Skip to main content
Erschienen in: Clinical and Translational Oncology 6/2019

Open Access 23.11.2018 | Review Article

The dual effect of morphine on tumor development

verfasst von: H. Tuerxun, J. Cui

Erschienen in: Clinical and Translational Oncology | Ausgabe 6/2019

Abstract

Morphine is a classic opioid drug used for reducing pain and is commonly prescribed as an effective drug to control cancer pain. Morphine has a direct role in the central nervous system to relieve pain, but because of its peripheral functions, morphine also has some side effects, such as nausea, constipation, and addiction (Gupta et al. in Sci World J 2015:10, 2015). In addition to its analgesic effect, the role of morphine in tumor development is an important question that has been investigated for many years with conflicting results. Numerous studies suggest that morphine has a role in both promoting and inhibiting tumor growth. In this extensive review, we attempt to comprehensively understand the effects of morphine and summarize both its positive and negative influences on various aspects of tumors, including tumor growth, angiogenesis, metastasis, inflammation, and immunomodulation.

Introduction

One of the most common symptoms of cancer is pain, with 50–80% of cancer patients experiencing some degree of pain. As tumors develop, they can cause severe pain by invading the surrounding nervous tissue, organs, and bones. This can cause a series of pathophysiological changes in the body, including changes in immune function.
Studies suggest [2] that pain can increase the release of corticotrophin-releasing hormone (CRH), resulting in immunosuppression. CRH acts on the neuroendocrine system, which leads to the redistribution of lymphocytes from circulating blood to lymphatic tissue. This causes lymphocytes to respond less to mitogens. Furthermore, immune cells synthesize and release β-endorphin, an endogenous opioid peptide that is an important immunomodulator and can rapidly decrease CD3 + , CD4 + , and CD4 +/CD8 + expression in peripheral blood, thereby leading to immunosuppression. Neuropeptides and other factors produced by the body as a result of cancer pain can inhibit the production and release of Il-2, which also leads to immune hypofunction.
Morphine is an analgesic for cancer pain which improves the quality of life of patients. Besides its analgesic effect, the clinical utility of morphine is limited by several adverse effects, including addiction, tolerance, immunosuppression, and constipation [3]. However, morphine is still regarded as the most effective medicine for patients with severe pain. How morphine influences tumor growth has been debated for years. Some studies indicate that morphine promotes tumor progression, while others suggest that morphine inhibits tumor progression.
This review attempts to clarify the dual function of morphine on tumor progression, including growth, angiogenesis, metastasis, inflammation, and immunomodulation (Fig. 1).

Effect of morphine on tumor cell growth

Many studies have investigated how morphine influences tumor cell growth. Some studies [4] reported that when morphine is administered at a concentration greater than 10 μM, tumor cell proliferation is inhibited. One study found that high concentrations of morphine reduced MCF-7 cell growth in nude mice [4]. A similar result was obtained when morphine treatment prevented neuroblastoma SH-SY5Y cells from differentiating [5]. Although many studies have shown that high concentrations of morphine can inhibit the proliferation of tumors, the mechanism remains unclear. The involvement of opioid receptors remains debatable. It was shown that opioids inhibit T47D cell growth, the mechanism of which was mediated through κ- and δ-opioid receptors [6]. Another aspect of morphine which may contribute to its antigrowth effect is via apoptosis promotion. Morphine activates c-JunN-terminal kinase, which generates reactive oxygen species (ROS), inducing the release of cytochrome c and caspase-9/3 by enhancing the pro-apoptotic protein Bim and reducing the anti-apoptotic protein Bcl-2 [7]. In addition to this mitochondrial pathway, morphine can also promote Fas-mediated apoptosis [8]. Apoptosis of MCF-7 cells was shown to be mediated by a novel δ-2 receptor, p53, and caspase-independent pathway [9]. Activation of the κ-opioid receptor (KOR) promotes apoptosis via a phospholipase C pathway in the CNE2 human epithelial tumor cell line [10]. There are obvious differences in the number of necrotic cells induced by morphine between different cell lines. For example, the number of necrotic cells in an MCF-7 cell line is higher than that in HL-60 and A549 cell lines [11]. This difference is largely dependent on how the cell lines react to morphine.
Despite this, there is also accumulating evidence for the growth-promoting effect of morphine. Sergeeva et al. [12] reported that morphine activates the proliferation of myeloid K562 and T-lymphoma Yurkat cells. Sabrina et al. [13] reported that clinically relevant concentrations of morphine can increase breast cancer progression. Recently, it was reported that morphine can inhibit cisplatin-induced apoptosis [14]. Some studies have confirmed that taking low doses of morphine can increase the growth of the tumor cell; however, the mechanism is still unclear. Some researchers have indicated that the μ-opioid receptor (MOR) may have a significant role in this mechanism. Mathew et al. [15] showed that compared to wild-type controls, μ-opioid receptor-knockout (MORKO) mice did not have significant tumor growth when injected with Lewis lung cancer cells. Further, after infusion of methylnaltrexone, a MOR antagonist, wild-type mice treated with Lewis lung cancer cells had a significant reduction, up to 90%, in tumor growth [15]. It has been pointed out that morphine can stimulate the mitogen-activated protein kinase (MAPK) or Erk pathways to regulate cell cycle progression by binding to the MOR [16]. Morphine can also stimulate the degranulation of mast cells to promote the release of neuropeptide substance P (SP) [17, 18], which has been shown to enhance the proliferation of tumor cells through the tachykinin 1 (NK-1) receptor [19]. The anti-apoptosis effect of morphine is involved. As naloxone cannot reverse this effect, a non-opioid receptor-mediated signaling pathway is implicated. Further reports suggest that morphine inhibits the generation of ROS and prevents the DOX-mediated activation of caspase-3, release of cytochrome c, and changes in Bax and Bcl-2 protein expression [20]. In addition, several researchers have focused their research efforts on Survivin, a member of the inhibitor of apoptosis family. It was shown that morphine can enhance renal cell carcinoma (RCC) growth by promoting the expression of Survivin [21].

