Skip to main content
Erschienen in: Cancer and Metastasis Reviews 2/2011

01.06.2011 | NON-THEMATIC REVIEW

Morphine and tumor growth and metastasis

verfasst von: Banafsheh Afsharimani, Peter Cabot, Marie-Odile Parat

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 2/2011

Einloggen, um Zugang zu erhalten

Abstract

Morphine is an analgesic widely used to alleviate cancer pain. In addition, the perioperative management of pain in cancer surgery patients most often includes opioids. However, there are reports that these drugs may alter cancer recurrence or metastasis. Several mechanisms have been proposed, such as the modulation of the immune response or cellular pathways that control the survival and migratory behavior of cancer cells. The published literature, however, presents some discrepancies, with reports suggesting that opioids may either promote or prevent the spread of cancer. It is of great importance to determine whether opioids, in particular the most widely used, morphine, may increase the risk of metastasis when used in cancer surgery. This review examines the available data on the effects of morphine which influence cancer metastasis or recurrence, including immunomodulation, tumor cell aggressiveness, and angiogenesis, with special emphasis on recently published clinical and laboratory based studies. We further discuss the parameters that may explain the difference between reports on the effects of morphine on cancer.
Literatur
1.
Zurück zum Zitat Sobel, H., & Bonorris, G. (1962). Effect of morphine on rats bearing Walker carcinosarcoma 256. Nature, 196, 896–897.PubMed Sobel, H., & Bonorris, G. (1962). Effect of morphine on rats bearing Walker carcinosarcoma 256. Nature, 196, 896–897.PubMed
2.
Zurück zum Zitat Kerros, C., Cavey, T., Sola, B., Jauzac, P., & Allouche, S. (2009). Somatostatin and opioid receptors do not regulate proliferation or apoptosis of the human multiple myeloma U266 cells. Journal of Experimental and Clinical Cancer Research, 28, 77. Kerros, C., Cavey, T., Sola, B., Jauzac, P., & Allouche, S. (2009). Somatostatin and opioid receptors do not regulate proliferation or apoptosis of the human multiple myeloma U266 cells. Journal of Experimental and Clinical Cancer Research, 28, 77.
3.
Zurück zum Zitat Biji, M., Lennon, F., Siegler, J., Mirzapoiazova, T., Mambetsariev, N., Sammani, S. et al. (2011). The novel role of the mu opioid receptor in lung cancer progression: A laboratory investigation. Anesthesia and Analgesia (in press). Biji, M., Lennon, F., Siegler, J., Mirzapoiazova, T., Mambetsariev, N., Sammani, S. et al. (2011). The novel role of the mu opioid receptor in lung cancer progression: A laboratory investigation. Anesthesia and Analgesia (in press).
4.
Zurück zum Zitat Nylund, G., Pettersson, A., Bengtsson, C., Khorram-Manesh, A., Nordgren, S., & Delbro, D. S. (2008). Functional expression of mu-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Digestive Diseases and Sciences, 53(2), 461–466.PubMed Nylund, G., Pettersson, A., Bengtsson, C., Khorram-Manesh, A., Nordgren, S., & Delbro, D. S. (2008). Functional expression of mu-opioid receptors in the human colon cancer cell line, HT-29, and their localization in human colon. Digestive Diseases and Sciences, 53(2), 461–466.PubMed
5.
Zurück zum Zitat Maneckjee, R., & Minna, J. D. (1990). Opioid and nicotine receptors affect growth regulation of human lung cancer cell lines. Proceedings of the National Academy of Sciences of the United States of America, 87(9), 3294–3298.PubMed Maneckjee, R., & Minna, J. D. (1990). Opioid and nicotine receptors affect growth regulation of human lung cancer cell lines. Proceedings of the National Academy of Sciences of the United States of America, 87(9), 3294–3298.PubMed
6.
Zurück zum Zitat Hatzoglou, A., Bakogeorgou, E., & Castanas, E. (1996). The antiproliferative effect of opioid receptor agonists on the T47D human breast cancer cell line, is partially mediated through opioid receptors. European Journal of Pharmacology, 296(2), 199–207.PubMed Hatzoglou, A., Bakogeorgou, E., & Castanas, E. (1996). The antiproliferative effect of opioid receptor agonists on the T47D human breast cancer cell line, is partially mediated through opioid receptors. European Journal of Pharmacology, 296(2), 199–207.PubMed
7.
Zurück zum Zitat Kampa, M., Bakogeorgou, E., Hatzoglou, A., Damianaki, A., Martin, P. M., & Castanas, E. (1997). Opioid alkaloids and casomorphin peptides decrease the proliferation of prostatic cancer cell lines (LNCaP, PC3 and DU145) through a partial interaction with opioid receptors. European Journal of Pharmacology, 335(2–3), 255–265.PubMed Kampa, M., Bakogeorgou, E., Hatzoglou, A., Damianaki, A., Martin, P. M., & Castanas, E. (1997). Opioid alkaloids and casomorphin peptides decrease the proliferation of prostatic cancer cell lines (LNCaP, PC3 and DU145) through a partial interaction with opioid receptors. European Journal of Pharmacology, 335(2–3), 255–265.PubMed
8.
Zurück zum Zitat Fichna, J., Krajewska, U., Rozalski, M., Mirowski, M., & Janecka, A. (2005). Characterization of the [125I]endomorphin-2 binding sites in the MCF7 breast cancer cell line. Peptides, 26(2), 295–299.PubMed Fichna, J., Krajewska, U., Rozalski, M., Mirowski, M., & Janecka, A. (2005). Characterization of the [125I]endomorphin-2 binding sites in the MCF7 breast cancer cell line. Peptides, 26(2), 295–299.PubMed
9.
Zurück zum Zitat Yin, D., Woodruff, M., Zhang, Y., Whaley, S., Miao, J., Ferslew, K., et al. (2006). Morphine promotes Jurkat cell apoptosis through pro-apoptotic FADD/P53 and anti-apoptotic PI3K/Akt/NF-kappaB pathways. Journal of Neuroimmunology, 174(1–2), 101–107.PubMed Yin, D., Woodruff, M., Zhang, Y., Whaley, S., Miao, J., Ferslew, K., et al. (2006). Morphine promotes Jurkat cell apoptosis through pro-apoptotic FADD/P53 and anti-apoptotic PI3K/Akt/NF-kappaB pathways. Journal of Neuroimmunology, 174(1–2), 101–107.PubMed
10.
Zurück zum Zitat Zhao, M., Zhou, G., Zhang, Y., Chen, T., Sun, X., Stuart, C., et al. (2009). Beta-arrestin2 inhibits opioid-induced breast cancer cell death through Akt and caspase-8 pathways. Neoplasma, 56(2), 108–113.PubMed Zhao, M., Zhou, G., Zhang, Y., Chen, T., Sun, X., Stuart, C., et al. (2009). Beta-arrestin2 inhibits opioid-induced breast cancer cell death through Akt and caspase-8 pathways. Neoplasma, 56(2), 108–113.PubMed
11.
