Open Access

Effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro

  • Authors:
    • Nai‑Bao Zhou
    • Kai‑Guo Wang
    • Zhi‑Jian Fu
  • View Affiliations

  • Published online on: August 16, 2019     https://doi.org/10.3892/ol.2019.10750
  • Pages: 4230-4236
  • 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

The combination of morphine and ketamine is considered safe and efficacious in many patients. However, a considerable number of immunomodulatory effects have been reported to be produced by both morphine and ketamine. The aim of the present study was to assess the direct effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro. Venous blood was obtained from patients with refractory cancer pain and peripheral blood mononuclear cells were isolated using the Ficoll‑Hypaque density gradient method. Anti‑CD3 beads were used to isolate T cells by positive selection. Subsequently, the T cells were treated with vehicle, 200 ng/ml of morphine or 200 ng/ml of morphine + 100 ng/ml ketamine for 24 h, following which the cells were stimulated with anti‑CD3 and anti‑CD28. Flow cytometric analysis of CD3+ T cells, and interleukin (IL)‑2 and interferon (IFN)‑γ in the supernatant, reverse transcription‑quantitative PCR analysis for the detection of IL‑2 and IFN‑γ and western blotting for the detection of p65 nuclear factor (NF)‑κB were performed. In vitro, the CD4+ and CD8+ T cell counts, CD4+/CD8+ ratio, secretion of IL‑2 and IFN‑γ in the supernatant, mRNA expression levels of IL‑2 and IFN‑γ and expression of p65 NF‑κB were significantly decreased following treatment with morphine and morphine + ketamine, compared with results in the control group (all P<0.05). However, there was no significant difference between treatment with morphine and that with morphine + ketamine. Treatment with morphine + ketamine in vitro decreased the immune functions of patients with refractory cancer pain, although the effect of treatment with morphine and a low dose of ketamine did not differ significantly from that with morphine treatment alone.

Introduction

Pain associated with cancer is a serious problem for patients, significantly affecting their quality of life and is difficult to treat. A considerable proportion of patients suffer from refractory cancer pain. Refractory pain is associated with the growth and metastasis of tumors (1,2), therefore, pain relief may be beneficial for improving the patient's quality of life while also supplementing cancer therapy. The World Health Organization has proposed that a three-step analgesic ladder is effective for treating cancer pain (3). For neuropathic pain in advanced cancer, the first-line therapeutic agent is typically an opioid, such as morphine (47), however, pain may not be fully relieved for many patients. In these cases, the combination of pharmacological agents may be necessary to relieve the refractory pain. The combination of morphine and ketamine has been considered safe and efficacious in numerous patients (813). Clinically, patients with refractory cancer pain report improved pain relief with fewer side effects when morphine and a low dose of ketamine were used compared with morphine alone.

A number of patients experience cancer pain driven by nociception and neuropathy (14) and neuropathic cancer pain driven by nociception is considered to be resistant to opioids (1517). For neuropathic pain, peripheral and central sensitization are the primary causes. Ketamine is an N-methyl-D aspartate (NMDA) antagonist and is also a non-opioid anesthetic agent. The primary clinical application of ketamine is for short diagnostic or surgical procedures (17). NMDA receptors are activated by the excitatory neurotransmitter glutamate in the spinal dorsal horn, which results in sensitization of the central nervous system (1821). Therefore, attenuating this sensitization may underlie the analgesic effects of ketamine (1921). Similarly, NMDA antagonism may be involved in the ketamine-mediated attenuation of acquired opioid tolerance (2022).

However, in laboratory animals and humans, morphine has been reported to produce numerous immunomodulatory effects in vivo and in vitro. As a potent immunomodulator, morphine has inhibitory and stimulatory effects on immune function, including altering the expression of CD4+ and CD8+ T cells, reducing the activity of natural killer cells (2325), altering the balance between Th1 and Th2 cells (26,27), increasing the production of interleukin (IL)-12 by peritoneal macrophages (28), increasing the production of IL-6 and tumor necrosis factor when induced by lipopolysaccharides, and increasing the activity of nuclear factor-κB (NF-κB) in macrophages and peripheral blood mononuclear cells (PBMCs) (29,30). In particular, in activated T cells, morphine has been shown to enhance the expression of IL-4 and suppress the expression of interferon (IFN)-γ and IL-2 (3135). For patients with cancer and an impaired immune system, the immunomodulatory effects of morphine may cause unwanted side effects during pain treatment. However, there are few reports regarding the effects of morphine on T cells in patients who suffer from refractory cancer pain. Similarly, there are few reports regarding the effect of ketamine on immune cells, particularly in T cells. Therefore, the aim of the present study was to assess the direct effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro.

