cells and modulating lipogenesis and lipolysis in adipocytes, highlighting its role in metabolic regulation[59]. In cancer, 5-HT metabolism is altered, contributing to tumor progression by enhancing cell proliferation, invasion, and angiogenesis through complex mechanisms, including receptor-mediated signaling and the serotonylation of proteins such as glyceraldehyde-3-phosphate dehydrogenase (GAPDH)[60]. In immune cells, particularly CD8+ T cells, 5-HT influences glycolytic metabolism by promoting the serotonylation of GAPDH, which shifts the metabolic phenotype toward glycolysis, supporting enhanced immune activity in the TME[61]. Moreover, 5-HT is also involved in regulating gluconeogenesis in the liver through its action on specific 5-HT receptors, further illustrating its capacity to control systemic metabolic processes[62]. Collectively, these findings underscore the multifaceted role of 5-HT in metabolic reprogramming, where it not only maintains systemic energy homeostasis but also adjusts metabolic pathways to support tumor progression and immune responses.
The β2-AR, activated by catecholamines such as NE, plays a critical role in suppressing CD8+ T-cell function. This suppression occurs through the inhibition of the metabolic reprogramming that is essential for T-cell activation and effector function. Under normal conditions, T-cell activation triggers an increase in glucose uptake and glycolysis, which are essential for providing the energy required for immune responses. However, activation of the β2-AR by NE reduces the expression of glucose transporter 1 on T-cells, impairing glucose uptake and glycolysis[55]. Moreover, β2-AR signaling also negatively impacts mitochondrial function, further dampening the metabolic capacity of CD8+ T-cells and weakening their ability to generate the energy necessary for an effective immune response. This metabolic blockade leads to a failure in T-cell activation, thereby promoting immune evasion within the TME[63]. In contrast, 5-HT has a dual role in the TME, which complicates its impact on immune responses. On one hand, 5-HT can enhance the activation and function of CD8+ T-cells by promoting glycolytic metabolism through a post-translational modification of GAPDH via serotonylation. This modification enhances GAPDH activity, which is crucial for the glycolytic shift required during T-cell activation and antitumor immune responses[61]. On the other hand, 5-HT also promotes immunosuppressive conditions within the TME, particularly when it accumulates in excess or is dysregulated. For instance, 5-HT interacts with the phosphoinositide 3-kinases/protein kinase B/mammalian target of rapamycin pathway in tumor cells, driving metabolic reprogramming that supports tumor growth and metastasis. This metabolic shift increases the production of immunosuppressive cytokines, such as interleukin-10 and transforming growth factor-β, which not only dampen T-cell activity but also promote the recruitment of MDSCs and Tregs to the TME, further hindering effective immune responses[64].
Based on the above discussion, we have uncovered the intricate and dynamic interplay between the neuro-immune axis and the metabolic network, highlighting the critical role of neural signals in orchestrating metabolic reprogramming within the TME. These findings point to a central insight: Neural regulation of metabolism not only fulfills the energetic demands of tumor cells but also reshapes the immune landscape, thereby profoundly influencing tumor immune evasion and progression. Notably, this regulatory mechanism exhibits a dual nature—capable of enhancing antitumor immune responses or, conversely, facilitating immunosuppression and tumor advancement. Thus, a deeper understanding of how neural signals precisely modulate metabolic pathways and immune function is essential. Such insight holds promise for elucidating the systemic underpinnings of tumor biology and for guiding the development of innovative therapeutic strategies targeting the neuro-metabolic-immune axis.
In recent years, although significant progress has been made in cancer treatment, especially in immunotherapy and targeted therapies, many patients still face the issue of poor treatment outcomes. Tumor immune evasion, therapy resistance, and the complexity of the TME remain major challenges in current treatment strategies. While conventional therapies such as chemotherapy and immune checkpoint inhibitors (ICIs) can sometimes effectively prolong patient survival, their effectiveness is still limited due to tumor cell adaptability and immune suppression. As a result, exploring new therapeutic targets and strategies, particularly targeting neural signaling pathways within the TME, has become an important task in cancer research.
Traditionally used for treating cardiovascular diseases, β-blockers (BBs) have been shown to inhibit epinephrine/NE-mediated immune evasion in several tumor models[14,15,58,65]. To identify relevant studies, we performed a focused literature search using the PubMed database. The search strategy used the following Boolean logic: (“propranolol” OR “beta-blocker”) AND (“cancer” OR “tumor”) AND “clinical trial”. Articles published before March 2025 in English were considered. While clinical trials were prioritized, one translational and preclinical study involving murine models was also included due to its mechanistic significance and clinical relevance. These studies are summarized in Table 1[66-72].
| Cancer | Study design | Drug | Main results | Sample size | Year |
| BC | Phase II RCT | Propranolol + etodolac | Reduced Ki-67, inflammatory cytokines, and pro-metastatic gene expression[62] | 38 | 2018 |
| MM | Phase I clinical trial | Propranolol + pembrolizumab | Safe with 78% objective response rate and biomarker modulation[63] | 9 | 2021 |
| BC | Phase II feasibility study | Propranolol + neoadjuvant chemotherapy | Feasible with 96% adherence, no major safety issues[64] | 10 | 2021 |
| CRC | Translational and preclinical study | Propranolol | Activated CD8+ T cells, suppressed AKT/MAPK signaling[65] | Unknow | 2020 |
| OC | Prospective pilot clinical trial | Propranolol + chemotherapy | Feasible with cytokine modulation and improved QOL[66] | 26 | 2019 |
| CRC | RCT | Propranolol + etodolac | Improved EMT and immune markers, and reduced recurrence trend[67] | 34 | 2020 |
| BC | Phase II RCT | Propranolol + etodolac | Reduced EMT and inflammation, and improved NK cell markers[68] | 38 | 2017 |
Synergistic effect of BBs with immunotherapy: ICIs have become a breakthrough in cancer treatment, yet many patients still do not benefit from them. Studies have found that SNS activation can suppress CD8+ T cell activity through β2-AR and promote the recruitment of immunosuppressive cells (such as MDSCs and Tregs), weakening the effects of immunotherapy[73]. It has been shown that BBs can counteract these effects by enhancing CD8+ T cell function and reducing immune suppression in the TME, thereby improving the effectiveness of ICIs[69]. Clinical studies have shown that BBs can improve the disease control rate and extend the duration of treatment in patients receiving ICI therapy. For example, in melanoma patients, propranolol combined with PD-1 inhibitors increased the objective response rate to 78%, which was significantly higher than the 46% observed with PD-1 inhibitors alone[67]. Additionally, BBs enhance T cell infiltration, reduce MDSCs, and improve the TME, further supporting their potential as immunotherapy enhancers[74].