Zurück zum Zitat Lin, X., Wang, Y. J., Li, Q., Hou, Y. Y., Hong, M. H., Cao, Y. L., et al. (2009). Chronic high-dose morphine treatment promotes SH-SY5Y cell apoptosis via c-Jun N-terminal kinase-mediated activation of mitochondria-dependent pathway. The FEBS Journal, 276(7), 2022–2036.PubMed Lin, X., Wang, Y. J., Li, Q., Hou, Y. Y., Hong, M. H., Cao, Y. L., et al. (2009). Chronic high-dose morphine treatment promotes SH-SY5Y cell apoptosis via c-Jun N-terminal kinase-mediated activation of mitochondria-dependent pathway. The FEBS Journal, 276(7), 2022–2036.PubMed
12.
Zurück zum Zitat Zagon, I. S., & McLaughlin, P. J. (2003). Opioids and the apoptotic pathway in human cancer cells. Neuropeptides, 37(2), 79–88.PubMed Zagon, I. S., & McLaughlin, P. J. (2003). Opioids and the apoptotic pathway in human cancer cells. Neuropeptides, 37(2), 79–88.PubMed
13.
Zurück zum Zitat Cadet, P., Rasmussen, M., Zhu, W., Tonnesen, E., Mantione, K. J., & Stefano, G. B. (2004). Endogenous morphinergic signaling and tumor growth. Frontier in Bioscience, 9, 3176–3186. Cadet, P., Rasmussen, M., Zhu, W., Tonnesen, E., Mantione, K. J., & Stefano, G. B. (2004). Endogenous morphinergic signaling and tumor growth. Frontier in Bioscience, 9, 3176–3186.
14.
Zurück zum Zitat Tegeder, I., Grosch, S., Schmidtko, A., Haussler, A., Schmidt, H., Niederberger, E., et al. (2003). G protein-independent G1 cell cycle block and apoptosis with morphine in adenocarcinoma cells: Involvement of p53 phosphorylation. Cancer Research, 63(8), 1846–1852.PubMed Tegeder, I., Grosch, S., Schmidtko, A., Haussler, A., Schmidt, H., Niederberger, E., et al. (2003). G protein-independent G1 cell cycle block and apoptosis with morphine in adenocarcinoma cells: Involvement of p53 phosphorylation. Cancer Research, 63(8), 1846–1852.PubMed
15.
Zurück zum Zitat Hatzoglou, A., Ouafik, L., Bakogeorgou, E., Thermos, K., & Castanas, E. (1995). Morphine cross-reacts with somatostatin receptor SSTR2 in the T47D human breast cancer cell line and decreases cell growth. Cancer Research, 55(23), 5632–5636.PubMed Hatzoglou, A., Ouafik, L., Bakogeorgou, E., Thermos, K., & Castanas, E. (1995). Morphine cross-reacts with somatostatin receptor SSTR2 in the T47D human breast cancer cell line and decreases cell growth. Cancer Research, 55(23), 5632–5636.PubMed
16.
Zurück zum Zitat Maneckjee, R., Biswas, R., & Vonderhaar, B. K. (1990). Binding of opioids to human MCF-7 breast cancer cells and their effects on growth. Cancer Research, 50(8), 2234–2238.PubMed Maneckjee, R., Biswas, R., & Vonderhaar, B. K. (1990). Binding of opioids to human MCF-7 breast cancer cells and their effects on growth. Cancer Research, 50(8), 2234–2238.PubMed
17.
Zurück zum Zitat Panagiotou, S., Bakogeorgou, E., Papakonstanti, E., Hatzoglou, A., Wallet, F., Dussert, C., et al. (1999). Opioid agonists modify breast cancer cell proliferation by blocking cells to the G2/M phase of the cycle: Involvement of cytoskeletal elements. Journal of Cellular Biochemistry, 73(2), 204–211.PubMed Panagiotou, S., Bakogeorgou, E., Papakonstanti, E., Hatzoglou, A., Wallet, F., Dussert, C., et al. (1999). Opioid agonists modify breast cancer cell proliferation by blocking cells to the G2/M phase of the cycle: Involvement of cytoskeletal elements. Journal of Cellular Biochemistry, 73(2), 204–211.PubMed
18.
Zurück zum Zitat Sueoka, E., Sueoka, N., Kai, Y., Okabe, S., Suganuma, M., Kanematsu, K., et al. (1998). Anticancer activity of morphine and its synthetic derivative, KT-90, mediated through apoptosis and inhibition of NF-kappaB activation. Biochemical and Biophysical Research Communications, 252(3), 566–570.PubMed Sueoka, E., Sueoka, N., Kai, Y., Okabe, S., Suganuma, M., Kanematsu, K., et al. (1998). Anticancer activity of morphine and its synthetic derivative, KT-90, mediated through apoptosis and inhibition of NF-kappaB activation. Biochemical and Biophysical Research Communications, 252(3), 566–570.PubMed
19.
Zurück zum Zitat Sueoka, N., Sueoka, E., Okabe, S., & Fujiki, H. (1996). Anti-cancer effects of morphine through inhibition of tumour necrosis factor-alpha release and mRNA expression. Carcinogenesis, 17(11), 2337–2341.PubMed Sueoka, N., Sueoka, E., Okabe, S., & Fujiki, H. (1996). Anti-cancer effects of morphine through inhibition of tumour necrosis factor-alpha release and mRNA expression. Carcinogenesis, 17(11), 2337–2341.PubMed
20.
Zurück zum Zitat Iglesias, M., Segura, M. F., Comella, J. X., & Olmos, G. (2003). [mu]-Opioid receptor activation prevents apoptosis following serum withdrawal in differentiated SH-SY5Y cells and cortical neurons via phosphatidylinositol 3-kinase. Neuropharmacology, 44(4), 482–492.PubMed Iglesias, M., Segura, M. F., Comella, J. X., & Olmos, G. (2003). [mu]-Opioid receptor activation prevents apoptosis following serum withdrawal in differentiated SH-SY5Y cells and cortical neurons via phosphatidylinositol 3-kinase. Neuropharmacology, 44(4), 482–492.PubMed
21.
Zurück zum Zitat Lin, X., Li, Q., Wang, Y. J., Ju, Y. W., Chi, Z. Q., Wang, M. W., et al. (2007). Morphine inhibits doxorubicin-induced reactive oxygen species generation and nuclear factor κB transcriptional activation in neuroblastoma SH-SY5Y cells. The Biochemical Journal, 406(2), 215–221.PubMed Lin, X., Li, Q., Wang, Y. J., Ju, Y. W., Chi, Z. Q., Wang, M. W., et al. (2007). Morphine inhibits doxorubicin-induced reactive oxygen species generation and nuclear factor κB transcriptional activation in neuroblastoma SH-SY5Y cells. The Biochemical Journal, 406(2), 215–221.PubMed
22.