Materials and methods

T cell culture, T cell activation and reagents

The entire protocol (SDTHEC201504001) was approved by the Ethics Committee of Shandong Cancer Hospital and Institute (Jinan, China; Chairperson Dr Jinming Yu) and conducted according to the principles of the Helsinki Declaration. Informed, written consent was obtained from all patients with cancer pain, and the patients were involved in the study for >3 months.

Blood was withdrawn following venipuncture. Peripheral blood mononuclear cells (PBMCs) were isolated using the Ficoll-Hypaque density gradient method according to the manufacturer's protocol (Cedarlane). The T cells were isolated by positive selection using anti-CD3 beads (Miltenyi Biotec, Inc.) according to the manufacturer's protocol, and confirmed by fluorescence-associated cell sorting (85% purity). A final concentration of 1×105 cells/ml were plated into 96-well plates, and the T cells were cultivated at 37°C and 5% CO2 in RPMI 1640 medium supplemented with 10% fetal calf serum, 100 U/ml penicillin and 100 mg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc.).

The T cells were treated with either vehicle (normal saline), 200 ng/ml of morphine (Humanwell Healthcare (Group) Co., Ltd.), or 200 ng/ml of morphine + 100 ng/ml ketamine (pure hydrochloride salt without preservatives, Sigma-Aldrich; Merck KGaA) for 24 h at 37°C. By determining the plasma concentration of morphine and ketamine in patients with refractory cancer pain in the clinic, and referring to other human and animal experiments using morphine and ketamine, a morphine concentration of 200 ng/ml and ketamine concentration of 100 ng/ml were established (3638). These concentrations are approximately equal to the doses of morphine and ketamine commonly used in patients with refractory cancer pain in the clinic. Subsequently, the cells were stimulated with anti-CD3 and anti-CD28 (R&D Systems, Inc.; mouse-anti-human, 5 mg/ml) for 24 h at 37°C. The groups were designated as the control group, morphine group and morphine + ketamine group. To activate T cells, it is necessary for the antigen-specific T cell receptor and a second co-receptor, such as CD28, to be sufficiently occupied (39). In the present study, anti-CD3 and anti-CD28 stimulation were used to investigate the effects of morphine and ketamine on the T cells.

Flow cytometric analysis of CD3+ T cells

Following experimental treatment, the T cells (1×105/ml) were harvested by centrifugation for 5 min at 300 × g room temperature, washed in phosphate-buffered saline and subsequently incubated with trichromatism monoclonal antibody reagent [CD4 fluorescein isothiocyanate/CD8 phycoerythrin/CD3 PerCP (BD Biosciences), 0.5 mg/ml] according to the manufacturer's protocol. Subsequently, the quantity and percentage of CD3/CD4/CD8T cells were measured using a flow cytometer (FACSCalibur; BD Biosciences).

Supernatant cytokine protein analysis

Cytokine-specific ELISA kits (R&D Systems, Inc.) were used to measure the protein concentrations of IL-2 and IFN-γ according to the manufacturer's protocol. The experiments were repeated three times.

Reverse transcription-quantitative PCR analysis

Total RNA was extracted from the T cells by lysis using guanidinium isothiocyanate and phenol acid extraction. A total of 1 µg RNA, 0.5 µl RNase H minus (Promega Corporation) and 1 µl Moloney murine leukemia virus reverse transcriptase were used for cDNA synthesis with the following thermocycling conditions: 95°C 5 min; 95°C 15 sec, 60°C 35 sec, 40 cycles; 72°C 5 min; 4°C terminal. For each PCR, 2 µl cDNA was used and the primers were obtained from BD Biosciences. The primer sequences were as follows: IL-2 forward, 5′-GAATGGAATTAATAATTACAAGAATCCC-3′ and reverse, 5′-TGTTTCAGATCCCTTTAGTTCCAG-3′; and IFN-γ forward, 5′-TCGGTAACTGACTTGAATGTCCA-3′ and reverse, 5′-TCCTTTTTCGCTTCCCTGTTTT-3′. The Light Cycler-Fast Start DNA Master SYBR Green I kit (Roche Diagnostics) was used for the RT-qPCR analysis of IL-2, and the Revert Aid™ First Strand cDNA Synthesis kit (Roche Diagnostics) was used for IFN-γ, according to the manufacturer protocols. The quantitative analysis of original template was performed by the change of fluorescence of the amplification product.