BBs enhance chemotherapy efficacy: Chemotherapy remains the cornerstone of cancer treatment, but tumor resistance limits its efficacy. Studies have shown that BBs can enhance chemotherapy sensitivity by reducing SNS activity[73]. In a triple-negative breast cancer mouse model, propranolol combined with anthracycline chemotherapy (e.g., doxorubicin) significantly reduced distant metastasis and lowered NE levels in the TME, inhibiting β2-AR-mediated pro-cancer signaling[75]. Furthermore, in a CRC model, propranolol reduced the expression of carbonic anhydrase IX in the TME, disrupting hypoxic adaptation and enhancing the effect of fluorouracil chemotherapy[76].
In recent years, several studies have revealed the negative regulatory role of 5-HT in tumors, making it a potential anti-tumor therapeutic target. In various mouse tumor models, peripheral depletion of 5-HT significantly inhibited tumor growth, prolonged survival, enhanced CD8+ T cell infiltration and effector function, and downregulated the expression of the immune-suppressive molecule PD-L1 on tumor cells. Drug interventions such as selective 5-HT reuptake inhibitors (e.g., fluoxetine) and tryptophan hydroxylase 1 inhibitors (e.g., telotristat) showed anti-tumor effects similar to genetic depletion and synergized with PD-1 antibodies to significantly improve immunotherapy response rates[77].
Moreover, research targeting the 5-HT receptor 5-hydroxytryptamine receptor 2B further deepens the potential of 5-HT-targeted strategies. In CRC, high expression of HTR2B is associated with poor prognosis, and antagonists such as SB204741 and GM-60186 can significantly inhibit cancer cell proliferation and migration by suppressing the extracellular regulated protein kinases signaling pathway, reducing cyclin D1 expression, and inducing apoptosis[78]. Another study indicated that HTR2B promotes CRC metastasis through the cAMP responsive element binding protein 1-zinc finger E-box binding homeobox 1 axis driving epithelial-mesenchymal transition, and the use of specific antagonists like RS127445 effectively suppressed metastatic foci formation[79].
Nerve growth factor (NGF) and its receptor TrkA play crucial roles in tumor immune evasion, therapy resistance, and tumor-neural interactions. Research suggests that NGF plays a significant role in tumor progression by promoting neurogenesis and neural reprogramming. This makes NGF a potential therapeutic target, especially in the context of nerve-cancer interactions[80].
In melanoma, NGF suppresses tumor cells’ response to immune factors such as interferon-gamma, reducing CD8+ T cell and natural killer cell infiltration in the TME, thus promoting immune evasion. Inhibiting NGF or its receptor TrkA significantly enhances the efficacy of immunotherapy, boosting T cell function and memory cell generation[81]. Furthermore, the chemotherapy drug doxorubicin upregulates NGF expression, promoting sympathetic nerve growth in tumors and exacerbating tumor growth and chemotherapy-induced neuropathic pain. Targeting NGF signaling not only enhances immune responses but also alleviates chemotherapy side effects[82].
In pancreatic cancer studies, high expression of the Trk receptor is associated with poor prognosis. Targeting the NGF signaling pathway can inhibit tumor neurogenesis, improve the immune microenvironment, and enhance chemotherapy efficacy[83]. Additionally, high expression of the NGF receptor (NGFR) in melanoma is closely linked to immune resistance. Cells with high NGFR expression reduce T cell infiltration and immune rejection, significantly weakening the treatment response. Inhibiting NGFR or its downstream pathways can restore immunotherapy sensitivity and enhance therapeutic effects[84].
Targeting neural signaling pathways in cancer therapy opens up new avenues for treatment. Its innovation lies in focusing not only on tumor cells themselves but also on regulating the TME and immune evasion mechanisms. This approach can enhance the effects of immunotherapy and increase chemotherapy sensitivity. However, this strategy also faces challenges, particularly the potential side effects from tumor heterogeneity and the widespread effects of the nervous system, which may limit its clinical applicability. Despite these challenges, the strategy of targeting neural signals holds great research potential, and as research progresses, it may become a critical complement to cancer treatment, driving an overall transformation in therapeutic approaches.
The complex interplay between the nervous system and cancer biology, as illustrated in this study, reveals a paradigm shift in our understanding of tumor progression. Traditionally viewed as a disease of uncontrolled cellular proliferation, cancer is now increasingly recognized as a systemic disorder influenced by neuroimmune and metabolic dynamics. However, while the evidence supports a robust role for neurotransmitter signaling in shaping tumor behavior and immune evasion, critical gaps and contradictions remain, warranting a deeper, more nuanced exploration.