Zurück zum Zitat Sergeeva, M. G., Grishina, Z. V., & Varfolomeyev, S. D. (1993). Morphine effect on proliferation of normal and tumor cells of immune origin. Immunology Letters, 36(2), 215–218.PubMed Sergeeva, M. G., Grishina, Z. V., & Varfolomeyev, S. D. (1993). Morphine effect on proliferation of normal and tumor cells of immune origin. Immunology Letters, 36(2), 215–218.PubMed
23.
Zurück zum Zitat Debruyne, D. J., Mareel, M. M., & Bracke, M. E. (2010). Opioids affect focal contact-mediated cell-substrate adhesion. European Journal of Cancer Prevention, 19(3), 227–238.PubMed Debruyne, D. J., Mareel, M. M., & Bracke, M. E. (2010). Opioids affect focal contact-mediated cell-substrate adhesion. European Journal of Cancer Prevention, 19(3), 227–238.PubMed
24.
Zurück zum Zitat Harimaya, Y., Koizumi, K., Andoh, T., Nojima, H., Kuraishi, Y., & Saiki, I. (2002). Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells. Cancer Letters, 187(1–2), 121–127.PubMed Harimaya, Y., Koizumi, K., Andoh, T., Nojima, H., Kuraishi, Y., & Saiki, I. (2002). Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells. Cancer Letters, 187(1–2), 121–127.PubMed
25.
Zurück zum Zitat Horvath, R. J., & DeLeo, J. A. (2009). Morphine enhances microglial migration through modulation of P2X4 receptor signaling. The Journal of Neuroscience, 29(4), 998–1005.PubMed Horvath, R. J., & DeLeo, J. A. (2009). Morphine enhances microglial migration through modulation of P2X4 receptor signaling. The Journal of Neuroscience, 29(4), 998–1005.PubMed
26.
Zurück zum Zitat Zagon, I. S., Rahn, K. A., & McLaughlin, P. J. (2007). Opioids and migration, chemotaxis, invasion, and adhesion of human cancer cells. Neuropeptides, 41(6), 441–452.PubMed Zagon, I. S., Rahn, K. A., & McLaughlin, P. J. (2007). Opioids and migration, chemotaxis, invasion, and adhesion of human cancer cells. Neuropeptides, 41(6), 441–452.PubMed
27.
Zurück zum Zitat Gach, K., Szemraj, J., Wyrebska, A., & Janecka, A. (2011). The influence of opioids on matrix metalloproteinase-2 and -9 secretion and mRNA levels in MCF-7 breast cancer cell line. Molecular Biology Reports (in press). Gach, K., Szemraj, J., Wyrebska, A., & Janecka, A. (2011). The influence of opioids on matrix metalloproteinase-2 and -9 secretion and mRNA levels in MCF-7 breast cancer cell line. Molecular Biology Reports (in press).
28.
Zurück zum Zitat Gach, K., Szemraj, J., Fichna, J., Piestrzeniewicz, M., Delbro, D. S., & Janecka, A. (2009). The influence of opioids on urokinase plasminogen activator on protein and mRNA level in MCF-7 breast cancer cell line. Chemical Biology & Drug Design, 74(4), 390–396. Gach, K., Szemraj, J., Fichna, J., Piestrzeniewicz, M., Delbro, D. S., & Janecka, A. (2009). The influence of opioids on urokinase plasminogen activator on protein and mRNA level in MCF-7 breast cancer cell line. Chemical Biology & Drug Design, 74(4), 390–396.
29.
Zurück zum Zitat Dunn, G. P., Old, L. J., & Schreiber, R. D. (2004). The immunobiology of cancer immunosurveillance and immunoediting. Immunity, 21(2), 137–148.PubMed Dunn, G. P., Old, L. J., & Schreiber, R. D. (2004). The immunobiology of cancer immunosurveillance and immunoediting. Immunity, 21(2), 137–148.PubMed
30.
Zurück zum Zitat Grivennikov, S. I., Greten, F. R., & Karin, M. (2010). Immunity, inflammation, and cancer. Cell, 140(6), 883–899.PubMed Grivennikov, S. I., Greten, F. R., & Karin, M. (2010). Immunity, inflammation, and cancer. Cell, 140(6), 883–899.PubMed
31.
Zurück zum Zitat Bindea, G., Mlecnik, B., Fridman, W. H., Pages, F., & Galon, J. (2010). Natural immunity to cancer in humans. Current Opinion in Immunology, 22(2), 215–222.PubMed Bindea, G., Mlecnik, B., Fridman, W. H., Pages, F., & Galon, J. (2010). Natural immunity to cancer in humans. Current Opinion in Immunology, 22(2), 215–222.PubMed
32.
Zurück zum Zitat McCarthy, L., Wetzel, M., Sliker, J. K., Eisenstein, T. K., & Rogers, T. J. (2001). Opioids, opioid receptors, and the immune response. Drug and Alcohol Dependence, 62(2), 111–123.PubMed McCarthy, L., Wetzel, M., Sliker, J. K., Eisenstein, T. K., & Rogers, T. J. (2001). Opioids, opioid receptors, and the immune response. Drug and Alcohol Dependence, 62(2), 111–123.PubMed
33.
Zurück zum Zitat Vallejo, R., de Leon-Casasola, O., & Benyamin, R. (2004). Opioid therapy and immunosuppression: A review. American Journal of Therapeutics, 11(5), 354–365.PubMed Vallejo, R., de Leon-Casasola, O., & Benyamin, R. (2004). Opioid therapy and immunosuppression: A review. American Journal of Therapeutics, 11(5), 354–365.PubMed
34.
Zurück zum Zitat Roy, S., Wang, J., Kelschenbach, J., Koodie, L., & Martin, J. (2006). Modulation of immune function by morphine: Implications for susceptibility to infection. Journal of Neuroimmune Pharmacology, 1(1), 77–89.PubMed Roy, S., Wang, J., Kelschenbach, J., Koodie, L., & Martin, J. (2006). Modulation of immune function by morphine: Implications for susceptibility to infection. Journal of Neuroimmune Pharmacology, 1(1), 77–89.PubMed
35.
Zurück zum Zitat Sacerdote, P., Bianchi, M., Gaspani, L., Manfredi, B., Maucione, A., Terno, G., et al. (2000). The effects of tramadol and morphine on immune responses and pain after surgery in cancer patients. Anesthesia and Analgesia, 90(6), 1411–1414.PubMed Sacerdote, P., Bianchi, M., Gaspani, L., Manfredi, B., Maucione, A., Terno, G., et al. (2000). The effects of tramadol and morphine on immune responses and pain after surgery in cancer patients. Anesthesia and Analgesia, 90(6), 1411–1414.PubMed
36.