Western blot analysis for activated p65 NF-κB

The T cells were lysed with RIPA protein lysis buffer (100 µg/ml Nonidet P-40, 150 mol/l NaCl and 50 mmol/l Tris-HCl supplemented with a protease inhibitor mixture). Following one freeze/thaw cycle, the lysates were centrifuged for 15 min at 12,000 g (2–8°C). A BCA protein concentration kit was used to determine protein concentration. Following electrophoresis using 10% SDS-PAGE, the resolved protein samples were transferred onto an ECL nitrocellulose membrane (EMD Millipore) by electroblotting. 5% (w/v) of skimmed milk powder was used for blocking for 1 h at room temperature. Subsequently, the nitrocellulose membranes were incubated overnight at 4°C with primary antibody [rabbit anti-NF-κB p65 antibody (cat. no., ab16502); dilution, 1:2,000; Abcam/rabbit anti-β-actin (cat. no., ab8227); dilution, 1:2,000; Abcam], and then incubated with the secondary goat anti-Rabbit horseradish peroxidase antibody (cat. no., ab205718; 1:2,000; Abcam) at room temperature for 2 h. Protein concentration was determined by ECL western blotting detection kit (cat. no., ab65623; Abcam). Image J version 1.8.0 (National Institutes of Health) was used for quantification of blots.

Statistical analysis

All variables were assessed in triplicate and all experiments were repeated at least three times. All data are expressed as the mean ± standard deviation. Statistical comparisons between experimental groups were performed, where appropriate, with SPSS version 17.0 (SPSS, Inc., Chicago, IL, USA). Statistical significance was analyzed using a paired t-test or one-way ANOVA followed by a non-parametric Student-Newman-Keuls test for multiple comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Flow cytometric analysis of CD3+ T cells

The counts of T cells expressing CD4 and CD8 were significantly decreased when the cells were treated with morphine and when treated with morphine + ketamine. The decrease in the count of CD4+ T cells was larger than that of CD8+ T cells, therefore, the ratio of CD4+/CD8+ was also significantly decreased by morphine and morphine + ketamine treatments (Fig. 1A-C; all P<0.05). However, the decreases in the CD4+ and CD8+ T cell counts and the CD4+/CD8+ ratio did not differ significantly when the cells were treated with morphine compared with those when the cells were treated with morphine + ketamine.

Supernatant cytokine protein analysis and RT-qPCR analysis

There were no significant differences in the concentrations of IL-2 and IFN-γ in the supernatants of the morphine group and the morphine + ketamine group compared with those in the control group (Fig. 2). However, the mRNA expression levels of IL-2 and IFN-γ were significantly decreased in the morphine group and the morphine + ketamine group compared with those in the control group (Fig. 3; P<0.05). The results of the morphine group did not differ significantly from those of the morphine + ketamine group.

Western blot analysis of activated p65 NF-κB

The morphine and morphine + ketamine treatments decreased the quantity of activated p65 NF-κB entering the nuclei of T cells compared with that in the control group (Fig. 4; P<0.05). The results in the morphine group did not differ significantly from those in the morphine + ketamine group.

Discussion

Patients with refractory cancer pain suffer from pain and consequent stress on a daily basis. Those individuals experiencing severe pain, stress or who become addicted to opioids are generally immunosuppressed resulting from activation of the neuroendocrine-immune network and the potent effect of neuroactive ligands on cells of the immune system (4045). Opioids have been demonstrated to modulate immune responses, and opioid receptors are expressed on immune cells (41). In vivo and in vitro, the therapeutic and chronic use of morphine affects the physiological function of the immune system, including innate and adaptive immunity (46,47).

T cells and the various subpopulations serve important roles in cell-mediated immunity. The ratios of the subpopulations of T cells are stably maintained to ensure an effective immune response. In the present study, CD4+ and CD8+ T cells, and the ratio of CD4+/CD8+ T cells were all decreased by morphine, which suggested an attenuation of the immune response.

IL-2 is a pivotal cytokine for T cell function, and a previous study demonstrated that chronic treatment with morphine can inhibit the mRNA expression and secretion of IL-2 (48). Similarly, in the present study on patients with refractory cancer pain, the mRNA expression levels of IL-2 and IL-2 were downregulated by morphine treatment. A previous study showed that the expression of IL-2 was regulated almost entirely at the transcriptional level and that the essential elements lie within 300 bp upstream of the start codon (49), which can be bound by several inducible transcription factors including NF-κB. It has been hypothesized that the production of IL-2 is inhibited with chronic morphine treatment at the transcriptional level and through epigenetic mechanisms (31,34,35,50,51).

As a cytokine modulating all phases of immune processes, IFN-γ is produced in large quantities in activated T cells (52) and serves an important role in host resistance to infection (53). Previous studies have demonstrated that chronic morphine treatment inhibited IFN-γ protein synthesis (32,54), similar to the results of the present study. Furthermore, the results of the present study demonstrated that the mRNA expression levels of IFN-γ were downregulated by morphine, suggesting that morphine modulated IFN-γ by altering the gene expression and protein synthesis of IFN-γ. It is hypothesized that transcription of the IFN-γ gene is coordinated by the transcription factor NF-κB (5557).