One of the most compelling—but underexplored—aspects of neuro-tumor interaction lies in its bidirectionality. While the nervous system clearly influences tumor progression, tumors themselves may remodel neural circuits through axonogenesis and neurogenesis[85]. This reciprocal feedback loop suggests that tumors can “educate” the nervous system to establish a more permissive niche. Yet, current therapeutic strategies predominantly target the outbound influence of neural signals on the tumor, neglecting the inbound modulatory potential of tumors on neural function. This raises provocative questions: Could tumors induce neuroplastic changes akin to those seen in chronic pain or depression? Could such changes be harnessed or reversed therapeutically?
Moreover, the duality of neurotransmitter functions introduces a significant therapeutic dilemma. For instance, both ACh and serotonin exhibit paradoxical roles—alternately promoting immune regulation and immune evasion depending on context, dosage, and receptor subtype expression[16,17,27-29]. These functional bifurcations challenge the binary classification of neurotransmitters as “pro-“ or “anti-“ tumorigenic and underscore the necessity of precision neuropharmacology. Rather than blunt inhibition or activation, future therapies may need to rely on spatiotemporal modulation of specific receptor subtypes within defined microenvironments.
Another dimension ripe for exploration is the intersection of neural signaling with metabolic fitness of immune cells. While the Warburg effect and glutamine addiction are well-characterized for tumor cells[30,36], emerging evidence suggests that neuro-mediated metabolic suppression of cytotoxic T cells is a major bottleneck in immunotherapy[55,63]. The β2-adrenergic suppression of glycolytic flux in CD8+ T cells illustrates how metabolic checkpoints are not merely intrinsic but can be exogenously imposed by stress-derived neurotransmitters[55]. This reframes adrenergic signaling not just as a passive contributor to immune suppression, but as an active metabolic switch—raising the possibility that targeting neuro-metabolic axes might revive “metabolically paralyzed” immune cells within the TME.
Importantly, this paper also raises a philosophical and practical debate regarding the therapeutic implications of targeting such deeply conserved and widely expressed systems as neurotransmitter pathways. The systemic effects of BBs or serotonin modulators, for example, might extend beyond tumors to influence cognition, mood, cardiovascular tone, and gut physiology[86-89]. This highlights a crucial ethical and clinical balancing act: Can we disrupt tumor-supportive neurobiology without disrupting systemic neurophysiology? The answer may lie in localized delivery methods, nanomedicine, or engineering of receptor-selective ligands that exhibit TME-specific activation.
Finally, the emerging convergence of cancer neuroscience with systems immunology and metabolism invites the construction of integrative models. Such models must go beyond correlative omics to map causal signaling hierarchies and feedback loops across the neuro-immune-metabolic triad. This interdisciplinary approach may unlock a new generation of therapeutic strategies—ones that do not merely kill tumor cells but restore systemic immune competence, recalibrate metabolic resilience, and rewire neurophysiological balance.
This paper underscores that neural regulation contributes to tumor immune evasion not only by directly modulating immune cells, but also by reprogramming tumor metabolism in ways that suppress immune responses. By proposing the concept of a neuro-metabolic-immune axis, this study offers a unified framework that integrates neural-immune interactions with metabolic modulation, thereby providing a more comprehensive understanding of how the nervous system shapes the TME. Based on current evidence, we also outline the mechanistic rationale for targeting neural signaling pathways as a feasible and promising therapeutic strategy. While emerging studies suggest the potential of targeting neural signaling pathways in cancer, several limitations remain: Clinical validation is still limited, the systemic physiological consequences of neural modulation are not fully understood, and this paper may not exhaustively capture the complexity and diversity of neuro-metabolic-immune interactions across all tumor types. Further in-depth and translational research is warranted to fully harness this axis for clinical benefit.
| 1. | Siegel RL, Kratzer TB, Giaquinto AN, Sung H, Jemal A. Cancer statistics, 2025. CA Cancer J Clin. 2025;75:10-45. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 161] [Cited by in RCA: 701] [Article Influence: 701.0] [Reference Citation Analysis (1)] |
| 2. | Dagogo-Jack I, Shaw AT. Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 2018;15:81-94. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1349] [Cited by in RCA: 2343] [Article Influence: 292.9] [Reference Citation Analysis (0)] |
| 3. | Bader JE, Voss K, Rathmell JC. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Mol Cell. 2020;78:1019-1033. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 566] [Cited by in RCA: 669] [Article Influence: 133.8] [Reference Citation Analysis (2)] |
| 4. | Hanahan D, Monje M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. Cancer Cell. 2023;41:573-580. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 21] [Cited by in RCA: 155] [Article Influence: 77.5] [Reference Citation Analysis (0)] |