Zurück zum Zitat Sacerdote, P. (2008). Opioid-induced immunosuppression. Current Opinion in Supportive and Palliative Care, 2(1), 14–18.PubMed Sacerdote, P. (2008). Opioid-induced immunosuppression. Current Opinion in Supportive and Palliative Care, 2(1), 14–18.PubMed
37.
Zurück zum Zitat Beilin, B., Shavit, Y., Hart, J., Mordashov, B., Cohn, S., Notti, I., et al. (1996). Effects of anesthesia based on large versus small doses of fentanyl on natural killer cell cytotoxicity in the perioperative period. Anesthesia and Analgesia, 82(3), 492–497.PubMed Beilin, B., Shavit, Y., Hart, J., Mordashov, B., Cohn, S., Notti, I., et al. (1996). Effects of anesthesia based on large versus small doses of fentanyl on natural killer cell cytotoxicity in the perioperative period. Anesthesia and Analgesia, 82(3), 492–497.PubMed
38.
Zurück zum Zitat Shavit, Y., Ben-Eliyahu, S., Zeidel, A., & Beilin, B. (2004). Effects of fentanyl on natural killer cell activity and on resistance to tumor metastasis in rats. Dose and timing study. Neuroimmunomodulation, 11(4), 255–260.PubMed Shavit, Y., Ben-Eliyahu, S., Zeidel, A., & Beilin, B. (2004). Effects of fentanyl on natural killer cell activity and on resistance to tumor metastasis in rats. Dose and timing study. Neuroimmunomodulation, 11(4), 255–260.PubMed
39.
Zurück zum Zitat Franchi, S., Panerai, A. E., & Sacerdote, P. (2007). Buprenorphine ameliorates the effect of surgery on hypothalamus–pituitary–adrenal axis, natural killer cell activity and metastatic colonization in rats in comparison with morphine or fentanyl treatment. Brain, Behavior, and Immunity, 21(6), 767–774.PubMed Franchi, S., Panerai, A. E., & Sacerdote, P. (2007). Buprenorphine ameliorates the effect of surgery on hypothalamus–pituitary–adrenal axis, natural killer cell activity and metastatic colonization in rats in comparison with morphine or fentanyl treatment. Brain, Behavior, and Immunity, 21(6), 767–774.PubMed
40.
Zurück zum Zitat Rahim, R. T., Meissler, J. J., Jr., Cowan, A., Rogers, T. J., Geller, E. B., Gaughan, J., et al. (2001). Administration of mu-, kappa- or delta2-receptor agonists via osmotic minipumps suppresses murine splenic antibody responses. International Immunopharmacology, 1(11), 2001–2009.PubMed Rahim, R. T., Meissler, J. J., Jr., Cowan, A., Rogers, T. J., Geller, E. B., Gaughan, J., et al. (2001). Administration of mu-, kappa- or delta2-receptor agonists via osmotic minipumps suppresses murine splenic antibody responses. International Immunopharmacology, 1(11), 2001–2009.PubMed
41.
Zurück zum Zitat Stefano, G. B., Burrill, J. D., Labur, S., Blake, J., & Cadet, P. (2005). Regulation of various genes in human leukocytes acutely exposed to morphine: Expression microarray analysis. Medical Science Monitor, 11(5), MS35–MS42.PubMed Stefano, G. B., Burrill, J. D., Labur, S., Blake, J., & Cadet, P. (2005). Regulation of various genes in human leukocytes acutely exposed to morphine: Expression microarray analysis. Medical Science Monitor, 11(5), MS35–MS42.PubMed
42.
Zurück zum Zitat Eisenstein, T. K., & Hilburger, M. E. (1998). Opioid modulation of immune responses: Effects on phagocyte and lymphoid cell populations. Journal of Neuroimmunology, 83(1–2), 36–44.PubMed Eisenstein, T. K., & Hilburger, M. E. (1998). Opioid modulation of immune responses: Effects on phagocyte and lymphoid cell populations. Journal of Neuroimmunology, 83(1–2), 36–44.PubMed
43.
Zurück zum Zitat Mathews, P. M., Froelich, C. J., Sibbitt, W. L., & Bankhurst, A. D. (1983). Enhancement of natural cytotoxicity by beta-endorphin. Journal of Immunology, 130(4), 1658–1662. Mathews, P. M., Froelich, C. J., Sibbitt, W. L., & Bankhurst, A. D. (1983). Enhancement of natural cytotoxicity by beta-endorphin. Journal of Immunology, 130(4), 1658–1662.
44.
Zurück zum Zitat Wei, G., Moss, J., & Yuan, C. S. (2003). Opioid-induced immunosuppression: Is it centrally mediated or peripherally mediated? Biochemical Pharmacology, 65(11), 1761–1766.PubMed Wei, G., Moss, J., & Yuan, C. S. (2003). Opioid-induced immunosuppression: Is it centrally mediated or peripherally mediated? Biochemical Pharmacology, 65(11), 1761–1766.PubMed
45.
Zurück zum Zitat Hall, D. M., Suo, J. L., & Weber, R. J. (1998). Opioid mediated effects on the immune system: Sympathetic nervous system involvement. Journal of Neuroimmunology, 83(1–2), 29–35.PubMed Hall, D. M., Suo, J. L., & Weber, R. J. (1998). Opioid mediated effects on the immune system: Sympathetic nervous system involvement. Journal of Neuroimmunology, 83(1–2), 29–35.PubMed
46.
Zurück zum Zitat Liang-Suo, J., Gomez-Flores, R., & Weber, R. J. (2002). Immunosuppression induced by central action of morphine is not blocked by mifepristone (RU 486). Life Sciences, 71(22), 2595–2602.PubMed Liang-Suo, J., Gomez-Flores, R., & Weber, R. J. (2002). Immunosuppression induced by central action of morphine is not blocked by mifepristone (RU 486). Life Sciences, 71(22), 2595–2602.PubMed
47.
Zurück zum Zitat Wang, J., Charboneau, R., Balasubramanian, S., Barke, R. A., Loh, H. H., & Roy, S. (2002). The immunosuppressive effects of chronic morphine treatment are partially dependent on corticosterone and mediated by the mu-opioid receptor. Journal of Leukocyte Biology, 71(5), 782–790.PubMed Wang, J., Charboneau, R., Balasubramanian, S., Barke, R. A., Loh, H. H., & Roy, S. (2002). The immunosuppressive effects of chronic morphine treatment are partially dependent on corticosterone and mediated by the mu-opioid receptor. Journal of Leukocyte Biology, 71(5), 782–790.PubMed
48.
Zurück zum Zitat Pruett, S. B., Han, Y. C., & Fuchs, B. A. (1992). Morphine suppresses primary humoral immune responses by a predominantly indirect mechanism. The Journal of Pharmacology and Experimental Therapeutics, 262(3), 923–928.PubMed Pruett, S. B., Han, Y. C., & Fuchs, B. A. (1992). Morphine suppresses primary humoral immune responses by a predominantly indirect mechanism. The Journal of Pharmacology and Experimental Therapeutics, 262(3), 923–928.PubMed
49.