As a central regulator of the immune response (58), NF-κB regulates the transcription of various genes, including a number of cytokines and genes encoding immunoreceptors (59,60), and it is sequestered in the cytoplasm in the majority of cell types (61). NF-κB is also vital for regulating the activation, proliferation and differentiation of T cells (62). Roy et al (30) demonstrated that morphine prevented the binding of NF-κB to its consensus DNA sequence in macrophages and activated T cells of patients who had received chronic morphine treatment.

In vivo, the pathways regulating the immunomodulatory effects of opioids transduced to the immune effector cells primarily include the following: i) Indirectly via the central nervous system, for example, by activation of the hypothalamic-pituitary-adrenal axis (63,64); ii) directly via atypical opioid receptors (25); and iii) directly via the classic µ- (preferential), δ- and κ-opioid receptor subtypes present on immune cells (65,66) that belong to the Gi protein-coupled receptor superfamily (67). Additionally, it has been shown that the activation of NF-κB was inhibited by elevated cAMP in T cells at the molecular level (6870). Therefore, morphine may bind to µ-opioid receptors and activate Gi protein-coupled receptors, resulting in the intracellular accumulation of cAMP, and thus leading to decreased NF-κB activation and the inhibition of IL-2 and IFN-γ transcription.

Ketamine is often administered epidurally or intravenously in combination with an opioid for the treatment of chronic cancer pain (71,72). For the clinical treatment of refractory cancer pain, ketamine is often used as an adjuvant analgesic; as ketamine alone is not used for refractory cancer pain, a group treated with ketamine only was not included in the present study. However, ketamine also affects immune cells. It has been suggested that ketamine inhibits the production and function of dendritic cells (73). Treatment with ketamine alone has also been shown to increase the ratios of Th1/Th2 and IFN-γ/IL-4 (74). However, in the present study, ketamine did not induce significant effects in any of the measurements, including CD4+ T cells, CD8+ T cells, the CD4+/CD8+ ratio, concentrations of IL-2 and IFN-γ in the supernatant, mRNA expression levels of IL-2 and IFN-γ and the activation of NF-κB. A possible explanation for the lack of effect of ketamine was the dose used, which may have been too low. In clinical use, the dose of ketamine used is variable. The epidural administration of ketamine is typically ~0.4 mg/ml (~1.5 mM) (7580), whereas intrathecal administration is typically <25 mg/ml (~93 mM) (8183). Another study demonstrated that the infusion dose of ketamine for cancer-associated treatment was 0.084–0.6 mg/kg/h (20). In the present study, a dose of 100 ng/ml of ketamine was considered to be ineffective.

In conclusion, in vitro treatment with morphine alone and in conjunction with ketamine inhibited the immune functions of patients with refractory cancer pain. Treatment decreased the counts of CD4+ and CD8+ T cells, CD4+/CD8+ ratio, concentrations of IL-2 and IFN-γ in the supernatant, mRNA expression levels of IL-2 and IFN-γ and the activation of NF-κB, but the effect of morphine in conjunction with ketamine did not differ significantly from the effect of morphine alone. The present study may provide evidence for clinicians to offer more relief of pain and less suppression of immune function to patients with refractory cancer pain.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the Natural Science Foundation (Shandong, China; grant no. ZR2011HM039).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

NBZ, KGW, ZJF designed the study and completed the organization and writing. NBZ conducted the experiments and analyzed the data.

Ethics approval and consent to participate

The entire protocol (SDTHEC201504001) was approved by the Ethics Committee of Shandong Cancer Hospital and Institute (Jinan, China; chairperson Dr Jinming Yu) and was conducted according to the principles of the Helsinki Declaration. Informed, written consent was obtained from patients with cancer pain.

Patient consent for publication

The patients consented to publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

Page GG, Ben-Eliyahu S, Yirmiya R and Liebeskind JC: Morphine attenuates surgery-induced enhancement of metastatic colonization in rats. Pain. 54:21–28. 1993. View Article : Google Scholar : PubMed/NCBI

2 

Sasamura T, Nakamura S, Iida Y, Fuji H, Murata J, Saiki I, Nojima H and Kuraishi Y: Morphine analgesia suppresses tumor growth and metastasis in a mouse model of cancer pain produced by orthotopic tumor inoculation. Eur J Pharmacol. 441:185–191. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Zech DF, Grond S, Lynch J, Hertel D and Lehmann KA: Validation of World Health Organization guidelines for cancer pain relief: A 10-year prospective study. Pain. 63:65–76. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Collins JJ, Grier HE, Kinney HC and Berde CB: Control of severe pain in children with terminal malignancy. J Pediatr. 126:653–657. 1995. View Article : Google Scholar : PubMed/NCBI