Effect of morphine on angiogenesis

The formation of new blood vessels is an essential part of tumor development. Because morphine is one of the most commonly used drugs for treating cancer pain, its effect on angiogenesis has attracted great attention from researchers. When a solid tumor grows, newly proliferating tumor cells are localized far away from their vascular supply. This low oxygen or hypoxic environmental stimulates tumor cells to secrete vascular endothelial growth factor (VEGF) [22], which promotes the formation of new blood vessels to sustain the tumor growth [23]. Balasubramanian et al. [24] reported that in rat cardiomyocytes and human umbilical vein endothelial cells, morphine inhibits the secretion of VEGF induced by hypoxia. Koodie et al. [25] observed that when morphine is administered at clinically relevant analgesic doses by continuous slow release implantation, tumor cell-induced angiogenesis is reduced. They also confirmed that this inhibitory effect was mediated by hypoxia-induced mitochondrial p38 MAPK pathway inhibition [25]. Others [26] have argued that morphine also inhibits angiogenesis by suppressing VEGF signaling via the KOR, as KOR knockout mice grafted with Lewis lung carcinoma or B16 melanoma had more proliferation and enhanced angiogenesis compared to wild type mice [26]. In addition to its direct effect, morphine can also suppress the migration of leukocytes and reduce angiogenesis [27].
Morphine not only inhibits angiogenesis, but also promotes the formation of new blood vessels. It was shown by Bimonte [13] that clinically relevant concentrations of morphine stimulates tumor angiogenesis. Singleton and Moss [28] found that morphine can transactivate the VEGF receptor and stimulate angiogenesis and further, angiogenesis was blocked after the application of a MOR antagonist [28]. The transactivation of the VEGF receptor by morphine may be mediated by adhesion molecules, such as ICAM-1 [29]. Another implicated pathway is the stimulation of the MAPK signaling pathway through G protein-coupled receptors and nitric oxide (NO). Chronic morphine therapy can increase the levels of nitric oxide synthase (NOS), NO, and cyclooxygenase-2 (COX-2) in mouse kidneys [30]. Similarly, 2 weeks of chronic morphine therapy stimulated COX-2, prostaglandin E2, and angiogenesis, which was accompanied by increased tumor weight and metastasis and reduced survival [31]. It could thus be postulated that morphine can upregulate COX-2, increase prostaglandin E2, and promote angiogenesis.

Effect of morphine on tumor invasion and metastasis

The most common reason why a patient fails to respond to cancer treatment is not because of the original tumor itself, but instead, because of metastasis from the primary one. The activation of urokinase plasminogen activator (uPA) and matrix metalloproteinases (MMPs) play an important role in the degradation of the extracellular matrix (ECM), which is regarded as an indispensable step in tumor migration [32, 33]. MMP is a type of endopeptidase which can remodel the constituents of the ECM [32]. It was reported that morphine reduces the migration and invasion of breast cancer cells by inhibiting MMPs [34]. The findings from Katarzyna et al. [35] indicate that morphine exerts its inhibitory effect on MMP-2 and MMP-9 by inhibiting the NO/NOS system. Adhesion is also important for metastasis. Min et al. [36] clarified that morphine can weaken endothelial cell adhesion molecules induced by LPS-stimulated colon cancer cells and reduce metastasis.
However, conflicting data on the effects of morphine on tumor metastasis were simultaneously published. As mentioned before, uPA plays a crucial role in the degradation of the ECM [33]. Gach et al. [37] reported that in MCF-7 breast cancer cells, morphine significantly increases the secretion of uPA. Similarly, Nylund et al. [38] showed that in HT-29 colon cancer cells, morphine induces the secretion of uPA. Naloxone can reverse this morphine-induced upregulation, proving that opioid receptors were involved in the process. Recently, it has been found that over-expressing Survivin can promote tumors to metastasize and increase genomic instability, thereby promoting tumor invasion and metastasis [39, 40]. It was also shown that morphine can enhance RCC growth by promoting Survivin expression [39, 40].

Effect of morphine on inflammation

The inflammatory response plays a crucial role in the different stages of tumor development and mainly affects the tumor through two direct pathways: (1) adjusting the antitumor immunity of the host and (2) creating a pro-tumorigenic microenvironment. Boettger et al. [41] reported that morphine can reduce the inflammatory response in the chronic antigen-induced arthritis model. Further research showed that morphine can modulate inflammatory cytokines to inhibit inflammation [42]. In addition, morphine can induce the activation of KOR to exert its anti-inflammatory response [43]. As such, morphine can prevent inflammation, inhibit tumor growth, and ameliorate addiction.
In contrast to its inhibiting effects on inflammation, morphine can also stimulate the degranulation of mast cells to promote the release of the neuropeptide SP [17, 18]. This then aggravates the inflammatory response, helps the tumor escape from the immune surveillance, induces addiction, and increases the infection rate.