Zurück zum Zitat Rahim, R. T., Meissler, J. J., Jr., Adler, M. W., & Eisenstein, T. K. (2005). Splenic macrophages and B cells mediate immunosuppression following abrupt withdrawal from morphine. Journal of Leukocyte Biology, 78(6), 1185–1191.PubMed Rahim, R. T., Meissler, J. J., Jr., Adler, M. W., & Eisenstein, T. K. (2005). Splenic macrophages and B cells mediate immunosuppression following abrupt withdrawal from morphine. Journal of Leukocyte Biology, 78(6), 1185–1191.PubMed
50.
Zurück zum Zitat Gaveriaux-Ruff, C., Matthes, H. W., Peluso, J., & Kieffer, B. L. (1998). Abolition of morphine-immunosuppression in mice lacking the mu-opioid receptor gene. Proceedings of the National Academy of Sciences of the United States of America, 95(11), 6326–6330.PubMed Gaveriaux-Ruff, C., Matthes, H. W., Peluso, J., & Kieffer, B. L. (1998). Abolition of morphine-immunosuppression in mice lacking the mu-opioid receptor gene. Proceedings of the National Academy of Sciences of the United States of America, 95(11), 6326–6330.PubMed
51.
Zurück zum Zitat Rahim, R. T., Meissler, J. J., Zhang, L., Adler, M. W., Rogers, T. J., & Eisenstein, T. K. (2003). Withdrawal from morphine in mice suppresses splenic macrophage function, cytokine production, and costimulatory molecules. Journal of Neuroimmunology, 144(1–2), 16–27.PubMed Rahim, R. T., Meissler, J. J., Zhang, L., Adler, M. W., Rogers, T. J., & Eisenstein, T. K. (2003). Withdrawal from morphine in mice suppresses splenic macrophage function, cytokine production, and costimulatory molecules. Journal of Neuroimmunology, 144(1–2), 16–27.PubMed
52.
Zurück zum Zitat Colacchio, T. A., Yeager, M. P., & Hildebrandt, L. W. (1994). Perioperative immunomodulation in cancer surgery. American Journal of Surgery, 167(1), 174–179.PubMed Colacchio, T. A., Yeager, M. P., & Hildebrandt, L. W. (1994). Perioperative immunomodulation in cancer surgery. American Journal of Surgery, 167(1), 174–179.PubMed
53.
Zurück zum Zitat Benish, M., Bartal, I., Goldfarb, Y., Levi, B., Avraham, R., Raz, A., et al. (2008). Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis. Annals of Surgical Oncology, 15(7), 2042–2052.PubMed Benish, M., Bartal, I., Goldfarb, Y., Levi, B., Avraham, R., Raz, A., et al. (2008). Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis. Annals of Surgical Oncology, 15(7), 2042–2052.PubMed
54.
Zurück zum Zitat Glasner, A., Avraham, R., Rosenne, E., Benish, M., Zmora, O., Shemer, S., et al. (2010). Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a beta-adrenergic antagonist and a cyclooxygenase-2 inhibitor. Journal of Immunology, 184(5), 2449–2457. Glasner, A., Avraham, R., Rosenne, E., Benish, M., Zmora, O., Shemer, S., et al. (2010). Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a beta-adrenergic antagonist and a cyclooxygenase-2 inhibitor. Journal of Immunology, 184(5), 2449–2457.
55.
Zurück zum Zitat Hua, S., & Cabot, P. J. (2010). Mechanisms of peripheral immune-cell-mediated analgesia in inflammation: Clinical and therapeutic implications. Trends in Pharmacological Sciences, 31(9), 427–433.PubMed Hua, S., & Cabot, P. J. (2010). Mechanisms of peripheral immune-cell-mediated analgesia in inflammation: Clinical and therapeutic implications. Trends in Pharmacological Sciences, 31(9), 427–433.PubMed
56.
Zurück zum Zitat Cabot, P. J., Carter, L., Gaiddon, C., Zhang, Q., Schafer, M., Loeffler, J. P., et al. (1997). Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. Journal of Clinical Investigation, 100(1), 142–148.PubMed Cabot, P. J., Carter, L., Gaiddon, C., Zhang, Q., Schafer, M., Loeffler, J. P., et al. (1997). Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. Journal of Clinical Investigation, 100(1), 142–148.PubMed
57.
Zurück zum Zitat Finley, M. J., Happel, C. M., Kaminsky, D. E., & Rogers, T. J. (2008). Opioid and nociceptin receptors regulate cytokine and cytokine receptor expression. Cellular Immunology, 252(1–2), 146–154.PubMed Finley, M. J., Happel, C. M., Kaminsky, D. E., & Rogers, T. J. (2008). Opioid and nociceptin receptors regulate cytokine and cytokine receptor expression. Cellular Immunology, 252(1–2), 146–154.PubMed
58.
Zurück zum Zitat Martin, J. L., Charboneau, R., Barke, R. A., & Roy, S. (2010). Chronic morphine treatment inhibits LPS-induced angiogenesis: Implications in wound healing. Cellular Immunology, 265(2), 139–145.PubMed Martin, J. L., Charboneau, R., Barke, R. A., & Roy, S. (2010). Chronic morphine treatment inhibits LPS-induced angiogenesis: Implications in wound healing. Cellular Immunology, 265(2), 139–145.PubMed
59.
Zurück zum Zitat Stefano, G. B., Hartman, A., Bilfinger, T. V., Magazine, H. I., Liu, Y., Casares, F., et al. (1995). Presence of the mu3 opiate receptor in endothelial cells. Coupling to nitric oxide production and vasodilation. Journal of Biological Chemistry, 270(51), 30290–30293.PubMed Stefano, G. B., Hartman, A., Bilfinger, T. V., Magazine, H. I., Liu, Y., Casares, F., et al. (1995). Presence of the mu3 opiate receptor in endothelial cells. Coupling to nitric oxide production and vasodilation. Journal of Biological Chemistry, 270(51), 30290–30293.PubMed
60.
Zurück zum Zitat Gupta, K., Kshirsagar, S., Chang, L., Schwartz, R., Law, P. Y., Yee, D., et al. (2002). Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth. Cancer Research, 62(15), 4491–4498.PubMed Gupta, K., Kshirsagar, S., Chang, L., Schwartz, R., Law, P. Y., Yee, D., et al. (2002). Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth. Cancer Research, 62(15), 4491–4498.PubMed
61.