5 

Hurwitz CA, Duncan J and Wolfe J: Caring for the child with cancer at the close of life: ‘There are people who make it and I'm hoping I'm one of them.’. JAMA. 292:2141–2149. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Jalmsell L, Kreicbergs U, Onelöv E, Steineck G and Henter JI: Symptoms affecting children with malignancies during the last month of life: A nationwide follow-up. Pediatrics. 117:1314–1320. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Wolfe J, Grier HE, Klar N, Levin SB, Ellenbogen JM, Salem-Schatz S, Emanuel EJ and Weeks JC: Symptoms and suffering at the end of life in children with cancer. N Engl J Med. 342:326–333. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Finkel JC, Pestieau SR and Quezado ZM: Ketamine as an adjuvant for treatment of cancer pain in children and adolescents. J Pain. 8:515–521. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Mercadante S, Lodi F, Sapio M, Calligara M and Serretta R: Longterm ketamine subcutaneous continuous infusion in neuropathic cancer pain. J Pain Symptom Manage. 10:564–568. 1995. View Article : Google Scholar : PubMed/NCBI

10 

Elsewaisy O, Slon B and Monagle J: Analgesic effect of subanesthetic intravenous ketamine in refractory neuropathic pain: A case report. Pain Med. 11:946–950. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Webster LR and Walker MJ: Safety and efficacy of prolonged outpatient ketamine infusions for neuropathic pain. Am J Ther. 13:300–305. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Visser E and Schug SA: The role of ketamine in pain management. Biomed Pharmacother. 60:341–348. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Subramaniam K, Subramaniam B and Steinbrook RA: Ketamine as adjuvant analgesic to opioids: A quantitative and qualitative systematic review. Anesth Analg. 99:482–495. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Grond S, Radbruch L, Meuser T, Sabatowski R, Loick G and Lehmann KA: Assessment and treatment of neuropathic cancer pain following WHO guidelines. Pain. 79:15–20. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Cherny NI, Thaler HT, Friedlander-Klar H, Lapin J, Foley KM, Houde R and Portenoy RK: Opioid responsiveness of cancer pain syndromes caused by neuropathic or nociceptive mechanisms: A combined analysis of controlled, single-dose studies. Neurology. 44:857–861. 1994. View Article : Google Scholar : PubMed/NCBI

16 

Martin LA and Hagen NA: Neuropathic pain in cancer patients: Mechanisms, syndromes, and clinical controversies. J Pain Symptom Manage. 14:99–117. 1997. View Article : Google Scholar : PubMed/NCBI

17 

Enarson MC, Hays H and Woodroffe MA: Clinical experience with oral ketamine. J Pain Symptom Manage. 17:384–386. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Coggeshall RE and Carlton SM: Receptor localization in the mammalian dorsal horn and primary afferent neurons. Brain Res Brain Res Rev. 24:28–66. 1997. View Article : Google Scholar : PubMed/NCBI

19 

Persson J, Axelsson G, Hallin RG and Gustafsson LL: Beneficial effects of ketamine in a chronic pain state with allodynia, possibly due to central sensitization. Pain. 60:217–222. 1995. View Article : Google Scholar : PubMed/NCBI

20 

Okon T: Ketamine: An introduction for the pain and palliative medicine physician. Pain Physician. 10:493–500. 2007.PubMed/NCBI

21 

Grande LA, O'Donnell BR, Fitzgibbon DR and Terman GW: Ultra-low dose ketamine and memantine treatment for pain in an opioid-tolerant oncology patient. Anesth Analg. 107:1380–1383. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Shimoyama N, Shimoyama M, Inturrisi CE and Elliott KJ: Ketamine attenuates and reverses morphine tolerance in rodents. Anesthesiology. 85:1357–1366. 1996. View Article : Google Scholar : PubMed/NCBI

23 

Weber RJ and Pert A: The periaqueductal gray matter mediates opiate-induced immunosuppression. Science. 245:188–190. 1989. View Article : Google Scholar : PubMed/NCBI

24 

Gavériaux-Ruff C, Matthes HW, Peluso J and Kieffer BL: Abolition of morphine-immunosuppression in mice lacking the mu-opioid receptor gene. Proc Natl Acad Sci USA. 95:6326–6330. 1998. View Article : Google Scholar : PubMed/NCBI

25 

Roy S, Barke RA and Loh HH: MU-opioid receptor-knockout mice: Role of mu-opioid receptor in morphine mediated immune functions. Brain Res Mol Brain Res. 61:190–194. 1998. View Article : Google Scholar : PubMed/NCBI