Effect of morphine on the immune system

A number of mechanisms for the effects of morphine on immunity have been described. Morphine regulates immune function by directly acting on immune cells and via indirect actions, which include interacting with the central nervous system (CNS) and promoting its release of immune mediators (Table 1).
Table 1
Morphine modules immune function by direct and indirect actions
 
The effects of morphine
Immune cells
 Innate cells
  Macrophages
Reduce the proliferation of macrophage progenitor cells
Reduce recruitment
Reduce Fcg R mediated phagocytosis
Reduce the release of NO
  NK cells
Suppress the activity of NK cells
 Adaptive cells
  B cells
Inhibit the function of macrophages and polymorphonuclear leukocytes
  T cells
Interfere with transcriptional activation of the IL-2 gene
Inhibit IFN-γ
CNS
 Acute administration
  PAG
Suppress NK cell cytotoxicity and lymphocyte proliferaiton
  NAS
Inhibit NK cell cytotoxicity
 Chronic administration
  HPA axis
Decrease NK cell cytotoxicity

Direct effects

Morphine induces immune suppression by directly regulating adaptive and innate cells, including macrophages, natural killer (NK) cells, B cells, and T cells.
For morphine to exert its direct effects on an immune cell, either the immune cell must express opioid receptors or morphine must be capable of working through non-opioid receptors [44].
Macrophages/phagocytic cells, together with NK cells, constitute the major part of innate immunity. Morphine reduces the number of macrophages that can be used to respond to infection by reducing the proliferation of macrophage progenitor cells [45] and inhibiting the recruitment of these cells [46]. In addition, macrophage phagocytosis can be inhibited [47] and macrophage bactericidal activity can be disrupted by reducing the release of NO [48]. Further studies have shown that deletion of the MOR gene completely eliminates the inhibitory effect of morphine on phagocytosis and bactericidal activity. Morphine substantially decreases the activity of NK cells. It is thought that morphine suppresses the activity of NK cells as a result of the activation of opioid receptors in the CNS [49].
Similar to its effect on the innate immune system, prolonged morphine treatment attenuates the adaptive immune response. T lymphocytes are the main cells involved in cellular immunity. They also regulate the activity of other cells through neuroendocrine mechanisms or cytokines. Chronic morphine treatment results in a decrease in cell viability, proliferative responses, T-helper cell function, and CD4/CD8 cell populations [50]. In addition, morphine has been shown to significantly reduce the production of IL-2, TNF-α, and IFN-γ in mouse spleen cell cultures [51]. Mechanistically, morphine can impair the proliferation of lymphocytes by interfering with the transcriptional activation of the IL-2 gene [52], as well as interfering with the activity of the IFN-γ promoter through two distinct cAMP-dependent pathways [50]. These effects are absent in MORKO mice, further demonstrating that the activation of MOR can lead to immunosuppressive effects. Contrary to T cell research, the effect of morphine on B-cell function is limited. B lymphocytes are involved in humoral immunity mainly by producing antibodies and memory cells. Morphine inhibits the function of macrophages and polymorphonuclear leukocytes via the neural immune circuit, thereby indirectly regulating B cell function [53].

Indirect effects

The indirect action of morphine on the immune system via the CNS is also supported by conflicting evidence [54]. Acute morphine administration releases biological amines into the periaqueductal gray to suppress NK cell cytotoxicity and lymphocyte proliferation [5456]. Additionally, morphine releases neuropeptide γ via D1 receptors into the nucleus accumbens shell to inhibit splenic NK cell cytotoxicity [57]. Chronic morphine administration increases the activity of the hypothalamic–pituitary–adrenal axis and the release of immunosuppressive glucocorticoids, both of which decrease the cytotoxicity of NK cells [58, 59].

Conclusions and perspectives

Current research shows that morphine plays a dual role in the regulation of tumors, including its effect on tumor growth, angiogenesis, metastasis, inflammation, and immunity. However, the mechanisms remain unclear. The expression of MOR in tumors may be the key to these mechanisms. Morphine has a role in tumor growth and metastasis by modulating apoptosis and VEGF signaling. Thus, opioid antagonists might become novel therapeutic options for tumor treatment.
The major factors responsible for the dual role of morphine in tumors is the dose of morphine and type of tumor. Generally speaking, at high concentrations, morphine inhibits tumor cell growth, angiogenesis, invasion, and metastasis. However, low daily doses of morphine stimulates tumor cell proliferation, angiogenesis, and immunosuppression. The mechanism of the concentration-dependent effect of morphine is not clear. In addition, the effect of morphine is dependent on the type of cancer, since different cancer cells overexpress different opioid receptors.
In vitro and animal studies are principally used to explore these mechanisms, because the environment can be carefully controlled. However, the findings from these studies cannot be completely extrapolated to humans, as the internal environment of the human body is complex. As a result, further research is necessary to fully understand the effects of morphine on the development of tumors in a human physiological environment. A small number of studies have found that greater morphine requirement are associated with shorter progression-free and shorter survival rate [60, 61]. Thus, further studies are needed to determine how to utilize the advantages of morphine and minimize the negative effects. Until there is more reliable evidence, it is currently necessary to use the adequate dose of morphine needed to relieve pain in cancer patients.

