Open Access

Research progress on the impact of opioids on the tumor immune microenvironment (Review)

  • Authors:
    • Yuancheng Zhou
    • Wenyu Li
    • Yuanji Chen
    • Xudong Hu
    • Chuanwang Miao
  • View Affiliations

  • Published online on: April 15, 2025     https://doi.org/10.3892/mco.2025.2848
  • Article Number: 53
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Opioids have been extensively used in cancer pain management because they can significantly improve the quality of life of patients with advanced cancer. However, recent evidence suggests that opioids can also modulate the tumor immune microenvironment by interacting with opioid receptors on immune cells, potentially regulating tumor progression and efficacy of cancer treatments. Notably, morphine can exhibit a dose‑dependent effect on tumor immunity in pancreatic cancer and renal cell models, with lower doses potentially promoting tumor migration and invasion of pancreatic cancer cells, whereas higher doses shows the effect of inhibiting migration and invasion through distinct molecular pathways. The present review therefore comprehensively explored the mechanisms by which opioids can regulate the tumor immune microenvironment, focusing on their effects on immune cells, oxidative stress and angiogenesis. It also examined the interactions between opioids and other analgesics, along with their potential impact on immune modulation. All relevant articles and materials were retrieved from PubMed using the key words ‘opioids’, ‘immune system’, ‘T cells’, ‘monocytes’, ’macrophages’, ‘lymphocytes’, ‘natural killer cell’, ‘immunotherapy’, ‘immune cell function’ and ‘dose dependent effect’. The immunosuppressive effects of opioids, particularly through the µ‑opioid receptor, can suppress the activity of natural killer cells, impair antigen presentation and promote the function of regulatory T cells (Tregs). These effects may contribute to tumor progression and metastasis. The severity of these immunosuppressive effects appears to be dose‑dependent and can vary among different tumor types. There is evidence to suggest that tumors with higher immune responsiveness will experience more pronounced suppression, including the reduction of tumor angiogenesis, resulting in a decrease in tumor volume and decrease in tumor metastases. Furthermore, the combination of opioids with other analgesics, such as non‑steroidal anti‑inflammatory drugs, has the potential to exacerbate immunosuppression, which can in turn increase the risk of infections. Therefore, although opioids are essential for pain management in patients with cancer, their potential to modulate the immune microenvironment and promote tumor progression requires careful consideration. Clinicians should evaluate the advantages and disadvantages of opioids, especially regarding emerging immunotherapies, to minimize their potential negative effects on the outcomes of cancer treatments. Future studies are recommended to prioritize the development of strategies that optimize pain management whilst preserving immune function, such as receptor‑specific opioid formulations or adjunctive therapies targeting immunosuppressive pathways.

1. Introduction

Opioids, such as morphine, fentanyl, oxycodone and methadone, are a class of psychoactive compounds targeting central and peripheral opioid receptors, are widely applied in the management of cancer-associated pain due to their potent analgesic effects (1-3), including the management of moderate to severe cancer pain and the management of pain at the site of primary and metastasis (4-6). The use of opioids have significantly improved the quality of life of patients with cancer (7,8). However, recent studies suggest that opioids may interact with immune cells within the tumor microenvironment, potentially affecting tumor progression and treatment responses (9,10).

Although there is ample evidence indicating certain immunomodulatory effects of opioids (10,11), including their effects of reducing T cell activity, suppressing the activity of NK cells and inhibiting B cell activity to reduce antibody production. Notably, these effects appear to vary across opioid types, with morphine showing the most pronounced effects. However, the specific mechanisms by which they can influence the tumor immune microenvironment remain to be fully elucidated. There are great knowledge gaps regarding the immunomodulatory effects of different opioids on various tumor models and outcomes of cancer treatments. Additionally, the interactions between opioids and novel immunotherapies, such as immune checkpoint inhibitors, require further exploration.

The present review aims to systematically evaluate the effects of opioids on the tumor immune microenvironment, emphasizing their potential immunomodulatory roles across different tumor types and their potential interactions with immunotherapies. The present review also integrates recent research findings to offer actionable insights for the clinical use of opioids, particularly in achieving effective pain management whilst preserving immune function in patients with cancer.

To the best of our knowledge, current literature has only rarely delved into the dose-dependent effects of opioids on the immune system, as they mainly instead focused on the effects of opioids on pain management. A number of the previous studies have mentioned the dose-dependent effects of opioids (11,12). In the present review, by elucidating the dose dependent effects and dual effect of opioids on the tumor immune microenvironment at different doses and cancer types, with particular attention to threshold concentrations observed in clinical and experimental practice, it explored the mechanisms by which opioids can regulate tumor progression under different conditions. In addition, the effects of opioids on immune cell function at different doses were discussed in detail. From this perspective, the present review hopes to reveal the potential immunosuppressive mechanism of opioids in patients with cancer.

2. Methods

The present review systematically retrieved the materials from PubMed (https://pubmed.ncbi.nlm.nih.gov), using key words ‘opioids’, ‘immune system’, ’T cells’, ’monocytes’, ’macrophages’, ‘lymphocytes’, ‘natural killer cell’, ‘immunotherapy’, ‘immune cell function’ and ‘dose dependent effect’.

Inclusion criteria: i) The present review included all original studies, systematic reviews and meta-analyses associated with cancer immunotherapy, opioid mechanisms of action and their effects on the immune system; ii) Studies involving the effects of opioids on the immune system, cancer growth or immunotherapy effects, including in vitro cell experiments, animal experiments and clinical studies, were all included; iii) studies must address the use of opioids and analyze their effects on immune cells [including T cells, macrophages, natural killer (NK) cells] or immune-related molecules (such as IL-2 and IFN-γ); iv) studies need to have control groups (such as individuals or cell/animal models not using opioids) to compare the effects of opioids on the immune system; v) at least one indicator of immune function (such as cytokine level, immune cell activity and tumor microenvironment change) must be reported; and vi) Literature between January 2000 and November 2024.

Exclusion criteria: i) The content of the study was not relevant, namely if the study did not explore the interaction between opioids and the immune system or if it placed too much focus on the analgesic effects of opioids but did not involve immune regulation; ii) insufficient or low-quality data, such as if the study did not provide complete experimental data, lacked control groups, had too small sample sizes or were of low methodological quality (such as drop-out rate >20%); and iii) non-English literatures, to avoid translation bias.

3. Mechanisms of opioids and their impact on the tumor immune microenvironment

Mechanisms of opioids

Opioids are commonly administered for the management of severe pain, because they can primarily target the µ, κ, δ and nociception (NOP) receptors (13). Amongst these, the µ receptor, with its two subtypes µ1 and µ2, is predominantly responsible for the key analgesic effects of opioids (14,15). In addition, they have also been implicated in opioid addiction (16). The µ1 receptor associated with central analgesia, euphoria and opioid use dependence (17). The µ2 receptor primarily mediates physiological processes, such as respiratory depression and inhibition of gastrointestinal motility (18). By contrast, the κ receptor and its three subtypes (κ1, κ2 and κ3), which are also involved in analgesia and sedation, exhibit relatively milder respiratory depression effects compared with these exerted by the µ receptor (19). The δ receptor is associated with anti-anxiety and antidepressant effects, whilst the NOP receptor is involved in regulating various responses, including reward and depression (20).

Introduction to the impact of opioids on the tumor immune microenvironment

Opioid receptors, including µ, δ, κ and NOP, are involved in regulating a number of physiological activities, such as analgesia, memory modulation, respiratory control and angiogenesis (21,22). A number of studies have suggested that opioids may contribute to the oxidative stress response within the breast cancer and human neuroblastoma tumor microenvironment (23-25), promoting the production of reactive oxygen species (ROS) (26,27). As a major mutagen, ROS can increase the risk of tumor development in opioid users (23,28). Additionally, opioids may exacerbate the oxidative imbalance, particularly in patients with cancer who frequently exhibit heightened oxidative stress due to concurrent inflammation (29). This suggests that opioids could cause greater harm to patients with cancer compared with healthy individuals. Table I presents a functional overview of opioid receptors in the tumor immune microenvironment.