26 

Roy S, Charboneau RG and Barke RA: Morphine synergizes with lipopolysaccharide in a chronic endotoxemia model. J Neuroimmunol. 95:107–114. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Sacerdote P, Gaspani L and Panerai AE: The opioid antagonist naloxone induces a shift from type 2 to type 1 cytokine pattern in normal and skin-grafted mice. Ann NY Acad Sci. 917:755–763. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Peng X, Mosser DM, Adler MW, Rogers TJ, Meissler JJ Jr and Eisenstein TK: Morphine enhances interleukin-12 and the production of other pro-inflammatory cytokines in mouse peritoneal macrophages. J Leukoc Biol. 68:723–728. 2000.PubMed/NCBI

29 

Chao CC, Molitor TW, Close K, Hu S and Peterson PK: Morphine inhibits the release of tumor necrosis factor in human peripheral blood mononuclear cell cultures. Int J Immunopharmacol. 15:447–453. 1993. View Article : Google Scholar : PubMed/NCBI

30 

Roy S, Cain KJ, Chapin RB, Charboneau RG and Barke RA: Morphine modulates NF kappa B activation in macrophages. Biochem Biophys Res Commun. 245:392–396. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Roy S, Chapin RB, Cain KJ, Charboneau RG, Ramakrishnan S and Barke RA: Morphine inhibits transcriptional activation of IL-2 in mouse thymocytes. Cell Immunol. 179:1–9. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Roy S, Balasubramanian S, Sumandeep S, Charboneau R, Wang J, Melnyk D, Beilman GJ, Vatassery R and Barke RA: Morphine directs T cells toward T(H2) differentiation. Surgery. 130:304–309. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Roy S, Wang J, Charboneau R, Loh HH and Barke RA: Morphine induces CD4+ T cell IL-4 expression through an adenylyl cyclase mechanism independent of the protein kinase A pathway. J Immunol. 175:6361–6367. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Wang J, Barke RA, Charboneau R, Loh HH and Roy S: Morphine negatively regulates interferon-gamma promoter activity in activated murine T cells through two distinct cyclic AMP-dependent pathways. J Biol Chem. 278:37622–37631. 2003. View Article : Google Scholar : PubMed/NCBI

35 

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. View Article : Google Scholar : PubMed/NCBI

36 

Pacifici GM: Metabolism and pharmacokinetics of morphine in neonates: A review. Clinics (Sao Paulo). 71:474–480. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Heiskanen T, Langel K, Gunnar T, Lillsunde P and Kalso EA: Opioid concentrations in oral fluid and plasma in cancer patients with pain. J Pain Symptom Manage. 50:524–532. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Khalili-Mahani N, Martini CH, Olofsen E, Dahan A and Niesters M: Effect of subanaesthetic ketamine on plasma and saliva cortisol secretion. Br J Anaesth. 115:68–75. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Zhang J, Salojin KV, Gao JX, Cameron MJ, Bergerot I and Delovitch TL: p38 mitogen-activated protein kinase mediates signal integration of TCR/CD28 costimulation in primary murine T cells. Immunol. 162:3819–3829. 1999.

40 

Rouveix B: Opiates and immune function: Consequences on infectious diseases with special reference to AIDS. Therapie. 47:503–512. 1992.PubMed/NCBI

41 

Sibinga NE and Goldstein A: Opioid peptides and opioid receptors in cells of the immune system. Annu Rev Immunol. 6:219–249. 1988. View Article : Google Scholar : PubMed/NCBI

42 

Weigent DA and Blalock JE: Interactions between the neuroendocrine and immune systems: Common hormones and receptors. Immunol Rev. 100:79–108. 1987. View Article : Google Scholar : PubMed/NCBI

43 

Nair MP, Schwartz SA, Polasani R, Hou J, Sweet A and Chadha KC: Immunoregulatory effects of morphine on human lymphocytes. Clin Diagn Lab Immunol. 4:127–132. 1997.PubMed/NCBI

44 

Kulkarni-Narla A, Walcheck B and Brown DR: Opioid receptors on bone marrow neutrophils modulate chemotaxis and CD11b/CD18 expression. Eur J Pharmacol. 414:289–294. 2001. View Article : Google Scholar : PubMed/NCBI

45 

McCarthy L, Wetzel M, Sliker JK, Eisenstein TK and Rogers TJ: Opioids, opioid receptors, and the immune response. Drug Alcohol Depend. 62:111–123. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Roy S and Loh HH: Effects of opioids on the immune system. Neurochem Res. 21:1375–1386. 1996. View Article : Google Scholar : PubMed/NCBI

47 

Bryant HU, Bernton EW and Holaday JW: Immunosuppressive effects of chronic morphine treatment in mice. Life Sci. 41:1731–1738. 1987. View Article : Google Scholar : PubMed/NCBI