Compliance with ethical standards

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

This article does not contain any studies with human participants performed by any of the authors.
No informed consent is required.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Gupta M, Msambichaka L, Ballas SK, Gupta K. Morphine for the treatment of pain in sickle cell disease. Sci World J. 2015;2015:10.CrossRef Gupta M, Msambichaka L, Ballas SK, Gupta K. Morphine for the treatment of pain in sickle cell disease. Sci World J. 2015;2015:10.CrossRef
2.
Zurück zum Zitat Shen LD, Yang J, Li YX, et al. Correlation between senile cancer pain and depression and its effect on immune function[J]. J Kunming Med Coll. 2011;2(3):31–4. Shen LD, Yang J, Li YX, et al. Correlation between senile cancer pain and depression and its effect on immune function[J]. J Kunming Med Coll. 2011;2(3):31–4.
3.
Zurück zum Zitat Grace Peter M, Maier Steven F, Watkins Linda R. Opioid-induced central immune signaling: implications for opioid analgesia. Headache. 2015;55(4):475–89.CrossRef Grace Peter M, Maier Steven F, Watkins Linda R. Opioid-induced central immune signaling: implications for opioid analgesia. Headache. 2015;55(4):475–89.CrossRef
4.
Zurück zum Zitat Tegeder I, Grosch S, Schmidtko A, Haussler A, Schmidt H, Niederberger E, Scholich K, Geisslinger G. G protein-independent G1 cell cycle block and apoptosis with morphine in adenocarcinoma cell:involvement of p53 phosphorylation. Cancer Res. 2003;63(8):1846–52.PubMed Tegeder I, Grosch S, Schmidtko A, Haussler A, Schmidt H, Niederberger E, Scholich K, Geisslinger G. G protein-independent G1 cell cycle block and apoptosis with morphine in adenocarcinoma cell:involvement of p53 phosphorylation. Cancer Res. 2003;63(8):1846–52.PubMed
5.
Zurück zum Zitat Gonzalez-Nunez Veronica, Noriega-Prieto Jose Antonio, Rodríguez Raquel E. Morphine modulates cell proliferation through mir133b & mir128 in the neuroblastoma SH-SY5Y cell line. Biochim Biophys Acta Mol Basis Dis. 2014;1842(4):566–72.CrossRef Gonzalez-Nunez Veronica, Noriega-Prieto Jose Antonio, Rodríguez Raquel E. Morphine modulates cell proliferation through mir133b & mir128 in the neuroblastoma SH-SY5Y cell line. Biochim Biophys Acta Mol Basis Dis. 2014;1842(4):566–72.CrossRef
6.
Zurück zum Zitat Hatzoglou A, Bakogeorgou E, Castanas E. The antiproliferative effect of opioid receptor agonists on the T47D human breast cancer cell line, is partially mediated through opioid receptors. Eur J Pharmacol. 1996;296(2):199–207.CrossRef Hatzoglou A, Bakogeorgou E, Castanas E. The antiproliferative effect of opioid receptor agonists on the T47D human breast cancer cell line, is partially mediated through opioid receptors. Eur J Pharmacol. 1996;296(2):199–207.CrossRef
7.
Zurück zum Zitat Lin X, Wang YJ, Li Q, Hou YY, Hong MH, Cao YL, Chi ZQ, Liu JG. Chronic high-dose morphine treatment promotes SH-SY5Y cell apoptosis via c-Jun N-terminal kinase-mediated activation of mitochondria-dependent pathway. FEBS J. 2009;276(7):2022–36.CrossRef Lin X, Wang YJ, Li Q, Hou YY, Hong MH, Cao YL, Chi ZQ, Liu JG. Chronic high-dose morphine treatment promotes SH-SY5Y cell apoptosis via c-Jun N-terminal kinase-mediated activation of mitochondria-dependent pathway. FEBS J. 2009;276(7):2022–36.CrossRef
8.
Zurück zum Zitat Yin D, Mufson RA, Wang R, Shi Y. Fas-mediated cell death promoted by opioids. Nature. 1999;397(6716):218.CrossRef Yin D, Mufson RA, Wang R, Shi Y. Fas-mediated cell death promoted by opioids. Nature. 1999;397(6716):218.CrossRef
9.
Zurück zum Zitat Pati ML, Hornick JR, Niso M, Berardi F, Spitzer D. Sigma-2 receptor agonist derivatives of 1-Cyclohexyl-4-[3-(5-methoxy-1,2,3,4-tetrahydronaphthalen-1-yl)propyl]piperazine (PB28) induce cell death via mitochondrial superoxide production and caspase activation in pancreatic cancer. BMC Cancer. 2017;17(1):51.CrossRef Pati ML, Hornick JR, Niso M, Berardi F, Spitzer D. Sigma-2 receptor agonist derivatives of 1-Cyclohexyl-4-[3-(5-methoxy-1,2,3,4-tetrahydronaphthalen-1-yl)propyl]piperazine (PB28) induce cell death via mitochondrial superoxide production and caspase activation in pancreatic cancer. BMC Cancer. 2017;17(1):51.CrossRef
10.
Zurück zum Zitat Diao CT, Li L, Lau SY, Wong TM, Wong NS. kappa-Opioid receptor potentiates apoptosis via a phospholipase C pathway in the CNE2 humanepithelial tumor cell line. Biochim Biophys Acta. 2000;1499(1–2):49–62.CrossRef Diao CT, Li L, Lau SY, Wong TM, Wong NS. kappa-Opioid receptor potentiates apoptosis via a phospholipase C pathway in the CNE2 humanepithelial tumor cell line. Biochim Biophys Acta. 2000;1499(1–2):49–62.CrossRef
11.
Zurück zum Zitat Hatsukari Ikusuke, Hitosugi Naoko, Ohno Rie, Hashimoto Ken, Nakamura Shinichi, Satoh Kazue, Nagasaka Hiroshi, Matsumoto Isao, Sakagami Hiroshi. Induction of apoptosis by morphine in human tumor cell lines in vitro. Anticancer Res. 2007;27(2):857–64.PubMed Hatsukari Ikusuke, Hitosugi Naoko, Ohno Rie, Hashimoto Ken, Nakamura Shinichi, Satoh Kazue, Nagasaka Hiroshi, Matsumoto Isao, Sakagami Hiroshi. Induction of apoptosis by morphine in human tumor cell lines in vitro. Anticancer Res. 2007;27(2):857–64.PubMed