| 5. | Zhang Z, Lv ZG, Lu M, Li H, Zhou J. Nerve-tumor crosstalk in tumor microenvironment: From tumor initiation and progression to clinical implications. Biochim Biophys Acta Rev Cancer. 2024;1879:189121. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 6. | Gysler SM, Drapkin R. Tumor innervation: peripheral nerves take control of the tumor microenvironment. J Clin Invest. 2021;131:e147276. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 125] [Article Influence: 31.3] [Reference Citation Analysis (0)] |
| 7. | Huang Q, Hu B, Zhang P, Yuan Y, Yue S, Chen X, Liang J, Tang Z, Zhang B. Neuroscience of cancer: unraveling the complex interplay between the nervous system, the tumor and the tumor immune microenvironment. Mol Cancer. 2025;24:24. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 13] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
| 8. | Winkler F, Venkatesh HS, Amit M, Batchelor T, Demir IE, Deneen B, Gutmann DH, Hervey-Jumper S, Kuner T, Mabbott D, Platten M, Rolls A, Sloan EK, Wang TC, Wick W, Venkataramani V, Monje M. Cancer neuroscience: State of the field, emerging directions. Cell. 2023;186:1689-1707. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 265] [Cited by in RCA: 255] [Article Influence: 127.5] [Reference Citation Analysis (0)] |
| 9. | Chen QY, Gao B, Tong D, Huang C. Crosstalk between extracellular vesicles and tumor-associated macrophage in the tumor microenvironment. Cancer Lett. 2023;552:215979. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 33] [Reference Citation Analysis (0)] |
| 10. | Wang S, Sun J, Dastgheyb RM, Li Z. Tumor-derived extracellular vesicles modulate innate immune responses to affect tumor progression. Front Immunol. 2022;13:1045624. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 21] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
| 11. | Sejda A, Sigorski D, Gulczyński J, Wesołowski W, Kitlińska J, Iżycka-Świeszewska E. Complexity of Neural Component of Tumor Microenvironment in Prostate Cancer. Pathobiology. 2020;87:87-99. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 31] [Article Influence: 6.2] [Reference Citation Analysis (0)] |
| 12. | Tan X, Sivakumar S, Bednarsch J, Wiltberger G, Kather JN, Niehues J, de Vos-Geelen J, Valkenburg-van Iersel L, Kintsler S, Roeth A, Hao G, Lang S, Coolsen ME, den Dulk M, Aberle MR, Koolen J, Gaisa NT, Olde Damink SWM, Neumann UP, Heij LR. Nerve fibers in the tumor microenvironment in neurotropic cancer-pancreatic cancer and cholangiocarcinoma. Oncogene. 2021;40:899-908. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 50] [Cited by in RCA: 77] [Article Influence: 19.3] [Reference Citation Analysis (0)] |
| 13. | Park HJ, Lee SC, Park SH. Norepinephrine stimulates M2 macrophage polarization via β2-adrenergic receptor-mediated IL-6 production in breast cancer cells. Biochem Biophys Res Commun. 2024;741:151087. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 14. | Sun J, Jia X, Zhang Z, Yang Y, Zhai C, Zhao B, Liu Y. Role of β-adrenergic signaling and the NLRP3 inflammasome in chronic intermittent hypoxia-induced murine lung cancer progression. Respir Res. 2024;25:347. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 15. | Yang J, Wei W, Zhang S, Jiang W. Chronic stress influences the macrophage M1-M2 polarization balance through β-adrenergic signaling in hepatoma mice. Int Immunopharmacol. 2024;138:112568. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 9] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 16. | Cox MA, Bassi C, Saunders ME, Nechanitzky R, Morgado-Palacin I, Zheng C, Mak TW. Beyond neurotransmission: acetylcholine in immunity and inflammation. J Intern Med. 2020;287:120-133. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 52] [Cited by in RCA: 117] [Article Influence: 23.4] [Reference Citation Analysis (0)] |
| 17. | Koarai A, Ichinose M. Possible involvement of acetylcholine-mediated inflammation in airway diseases. Allergol Int. 2018;67:460-466. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 26] [Cited by in RCA: 41] [Article Influence: 5.9] [Reference Citation Analysis (0)] |
| 18. | Darragh LB, Nguyen A, Pham TT, Idlett-Ali S, Knitz MW, Gadwa J, Bukkapatnam S, Corbo S, Olimpo NA, Nguyen D, Van Court B, Neupert B, Yu J, Ross RB, Corbisiero M, Abdelazeem KNM, Maroney SP, Galindo DC, Mukdad L, Saviola A, Joshi M, White R, Alhiyari Y, Samedi V, Van Bokhoven A, St John M, Karam SD. Sensory nerve release of CGRP increases tumor growth in HNSCC by suppressing TILs. Med. 2024;5:254-270.e8. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 20] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
| 19. | Sánchez ML, Rodríguez FD, Coveñas R. Neuropeptide Y Peptide Family and Cancer: Antitumor Therapeutic Strategies. Int J Mol Sci. 2023;24:9962. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 22] [Reference Citation Analysis (0)] |
| 20. | Nagashima H, Mahlakõiv T, Shih HY, Davis FP, Meylan F, Huang Y, Harrison OJ, Yao C, Mikami Y, Urban JF Jr, Caron KM, Belkaid Y, Kanno Y, Artis D, O'Shea JJ. Neuropeptide CGRP Limits Group 2 Innate Lymphoid Cell Responses and Constrains Type 2 Inflammation. Immunity. 2019;51:682-695.e6. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 115] [Cited by in RCA: 233] [Article Influence: 38.8] [Reference Citation Analysis (0)] |
| 21. | Yuan K, Zheng J, Shen X, Wu Y, Han Y, Jin X, Huang X. Sensory nerves promote corneal inflammation resolution via CGRP mediated transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway. Int Immunopharmacol. 2022;102:108426. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 40] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