Table I

A summary of the functions of opioid receptors in the tumor immune microenvironment.

Table I

A summary of the functions of opioid receptors in the tumor immune microenvironment.

Opioid receptorMain functionImpact on Tumor Immune MicroenvironmentKey studies/findings(Refs.)
µCentral analgesia, respiratory depressionPromotes tumor growth by enhancing angiogenesis and suppressing immune cell activity (such as natural killer cells and T cells)Morphine binding to µ receptors increases tumor invasiveness by modulating macrophage activity and promoting M2 polarization(12,34,37)
κAnalgesia, mild respiratory depressionInhibits angiogenesis by reducing VEGFR2 and neurophilin 1 expression, thereby potentially suppressing tumor growthκ receptor agonists exhibit anti-angiogenic properties, suggesting potential therapeutic applications in cancer treatment(51-53)
δAnti-anxiety, antidepressant effectsMay promote the proliferation and metastasis of cancer cells through pathways, such as Janus kinase/STAT, in specific cancer typesδ receptor activation can stimulate the metastasis and progression of breast cancer cells(50)
Nociception receptorsRegulates reward, depression, anxietyModulates various physiological responses, including immune suppression, though its direct impact on tumors is less studiedInvolved in complex signaling pathways that may indirectly influence tumor immune microenvironment(18)

Opioids can modulate immune functions through both direct and indirect pathways, influencing the body's response to tumor cells (9,30). Direct effects occur when opioids bind to opioid receptors on immune cells, directly regulating their physiological activity, which is primarily mediated through µ receptors. By contrast, indirect effects are mediated through the inhibitory effects of opioids on the central nervous system, affecting various receptors within the sympathetic nervous system. They can involve D1 receptors, ultimately affecting peripheral Y1 receptors to inhibit spleen NK cell cytotoxicity (9,30). Previous animal studies and studies on human CD3+ T cell and leukemic T-cell lines have demonstrated that µ receptor activation can suppress the activity of NK cells and inhibit the mitogenic responses of T cells (31-33). In addition, it can also suppress the mitogen responses of B cells in mice following an in vivo administration (34).

Existing evidence suggests that opioids may have dual effects on tumor progression, depending on the concentrations applied and the tumor cell heterogeneity profile (35,36). Previous studies on morphine have indicated that µ receptor expression in Lewis lung carcinoma cells may be crucial to these mechanisms, influencing tumor growth and metastasis by regulating apoptosis and VEGF signaling. As a result, opioid antagonists may represent a novel therapeutic option for cancer treatment (9,12,34). The following sections discuss the mechanisms of various opioid receptors and their potential effects on the immune system.

µ receptor

µ receptor agonists are considered immunosuppressive agents that can hinder the body's ability to combat cancer cells, particularly by promoting angiogenesis (37). Morphine, as a µ receptor agonist, also shows an agonistic effect on δ and κ receptors. However, it shows a low drug selectivity on δ and κ receptors compared with µ receptors (38). Therefore, morphine is primarily the predominant µ receptor agonist. Previous studies have shown that activation of morphine-induced µ receptors can promote breast cancer cell proliferation both in vivo and in vitro (37,39). Morphine can also regulate the polarization of macrophages toward an M1 or M2 (alternative) activation state in vitro. A previous study on murine bone marrow cells documented that morphine can inhibit macrophage M1 polarization in vitro (40). In addition, morphine was found to inhibit M2 macrophage (alternative) activation in vitro, including the processes of antigen uptake and caused morphological changes, such as decrease in perimeter, area and aspect ratio, increase in roundness, circularity and solidity (40). An experimental study on breast cancer indicated that morphine can decrease macrophage M2 polarization through blocking the activation of IL-4, which may lead to breast cancer progression (41). However, other studies on morphine have indicated that it can inhibit tumor invasiveness by modulating the production of macrophage proteases in the tumor microenvironment. Specifically, morphine affects the production of certain proteins such as inhibiting the production of MMP-9 and MMP-2 by inhibiting the nitric oxide (NO)/NO synthase (NOS) system] in MCF-7 breast cancer cells and pancreatic cancer (42,43). The NO/NOS system serves an important role in tumor progression, NO, as an important bioregulatory mediator in the human body, is produced in the body by the action of NOS, exhibiting the function of inducing extracellular matrix degradation (44,45). MMP-9 breaks down the extracellular matrix (ECM) and the basement membrane, which are structural components that provide physical barriers to multiple tumor cells, including giant cell tumor of bone (GCTB), non-small cell small lung cancer (NSCLC) and breast cancer. By degrading these barriers, MMP-9 enables tumor cells to invade surrounding tissues and spread to other parts of the body, increasing tumor invasiveness and metastasis (46,47). Therefore, by inhibiting the production of MMP-9 by inhibiting the NO/NOS system, morphine may exhibit an inhibitory function against tumor metastasis and invasiveness.

Previous studies have demonstrated that morphine can exert a dual effect on the proliferation of pancreatic cancer cells. In murine experiments, the tumor volume was found to be significantly increased in the low-dose morphine group (at doses of 0.5 mg/kg), whereas it was significantly smaller in the high-dose morphine group (at doses of 5 mg/kg) compared with that in the control group. The underlying mechanism was proposed to be mediated through the p38/JNK pathway. Specifically, low concentrations of morphine in this concentration led to an increase in phosphorylated JNK, whilst the phosphorylation of p38 was reduced. By contrast, higher concentrations of morphine reversed these aforementioned processes, resulting in increased p38 activation and decreased JNK activation (48). Another previous in vitro study reported that in renal cell carcinoma, when using morphine at a dose of 2 nmol to 3.5 µmol in serum concentrations, converted to clinical dosage at 10-2,450 mg/day, it exerted no significant proliferative effects. However, after increasing the dose to 50 µmol in serum concentrations, which equated to a commonly used clinical dose of 35 mg, morphine promoted the migratory ability of renal cell carcinoma cells (49). Therefore, it is essential to carefully consider the pharmacokinetic and tolerance effects of morphine during its administration. A clinical trial on patients with cervical cancer have been previously performed assessing the effects of morphine combined with ketamine (morphine, 1 mg/kg/day; ketamine, 1 mg/kg/day) and morphine alone (1 mg/kg/day) for pain management, where both groups yielded a decrease in CD4+ and CD4+/CD8+ ratios. This was proposed to be achieved by modulating T cell activation and cytokine expression, specifically by inhibiting the secretion of IL-2, IFN-γ and IL-17, through the Janus kinase 3/STAT5 pathway (50).

Animal models of NSCLC have shown that morphine (0.1 µg/µl) can promote the proliferation, migration and invasion of H460 cell-induced NSCLC by activating the Src/PI3K/AKT/mTOR signaling pathway, inhibiting the G2 phase of the cell cycle and suppressing apoptosis (51). Furthermore, another in vitro and in vivo study has demonstrated that, morphine (1.0 µmol/l) pre-treatment under hypoxic conditions can inhibit the production of the pro-angiogenic factor VEGF in Lewis lung carcinoma cells in vitro. In in vivo experiments, morphine exhibited an inhibitory effect on angiogenesis. This effect was mediated by morphine's interaction with hypoxia-inducible factor 1, which binds to its hypoxia response element, resulting in the suppression of VEGF production by µ receptor. Another murine study previously reported that morphine (plasma levels maintained between 250-400 ng/ml) can significantly reduce the blood vessel density, branching and length compared with those in the placebo group (52). This suggests that the MOR receptor-mediated processes can inhibit angiogenesis within the tumor microenvironment under certain conditions, thereby suppressing tumor growth. Morphine exhibit a dual effect on promote or inhibit tumor progression in different cancer models, including lung cancer model and breast cancer model, which may be due to the different downstream proteins activated by µ receptors on the surface of different cancer cells, the mechanism underlying the differences in performance among each cancer cell type warrants further exploration.