Zurück zum Zitat Fimiani, C., Mattocks, D., Cavani, F., Salzet, M., Deutsch, D. G., Pryor, S., et al. (1999). Morphine and anandamide stimulate intracellular calcium transients in human arterial endothelial cells: Coupling to nitric oxide release. Cellular Signalling, 11(3), 189–193.PubMed Fimiani, C., Mattocks, D., Cavani, F., Salzet, M., Deutsch, D. G., Pryor, S., et al. (1999). Morphine and anandamide stimulate intracellular calcium transients in human arterial endothelial cells: Coupling to nitric oxide release. Cellular Signalling, 11(3), 189–193.PubMed
62.
Zurück zum Zitat Hsiao, P. N., Chang, M. C., Cheng, W. F., Chen, C. A., Lin, H. W., Hsieh, C. Y., et al. (2009). Morphine induces apoptosis of human endothelial cells through nitric oxide and reactive oxygen species pathways. Toxicology, 256(1–2), 83–91.PubMed Hsiao, P. N., Chang, M. C., Cheng, W. F., Chen, C. A., Lin, H. W., Hsieh, C. Y., et al. (2009). Morphine induces apoptosis of human endothelial cells through nitric oxide and reactive oxygen species pathways. Toxicology, 256(1–2), 83–91.PubMed
63.
Zurück zum Zitat Chen, C., Farooqui, M., & Gupta, K. (2006). Morphine stimulates vascular endothelial growth factor-like signaling in mouse retinal endothelial cells. Current Neurovascular Research, 3(3), 171–180.PubMed Chen, C., Farooqui, M., & Gupta, K. (2006). Morphine stimulates vascular endothelial growth factor-like signaling in mouse retinal endothelial cells. Current Neurovascular Research, 3(3), 171–180.PubMed
64.
Zurück zum Zitat Singleton, P. A., Lingen, M. W., Fekete, M. J., Garcia, J. G. N., & Moss, J. (2006). Methylnaltrexone inhibits opiate and VEGF-induced angiogenesis: Role of receptor transactivation. Microvascular Research, 72(1–2), 3–11.PubMed Singleton, P. A., Lingen, M. W., Fekete, M. J., Garcia, J. G. N., & Moss, J. (2006). Methylnaltrexone inhibits opiate and VEGF-induced angiogenesis: Role of receptor transactivation. Microvascular Research, 72(1–2), 3–11.PubMed
65.
Zurück zum Zitat Belcheva, M. M., Haas, P. D., Tan, Y., Heaton, V. M., & Coscia, C. J. (2002). The fibroblast growth factor receptor is at the site of convergence between mu-opioid receptor and growth factor signaling pathways in rat C6 glioma cells. The Journal of Pharmacology and Experimental Therapeutics, 303(3), 909–918.PubMed Belcheva, M. M., Haas, P. D., Tan, Y., Heaton, V. M., & Coscia, C. J. (2002). The fibroblast growth factor receptor is at the site of convergence between mu-opioid receptor and growth factor signaling pathways in rat C6 glioma cells. The Journal of Pharmacology and Experimental Therapeutics, 303(3), 909–918.PubMed
66.
Zurück zum Zitat Ustun, F., Durmus-Altun, G., Altaner, S., Tuncbilek, N., Uzal, C., & Berkarda, S. (2011). Evaluation of morphine effect on tumour angiogenesis in mouse breast tumour model, EATC. Medical Oncology (in press). Ustun, F., Durmus-Altun, G., Altaner, S., Tuncbilek, N., Uzal, C., & Berkarda, S. (2011). Evaluation of morphine effect on tumour angiogenesis in mouse breast tumour model, EATC. Medical Oncology (in press).
67.
Zurück zum Zitat Poonawala, T., Levay-Young, B. K., Hebbel, R. P., & Gupta, K. (2005). Opioids heal ischemic wounds in the rat. Wound Repair and Regeneration, 13(2), 165–174.PubMed Poonawala, T., Levay-Young, B. K., Hebbel, R. P., & Gupta, K. (2005). Opioids heal ischemic wounds in the rat. Wound Repair and Regeneration, 13(2), 165–174.PubMed
68.
Zurück zum Zitat Singleton, P. A., Mambetsariev, N., Lennon, F. E., Mathew, B., Siegler, J. H., & Moreno-Vinasco, L. (2010). Methylnaltrexone potentiates the anti-angiogenic effects of mTOR inhibitors. J Angiogenes Res, 2(1), 5.PubMed Singleton, P. A., Mambetsariev, N., Lennon, F. E., Mathew, B., Siegler, J. H., & Moreno-Vinasco, L. (2010). Methylnaltrexone potentiates the anti-angiogenic effects of mTOR inhibitors. J Angiogenes Res, 2(1), 5.PubMed
69.
Zurück zum Zitat Dai, X., Song, H. J., Cui, S. G., Wang, T., Liu, Q., & Wang, R. (2010). The stimulative effects of endogenous opioids on endothelial cell proliferation, migration and angiogenesis in vitro. European Journal of Pharmacology, 628(1–3), 42–50.PubMed Dai, X., Song, H. J., Cui, S. G., Wang, T., Liu, Q., & Wang, R. (2010). The stimulative effects of endogenous opioids on endothelial cell proliferation, migration and angiogenesis in vitro. European Journal of Pharmacology, 628(1–3), 42–50.PubMed
70.
Zurück zum Zitat Farooqui, M., Li, Y., Rogers, T., Poonawala, T., Griffin, R. J., Song, C. W., et al. (2007). COX-2 inhibitor celecoxib prevents chronic morphine-induced promotion of angiogenesis, tumour growth, metastasis and mortality, without compromising analgesia. British Journal of Cancer, 97(11), 1523–1531.PubMed Farooqui, M., Li, Y., Rogers, T., Poonawala, T., Griffin, R. J., Song, C. W., et al. (2007). COX-2 inhibitor celecoxib prevents chronic morphine-induced promotion of angiogenesis, tumour growth, metastasis and mortality, without compromising analgesia. British Journal of Cancer, 97(11), 1523–1531.PubMed
71.
Zurück zum Zitat Radisavljevic, Z., Avraham, H., & Avraham, S. (2000). Vascular endothelial growth factor up-regulates ICAM-1 expression via the phosphatidylinositol 3 OH-kinase/AKT/Nitric oxide pathway and modulates migration of brain microvascular endothelial cells. The Journal of Biological Chemistry, 275(27), 20770–20774.PubMed Radisavljevic, Z., Avraham, H., & Avraham, S. (2000). Vascular endothelial growth factor up-regulates ICAM-1 expression via the phosphatidylinositol 3 OH-kinase/AKT/Nitric oxide pathway and modulates migration of brain microvascular endothelial cells. The Journal of Biological Chemistry, 275(27), 20770–20774.PubMed
72.