12.
Zurück zum Zitat Sergeeva MG, Grishina ZV, Varfolomeyev SD. Morphine effect on proliferation of normal and tumor cells of immune origin. Immunol Lett. 1993;36(2):215–8.CrossRef Sergeeva MG, Grishina ZV, Varfolomeyev SD. Morphine effect on proliferation of normal and tumor cells of immune origin. Immunol Lett. 1993;36(2):215–8.CrossRef
13.
Zurück zum Zitat Bimonte Sabrina, Barbieri Antonio, Rea Domenica, Palma Giuseppe, Luciano Antonio, Cuomo Arturo, Arra Claudio, Izzo Francesco. Morphine Promotes Tumor Angiogenesis and Increases Breast Cancer Progression. Biomed Res Int. 2015;2015:161508.PubMedPubMedCentral Bimonte Sabrina, Barbieri Antonio, Rea Domenica, Palma Giuseppe, Luciano Antonio, Cuomo Arturo, Arra Claudio, Izzo Francesco. Morphine Promotes Tumor Angiogenesis and Increases Breast Cancer Progression. Biomed Res Int. 2015;2015:161508.PubMedPubMedCentral
14.
Zurück zum Zitat Cao LH, Li HT, Lin WQ, et al. Morphine, a potential antagonist of cisplatin cytotoxicity, inhibits cisplatin induced apoptosis and suppression of tumor growth in nasopharyngeal carcinoma xenografts. Sci Rep. 2016;6:18706.CrossRef Cao LH, Li HT, Lin WQ, et al. Morphine, a potential antagonist of cisplatin cytotoxicity, inhibits cisplatin induced apoptosis and suppression of tumor growth in nasopharyngeal carcinoma xenografts. Sci Rep. 2016;6:18706.CrossRef
15.
Zurück zum Zitat Mathew B, Lennon FE, Siegler J, Mambetsariev N, et al. The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation. Anesth Analg. 2011;112(3):558–67.CrossRef Mathew B, Lennon FE, Siegler J, Mambetsariev N, et al. The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation. Anesth Analg. 2011;112(3):558–67.CrossRef
16.
Zurück zum Zitat Fujioka N, Nguyen J, Chen C, Li Y, Pasrija T, Niehans G, Johnson K, Gupta V, Kratzke R, Kalpna G. Morphine-induced epidermal growth factor pathway activation in non-small cell lung cell. Anesth Analg. 2011;113(6):1353–64.CrossRef Fujioka N, Nguyen J, Chen C, Li Y, Pasrija T, Niehans G, Johnson K, Gupta V, Kratzke R, Kalpna G. Morphine-induced epidermal growth factor pathway activation in non-small cell lung cell. Anesth Analg. 2011;113(6):1353–64.CrossRef
17.
Zurück zum Zitat Vincent L, Vang D, Nguyen J, et al. Mast cell activation contributes to sickle cell pathobiology and pain in mice. Blood. 2013;122:1853–62.CrossRef Vincent L, Vang D, Nguyen J, et al. Mast cell activation contributes to sickle cell pathobiology and pain in mice. Blood. 2013;122:1853–62.CrossRef
18.
Zurück zum Zitat Aich A, Afrin LB, Gupta K. Mast cell-mediated mechanisms of nociception. Int J Mol Sci. 2015;16:29069–92.CrossRef Aich A, Afrin LB, Gupta K. Mast cell-mediated mechanisms of nociception. Int J Mol Sci. 2015;16:29069–92.CrossRef
19.
Zurück zum Zitat Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in cancer progression. Peptides. 2013;48:1–9.CrossRef Muñoz M, Coveñas R. Involvement of substance P and the NK-1 receptor in cancer progression. Peptides. 2013;48:1–9.CrossRef
20.
Zurück zum Zitat Lin X, Li Q, Wang YJ, Ju YW, Chi ZQ, Wang MW, Liu JG. Morphine inhibits doxorubicin-induced reactive oxygen species generation and nuclear factor kappaB transcriptional activation in neuroblastoma SH-SY5Y cells. Biochem J. 2007;406(2):215–21.CrossRef Lin X, Li Q, Wang YJ, Ju YW, Chi ZQ, Wang MW, Liu JG. Morphine inhibits doxorubicin-induced reactive oxygen species generation and nuclear factor kappaB transcriptional activation in neuroblastoma SH-SY5Y cells. Biochem J. 2007;406(2):215–21.CrossRef
21.
Zurück zum Zitat Ma Y, Ren Z, Ma S, Yan W, He M, Wang D, Ding P. Morphine enhances renal cell carcinoma aggressiveness through promotes survivin level. Ren Fail. 2017;39(1):258–64.CrossRef Ma Y, Ren Z, Ma S, Yan W, He M, Wang D, Ding P. Morphine enhances renal cell carcinoma aggressiveness through promotes survivin level. Ren Fail. 2017;39(1):258–64.CrossRef
22.
Zurück zum Zitat Folkman J, D’Amore PA. Blood vessel formation: what is its molecular basis? Cell. 1996;87(7):1153–5.CrossRef Folkman J, D’Amore PA. Blood vessel formation: what is its molecular basis? Cell. 1996;87(7):1153–5.CrossRef
23.
Zurück zum Zitat Brekken RA, Thorpe PE. Vascular endothelial growth factor and vascular targeting of solid tumors. Anticancer Res. 2001;21(6b):4221–9.PubMed Brekken RA, Thorpe PE. Vascular endothelial growth factor and vascular targeting of solid tumors. Anticancer Res. 2001;21(6b):4221–9.PubMed
24.
Zurück zum Zitat Balasubramanian S, Ramakrishnan S, Charboneau R, Wang J, Barke RA, Roy S. Morphine sulfate inhibits hypoxia-induced vascular endothelial growth factor expression in endothelial cells and cardiac myocytes. J Mol Cell Cardiol. 2001;33:2179–87.CrossRef Balasubramanian S, Ramakrishnan S, Charboneau R, Wang J, Barke RA, Roy S. Morphine sulfate inhibits hypoxia-induced vascular endothelial growth factor expression in endothelial cells and cardiac myocytes. J Mol Cell Cardiol. 2001;33:2179–87.CrossRef