δ receptor

A previous study has addressed the δ receptor's role in breast cancer progression, with the majority primarily focusing on breast cancer. A mouse study previously indicated that δ receptor agonist (D-Ala2,D-Leu5)-Enkephalin treatment promoted lung metastasis by breast cancer cells, through the migratory Janus kinase 1/2-STAT3 axis and by enhancing epithelial-mesenchymal transition (53). Subsequent studies on human breast cancer cells have shown that expression of the δ receptor was increased in breast cancer cells, where the inhibition of the δ receptor was found to suppress cell proliferation, suggesting its role in the proliferation process (53,54). In addition, δ receptor was found to underly the progression of breast cancer by activating the protein kinase C/ERK signaling pathway. Therefore, the δ receptor inhibitors were proposed to serve as an adjunctive treatment for patients with breast cancer (53).

κ receptors

The role of the κ receptor in the tumor microenvironment primarily involved inhibiting angiogenesis, particularly the transformation of vascular progenitor cells into vascular endothelial cells. κ receptor agonists were found to inhibit VEGFR2 expression, thereby suppressing endothelial cell migration and angiogenesis in Lewis lung carcinoma and breast cancer (22,52,53).

Previous studies have shown that the κ receptor is highly expressed in endothelial progenitor cells and endothelial cells, where its activation can reduce the expression of VEGFR2 and neuropilin 1, which are necessary for angiogenesis, by decreasing cAMP production through inhibiting the cAMP/protein kinase A (PKA) pathway (22,52,53). In murine tumor models, κ receptor knockout mice exhibited enhanced tumor proliferation and angiogenesis following lung cancer cell transplantation compared with those in wild-type mice. Consequently, κ receptor agonists may inhibit tumor growth by suppressing angiogenesis (22,52,53).

The anti-angiogenic properties of κ receptor hold potential therapeutic value in cancer treatment. However, κ receptor agonists, such as butorphanol, nalbuphine and dynorphins, pose a risk of drug dependence and tolerance (55,56), necessitating careful consideration in their development and application, particularly for the combined treatment of cancer pain and anti-angiogenesis.

Additionally, another previous study on breast cancer cells found that the expression of κ receptor was significantly increased in breast cancer cells compared with that in non-tumor cells (normal human mammary epithelial cells). The inhibition of PI3K/AKT signaling by using inhibitors Recilisib and Buparlisib, or knockdown of the κ receptor, was found to reduce the viability and migration of breast cancer cells. These findings suggested that κ receptor may serve as a potential breast cancer growth antagonist (57).

4. Impact of opioids on the immune system

Previous studies have indicated that opioids can suppress the immune system, where in vitro studies have shown that opioids can damage immune cells, including macrophages and T cells (12,30). Epidemiological studies have also suggested that long-term opioid use may increase the risk of infections (30,34,58). The effects of opioids on immune cells are summarized in Fig. 1 and Table II.

Table II

A summary of the effects of opioids on different types of lymphocytes within the tumor immune microenvironment.

Table II

A summary of the effects of opioids on different types of lymphocytes within the tumor immune microenvironment.

OpioidLymphocyteEffect Mechanism/Outcome(Refs.)
MorphineT CellsDecreaseInhibits IL-2 production and receptor expression; reduces T cell activity and immune response(100,101)
 CD8+ T CellsDecreaseSuppresses IFN-γ production, inhibits STAT-1 pathway activation and leads to impaired antiviral response(102,103)
 NK cellsDecreaseSuppresses the activity of NK cells through central opioid receptors(102,103)
 B CellsDecreaseSuppresses mitogen-induced responses and reduces antibody production(78,104)
FentanylNK CellsDecreaseProlonged post-operative suppression, particularly in cancer surgery(30)
BuprenorphineT CellsNo Effect/reversed suppressionShows no effect or reverses postoperative suppressive effects(9)
TramadolNK CellsIncrease/reversed SuppressionIncreases the activity of NK cells or reverses the suppressive effects of surgery(30)

[i] NK, Natural killer.

Impact on T cells

Animal and cell studies have demonstrated that morphine can reduce the activity of T cells in mice by inhibiting the lymphocyte IL-2 production and IL-2 receptor expression in the spleen, which in turn reduces T cell activity and impairs immune function (32,59). In addition, cell experiments have demonstrated that morphine can reduce lactate dehydrogenase (LDH) release and the number of CD8+ T cells, whilst increasing the ratio of CD4+/CD8+ T cells (32,59). Another previous study on the effects of morphine on the anti-human immunodeficiency virus properties of CD8+ T cells has indicated that morphine can inhibit the activity of CD8+ T cells by directly binding to µ receptors, which can downregulate CD8+ T cell activity. In addition, morphine can reduce the secretion of IFN-γ by CD8+ T cells, thereby impairing their function. Morphine was also found to reduce the production of IFN-γ by CD8+ T cells and inhibits CD8 T cell-induced expression of the STAT1, further impairing IFN-γ signaling to compromise CD8 T cell activity, resulting in immune dysfunction.

Impact on monocytes and macrophages

Morphine can inhibit the macrophage-mediated clearance of pathogens (such as Streptococcus pneumoniae) by suppressing the toll-like receptor (TLR)9/NFκB signaling pathway, which increases the risk of infections (60). It can also reduce the production of IL-23 by dendritic cells and macrophages by inhibiting the TLR3 and nucleotide-binding oligomerization domain-containing protein 2 signaling pathways, weakening the body's defense mechanisms (60,61). In addition, recent studies suggest that morphine can significantly increase the expression of TLR4, p65, NF-κB and cytokines (IL-6 and TNF-α) in microglia and macrophages (62,63).

Numerous studies have demonstrated that morphine, as an opioid receptor agonist, can directly downregulate various phagocytic activity and enhance cell apoptosis in a number of cell types, including mouse peritoneal macrophages, bone marrowderived macrophage (BMDM) and peripheral blood monocytes (64,65). In particular, in BMDM, morphine can enhance LPS-induced macrophage apoptosis through a peroxisome proliferator-activated receptor γ-dependent mechanism (65). This interaction inhibits the production of ROS, including superoxide and peroxide, resulting in decreased superoxide anion production. Since these intermediates are involved in the bactericidal mechanism of phagocytes, morphine treatment resulted in decreased phagocytic efficacy and reduced macrophage bactericidal activity (40). Morphine has also been found to inhibit the formation of macrophage colonies in soft agar from cultured mouse bone marrow progenitors (34).

Impact on NK cells

In rat models, the proliferation of NK cells was reported to be significantly suppressed in mice treated with morphine compared with that in controls. Administration of a nucleus accumbens shell blocker reversed the suppressive effects of morphine on the proliferation of NK cells in mice, suggesting that the nucleus accumbens serves a critical role in regulating peripheral immune responses. This process is likely mediated by the activation of sympathetic pathways and dopaminergic neurohumoral immunomodulation, with morphine indirectly affecting NK cells (34,66,67). A previous study demonstrated that morphine can inhibit lymphocyte proliferation and NK cell activity in vitro (68), whereas tramadol exhibits the opposite effects (67,68). This effect of tramadol was due to the (+)-enantiomer of tramadol inhibiting serotonin (5-HT) reuptake, increase extracellular 5-HT concentrations in the synaptic cleft, prolonging its combining time with 5-HT receptors and amplifying 5-HT signaling, which can promote immune activation, increase NK cell activity and IL-2 production (67). These findings suggest that different opioids may have distinct impact on the immune system.

Contradictory research findings

Opioids are recognized for their inhibitory effects on immune cell function (12,34), including the suppression of lymphocytes, T cells and monocytes, which may in turn adversely affect the immune system and promote tumor growth. However, the literature also presents significant contradictions regarding these effects. These conflicting findings highlight the dual effects of opioids.