48 

Wang J, Charboneau R, Balasubramanian S, Barke RA, Loh HH and Roy S: Morphine modulates lymph node-derived T lymphocyte function: Role of caspase-3, −8 and nitric oxide. J Leukocyte Biol. 70:527–536. 2001.PubMed/NCBI

49 

Jain J, Loh C and Rao A: Transcriptional regulation of the IL-2 gene. Curr Opin Immunol. 7:333–342. 1995. View Article : Google Scholar : PubMed/NCBI

50 

Wang JH, 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. View Article : Google Scholar : PubMed/NCBI

51 

Thomas PT, House RV and Bhargava HN: Direct cellular immunomodulation produced by diacetylmorphine (heroin) or methadone. Gen Pharmacol. 26:123–130. 1995. View Article : Google Scholar : PubMed/NCBI

52 

Young HA and Hardy KJ: Interferon-gamma: Producer cells, activation stimuli, and molecular genetic regulation. Pharmacol Ther. 45:137–151. 1990. View Article : Google Scholar : PubMed/NCBI

53 

Reuben JM, Lee BN, Paul M, Kline MW, Cron SG, Abramson S, Lewis D, Kozinetz CA and Shearer WT: Magnitude of IFN-gamma production in HIV-1-infected children is associated with virus suppression. J Allergy Clin Immunol. 110:255–261. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Beyers AD, van Rie A, Adams J, Fenhalls G, Gie R and Beyers N: Signals that regulate the host response to Mycobacterium tuberculosis. Novartis Found Symp. 217:145–159. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Sweetser MT, Hoey T, Sun YL, Weaver WM, Price GA and Wilson CB: The roles of nuclear factor of activated T cells and ying-yang 1 in activation-induced expression of the interferon-gamma promoter in T cells. J Biol Chem. 273:34775–34783. 1998. View Article : Google Scholar : PubMed/NCBI

56 

Cippitelli M, Ye J, Viggiano V, Sica A, Ghosh P, Gulino A, Santoni A and Young HA: Retinoic acid-induced transcriptional modulation of the human interferon-gamma promoter. J Biol Chem. 271:26783–26793. 1996. View Article : Google Scholar : PubMed/NCBI

57 

Kaminuma O, Elly C, Tanaka Y, Mori A, Liu YC, Altman A and Miyatake S: Vav-induced activation of the human IFN-gamma gene promoter is mediated by upregulation of AP-1 activity. FEBS Lett. 514:153–158. 2002. View Article : Google Scholar : PubMed/NCBI

58 

Sen R and Baltimore D: Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 46:705–716. 1986. View Article : Google Scholar : PubMed/NCBI

59 

Pahl HL: Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 18:6853–6866. 1999. View Article : Google Scholar : PubMed/NCBI

60 

Pannen BH and Robotham JL: The acute-phase response. New Horiz. 3:183–197. 1995.PubMed/NCBI

61 

Baeuerle PA and Baltimore D: Activation of DNA-binding activity in an apparently cytoplasmic precursor of the NF-kappa B transcription factor. Cell. 53:211–217. 1988. View Article : Google Scholar : PubMed/NCBI

62 

Ghosh S, May MJ and Kopp EB: NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 16:225–260. 1998. View Article : Google Scholar : PubMed/NCBI

63 

Cabot PJ, Carter L, Schafer M and Stein C: Methionine-enkephalin-and Dynorphin A-release from immune cells and control of inflammatory pain. Pain. 93:207–212. 2001. View Article : Google Scholar : PubMed/NCBI

64 

Roy S, Wang JH, Balasubramanian S, Sumandeep, Charboneau R, Barke R and Loh HH: Role of hypothalamic-pituitary axis in morphine-induced alteration in thymic cell distribution using mu-opioid receptor knockout mice. J Neuroimmunol. 116:147–155. 2001. View Article : Google Scholar : PubMed/NCBI

65 

Kieffer BL: Recent advances in molecular recognition and signal transduction of active peptides: Receptors for opioid peptides. Cell Mol Neurobiol. 15:615–635. 1995. View Article : Google Scholar : PubMed/NCBI

66 

Pol O and Puig MM: Expression of opioid receptors during peripheral inflammation. Curr Top Med Chem. 4:51–61. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Rogers TJ and Peterson PK: Opioid G protein-coupled receptors: Signals at the crossroads of inflammation. Trends Immunol. 24:116–121. 2003. View Article : Google Scholar : PubMed/NCBI

68 

Haraguchi S, Good RA and Day NK: Immunosuppressive retroviral peptides: cAMP and cytokine patterns. Immunol Today. 16:595–603. 1995. View Article : Google Scholar : PubMed/NCBI