25.
Zurück zum Zitat Koodie L, Ramakrishnan S, Roy S. Morphine suppresses tumor angiogenesis through a HIF-1alpha/p38MAPK pathway. Am J Pathol. 2010;177(2):984–97.CrossRef Koodie L, Ramakrishnan S, Roy S. Morphine suppresses tumor angiogenesis through a HIF-1alpha/p38MAPK pathway. Am J Pathol. 2010;177(2):984–97.CrossRef
26.
Zurück zum Zitat Yamamizu K, Hamada Y, Narita M. j Opioid receptor ligands regulate angiogenesis in development and in tumours. Br J Pharmacol. 2015;172(2):268–76.CrossRef Yamamizu K, Hamada Y, Narita M. j Opioid receptor ligands regulate angiogenesis in development and in tumours. Br J Pharmacol. 2015;172(2):268–76.CrossRef
27.
Zurück zum Zitat Koodie L, Yuan H, Pumper JA, et al. Morphine inhibits migration of tumor-infiltrating leukocytes and suppresses angiogenesis associated with tumor growth in mice. Am J Pathol. 2014;184:1073–84.CrossRef Koodie L, Yuan H, Pumper JA, et al. Morphine inhibits migration of tumor-infiltrating leukocytes and suppresses angiogenesis associated with tumor growth in mice. Am J Pathol. 2014;184:1073–84.CrossRef
28.
Zurück zum Zitat Singleton PA, Moss J. Effect of perioperative opioids on cancer recurrence: a hypothesis. Future Oncol. 2010;6(8):1237–42.CrossRef Singleton PA, Moss J. Effect of perioperative opioids on cancer recurrence: a hypothesis. Future Oncol. 2010;6(8):1237–42.CrossRef
29.
Zurück zum Zitat Kevil CG, Orr AW, Langston W, Mickett K, Murphy-Ullrich J, Patel RP, et al. Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. J Biol Chem. 2004;279(18):19230–8.CrossRef Kevil CG, Orr AW, Langston W, Mickett K, Murphy-Ullrich J, Patel RP, et al. Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. J Biol Chem. 2004;279(18):19230–8.CrossRef
30.
Zurück zum Zitat Leo S, Nuydens R, Meert TF. Opioid-induced proliferation of vascular endothelial cells. J Pain Res. 2009;2:59–66.PubMedPubMedCentral Leo S, Nuydens R, Meert TF. Opioid-induced proliferation of vascular endothelial cells. J Pain Res. 2009;2:59–66.PubMedPubMedCentral
31.
Zurück zum Zitat Farooqui M, Li Y, Rogers T, Poonawala T, Griffin RJ, Song CW, et al. COX-2 inhibitor celecoxib prevents chronic morphine-induced promotion of angiogenesis, tumour growth, metastasis and mortality, without compromising analgesia. Br J Cancer. 2007;97(11):1523–31.CrossRef Farooqui M, Li Y, Rogers T, Poonawala T, Griffin RJ, Song CW, et al. COX-2 inhibitor celecoxib prevents chronic morphine-induced promotion of angiogenesis, tumour growth, metastasis and mortality, without compromising analgesia. Br J Cancer. 2007;97(11):1523–31.CrossRef
32.
Zurück zum Zitat Yamamoto Kazuhiro, Murphy Gillian, Troeberg Linda. Extracellular regulation of metalloproteinases. Matrix Biol. 2015;44–46:255–63.CrossRef Yamamoto Kazuhiro, Murphy Gillian, Troeberg Linda. Extracellular regulation of metalloproteinases. Matrix Biol. 2015;44–46:255–63.CrossRef
33.
Zurück zum Zitat Gouri Adel, Dekaken Aoulia, El Bairi Khalid, Aissaoui Arifa, Laabed Nihad, Chefrour Mohamed, Ciccolini Joseph, Milano Gérard, Benharkat Sadek. Plasminogen activator system and breast cancer: potential role in therapy decision making and precision medicine. Biomarker Insight. 2016;11:105–11.CrossRef Gouri Adel, Dekaken Aoulia, El Bairi Khalid, Aissaoui Arifa, Laabed Nihad, Chefrour Mohamed, Ciccolini Joseph, Milano Gérard, Benharkat Sadek. Plasminogen activator system and breast cancer: potential role in therapy decision making and precision medicine. Biomarker Insight. 2016;11:105–11.CrossRef
34.
Zurück zum Zitat Xie Nan, Khabbazi Samira, Nassar Zeyad D, Gregory Kye, Vithanage Tharindu, Anand-Apte Bela, Cabot Peter J, Sturgess David, Shaw Paul N, Parat Marie-Odile. Morphine alters the circulating proteolytic profile in mice: functional consequences on cellular migration and invasion. FASEB J. 2017;31(12):5208–16.CrossRef Xie Nan, Khabbazi Samira, Nassar Zeyad D, Gregory Kye, Vithanage Tharindu, Anand-Apte Bela, Cabot Peter J, Sturgess David, Shaw Paul N, Parat Marie-Odile. Morphine alters the circulating proteolytic profile in mice: functional consequences on cellular migration and invasion. FASEB J. 2017;31(12):5208–16.CrossRef
35.
Zurück zum Zitat Gach Katarzyna, Szemraj Janusz, Wyrębska Anna, Janecka Anna. The influence of opioids on matrix metalloproteinase-2 and -9 secretion and mRNA levels in MCF-7 breast cancer cell line. Mol Biol Rep. 2011;38(2):1231–6.CrossRef Gach Katarzyna, Szemraj Janusz, Wyrębska Anna, Janecka Anna. The influence of opioids on matrix metalloproteinase-2 and -9 secretion and mRNA levels in MCF-7 breast cancer cell line. Mol Biol Rep. 2011;38(2):1231–6.CrossRef
36.