Dual role of µ receptors. µ receptor agonists may promote tumor growth under certain conditions. The activation of morphine-induced µ receptors can promote breast cancer cell proliferation both in vivo and in vitro (37,39). In addition, low concentrations of morphine in pancreatic cancer led to an increase in phosphorylated JNK, whilst the phosphorylation of p38 was reduced, resulting in an increment in tumor volume (48). However, morphine pre-treatment under hypoxic conditions inhibits VEGF production, which can suppress tumor growth (51,69). By inhibiting angiogenesis and production of M2-polarized macrophages, whilst inhibiting the production of MMP9 by macrophages, morphine can also suppress tumor growth (42,43).

Dual effect of κ receptors. Although κ receptor agonists can inhibit angiogenesis and tumor growth through the cAMP/PKA pathway, they may promote metastasis of breast cancer when activated in breast cancer cells (57,70).

The expression and functional profiles of different receptors, including µ and κ receptors, may vary in different types of tumors, leading to differences in anti-tumor or pro-tumor effects. The existing contradictory information are summarized in Table III.

Table III

A summary of the existing conflict effects of opioids.

Table III

A summary of the existing conflict effects of opioids.

Conflicts existingPossible explanations(Refs.)
µ receptors may promote or inhibit tumor growthThe receptor activation effect is dose-dependent; Anoxic conditions also influence the effect of µ receptor agonists(48)
κ receptors can reduce angiogenesis to reduce tumor growth, but they are highly expressed in breast cancer cellsκ receptor agonists can reduce VEGF expression and inhibit angiogenesis in endothelial cells; but in breast cancer, κ receptors can promote tumor growth through PI3K/AKT pathway(30,57)

5. Opioids in different cancer types

Effects of opioids on breast cancer

Previous studies of opioid use in breast cancer have shown that the use of fentanyl, a δ receptor agonist, as anesthetics can reduce NK cell cytotoxicity compared with sevoflurane, during surgery. The use of δ receptor agonists in breast cancer can also promote the metastasis of breast cancer cells (53,71). However, activation of κ receptor has been documented to exhibit an inhibitory effect on estrogen receptor (ER)-positive breast cancer through the κ receptor/ER/X-box binding protein 1 pathway (72).

Effects of opioids on lung cancer

Opioids can inhibit lung cancer angiogenesis through different pathways. In Lewis lung carcinoma murine experiments, κ receptor knockout mice showed increased proliferation and enhanced tumor angiogenesis compared with those in wild-type mice, through the VEGF signaling pathway, indicating that κ receptor may inhibit the blood supply and growth of lung cancer (70). However, in lung cancer cell experiments, morphine may suppress the immune function by upregulating maelstrom spermatogenic transposon silencer (MAEL) expression, increasing the levels of Programmed death-ligand (PD-L)1, TGF-β and IL-10, whilst decreasing IL-2 levels (59).

Effects of opioids on liver cancer

Morphine, as a µ receptor agonist, can suppress the metastasis and growth of liver cancer cells by upregulating the expression of opioid growth factor receptor and downregulation of µ-opioid receptor and MMP-9 expression (73). Murine experiments have previously reported that tramadol can suppress the growth of orthotopic liver tumors by promoting M1 macrophage polarization in the tumor microenvironment (74).

6. Interactions between opioids and common analgesics

Anesthesia during cancer surgery rarely involves a single anesthetic, such that cancer pain management typically requires the application of multiple drugs (75). There are complex pharmacokinetic interactions between various anesthetics, which significantly affect the patient's postoperative recovery. Opioids can inhibit the proliferation of T cells and hinder the function of B cells through µ receptors and by upregulating the production of proinflammatory cytokines, such as IL-1β and IL-6. By promoting the production of cAMP, µ receptor agonists can inhibit T cell receptor signaling through the PKA/Csk-binding phosphoprotein associated with glycosphingolipid-enriched microdomains/C-terminal Src kinase-leukocyte-specific protein tyrosine kinase pathway, resulting in a decrease in IL-2, which can inhibit the proliferation of T cells (76,77). In addition, they can inhibit the activity of NK cells through opioid receptors in the central nervous system (78). They can also stimulate the metabolism of benzodiazepines, potentially leading to respiratory depression when combined with benzodiazepines (79). When co-administered with certain general anesthetics, such as isoflurane and propofol, opioids may enhance respiratory depression, which causes severe respiratory depression and apnea (80). By contrast, co-administration with NSAIDs may affect drug metabolism through the liver, increasing the risk of gastrointestinal injury and bleeding (81). Combining opioids with alcohol can increase central nervous system depression, potentially leading to confusion and respiratory depression (82). Since patients with cancer may also take antidepressants, co-administration with opioids can result in respiratory depression, confusion, mood instability, drowsiness and other mental symptoms, such as anxiety, depression and hallucinations (specifically false sensory perceptions in the absence of external stimuli) (83). Combining opioids with certain sedatives, including benzodiazepines, may cause hypotension and bradycardia, increasing the cardiovascular burden, especially in elderly patients or those with a history of cardiovascular disease (84). Therefore, careful monitoring of cardiovascular function and the timely adjustment of medication may prevent the aforementioned complications (85).

Clinical scenarios for the combined use of opioids

As common analgesics in clinical settings, opioids have the potential to be combined with non-traditional cancer therapies. Emerging non-traditional cancer therapies, such as immunotherapy, primarily involve immune checkpoint inhibitors, adoptive cell therapy and cancer vaccines (86-88). Cancer immunotherapy is fundamentally based on the inhibition of immune escape mechanisms utilized by tumor cells. Currently, cancer immunotherapies primarily focus on immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) (89). The application of PD-1 or its ligands, PD-L1 and PD-L2 monoclonal antibodies, selectively blocks the interaction between tumor cells and T cells. This restores antitumor immunity and enhances cytotoxic T cell-mediated tumor destruction, which ultimately causes tumor eradication (90). CTLA-4 monoclonal antibodies inhibit the co-stimulatory interaction between CTLA-4 molecules on regulatory T cells and Fc, which induces the death of regulatory T cells, reduces negative T cell regulation and enhances T cell-mediated immune responses (91). Immune checkpoint inhibitor therapy allows T cells to effectively infiltrate into tumor sites and form long-lasting immune responses by activating the body's T-cell immunity (87,92,93).

However, the modulation of immune activity, upregulation of T cell activity and reduction of Treg cell-mediated immunosuppressive activity can increase the risk of autoimmune complications and inflammation, such as checkpoint inhibitor pneumonitis induced by anti-PD-1/PDL-1 immune checkpoint inhibitors (94). To improve survival rates and quality of life, it is common to co-administer opioids and immunosuppressants to patients who commonly experience abdominal and lesion pain (94). Opioids can damage T cells and macrophages, where retrospective analyses of PD-1/PD-L1 concomitant medications have shown that patients with melanoma and NSCLC who receive opioids along with immunotherapy exhibit significantly lower overall survival rates, objective response rates and progression-free survival rates compared with patients who receive immunotherapy without opioids. This combination carries a higher risk of disease progression, gut microbiota dysbiosis and gut barrier dysfunction (95). Therefore, it is necessary to closely monitor changes in gastrointestinal function and inflammation markers when the treatment plan involves a combination of opioid administration and immunotherapy. Alternative treatments should be promptly adopted in the case of impaired gastrointestinal function.

Opioids can damage immune components, such as macrophages and T lymphocytes. However, considering the dose-dependent effects of opioids on the immune system, further studies are needed to explore how opioids can affect the tumor immune microenvironment, which differs from the normal immune composition (96). The long-term effects of opioids on the immune system and the complex immune system within the tumor microenvironment require further investigation.