Zurück zum Zitat Kevil, C. G., Orr, A. W., Langston, W., Mickett, K., Murphy-Ullrich, J., Patel, R. P., et al. (2004). Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. The Journal of Biological Chemistry, 279(18), 19230–19238.PubMed Kevil, C. G., Orr, A. W., Langston, W., Mickett, K., Murphy-Ullrich, J., Patel, R. P., et al. (2004). Intercellular adhesion molecule-1 (ICAM-1) regulates endothelial cell motility through a nitric oxide-dependent pathway. The Journal of Biological Chemistry, 279(18), 19230–19238.PubMed
73.
Zurück zum Zitat Wu, Y., Ip, J. E., Huang, J., Zhang, L., Matsushita, K., Liew, C. C., et al. (2006). Essential role of ICAM-1/CD18 in mediating EPC recruitment, angiogenesis, and repair to the infarcted myocardium. Circulation Research, 99(3), 315–322.PubMed Wu, Y., Ip, J. E., Huang, J., Zhang, L., Matsushita, K., Liew, C. C., et al. (2006). Essential role of ICAM-1/CD18 in mediating EPC recruitment, angiogenesis, and repair to the infarcted myocardium. Circulation Research, 99(3), 315–322.PubMed
74.
Zurück zum Zitat Nair, M., Mahajan, S., & Reynolds, J. (2006). Opiates upregulate adhesion molecule expression in brain microvascular endothelial cells: Implications for altered blood brain barrier permeability. American Journal of Infectious Diseases, 2(2), 58–66. Nair, M., Mahajan, S., & Reynolds, J. (2006). Opiates upregulate adhesion molecule expression in brain microvascular endothelial cells: Implications for altered blood brain barrier permeability. American Journal of Infectious Diseases, 2(2), 58–66.
75.
Zurück zum Zitat Pasi, A., Qu, B. X., Steiner, R., Senn, H. J., Bar, W., & Messiha, F. S. (1991). Angiogenesis: Modulation with opioids. General Pharmacology, 22(6), 1077–1079.PubMed Pasi, A., Qu, B. X., Steiner, R., Senn, H. J., Bar, W., & Messiha, F. S. (1991). Angiogenesis: Modulation with opioids. General Pharmacology, 22(6), 1077–1079.PubMed
76.
Zurück zum Zitat Blebea, J., Mazo, J. E., Kihara, T. K., Vu, J. H., McLaughlin, P. J., Atnip, R. G., et al. (2000). Opioid growth factor modulates angiogenesis. Journal of Vascular Surgery, 32(2), 364–373.PubMed Blebea, J., Mazo, J. E., Kihara, T. K., Vu, J. H., McLaughlin, P. J., Atnip, R. G., et al. (2000). Opioid growth factor modulates angiogenesis. Journal of Vascular Surgery, 32(2), 364–373.PubMed
77.
Zurück zum Zitat Lam, C. F., Liu, Y. C., Tseng, F. L., Sung, Y. H., Huang, C. C., Jiang, M. J., et al. (2007). High-dose morphine impairs vascular endothelial function by increased production of superoxide anions. Anesthesiology, 106(3), 532–537.PubMed Lam, C. F., Liu, Y. C., Tseng, F. L., Sung, Y. H., Huang, C. C., Jiang, M. J., et al. (2007). High-dose morphine impairs vascular endothelial function by increased production of superoxide anions. Anesthesiology, 106(3), 532–537.PubMed
78.
Zurück zum Zitat Lam, C. F., Chang, P. J., Huang, Y. S., Sung, Y. H., Huang, C. C., Lin, M. W., et al. (2008). Prolonged use of high-dose morphine impairs angiogenesis and mobilization of endothelial progenitor cells in mice. Anesthesia and Analgesia, 107(2), 686–692.PubMed Lam, C. F., Chang, P. J., Huang, Y. S., Sung, Y. H., Huang, C. C., Lin, M. W., et al. (2008). Prolonged use of high-dose morphine impairs angiogenesis and mobilization of endothelial progenitor cells in mice. Anesthesia and Analgesia, 107(2), 686–692.PubMed
79.
Zurück zum Zitat Martin, J. L., Koodie, L., Krishnan, A. G., Charboneau, R., Barke, R. A., & Roy, S. (2010). Chronic morphine administration delays wound healing by inhibiting immune cell recruitment to the wound site. The American Journal of Pathology, 176(2), 786–799.PubMed Martin, J. L., Koodie, L., Krishnan, A. G., Charboneau, R., Barke, R. A., & Roy, S. (2010). Chronic morphine administration delays wound healing by inhibiting immune cell recruitment to the wound site. The American Journal of Pathology, 176(2), 786–799.PubMed
80.
Zurück zum Zitat Balasubramanian, S., Ramakrishnan, S., Charboneau, R., Wang, J., Barke, R. A., & Roy, S. (2001). Morphine sulfate inhibits hypoxia-induced vascular endothelial growth factor expression in endothelial cells and cardiac myocytes. Journal of Molecular and Cellular Cardiology, 33(12), 2179–2187.PubMed Balasubramanian, S., Ramakrishnan, S., Charboneau, R., Wang, J., Barke, R. A., & Roy, S. (2001). Morphine sulfate inhibits hypoxia-induced vascular endothelial growth factor expression in endothelial cells and cardiac myocytes. Journal of Molecular and Cellular Cardiology, 33(12), 2179–2187.PubMed
81.
Zurück zum Zitat Roy, S., Balasubramanian, S., Wang, J., Chandrashekhar, Y., Charboneau, R., & Barke, R. (2003). Morphine inhibits VEGF expression in myocardial ischemia. Surgery, 134(2), 336–344.PubMed Roy, S., Balasubramanian, S., Wang, J., Chandrashekhar, Y., Charboneau, R., & Barke, R. (2003). Morphine inhibits VEGF expression in myocardial ischemia. Surgery, 134(2), 336–344.PubMed
82.
Zurück zum Zitat Koodie, L., Ramakrishnan, S., & Roy, S. (2010). Morphine suppresses tumor angiogenesis through a HIF-1alpha/p38MAPK pathway. The American Journal of Pathology, 177(2), 984–997.PubMed Koodie, L., Ramakrishnan, S., & Roy, S. (2010). Morphine suppresses tumor angiogenesis through a HIF-1alpha/p38MAPK pathway. The American Journal of Pathology, 177(2), 984–997.PubMed
83.
Zurück zum Zitat Faramarzi, N., Abbasi, A., Tavangar, S. M., Mazouchi, M., & Dehpour, A. R. (2009). Opioid receptor antagonist promotes angiogenesis in bile duct ligated rats. Journal of Gastroenterology and Hepatology, 24(7), 1226–1229.PubMed Faramarzi, N., Abbasi, A., Tavangar, S. M., Mazouchi, M., & Dehpour, A. R. (2009). Opioid receptor antagonist promotes angiogenesis in bile duct ligated rats. Journal of Gastroenterology and Hepatology, 24(7), 1226–1229.PubMed
84.