| 22. | Zheng R, Wang S, Wang J, Zhou M, Shi Q, Liu B. Neuromedin U regulates the anti-tumor activity of CD8(+) T cells and glycolysis of tumor cells in the tumor microenvironment of pancreatic ductal adenocarcinoma in an NMUR1-dependent manner. Cancer Sci. 2024;115:334-346. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 23. | Sigorski D, Sejda A, Abualsaud N, Krawczyk E, Izycka-Swieszewska E, Kitlinska J. Neuropeptide Y in cancer-biological functions and potential clinical implications. Cancer Metastasis Rev. 2025;44:21. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 24. | Zhu F, Yu D, Qin X, Qian Y, Ma J, Li W, Liu Q, Wang C, Zhang Y, Li Y, Jiang D, Wang S, Xia P. The neuropeptide CGRP enters the macrophage cytosol to suppress the NLRP3 inflammasome during pulmonary infection. Cell Mol Immunol. 2023;20:264-276. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 18] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 25. | Quintero-Villegas A, Valdés-Ferrer SI. Role of 5-HT(7) receptors in the immune system in health and disease. Mol Med. 2019;26:2. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 44] [Cited by in RCA: 59] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
| 26. | Wu H, Denna TH, Storkersen JN, Gerriets VA. Beyond a neurotransmitter: The role of serotonin in inflammation and immunity. Pharmacol Res. 2019;140:100-114. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 99] [Cited by in RCA: 232] [Article Influence: 38.7] [Reference Citation Analysis (0)] |
| 27. | Balakrishna P, George S, Hatoum H, Mukherjee S. Serotonin Pathway in Cancer. Int J Mol Sci. 2021;22:1268. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 47] [Cited by in RCA: 87] [Article Influence: 21.8] [Reference Citation Analysis (0)] |
| 28. | Irving H, Turek I, Kettle C, Yaakob N. Tapping into 5-HT(3) Receptors to Modify Metabolic and Immune Responses. Int J Mol Sci. 2021;22:11910. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 14] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
| 29. | Karmakar S, Lal G. Role of serotonin receptor signaling in cancer cells and anti-tumor immunity. Theranostics. 2021;11:5296-5312. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 25] [Cited by in RCA: 105] [Article Influence: 26.3] [Reference Citation Analysis (0)] |
| 30. | Vaupel P, Schmidberger H, Mayer A. The Warburg effect: essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 2019;95:912-919. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 226] [Cited by in RCA: 600] [Article Influence: 100.0] [Reference Citation Analysis (0)] |
| 31. | Fukushi A, Kim HD, Chang YC, Kim CH. Revisited Metabolic Control and Reprogramming Cancers by Means of the Warburg Effect in Tumor Cells. Int J Mol Sci. 2022;23:10037. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 106] [Cited by in RCA: 130] [Article Influence: 43.3] [Reference Citation Analysis (0)] |
| 32. | Fendt SM. 100 years of the Warburg effect: A cancer metabolism endeavor. Cell. 2024;187:3824-3828. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 61] [Reference Citation Analysis (0)] |
| 33. | Yoo HC, Yu YC, Sung Y, Han JM. Glutamine reliance in cell metabolism. Exp Mol Med. 2020;52:1496-1516. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 452] [Cited by in RCA: 628] [Article Influence: 125.6] [Reference Citation Analysis (0)] |
| 34. | Yang L, Venneti S, Nagrath D. Glutaminolysis: A Hallmark of Cancer Metabolism. Annu Rev Biomed Eng. 2017;19:163-194. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 303] [Cited by in RCA: 327] [Article Influence: 40.9] [Reference Citation Analysis (0)] |
| 35. | Ma G, Zhang Z, Li P, Zhang Z, Zeng M, Liang Z, Li D, Wang L, Chen Y, Liang Y, Niu H. Reprogramming of glutamine metabolism and its impact on immune response in the tumor microenvironment. Cell Commun Signal. 2022;20:114. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 122] [Reference Citation Analysis (0)] |
| 36. | Cluntun AA, Lukey MJ, Cerione RA, Locasale JW. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer. 2017;3:169-180. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 411] [Cited by in RCA: 560] [Article Influence: 70.0] [Reference Citation Analysis (0)] |
| 37. | Bian X, Liu R, Meng Y, Xing D, Xu D, Lu Z. Lipid metabolism and cancer. J Exp Med. 2021;218:e20201606. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 582] [Cited by in RCA: 602] [Article Influence: 150.5] [Reference Citation Analysis (0)] |
| 38. | Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol. 2023;16:103. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 190] [Article Influence: 95.0] [Reference Citation Analysis (1)] |
| 39. | Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev Cell. 2021;56:1363-1393. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 48] [Cited by in RCA: 420] [Article Influence: 105.0] [Reference Citation Analysis (1)] |
| 40. | Yang K, Wang X, Song C, He Z, Wang R, Xu Y, Jiang G, Wan Y, Mei J, Mao W. The role of lipid metabolic reprogramming in tumor microenvironment. Theranostics. 2023;13:1774-1808. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 85] [Cited by in RCA: 104] [Article Influence: 52.0] [Reference Citation Analysis (0)] |
| 41. | Liu X, Lv M, Zhang W, Zhan Q. Dysregulation of cholesterol metabolism in cancer progression. Oncogene. 2023;42:3289-3302. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 44] [Article Influence: 22.0] [Reference Citation Analysis (0)] |