7. Therapeutic strategies and future directions

Opioids are used in cancer treatment to alleviate patients' pain and improve their quality of life. However, the efficiency of opioids depends on multiple factors, such as the type applied and severity of pain, the patient's overall health status, potential side effects and drug interactions (97). Clinical treatment typically follows the pain management guidelines published by the World Health Organization (WHO) (97). It is necessary to conduct comprehensive pain assessments and select appropriate dosages of analgesics based on the patient's condition and response. The choice of analgesics can be guided by the WHO analgesic ladder. If a single drug is insufficient to control pain, it is recommended to use a combination of multiple drugs along with non-pharmacological treatments, such as physical therapy, psychological support and alternative therapies such as traditional Chinese medicine, to alleviate the patient’s pain. Opioids may cause various side effects, such as constipation, vomiting, drowsiness and respiratory depression (98). Therefore, it is necessary to regularly monitor the patient's pain status, drug efficacy and side effects during the clinical treatment regimen. It is important to take the necessary measures to control these side effects and if necessary, to change the medications or increase their dosage to improve the patient's quality of life. Additionally, since cancer patients may take multiple drugs simultaneously, drug interactions must be carefully monitored.

The study of opioids represents a dynamic and advancing area of research. Researchers are actively pursuing novel drug targets to create innovative non-opioid analgesics and mitigate the adverse effects associated with the administration of opioids. To mitigate the risk of drug abuse and potential addiction, studies are investigating alternative therapies to overcome challenges, such as opioid abuse and dependence (99). In addition, it is crucial to develop selective opioid receptor antagonists to inhibit the potential immunosuppressive effects of opioids. κ receptor activation can exert anti-tumor effects by reducing VEGF production and inhibiting tumor angiogenesis (70). However, the use of receptor antagonists to block κ receptors on breast cancer cells can hinder breast cancer progression and reduce the risk of metastasis (70).

Additionally, advances in drug delivery systems, such as the use of nanotechnology or targeted delivery, can increase local drug concentration, reduce systemic side effects and enhance efficacy. Studies on genetic variations in analgesic responses may facilitate the development of personalized pain management strategies. Before clinical application, the efficacy, safety, tolerance and dependence of analgesics should be extensively investigated to prevent and mitigate their possible side effects.

8. Conclusion

Although opioids are essential for managing cancer-related pain, they serve a complex and dual role within the tumor immune microenvironment. The suppression of immune cell activity and promotion of angiogenesis is of particular importance in cancer, since they may undermine the effectiveness of antitumor therapies. Therefore, a deeper understanding of these mechanisms is crucial for optimizing the use of opioids in cancer treatment to ensure that pain management does not inadvertently compromise treatment outcomes. The following strategies and future research keynotes should be considered to achieve a balance between effective analgesia and optimal cancer pain management.

Personalized pain management plans

It is necessary to develop individualized analgesic plans under the WHO pain management guidelines to minimize opioid dosages whilst ensuring effective pain relief. This requires further development and integration of non-opioid analgesics, adjuvant therapies and non-pharmacological interventions to reduce the dependence on opioids.

Regular monitoring and assessment

Regular assessments of pain intensity, functional capacity and possible opioid-associated side effects are essential, where treatment strategies must be modified as needed to achieve effective pain management whilst minimizing immune function disruption.

Opioid-immune system interactions

The specific pathways through which different opioid receptors influence immune cell function, angiogenesis and tumor progression need to be investigated to inform safer analgesic choices.

Opioid use in conjunction with immunotherapies

The interaction of opioids with immune checkpoint inhibitors and other immunotherapies should be examined to determine their impact on treatment efficacy and patient outcomes.

Development of novel analgesics

It is necessary to explore and develop novel pain management agents that provide effective analgesia with minimal immunosuppressive effects to preserve the integrity of cancer therapies.

Acknowledgements

Not applicable.

Funding

Funding: The present review was supported by Shandong Natural Science Foundation Youth Program (grant nos. ZR2022QH227 and YXH2024YS029) and Research Project of Shandong Medical Association (grant no. YXH2024YS029).

Availability of data and materials

Not applicable.

Authors' contributions

YZ and WL were involved in the conception and design of the present review, collection and assembly of data. XH and CM contributed substantially to the conception and design of the study. CM also performed data analysis and data visualization. YC contributed to the refinement of the manuscript's narrative and logic critically reviewed the content. All authors were involved in drafting and revising the manuscript. All authors have reviewed and approved the final version of the submitted manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Trescot AM: Review of the role of opioids in cancer pain. J Natl Compr Canc Netw. 8:1087–1094. 2010.PubMed/NCBI View Article : Google Scholar

2 

Paice JA, Bohlke K, Barton D, Craig DS, El-Jawahri A, Hershman DL, Kong LR, Kurita GP, LeBlanc TW, Mercadante S, et al: Use of opioids for adults with pain from cancer or cancer treatment: ASCO guideline. J Clin Oncol. 41:914–930. 2023.PubMed/NCBI View Article : Google Scholar

3 

Fallon M, Giusti R, Aielli F, Hoskin P, Rolke R, Sharma M and Ripamonti CI: ESMO Guidelines Committee. Management of cancer pain in adult patients: ESMO clinical practice guidelines. Ann Oncol. 29 (Suppl 4):iv166–iv191. 2018.PubMed/NCBI View Article : Google Scholar

4 

Novy DM, Nelson DV, Koyyalagunta D, Cata JP, Gupta P and Gupta K: Pain, opioid therapy, and survival: A needed discussion. Pain. 161:496–501. 2020.PubMed/NCBI View Article : Google Scholar

5 

Satija A, Ahmed SM, Gupta R, Ahmed A, Rana SP, Singh SP, Mishra S and Bhatnagar S: Breast cancer pain management-A review of current & novel therapies. Indian J Med Res. 139:216–225. 2014.PubMed/NCBI

6 

Koulouris AI, Banim P and Hart AR: Pain in patients with pancreatic cancer: Prevalence, mechanisms, management and future developments. Dig Dis Sci. 62:861–870. 2017.PubMed/NCBI View Article : Google Scholar

7 

Montazeri A: Quality of life data as prognostic indicators of survival in cancer patients: an overview of the literature from 1982 to 2008. Health Qual Life Outcomes. 7(102)2009.PubMed/NCBI View Article : Google Scholar

8 

Mestdagh F, Steyaert A and Lavand'homme P: Cancer pain management: A narrative review of current concepts, strategies, and techniques. Curr Oncol. 30:6838–6858. 2023.PubMed/NCBI View Article : Google Scholar

9 

Bradley A and Boland JW: Effects of opioids on immune and endocrine function in patients with cancer pain. Curr Treat Options Oncol. 24:867–879. 2023.PubMed/NCBI View Article : Google Scholar

10 

Li Y, Sun L, Zhou Q, Lee AJ, Wang L, Zhang R and Wang S: Effects of opioid drugs on immune function in cancer patients. Biomed Pharmacother. 175(116665)2024.PubMed/NCBI View Article : Google Scholar

11 

Malafoglia V, Ilari S, Vitiello L, Tenti M, Balzani E, Muscoli C, Raffaeli W and Bonci A: The interplay between chronic pain, opioids, and the immune system. Neuroscientist. 28:613–627. 2022.PubMed/NCBI View Article : Google Scholar

12 

Plein LM and Rittner HL: Opioids and the immune system-friend or foe. Br J Pharmacol. 175:2717–2725. 2018.PubMed/NCBI View Article : Google Scholar

13 

Wang Y, Zhuang Y, DiBerto JF, Zhou XE, Schmitz GP, Yuan Q, Jain MK, Liu W, Melcher K, Jiang Y, et al: Structures of the entire human opioid receptor family. Cell. 186:413–427.e17. 2023.PubMed/NCBI View Article : Google Scholar

14 

Stein C: Opioid receptors. Annu Rev Med. 67:433–451. 2016.PubMed/NCBI View Article : Google Scholar

15 

Faouzi A, Varga BR and Majumdar S: Biased opioid ligands. Molecules. 25(4257)2020.PubMed/NCBI View Article : Google Scholar

16 

Wassum KM, Ostlund SB, Maidment NT and Balleine BW: Distinct opioid circuits determine the palatability and the desirability of rewarding events. Proc Natl Acad Sci USA. 106:12512–12517. 2009.PubMed/NCBI View Article : Google Scholar