Zurück zum Zitat Min Too Jae, Park Sang-Hee, Ji Yi-Hwa, Lee Yoon-Sook, Kim Tae Woo, Kim Jae Hwan, Kim Woon-Young, Park Young-Cheol. Morphine attenuates endothelial cell adhesion molecules induced by the supernatant of LPS-stimulated colon cancer cells. J Korean Med Sci. 2011;26(6):747–52.CrossRef Min Too Jae, Park Sang-Hee, Ji Yi-Hwa, Lee Yoon-Sook, Kim Tae Woo, Kim Jae Hwan, Kim Woon-Young, Park Young-Cheol. Morphine attenuates endothelial cell adhesion molecules induced by the supernatant of LPS-stimulated colon cancer cells. J Korean Med Sci. 2011;26(6):747–52.CrossRef
37.
Zurück zum Zitat Gach K, Szemraj J, Fichna J, Piestrzeniewicz M, Delbro DS, Janecka A. The influence of opioids on urokinase plasminogen activator on protein and mRNA level in MCF-7 breast cancer cell line. Chem Biol Drug Des. 2009;74(4):390–6.CrossRef Gach K, Szemraj J, Fichna J, Piestrzeniewicz M, Delbro DS, Janecka A. The influence of opioids on urokinase plasminogen activator on protein and mRNA level in MCF-7 breast cancer cell line. Chem Biol Drug Des. 2009;74(4):390–6.CrossRef
38.
Zurück zum Zitat Nylund G, Pettersson A, Bengtsson C, Khorram-Manesh A, Nordgren S, Delbro DS. Functional expression of mu-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Dig Dis Sci. 2008;53(2):461–6.CrossRef Nylund G, Pettersson A, Bengtsson C, Khorram-Manesh A, Nordgren S, Delbro DS. Functional expression of mu-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Dig Dis Sci. 2008;53(2):461–6.CrossRef
39.
Zurück zum Zitat Liu S, Qi L, Yu Q, et al. Survivin and HLA-I expression predicts survival of patients with clear cell renal cell carcinoma. Tumour Biol. 2014;35:8281–8.CrossRef Liu S, Qi L, Yu Q, et al. Survivin and HLA-I expression predicts survival of patients with clear cell renal cell carcinoma. Tumour Biol. 2014;35:8281–8.CrossRef
40.
Zurück zum Zitat Chen X, Chen XG, Hu X, et al. MiR-34a and miR-203 inhibit survivin expression to control cell proliferation and survival in human osteosarcoma cells. J Cancer. 2016;7:1057–65.CrossRef Chen X, Chen XG, Hu X, et al. MiR-34a and miR-203 inhibit survivin expression to control cell proliferation and survival in human osteosarcoma cells. J Cancer. 2016;7:1057–65.CrossRef
41.
Zurück zum Zitat Boettger MK, Weber K, Gajda M, Brauer R, Schaible HG. Spinally applied ketamine or morphine attenuate peripheral inflammation and hyperalgesia in acute and chronic phases of experimental arthritis. Brain Behav Immun. 2010;24(3):474–85.CrossRef Boettger MK, Weber K, Gajda M, Brauer R, Schaible HG. Spinally applied ketamine or morphine attenuate peripheral inflammation and hyperalgesia in acute and chronic phases of experimental arthritis. Brain Behav Immun. 2010;24(3):474–85.CrossRef
42.
Zurück zum Zitat Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schafer M, Loeffler JP, Stein C. Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100(1):142–8.CrossRef Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schafer M, Loeffler JP, Stein C. Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100(1):142–8.CrossRef
43.
Zurück zum Zitat Finley MJ, Happel CM, Kaminsky DE, Rogers TJ. Opioid and nociceptin receptors regulate cytokine and cytokine receptor expression. Cell Immunol. 2008;252(1–2):146–54.CrossRef Finley MJ, Happel CM, Kaminsky DE, Rogers TJ. Opioid and nociceptin receptors regulate cytokine and cytokine receptor expression. Cell Immunol. 2008;252(1–2):146–54.CrossRef
44.
Zurück zum Zitat Sabita R, Jinghua W, Jennifer K, Lisa K, Josephine M. Modulation of immune function by morphine: implications for susceptibility to infection. J Neuroimmun Pharmacol. 2006;1:77–89.CrossRef Sabita R, Jinghua W, Jennifer K, Lisa K, Josephine M. Modulation of immune function by morphine: implications for susceptibility to infection. J Neuroimmun Pharmacol. 2006;1:77–89.CrossRef
45.
Zurück zum Zitat Roy S, Wang J, Kelschenbach J, Koodie L, Martin J. Modulation of immune function by morphine: implications for susceptibility to infection. J Neuroimmune Pharmacol. 2006;1:77–89.CrossRef Roy S, Wang J, Kelschenbach J, Koodie L, Martin J. Modulation of immune function by morphine: implications for susceptibility to infection. J Neuroimmune Pharmacol. 2006;1:77–89.CrossRef
46.
Zurück zum Zitat Grimm MC, Ben-Baruch A, Taub DD, Howard OM, Resau JH, Wang JM, Ali H, Richardson R, Snyderman R, Oppenheim JJ. Opiates transdeactivate chemokine receptors: delta and mu opiate receptor-mediated heterologous desensitization. J Exp Med. 1998;188:317–25.CrossRef Grimm MC, Ben-Baruch A, Taub DD, Howard OM, Resau JH, Wang JM, Ali H, Richardson R, Snyderman R, Oppenheim JJ. Opiates transdeactivate chemokine receptors: delta and mu opiate receptor-mediated heterologous desensitization. J Exp Med. 1998;188:317–25.CrossRef
47.
Zurück zum Zitat Ninkovic J, Roy S. Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids. 2013;45:9–24.CrossRef Ninkovic J, Roy S. Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids. 2013;45:9–24.CrossRef
48.