| 42. | Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 2021;21:151-161. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 324] [Cited by in RCA: 526] [Article Influence: 131.5] [Reference Citation Analysis (0)] |
| 43. | Wang ZH, Peng WB, Zhang P, Yang XP, Zhou Q. Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine. 2021;73:103627. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 278] [Article Influence: 69.5] [Reference Citation Analysis (0)] |
| 44. | Wang JX, Choi SYC, Niu X, Kang N, Xue H, Killam J, Wang Y. Lactic Acid and an Acidic Tumor Microenvironment suppress Anticancer Immunity. Int J Mol Sci. 2020;21:8363. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 52] [Cited by in RCA: 268] [Article Influence: 53.6] [Reference Citation Analysis (0)] |
| 45. | Shi W, Cassmann TJ, Bhagwate AV, Hitosugi T, Ip WKE. Lactic acid induces transcriptional repression of macrophage inflammatory response via histone acetylation. Cell Rep. 2024;43:113746. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 62] [Article Influence: 62.0] [Reference Citation Analysis (0)] |
| 46. | Yang Y, Pei T, Liu C, Cao M, Hu X, Yuan J, Chen F, Guo B, Hong Y, Liu J, Li B, Li X, Wang H. Glutamine metabolic competition drives immunosuppressive reprogramming of intratumour GPR109A(+) myeloid cells to promote liver cancer progression. Gut. 2025;74:255-269. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 27] [Article Influence: 27.0] [Reference Citation Analysis (0)] |
| 47. | Guo C, You Z, Shi H, Sun Y, Du X, Palacios G, Guy C, Yuan S, Chapman NM, Lim SA, Sun X, Saravia J, Rankin S, Dhungana Y, Chi H. SLC38A2 and glutamine signalling in cDC1s dictate anti-tumour immunity. Nature. 2023;620:200-208. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 171] [Cited by in RCA: 164] [Article Influence: 82.0] [Reference Citation Analysis (0)] |
| 48. | Wang Y, Wang D, Yang L, Zhang Y. Metabolic reprogramming in the immunosuppression of tumor-associated macrophages. Chin Med J (Engl). 2022;135:2405-2416. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 16] [Cited by in RCA: 29] [Article Influence: 9.7] [Reference Citation Analysis (0)] |
| 49. | Liu PS, Wang H, Li X, Chao T, Teav T, Christen S, Di Conza G, Cheng WC, Chou CH, Vavakova M, Muret C, Debackere K, Mazzone M, Huang HD, Fendt SM, Ivanisevic J, Ho PC. α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol. 2017;18:985-994. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 444] [Cited by in RCA: 886] [Article Influence: 110.8] [Reference Citation Analysis (0)] |
| 50. | Fu Q, Xu L, Wang Y, Jiang Q, Liu Z, Zhang J, Zhou Q, Zeng H, Tong S, Wang T, Qi Y, Hu B, Fu H, Xie H, Zhou L, Chang Y, Zhu Y, Dai B, Zhang W, Xu J. Tumor-associated Macrophage-derived Interleukin-23 Interlinks Kidney Cancer Glutamine Addiction with Immune Evasion. Eur Urol. 2019;75:752-763. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 94] [Cited by in RCA: 157] [Article Influence: 26.2] [Reference Citation Analysis (0)] |
| 51. | Leone RD, Zhao L, Englert JM, Sun IM, Oh MH, Sun IH, Arwood ML, Bettencourt IA, Patel CH, Wen J, Tam A, Blosser RL, Prchalova E, Alt J, Rais R, Slusher BS, Powell JD. Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion. Science. 2019;366:1013-1021. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 679] [Cited by in RCA: 824] [Article Influence: 137.3] [Reference Citation Analysis (0)] |
| 52. | Yu W, Lei Q, Yang L, Qin G, Liu S, Wang D, Ping Y, Zhang Y. Contradictory roles of lipid metabolism in immune response within the tumor microenvironment. J Hematol Oncol. 2021;14:187. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 147] [Cited by in RCA: 159] [Article Influence: 39.8] [Reference Citation Analysis (0)] |
| 53. | Lim SA, Wei J, Nguyen TM, Shi H, Su W, Palacios G, Dhungana Y, Chapman NM, Long L, Saravia J, Vogel P, Chi H. Lipid signalling enforces functional specialization of T(reg) cells in tumours. Nature. 2021;591:306-311. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 68] [Cited by in RCA: 301] [Article Influence: 75.3] [Reference Citation Analysis (0)] |
| 54. | Tang Y, Chen Z, Zuo Q, Kang Y. Regulation of CD8+ T cells by lipid metabolism in cancer progression. Cell Mol Immunol. 2024;21:1215-1230. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 22] [Reference Citation Analysis (0)] |
| 55. | Qiao G, Bucsek MJ, Winder NM, Chen M, Giridharan T, Olejniczak SH, Hylander BL, Repasky EA. β-Adrenergic signaling blocks murine CD8(+) T-cell metabolic reprogramming during activation: a mechanism for immunosuppression by adrenergic stress. Cancer Immunol Immunother. 2019;68:11-22. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 93] [Cited by in RCA: 116] [Article Influence: 19.3] [Reference Citation Analysis (0)] |
| 56. | Guan Y, Yao W, Yu H, Feng Y, Zhao Y, Zhan X, Wang Y. Chronic stress promotes colorectal cancer progression by enhancing glycolysis through β2-AR/CREB1 signal pathway. Int J Biol Sci. 2023;19:2006-2019. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 34] [Reference Citation Analysis (0)] |
| 57. | Fitzgerald PJ. Norepinephrine release may play a critical role in the Warburg effect: an integrative model of tumorigenesis. Neoplasma. 2020;67:947-957. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 10] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