17 

Pasternak GW and Pan YX: Mu Opioids and their receptors: Evolution of a concept. Pharmacol Rev. 65:1257–1317. 2013.PubMed/NCBI View Article : Google Scholar

18 

Streicher JM and Bilsky EJ: Peripherally acting µ-opioid receptor antagonists for the treatment of opioid-related side effects: Mechanism of action and clinical implications. J Pharm Pract. 31:658–669. 2018.PubMed/NCBI View Article : Google Scholar

19 

Zhou Q, Zhang Z, Long S, Li W, Wang B and Liang N: Opioids in cancer: The κopioid receptor (Review). Mol Med Rep. 25(44)2022.PubMed/NCBI View Article : Google Scholar

20 

Peppin JF and Raffa RB: Delta opioid agonists: A concise update on potential therapeutic applications. J Clin Pharm Ther. 40:155–166. 2015.PubMed/NCBI View Article : Google Scholar

21 

Dai ZH, Xu X, Chen WQ, Nie LN, Liu Y, Sui N and Liang J: The role of hippocampus in memory reactivation: An implication for a therapeutic target against opioid use disorder. Curr Addict Rep. 9:67–79. 2022.PubMed/NCBI View Article : Google Scholar

22 

Yamamizu K, Hamada Y and Narita M: κ Opioid receptor ligands regulate angiogenesis in development and in tumours. Br J Pharmacol. 172:268–276. 2015.PubMed/NCBI View Article : Google Scholar

23 

Jelic MD, Mandic AD, Maricic SM and Srdjenovic BU: Oxidative stress and its role in cancer. J Cancer Res Ther. 17:22–28. 2021.PubMed/NCBI View Article : Google Scholar

24 

Rullo L, Caputi FF, Losapio LM, Morosini C, Posa L, Canistro D, Vivarelli F, Romualdi P and Candeletti S: Effects of different opioid drugs on oxidative status and proteasome activity in SH-SY5Y cells. Molecules. 27(8321)2022.PubMed/NCBI View Article : Google Scholar

25 

de Almeida AS, Pereira GC, Brum EDS, Silva CR, Antoniazzi CTD, Ardisson-Araújo D, Oliveira SM and Trevisan G: Role of TRPA1 expressed in bone tissue and the antinociceptive effect of the TRPA1 antagonist repeated administration in a breast cancer pain model. Life Sci. 276(119469)2021.PubMed/NCBI View Article : Google Scholar

26 

Skrabalova J, Drastichova Z and Novotny J: Morphine as a potential oxidative stress-causing agent. Mini Rev Org Chem. 10:367–372. 2013.PubMed/NCBI View Article : Google Scholar

27 

Schattauer SS, Bedini A, Summers F, Reilly-Treat A, Andrews MM, Land BB and Chavkin C: Reactive oxygen species (ROS) generation is stimulated by κ opioid receptor activation through phosphorylated c-Jun N-terminal kinase and inhibited by p38 mitogen-activated protein kinase (MAPK) activation. J Biol Chem. 294:16884–16896. 2019.PubMed/NCBI View Article : Google Scholar

28 

Reuter S, Gupta SC, Chaturvedi MM and Aggarwal BB: Oxidative stress, inflammation, and cancer: How are they linked? Free Radic Biol Med. 49:1603–1616. 2010.PubMed/NCBI View Article : Google Scholar

29 

Waris G and Ahsan H: Reactive oxygen species: Role in the development of cancer and various chronic conditions. J Carcinog. 5(14)2006.PubMed/NCBI View Article : Google Scholar

30 

Boland JW and Pockley AG: Influence of opioids on immune function in patients with cancer pain: From bench to bedside. Br J Pharmacol. 175:2726–2736. 2018.PubMed/NCBI View Article : Google Scholar

31 

Mizota T, Tsujikawa H, Shoda T and Fukuda K: Dual modulation of the T-cell receptor-activated signal transduction pathway by morphine in human T lymphocytes. J Anesth. 27:80–87. 2013.PubMed/NCBI View Article : Google Scholar

32 

Gomez-Flores R and Weber RJ: Inhibition of interleukin-2 production and downregulation of IL-2 and transferrin receptors on rat splenic lymphocytes following PAG morphine administration: A role in natural killer and T cell suppression. J Interferon Cytokine Res. 19:625–630. 1999.PubMed/NCBI View Article : Google Scholar

33 

Chen C, Farooqui M and Gupta K: Morphine stimulates vascular endothelial growth factor-like signaling in mouse retinal endothelial cells. Curr Neurovasc Res. 3:171–180. 2006.PubMed/NCBI View Article : Google Scholar

34 

Eisenstein TK: The role of opioid receptors in immune system function. Front Immunol. 10(2904)2019.PubMed/NCBI View Article : Google Scholar

35 

Tuerxun H and Cui J: The dual effect of morphine on tumor development. Clin Transl Oncol. 21:695–701. 2019.PubMed/NCBI View Article : Google Scholar

36 

Zhang XY, Liang YX, Yan Y, Dai Z and Chu HC: Morphine: Double-faced roles in the regulation of tumor development. Clin Transl Oncol. 20:808–814. 2018.PubMed/NCBI View Article : Google Scholar

37 

Zhang H, Zhou D, Gu J, Qu M, Guo K, Chen W and Miao C: Targeting the mu-Opioid receptor for cancer treatment. Curr Oncol Rep. 23(111)2021.PubMed/NCBI View Article : Google Scholar

38 

McDonald J and Lambert DG: Opioid receptors. BJA Education. 15:219–224. 2015.

39 

Bimonte S, Barbieri A, Rea D, Palma G, Luciano A, Cuomo A, Arra C and Izzo F: Morphine promotes tumor angiogenesis and increases breast cancer progression. Biomed Res Int. 2015(161508)2015.PubMed/NCBI View Article : Google Scholar

40 

Malik JA, Affan Khan M, Lamba T, Adeel Zafar M, Nanda S, Owais M and Agrewala JN: Immunosuppressive effects of morphine on macrophage polarization and function. Eur J Pharmacol. 975(176637)2024.PubMed/NCBI View Article : Google Scholar

41 

Tariq M, Zhang J, Liang G, Ding L, He Q and Yang B: Macrophage polarization: Anti-cancer strategies to target tumor-associated macrophage in breast cancer. J Cell Biochem. 118:2484–2501. 2017.PubMed/NCBI View Article : Google Scholar

42 

Gach K, Szemraj J, Wyrębska A and Janecka A: The influence of opioids on matrix metalloproteinase-2 and -9 secretion and mRNA levels in MCF-7 breast cancer cell line. Mol Biol Rep. 38:1231–1236. 2011.PubMed/NCBI View Article : Google Scholar

43 

Malsy M, Hackl C, Graf B, Bitzinger D and Bundscherer A: The effects of analgesics on the migration of pancreatic cancer cells. In Vivo. 36:576–581. 2022.PubMed/NCBI View Article : Google Scholar

44 

O'Sullivan S, Medina C, Ledwidge M, Radomski MW and Gilmer JF: Nitric oxide-matrix metaloproteinase-9 interactions: Biological and pharmacological significance: NO and MMP-9 interactions. Biochim Biophys Acta. 1843:603–617. 2014.PubMed/NCBI View Article : Google Scholar

45 

Nikitovic D, Corsini E, Kouretas D, Tsatsakis A and Tzanakakis G: ROS-major mediators of extracellular matrix remodeling during tumor progression. Food Chem Toxicol. 61:178–186. 2013.PubMed/NCBI View Article : Google Scholar

46 

Huang H: Matrix metalloproteinase-9 (MMP-9) as a cancer biomarker and MMP-9 biosensors: Recent advances. Sensors (Basel). 18(3249)2018.PubMed/NCBI View Article : Google Scholar