69 

Jimenez JL, Punzon C, Navarro J, Munoz-Fernandez MA and Fresno MJ: Phosphodiesterase 4 inhibitors prevent cytokine secretion by T lymphocytes by inhibiting nuclear factor-kappaB and nuclear factor of activated T cells activation. J Pharmacol Exp Ther. 299:753–759. 2001.PubMed/NCBI

70 

Chen D and Rothenberg EV: Interleukin 2 transcription factors as molecular targets of cAMP inhibition: Delayed inhibition kinetics and combinatorial transcription roles. J Exp Med. 179:931–942. 1994. View Article : Google Scholar : PubMed/NCBI

71 

Walker SM, Goudas LC, Cousins MJ and Carr DB: Combination spinal analgesic chemotherapy: A systematic review. Anesth Analg. 95:674–715. 2002. View Article : Google Scholar : PubMed/NCBI

72 

Hocking G and Cousins MJ: Ketamine in chronic pain management: An evidence-based review. Anesth Analg. 97:1730–1739. 2003. View Article : Google Scholar : PubMed/NCBI

73 

Ohta N, Ohashi Y and Fujino Y: Ketamine inhibits maturation of bone marrow-derived dendritic cells and priming of the Th1-type immune response. Anesth Analg. 109:793–800. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Gao M, Jin W, Qian Y, Ji L, Feng G and Sun J: Effect of N-methyl-D-aspartate receptor antagonist on T helper cell differentiation induced by phorbol-myristate-acetate and ionomycin. Cytokine. 56:458–465. 2011. View Article : Google Scholar : PubMed/NCBI

75 

Chia YY, Liu K, Liu YC, Chang HC and Wong CS: Adding ketamine in a multimodal patient-controlled epidural regimen reduces postoperative pain and analgesic consumption. Anesth Analg. 86:1245–1249. 1998. View Article : Google Scholar : PubMed/NCBI

76 

Tan PH, Kuo MC, Kao PF, Chia YY and Liu K: Patient-controlled epidural analgesia with morphine or morphine plus ketamine for postoperative pain relief. Eur J Anaesthesiol. 16:820–825. 1999. View Article : Google Scholar : PubMed/NCBI

77 

Choudhuri AH, Dharmani P, Kumarl N and Prakash A: Comparison of caudal epidural bupivacaine with bupivacaine plus tramadol and bupivacaine plus ketamine for postoperative analgesia in children. Anaesth Intensive Care. 36:174–179. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Gunes Y, Secen M, Ozcengiz D, Gündüz M, Balcioglu O and Isik G: Comparison of caudal ropivacaine, ropivacaine plus ketamine and ropivacaine plus tramadol administration for postoperative analgesia in children. Paediatr Anaesth. 14:557–563. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Ozbek H, Bilen A, Ozcengiz D, Gunes Y, Ozalevli M and Akman H: The comparison of caudal ketamine, alfentanil and ketamine plus alfentanil administration for postoperative analgesia in children. Paediatr Anaesth. 12:610–616. 2002. View Article : Google Scholar : PubMed/NCBI

80 

Gunduz M, Ozalevli M, Ozbek H and Ozcengiz D: Comparison of caudal ketamine with lidocaine or tramadol administration for postoperative analgesia of hypospadias surgery in children. Paediatr Anaesth. 16:158–163. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Vranken JH, Troost D, Wegener JT, Kruis MR and van der Vegt MH: Neuropathological findings after continuous intrathecal administration of S(+)-ketamine for the management of neuropathic cancer pain. Pain. 117:231–235. 2005. View Article : Google Scholar : PubMed/NCBI

82 

Sator-Katzenschlager S, Deusch E, Maier P, Spacek A and Kress HG: The long-term antinociceptive effect of intrathecal S(+)-ketamine in a patient with established morphine tolerance. Anesth Analg. 93:1032–1034. 2001. View Article : Google Scholar : PubMed/NCBI

83 

Benrath J, Scharbert G, Gustorff B, Adams HA and Kress HG: Longterm intrathecal S(+)-ketamine in a patient with cancer-related neuropathic pain. Br J Anaesth. 95:247–279. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 18 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou NB, Wang KG and Fu ZJ: Effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro. Oncol Lett 18: 4230-4236, 2019
APA
Zhou, N., Wang, K., & Fu, Z. (2019). Effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro. Oncology Letters, 18, 4230-4236. https://doi.org/10.3892/ol.2019.10750
MLA
Zhou, N., Wang, K., Fu, Z."Effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro". Oncology Letters 18.4 (2019): 4230-4236.
Chicago
Zhou, N., Wang, K., Fu, Z."Effect of morphine and a low dose of ketamine on the T cells of patients with refractory cancer pain in vitro". Oncology Letters 18, no. 4 (2019): 4230-4236. https://doi.org/10.3892/ol.2019.10750