| 58. | Zhou Y, Chu P, Wang Y, Li N, Gao Q, Wang S, Wei J, Xue G, Zhao Y, Jia H, Song J, Zhang Y, Pang Y, Zhu H, Sun J, Ma S, Su C, Hu B, Zhao Z, Zhang H, Lu J, Wang J, Wang H, Sun Z, Fang D. Epinephrine promotes breast cancer metastasis through a ubiquitin-specific peptidase 22-mediated lipolysis circuit. Sci Adv. 2024;10:eado1533. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 59. | Moon JH, Oh CM, Kim H. Serotonin in the regulation of systemic energy metabolism. J Diabetes Investig. 2022;13:1639-1645. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 24] [Reference Citation Analysis (0)] |
| 60. | Ye D, Xu H, Tang Q, Xia H, Zhang C, Bi F. The role of 5-HT metabolism in cancer. Biochim Biophys Acta Rev Cancer. 2021;1876:188618. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 11] [Cited by in RCA: 25] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 61. | Wang X, Fu SQ, Yuan X, Yu F, Ji Q, Tang HW, Li RK, Huang S, Huang PQ, Qin WT, Zuo H, Du C, Yao LL, Li H, Li J, Li DX, Yang Y, Xiao SY, Tulamaiti A, Wang XF, Dai CH, Zhang X, Jiang SH, Hu LP, Zhang XL, Zhang ZG. A GAPDH serotonylation system couples CD8(+) T cell glycolytic metabolism to antitumor immunity. Mol Cell. 2024;84:760-775.e7. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 39] [Reference Citation Analysis (0)] |
| 62. | El-Merahbi R, Löffler M, Mayer A, Sumara G. The roles of peripheral serotonin in metabolic homeostasis. FEBS Lett. 2015;589:1728-1734. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 130] [Cited by in RCA: 171] [Article Influence: 17.1] [Reference Citation Analysis (0)] |
| 63. | Farooq MA, Ajmal I, Hui X, Chen Y, Ren Y, Jiang W. β2-Adrenergic Receptor Mediated Inhibition of T Cell Function and Its Implications for CAR-T Cell Therapy. Int J Mol Sci. 2023;24:12837. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 13] [Reference Citation Analysis (0)] |
| 64. | Zheng Y, Li L, Shen Z, Wang L, Niu X, Wei Y, Sun S, Zhao J. Mechanisms of neural infiltration-mediated tumor metabolic reprogramming impacting immunotherapy efficacy in non-small cell lung cancer. J Exp Clin Cancer Res. 2024;43:284. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
| 65. | Fnu T, Shi P, Zhang W, Chung SSW, Damoci CB, Fang Y, Chen QY, Saqi A, Huang Y, Wu X, Lu C, Yang D, Wang TC, Que J. Sympathetic Neurons Promote Small Cell Lung Cancer through the β2-Adrenergic Receptor. Cancer Discov. 2025;15:616-632. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 10] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
| 66. | Haldar R, Shaashua L, Lavon H, Lyons YA, Zmora O, Sharon E, Birnbaum Y, Allweis T, Sood AK, Barshack I, Cole S, Ben-Eliyahu S. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 51] [Cited by in RCA: 63] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 67. | Gandhi S, Pandey MR, Attwood K, Ji W, Witkiewicz AK, Knudsen ES, Allen C, Tario JD, Wallace PK, Cedeno CD, Levis M, Stack S, Funchain P, Drabick JJ, Bucsek MJ, Puzanov I, Mohammadpour H, Repasky EA, Ernstoff MS. Phase I Clinical Trial of Combination Propranolol and Pembrolizumab in Locally Advanced and Metastatic Melanoma: Safety, Tolerability, and Preliminary Evidence of Antitumor Activity. Clin Cancer Res. 2021;27:87-95. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 27] [Cited by in RCA: 88] [Article Influence: 17.6] [Reference Citation Analysis (0)] |
| 68. | Hopson MB, Lee S, Accordino M, Trivedi M, Maurer M, Crew KD, Hershman DL, Kalinsky K. Phase II study of propranolol feasibility with neoadjuvant chemotherapy in patients with newly diagnosed breast cancer. Breast Cancer Res Treat. 2021;188:427-432. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 18] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 69. | Liao P, Song K, Zhu Z, Liu Z, Zhang W, Li W, Hu J, Hu Q, Chen C, Chen B, McLeod HL, Pei H, Chen L, He Y. Propranolol Suppresses the Growth of Colorectal Cancer Through Simultaneously Activating Autologous CD8(+) T Cells and Inhibiting Tumor AKT/MAPK Pathway. Clin Pharmacol Ther. 2020;108:606-615. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 31] [Article Influence: 6.2] [Reference Citation Analysis (0)] |
| 70. | Ramondetta LM, Hu W, Thaker PH, Urbauer DL, Chisholm GB, Westin SN, Sun Y, Ramirez PT, Fleming N, Sahai SK, Nick AM, Arevalo JMG, Dizon T, Coleman RL, Cole SW, Sood AK. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 29] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
| 71. | Haldar R, Ricon-Becker I, Radin A, Gutman M, Cole SW, Zmora O, Ben-Eliyahu S. Perioperative COX2 and β-adrenergic blockade improves biomarkers of tumor metastasis, immunity, and inflammation in colorectal cancer: A randomized controlled trial. Cancer. 2020;126:3991-4001. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 37] [Cited by in RCA: 77] [Article Influence: 15.4] [Reference Citation Analysis (0)] |
| 72. | Shaashua L, Shabat-Simon M, Haldar R, Matzner P, Zmora O, Shabtai M, Sharon E, Allweis T, Barshack I, Hayman L, Arevalo J, Ma J, Horowitz M, Cole S, Ben-Eliyahu S. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 148] [Cited by in RCA: 203] [Article Influence: 25.4] [Reference Citation Analysis (0)] |
| 73. | Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther. 2024;264:108750. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 74. | Mellgard G, Patel VG, Zhong X, Joshi H, Qin Q, Wang B, Parikh A, Jun T, Alerasool P, Garcia P, Gogerly-Moragoda M, Leiter A, Gallagher EJ, Oh WK, Galsky MD, Tsao CK. Effect of concurrent beta-blocker use in patients receiving immune checkpoint inhibitors for advanced solid tumors. J Cancer Res Clin Oncol. 2023;149:2833-2841. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 13] [Article Influence: 6.5] [Reference Citation Analysis (0)] |