47 

Mehner C, Hockla A, Miller E, Ran S, Radisky DC and Radisky ES: Tumor cell-produced matrix metalloproteinase 9 (MMP-9) drives malignant progression and metastasis of basal-like triple negative breast cancer. Oncotarget. 5:2736–2749. 2014.PubMed/NCBI View Article : Google Scholar

48 

Ning J, Chen X, Li Q, Yang D, Xie C, Qin S and Jiang H: Bidirectional effects of morphine on pancreatic cancer progression via the p38/JNK pathway. Sci Rep. 14(24233)2024.PubMed/NCBI View Article : Google Scholar

49 

Ma Y, Ren Z, Ma S, Yan W, He M, Wang D and Ding P: Morphine enhances renal cell carcinoma aggressiveness through promotes survivin level. Ren Fail. 39:258–264. 2016.PubMed/NCBI View Article : Google Scholar

50 

Jiang Y, Li T, Qian Y, Zuo X and Liu J: Morphine in combination with ketamine improves cervical cancer pain and suppresses immune function via the JAK3/STAT5 pathway. Pain Res Manag. 2022(9364365)2022.PubMed/NCBI View Article : Google Scholar

51 

Liu X, Yang J, Yang C, Huang X, Han M, Kang F and Li J: Morphine promotes the malignant biological behavior of non-small cell lung cancer cells through the MOR/Src/mTOR pathway. Cancer Cell Int. 21(622)2021.PubMed/NCBI View Article : Google Scholar

52 

Koodie L, Ramakrishnan S and Roy S: Morphine suppresses tumor angiogenesis through a HIF-1α/p38MAPK pathway. Am J Pathol. 177:984–997. 2010.PubMed/NCBI View Article : Google Scholar

53 

Tripolt S, Neubauer HA, Knab VM, Elmer DP, Aberger F, Moriggl R and Fux DA: Opioids drive breast cancer metastasis through the δ-opioid receptor and oncogenic STAT3. Neoplasia. 23:270–279. 2021.PubMed/NCBI View Article : Google Scholar

54 

Wei YC, Zhang B, Li X, Liu XM, Zhang J, Lei B, Li B, Zhai R, Chen Q and Li Y: Upregulation and activation of δopioid receptors promotes the progression of human breast cancer. Oncol Rep. 36:2579–2586. 2016.PubMed/NCBI View Article : Google Scholar

55 

Wang S: Historical review: Opiate addiction and opioid receptors. Cell Transplant. 28:233–238. 2019.PubMed/NCBI View Article : Google Scholar

56 

Leconte C, Mongeau R and Noble F: Traumatic stress-induced vulnerability to addiction: Critical role of the dynorphin/kappa opioid receptor system. Front Pharmacol. 13(856672)2022.PubMed/NCBI View Article : Google Scholar

57 

Li H, Ma Z and Lei Y: The expression of kappa-opioid receptor promotes the migration of breast cancer cells in vitro. BMC Anesthesiol. 21(210)2021.PubMed/NCBI View Article : Google Scholar

58 

Eisenstein TK: Opioids and the immune system: What is their mechanism of action? Br J Pharmacol. 164:1826–1828. 2011.PubMed/NCBI View Article : Google Scholar

59 

Wang Q, Liu Z, Tang S and Wu Z: Morphine suppresses the immune function of lung cancer by up-regulating MAEL expression. BMC Pharmacol Toxicol. 23(92)2022.PubMed/NCBI View Article : Google Scholar

60 

Wang J, Barke RA, Charboneau R, Schwendener R and Roy S: Morphine induces defects in early response of alveolar macrophages to streptococcus pneumoniae by modulating TLR9-NF-kappa B signaling1. J Immunol. 180:3594–3600. 2008.PubMed/NCBI View Article : Google Scholar

61 

Wang J, Ma J, Charboneau R, Barke R and Roy S: morphine inhibits murine dendritic cell IL-23 production by modulating toll-like receptor 2 and Nod2 signaling. J Biol Chem. 286:10225–10232. 2011.PubMed/NCBI View Article : Google Scholar

62 

Xie F, Kitagawa Y, Ogata H, Yasuhara S, You Z and Jeevendra Martyn JA: Morphine induces inflammatory responses via both TLR4 and cGAS-STING signaling pathways. Cytokine. 183(156737)2024.PubMed/NCBI View Article : Google Scholar

63 

Malik JA and Agrewala JN: Morphine acts via TLR4 resulting in neuroinflammation and immunosuppression. Med Hypotheses. 186(111335)2024.

64 

Singhal PC, Sharma P, Kapasi AA, Reddy K, Franki N and Gibbons N: Morphine enhances macrophage apoptosis. J Immunol. 160:1886–1893. 1998.PubMed/NCBI

65 

Lin M, Deng K, Li Y and Wan J: Morphine enhances LPSinduced macrophage apoptosis through a PPARγdependent mechanism. Exp Ther Med. 22(714)2021.PubMed/NCBI View Article : Google Scholar

66 

Saurer TB, Carrigan KA, Ijames SG and Lysle DT: Suppression of natural killer cell activity by morphine is mediated by the nucleus accumbens shell. J Neuroimmunol. 173:3–11. 2006.PubMed/NCBI View Article : Google Scholar

67 

Saeed I, La Caze A, Hollmann MW, Shaw PN and Parat MO: New Insights on Tramadol and Immunomodulation. Curr Oncol Rep. 23(123)2021.PubMed/NCBI View Article : Google Scholar

68 

Maher DP, Walia D and Heller NM: Suppression of human natural killer cells by different classes of opioids. Anesth Analg. 128:1013–1021. 2019.PubMed/NCBI View Article : Google Scholar

69 

Khabbazi S, Goumon Y and Parat MO: Morphine modulates interleukin-4- or breast cancer cell-induced pro-metastatic activation of macrophages. Sci Rep. 5(11389)2015.PubMed/NCBI View Article : Google Scholar

70 

Yamamizu K, Furuta S, Hamada Y, Yamashita A, Kuzumaki N and Narita M, Doi K, Katayama S, Nagase H, Yamashita JK and Narita M: к Opioids inhibit tumor angiogenesis by suppressing VEGF signaling. Sci Rep. 3(3213)2013.PubMed/NCBI View Article : Google Scholar

71 

Cho JS, Lee MH, Kim SI, Park S, Park HS, Oh E, Lee JH and Koo BN: The effects of perioperative anesthesia and analgesia on immune function in patients undergoing breast cancer resection: A prospective randomized study. Int J Med Sci. 14:970–976. 2017.PubMed/NCBI View Article : Google Scholar

72 

Shi Y, Zhang Y, Yu S, Luo J, Pan Z, Wang X and Tian J: Activation of kappa opioid receptor (KOR) inhibits estrogen receptor (ER)-positive breast cancer through the KOR-ER-XBP1 pathway. Biomed Pharmacother. 167(115462)2023.PubMed/NCBI View Article : Google Scholar

73 

Zhang HW, Wang F, Zhou YQ, Xu SP, Yu SY and Zhang ZG: Morphine suppresses liver cancer cell tumor properties in vitro and in vivo. Front Oncol. 11(666446)2021.PubMed/NCBI View Article : Google Scholar

74 

Wang L, Guo W, Guan H, Yan N, Cai X and Zhu L: Tramadol suppresses growth of orthotopic liver tumors via promoting M1 macrophage polarization in the tumor microenvironment. Naunyn-Schmiedebergs Arch Pharmacol. 397:4205–4218. 2024.PubMed/NCBI View Article : Google Scholar

75 

Schwenk ES and Mariano ER: Designing the ideal perioperative pain management plan starts with multimodal analgesia. Korean J Anesthesiol. 71:345–352. 2018.PubMed/NCBI View Article : Google Scholar

76 

Morgan EL: Regulation of human B lymphocyte activation by opioid peptide hormones. Inhibition of IgG production by opioid receptor class (µ-, kappa-, and delta-) selective agonists. J Neuroimmunol. 65:21–30. 1996.PubMed/NCBI View Article : Google Scholar