Zurück zum Zitat Menzebach A, Hirsch J, Nost R, Mogk M, Hempelmann G, Welters ID. Morphine inhibits complement receptor expression, phagocytosis and oxidative burst by a nitric oxide dependent mechanism. Anasthesiol Intensivmed Notfallmed Schmerzther. 2004;39:204–11.CrossRef Menzebach A, Hirsch J, Nost R, Mogk M, Hempelmann G, Welters ID. Morphine inhibits complement receptor expression, phagocytosis and oxidative burst by a nitric oxide dependent mechanism. Anasthesiol Intensivmed Notfallmed Schmerzther. 2004;39:204–11.CrossRef
49.
Zurück zum Zitat Tabellini G, Borsani E, Benassi M, et al. Effects of opioid therapy on human natural killer cells. Int Immunopharmacol. 2014;18:169–74.CrossRef Tabellini G, Borsani E, Benassi M, et al. Effects of opioid therapy on human natural killer cells. Int Immunopharmacol. 2014;18:169–74.CrossRef
50.
Zurück zum Zitat Ninković Jana, Roy Sabita. Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids. 2013;45(1):9–24.CrossRef Ninković Jana, Roy Sabita. Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids. 2013;45(1):9–24.CrossRef
51.
Zurück zum Zitat Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C. Pain control in inflammation governed by selectins. Nat Med. 1998;4:1425–8.CrossRef Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C. Pain control in inflammation governed by selectins. Nat Med. 1998;4:1425–8.CrossRef
52.
Zurück zum Zitat Roy S, Chapin RB, Cain KJ, Charboneau RG, Ramakrishnan S, Barke RA. Morphine inhibits transcriptional activation of IL-2 in mouse thymocytes. Cell Immunol. 1997;179:1–9.CrossRef Roy S, Chapin RB, Cain KJ, Charboneau RG, Ramakrishnan S, Barke RA. Morphine inhibits transcriptional activation of IL-2 in mouse thymocytes. Cell Immunol. 1997;179:1–9.CrossRef
53.
Zurück zum Zitat Liang Xuan, Liu Renyu, Chen Chunhua, Ji Fang, Li Tianzuo. Opioid system modulates the immune function: a review. Trans Perioperat Pain Med. 2016;1(1):5–13. Liang Xuan, Liu Renyu, Chen Chunhua, Ji Fang, Li Tianzuo. Opioid system modulates the immune function: a review. Trans Perioperat Pain Med. 2016;1(1):5–13.
54.
Zurück zum Zitat Weber RJ, Pert A. The periaqueductal gray matter mediates opiate-induced immunosuppression. Science. 1989;245:188–90.CrossRef Weber RJ, Pert A. The periaqueductal gray matter mediates opiate-induced immunosuppression. Science. 1989;245:188–90.CrossRef
55.
Zurück zum Zitat Irwin M, Hauger RL, Brown M, Britton KT. CRF activates autonomic nervous system and reduces natural killer cytotoxicity. Am J Physiol. 1988;255(5 Pt 2):R744–7.PubMed Irwin M, Hauger RL, Brown M, Britton KT. CRF activates autonomic nervous system and reduces natural killer cytotoxicity. Am J Physiol. 1988;255(5 Pt 2):R744–7.PubMed
56.
Zurück zum Zitat Hernandez MC, Flores LR, Bayer BM. Immunosuppression by morphine is mediated by central pathways. J Pharmacol Exp Ther. 1993;267:1336–41.PubMed Hernandez MC, Flores LR, Bayer BM. Immunosuppression by morphine is mediated by central pathways. J Pharmacol Exp Ther. 1993;267:1336–41.PubMed
57.
Zurück zum Zitat Saurer TB, Ijames SG, Lysle DT. Neuropeptide Y Y1 receptors mediate morphine-induced reductions of natural killer cell activity. J Neuroimmunol. 2006;177:18–26.CrossRef Saurer TB, Ijames SG, Lysle DT. Neuropeptide Y Y1 receptors mediate morphine-induced reductions of natural killer cell activity. J Neuroimmunol. 2006;177:18–26.CrossRef
58.
Zurück zum Zitat Fecho K, Maslonek KA, Dykstra LA, Lysle DT. Evidence for sympathetic and adrenal involvement in the immunomodulatory effects of acute morphine treatment in rats. J Pharmacol Exp Ther. 1996;277:633–45.PubMed Fecho K, Maslonek KA, Dykstra LA, Lysle DT. Evidence for sympathetic and adrenal involvement in the immunomodulatory effects of acute morphine treatment in rats. J Pharmacol Exp Ther. 1996;277:633–45.PubMed
59.
Zurück zum Zitat Mellon RD, Bayer BM. Role of central opioid receptor subtypes in morphine-induced alterations in peripheral lymphocyte activity. Brain Res. 1998;789:56–67.CrossRef Mellon RD, Bayer BM. Role of central opioid receptor subtypes in morphine-induced alterations in peripheral lymphocyte activity. Brain Res. 1998;789:56–67.CrossRef
60.
Zurück zum Zitat Zylla D, Gourley BL, Vang D, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer. 2013;119:4103–10.CrossRef Zylla D, Gourley BL, Vang D, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer. 2013;119:4103–10.CrossRef
61.
Zurück zum Zitat Zylla D, Ma Kuskowski, Gupta K, Gupta P. Association of opioid requirement and cancer pain with survival in advanced non-small cell lung cancer. Br J Anaesth. 2014;113:1–8.CrossRef Zylla D, Ma Kuskowski, Gupta K, Gupta P. Association of opioid requirement and cancer pain with survival in advanced non-small cell lung cancer. Br J Anaesth. 2014;113:1–8.CrossRef
Metadaten
Titel
The dual effect of morphine on tumor development
verfasst von
H. Tuerxun
J. Cui
Publikationsdatum
23.11.2018
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 6/2019
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-018-1974-5

Weitere Artikel der Ausgabe 6/2019

Clinical and Translational Oncology 6/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.