| 75. | Chang A, Botteri E, Gillis RD, Löfling L, Le CP, Ziegler AI, Chung NC, Rowe MC, Fabb SA, Hartley BJ, Nowell CJ, Kurozumi S, Gandini S, Munzone E, Montagna E, Eikelis N, Phillips SE, Honda C, Masuda K, Katayama A, Oyama T, Cole SW, Lambert GW, Walker AK, Sloan EK. Beta-blockade enhances anthracycline control of metastasis in triple-negative breast cancer. Sci Transl Med. 2023;15:eadf1147. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 53] [Reference Citation Analysis (0)] |
| 76. | Puzderova B, Belvoncikova P, Grossmannova K, Csaderova L, Labudova M, Fecikova S, Pastorek J, Barathova M. Propranolol, Promising Chemosensitizer and Candidate for the Combined Therapy through Disruption of Tumor Microenvironment Homeostasis by Decreasing the Level of Carbonic Anhydrase IX. Int J Mol Sci. 2023;24:11094. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 77. | Schneider MA, Heeb L, Beffinger MM, Pantelyushin S, Linecker M, Roth L, Lehmann K, Ungethüm U, Kobold S, Graf R, van den Broek M, Vom Berg J, Gupta A, Clavien PA. Attenuation of peripheral serotonin inhibits tumor growth and enhances immune checkpoint blockade therapy in murine tumor models. Sci Transl Med. 2021;13:eabc8188. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 9] [Cited by in RCA: 100] [Article Influence: 25.0] [Reference Citation Analysis (0)] |
| 78. | Lee JY, Park S, Park EJ, Pagire HS, Pagire SH, Choi BW, Park M, Fang S, Ahn JH, Oh CM. Inhibition of HTR2B-mediated serotonin signaling in colorectal cancer suppresses tumor growth through ERK signaling. Biomed Pharmacother. 2024;179:117428. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 79. | Li T, Wei L, Zhang X, Fu B, Zhou Y, Yang M, Cao M, Chen Y, Tan Y, Shi Y, Wu L, Xuan C, Du Q, Hu R. Serotonin Receptor HTR2B Facilitates Colorectal Cancer Metastasis via CREB1-ZEB1 Axis-Mediated Epithelial-Mesenchymal Transition. Mol Cancer Res. 2024;22:538-554. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 11] [Article Influence: 11.0] [Reference Citation Analysis (0)] |
| 80. | Silverman DA, Martinez VK, Dougherty PM, Myers JN, Calin GA, Amit M. Cancer-Associated Neurogenesis and Nerve-Cancer Cross-talk. Cancer Res. 2021;81:1431-1440. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 107] [Cited by in RCA: 143] [Article Influence: 35.8] [Reference Citation Analysis (0)] |
| 81. | Yin T, Wang G, Wang L, Mudgal P, Wang E, Pan CC, Alexander PB, Wu H, Cao C, Liang Y, Tan L, Huang D, Chong M, Chen R, Lim BJW, Xiang K, Xue W, Wan L, Hu H, Loh YH, Wang XF, Li QJ. Breaking NGF-TrkA immunosuppression in melanoma sensitizes immunotherapy for durable memory T cell protection. Nat Immunol. 2024;25:268-281. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 22] [Article Influence: 22.0] [Reference Citation Analysis (0)] |
| 82. | Zuo S, Wang Z, Jiang X, Zhao Y, Wen P, Wang J, Li J, Tanaka M, Dan S, Zhang Y, Wang Z. Regulating tumor innervation by nanodrugs potentiates cancer immunochemotherapy and relieve chemotherapy-induced neuropathic pain. Biomaterials. 2024;309:122603. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 83. | Qin J, Liu J, Wei Z, Li X, Chen Z, Li J, Zheng W, Liu H, Xu S, Yong T, Zhao B, Gou S, Ju S, Teng GJ, Yang X, Gan L. Targeted intervention in nerve-cancer crosstalk enhances pancreatic cancer chemotherapy. Nat Nanotechnol. 2025;20:311-324. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 12] [Article Influence: 12.0] [Reference Citation Analysis (0)] |
| 84. | Boshuizen J, Vredevoogd DW, Krijgsman O, Ligtenberg MA, Blankenstein S, de Bruijn B, Frederick DT, Kenski JCN, Parren M, Brüggemann M, Madu MF, Rozeman EA, Song JY, Horlings HM, Blank CU, van Akkooi ACJ, Flaherty KT, Boland GM, Peeper DS. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat Commun. 2020;11:3946. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 44] [Cited by in RCA: 75] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
| 85. | Mi J, Zhang W, Ren Y, Zhu L, Yang B, Wang H, Duan L. Neuronal guanine nucleotide exchange factor promotes the axonal growth and cancer cell proliferation via Ephrin-A3/EphA2 axis in lung adenocarcinoma. J Transl Med. 2025;23:246. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
| 86. | Archer C, Wiles N, Kessler D, Turner K, Caldwell DM. Beta-blockers for the treatment of anxiety disorders: A systematic review and meta-analysis. J Affect Disord. 2025;368:90-99. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 9] [Reference Citation Analysis (0)] |
| 87. | Behlke LM, Lenze EJ, Carney RM. The Cardiovascular Effects of Newer Antidepressants in Older Adults and Those With or At High Risk for Cardiovascular Diseases. CNS Drugs. 2020;34:1133-1147. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 20] [Cited by in RCA: 64] [Article Influence: 12.8] [Reference Citation Analysis (0)] |
| 88. | Fung TC, Vuong HE, Luna CDG, Pronovost GN, Aleksandrova AA, Riley NG, Vavilina A, McGinn J, Rendon T, Forrest LR, Hsiao EY. Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut. Nat Microbiol. 2019;4:2064-2073. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 170] [Cited by in RCA: 310] [Article Influence: 51.7] [Reference Citation Analysis (0)] |
| 89. | Munley JA, Kelly LS, Park G, Pons EE, Apple CG, Kannan KB, Bible LE, Efron PA, Nagpal R, Mohr AM. Nonselective beta blockade enhances gut microbiome diversity in a rodent model of trauma, hemorrhage, and chronic stress. J Trauma Acute Care Surg. 2025;98:309-318. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