77 

Börner C, Warnick B, Smida M, Hartig R, Lindquist JA, Schraven B, Höllt V and Kraus J: Mechanisms of opioid-mediated inhibition of human T cell receptor signaling. J Immunol. 183:882–889. 2009.PubMed/NCBI View Article : Google Scholar

78 

Beilin B, Shavit Y, Trabekin E, Mordashev B, Mayburd E, Zeidel A and Bessler H: The effects of postoperative pain management on immune response to surgery. Anesth Analg. 93:822–827. 2003.PubMed/NCBI View Article : Google Scholar

79 

Dahan A, van Lemmen M, Jansen S, Simons P and van der Schrier R: Buprenorphine: A treatment and cause of opioid-induced respiratory depression. Br J Anaesth. 128:402–404. 2022.PubMed/NCBI View Article : Google Scholar

80 

Kiyatkin EA: Respiratory depression and brain hypoxia induced by opioid drugs: Morphine, oxycodone, heroin, and fentanyl. Neuropharmacology. 151:219–226. 2019.PubMed/NCBI View Article : Google Scholar

81 

Abdel Shaheed C, Beardsley J, Day RO and McLachlan AJ: Immunomodulatory effects of pharmaceutical opioids and antipyretic analgesics: Mechanisms and relevance to infection. Br J Clin Pharmacol. 88:3114–3131. 2022.PubMed/NCBI View Article : Google Scholar

82 

Saunders KW, Von Korff M, Campbell CI, Banta-Green CJ, Sullivan MD, Merrill JO and Weisner C: Concurrent use of alcohol and sedatives among persons prescribed chronic opioid therapy: Prevalence and risk factors. J Pain. 13:266–275. 2012.PubMed/NCBI View Article : Google Scholar

83 

Leppert W: Pain management in patients with cancer: Focus on opioid analgesics. Curr Pain Headache Rep. 15:271–279. 2011.PubMed/NCBI View Article : Google Scholar

84 

Boon M, van Dorp E, Broens S and Overdyk F: Combining opioids and benzodiazepines: Effects on mortality and severe adverse respiratory events. Ann Palliat Med. 9:542–557. 2020.PubMed/NCBI View Article : Google Scholar

85 

Chen A and Ashburn MA: Cardiac effects of opioid therapy. Pain Med. 16 (Suppl 1):S27–S31. 2015.PubMed/NCBI View Article : Google Scholar

86 

Li Y, Wang M, Peng X, Yang Y, Chen Q, Liu J, She Q, Tan J, Lou C, Liao Z and Li X: mRNA vaccine in cancer therapy: Current advance and future outlook. Clin Transl Med. 13(e1384)2023.PubMed/NCBI View Article : Google Scholar

87 

Szeto GL and Finley SD: Integrative approaches to cancer immunotherapy. Trends Cancer. 5:400–410. 2019.PubMed/NCBI View Article : Google Scholar

88 

Wang Z and Cao YJ: Adoptive cell therapy targeting neoantigens: A frontier for cancer research. Front Immunol. 11(176)2020.PubMed/NCBI View Article : Google Scholar

89 

Topalian SL, Taube JM and Pardoll DM: Neoadjuvant checkpoint blockade for cancer immunotherapy. Science. 367(eaax0182)2020.PubMed/NCBI View Article : Google Scholar

90 

Wang SJ, Dougan SK and Dougan M: Immune mechanisms of toxicity from checkpoint inhibitors. Trends Cancer. 9:543–553. 2023.PubMed/NCBI View Article : Google Scholar

91 

Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z and Cheng Q: Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res. 40(184)2021.PubMed/NCBI View Article : Google Scholar

92 

Waldman AD, Fritz JM and Lenardo MJ: A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 20:651–668. 2020.PubMed/NCBI View Article : Google Scholar

93 

Shiravand Y, Khodadadi F, Kashani SMA, Hosseini-Fard SR, Hosseini S, Sadeghirad H, Ladwa R, O'Byrne K and Kulasinghe A: Immune checkpoint inhibitors in cancer therapy. Curr Oncol. 29:3044–3060. 2022.PubMed/NCBI View Article : Google Scholar

94 

Suresh K, Naidoo J, Lin CT and Danoff S: Immune checkpoint immunotherapy for non-small cell lung cancer: Benefits and pulmonary toxicities. Chest. 154:1416–1423. 2018.PubMed/NCBI View Article : Google Scholar

95 

Acharya C, Betrapally NS, Gillevet PM, Sterling RK, Akbarali H, White MB, Ganapathy D, Fagan A, Sikaroodi M and Bajaj JS: Chronic opioid use is associated with altered gut microbiota and predicts readmissions in patients with cirrhosis. Aliment Pharmacol Ther. 45:319–331. 2017.PubMed/NCBI View Article : Google Scholar

96 

Sacerdote P, Bianchi M, Gaspani L, Manfredi B, Maucione A, Terno G, Ammatuna M and Panerai AE: The effects of tramadol and morphine on immune responses and pain after surgery in cancer patients. Anesth Analg. 90:1411–1414. 2000.PubMed/NCBI View Article : Google Scholar

97 

Swarm RA, Paice JA, Anghelescu DL, Are M, Bruce JY, Buga S, Chwistek M, Cleeland C, Craig D, Gafford E, et al: Adult cancer pain, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 17:977–1077. 2019.PubMed/NCBI View Article : Google Scholar

98 

Mercadante S: Opioid analgesics adverse effects: The other side of the coin. Curr Pharm Des. 25:3197–3202. 2019.PubMed/NCBI View Article : Google Scholar

99 

Weir A: Opioid-free anaesthesia. Br J Hosp Med (Lond). 85:1–2. 2024.PubMed/NCBI View Article : Google Scholar

100 

Chen SH, Chen SS, Wang YP and Chen LK: Effects of systemic and neuraxial morphine on the immune system. Medicine (Baltimore). 98(e15375)2019.PubMed/NCBI View Article : Google Scholar

101 

Wang J, Barke RA and Roy S: Transcriptional and epigenetic regulation of interleukin-2 gene in activated T cells by morphine. J Biol Chem. 282:7164–7171. 2007.PubMed/NCBI View Article : Google Scholar

102 

Amodeo G, Bugada D, Franchi S, Moschetti G, Grimaldi S, Panerai A, Allegri M and Sacerdote P: Immune function after major surgical interventions: The effect of postoperative pain treatment. J Pain Res. 11:1297–1305. 2018.PubMed/NCBI View Article : Google Scholar

103 

Wang Y, Wang X, Ye L, Li J, Song L, Fulambarkar N and Ho W: Morphine suppresses IFN signaling pathway and enhances AIDS virus infection. PLoS One. 7(e31167)2012.PubMed/NCBI View Article : Google Scholar

104 

Vassou D, Bakogeorgou E, Kampa M, Dimitriou H, Hatzoglou A and Castanas E: Opioids modulate constitutive B-lymphocyte secretion. Int Immunopharmacol. 8:634–644. 2008.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2025
Volume 22 Issue 6

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou Y, Li W, Chen Y, Hu X and Miao C: Research progress on the impact of opioids on the tumor immune microenvironment (Review). Mol Clin Oncol 22: 53, 2025.
APA
Zhou, Y., Li, W., Chen, Y., Hu, X., & Miao, C. (2025). Research progress on the impact of opioids on the tumor immune microenvironment (Review). Molecular and Clinical Oncology, 22, 53. https://doi.org/10.3892/mco.2025.2848
MLA
Zhou, Y., Li, W., Chen, Y., Hu, X., Miao, C."Research progress on the impact of opioids on the tumor immune microenvironment (Review)". Molecular and Clinical Oncology 22.6 (2025): 53.
Chicago
Zhou, Y., Li, W., Chen, Y., Hu, X., Miao, C."Research progress on the impact of opioids on the tumor immune microenvironment (Review)". Molecular and Clinical Oncology 22, no. 6 (2025): 53. https://doi.org/10.3892/mco.2025.2848