Zurück zum Zitat Beilin, B., Shavit, Y., Trabekin, E., Mordashev, B., Mayburd, E., Zeidel, A., et al. (2003). The effects of postoperative pain management on immune response to surgery. Anesthesia and Analgesia, 97(3), 822–827.PubMed Beilin, B., Shavit, Y., Trabekin, E., Mordashev, B., Mayburd, E., Zeidel, A., et al. (2003). The effects of postoperative pain management on immune response to surgery. Anesthesia and Analgesia, 97(3), 822–827.PubMed
85.
Zurück zum Zitat Bar-Yosef, S., Melamed, R., Page, G. G., Shakhar, G., Shakhar, K., & Ben-Eliyahu, S. (2001). Attenuation of the tumor-promoting effect of surgery by spinal blockade in rats. Anesthesiology, 94(6), 1066–1073.PubMed Bar-Yosef, S., Melamed, R., Page, G. G., Shakhar, G., Shakhar, K., & Ben-Eliyahu, S. (2001). Attenuation of the tumor-promoting effect of surgery by spinal blockade in rats. Anesthesiology, 94(6), 1066–1073.PubMed
86.
Zurück zum Zitat Wada, H., Seki, S., Takahashi, T., Kawarabayashi, N., Higuchi, H., Habu, Y., et al. (2007). Combined spinal and general anesthesia attenuates liver metastasis by preserving TH1/TH2 cytokine balance. Anesthesiology, 106(3), 499–506.PubMed Wada, H., Seki, S., Takahashi, T., Kawarabayashi, N., Higuchi, H., Habu, Y., et al. (2007). Combined spinal and general anesthesia attenuates liver metastasis by preserving TH1/TH2 cytokine balance. Anesthesiology, 106(3), 499–506.PubMed
87.
Zurück zum Zitat Exadaktylos, A. K., Buggy, D. J., Moriarty, D. C., Mascha, E., & Sessler, D. I. (2006). Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis? Anesthesiology, 105(4), 660–664.PubMed Exadaktylos, A. K., Buggy, D. J., Moriarty, D. C., Mascha, E., & Sessler, D. I. (2006). Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis? Anesthesiology, 105(4), 660–664.PubMed
88.
Zurück zum Zitat Biki, B., Mascha, E., Moriarty, D. C., Fitzpatrick, J. M., Sessler, D. I., & Buggy, D. J. (2008). Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: A retrospective analysis. Anesthesiology, 109(2), 180–187.PubMed Biki, B., Mascha, E., Moriarty, D. C., Fitzpatrick, J. M., Sessler, D. I., & Buggy, D. J. (2008). Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: A retrospective analysis. Anesthesiology, 109(2), 180–187.PubMed
89.
Zurück zum Zitat Christopherson, R., James, K. E., Tableman, M., Marshall, P., & Johnson, F. E. (2008). Long-term survival after colon cancer surgery: A variation associated with choice of anesthesia. Anesthesia and Analgesia, 107(1), 325–332.PubMed Christopherson, R., James, K. E., Tableman, M., Marshall, P., & Johnson, F. E. (2008). Long-term survival after colon cancer surgery: A variation associated with choice of anesthesia. Anesthesia and Analgesia, 107(1), 325–332.PubMed
90.
Zurück zum Zitat Gottschalk, A., Ford, J. G., Regelin, C. C., You, J., Mascha, E. J., Sessler, D. I., et al. (2010). Association between epidural analgesia and cancer recurrence after colorectal cancer surgery. Anesthesiology, 113(1), 27–34.PubMed Gottschalk, A., Ford, J. G., Regelin, C. C., You, J., Mascha, E. J., Sessler, D. I., et al. (2010). Association between epidural analgesia and cancer recurrence after colorectal cancer surgery. Anesthesiology, 113(1), 27–34.PubMed
91.
Zurück zum Zitat Tsui, B. C., Rashiq, S., Schopflocher, D., Murtha, A., Broemling, S., Pillay, J., et al. (2010). Epidural anesthesia and cancer recurrence rates after radical prostatectomy. Canadian Journal of Anaesthesia, 57(2), 107–112.PubMed Tsui, B. C., Rashiq, S., Schopflocher, D., Murtha, A., Broemling, S., Pillay, J., et al. (2010). Epidural anesthesia and cancer recurrence rates after radical prostatectomy. Canadian Journal of Anaesthesia, 57(2), 107–112.PubMed
92.
Zurück zum Zitat Sessler, D. I., Ben-Eliyahu, S., Mascha, E. J., Parat, M. O., & Buggy, D. J. (2008). Can regional analgesia reduce the risk of recurrence after breast cancer? Methodology of a multicenter randomized trial. Contemp Clin Trials, 29(4), 517–526.PubMed Sessler, D. I., Ben-Eliyahu, S., Mascha, E. J., Parat, M. O., & Buggy, D. J. (2008). Can regional analgesia reduce the risk of recurrence after breast cancer? Methodology of a multicenter randomized trial. Contemp Clin Trials, 29(4), 517–526.PubMed
93.
Zurück zum Zitat O’Riain, S. C., Buggy, D. J., Kerin, M. J., Watson, R. W., & Moriarty, D. C. (2005). Inhibition of the stress response to breast cancer surgery by regional anesthesia and analgesia does not affect vascular endothelial growth factor and prostaglandin E2. Anesthesia and Analgesia, 100(1), 244–249.PubMed O’Riain, S. C., Buggy, D. J., Kerin, M. J., Watson, R. W., & Moriarty, D. C. (2005). Inhibition of the stress response to breast cancer surgery by regional anesthesia and analgesia does not affect vascular endothelial growth factor and prostaglandin E2. Anesthesia and Analgesia, 100(1), 244–249.PubMed
94.
Zurück zum Zitat Looney, M., Doran, P., & Buggy, D. J. (2010). Effect of anesthetic technique on serum vascular endothelial growth factor C and transforming growth factor beta in women undergoing anesthesia and surgery for breast cancer. Anesthesiology, 113(5), 1118–1125.PubMed Looney, M., Doran, P., & Buggy, D. J. (2010). Effect of anesthetic technique on serum vascular endothelial growth factor C and transforming growth factor beta in women undergoing anesthesia and surgery for breast cancer. Anesthesiology, 113(5), 1118–1125.PubMed
95.
Zurück zum Zitat Deegan, C. A., Murray, D., Doran, P., Moriarty, D. C., Sessler, D. I., Mascha, E., et al. (2010). Anesthetic technique and the cytokine and matrix metalloproteinase response to primary breast cancer surgery. Regional Anesthesia and Pain Medicine, 35(6), 490–495.PubMed Deegan, C. A., Murray, D., Doran, P., Moriarty, D. C., Sessler, D. I., Mascha, E., et al. (2010). Anesthetic technique and the cytokine and matrix metalloproteinase response to primary breast cancer surgery. Regional Anesthesia and Pain Medicine, 35(6), 490–495.PubMed
96.
Zurück zum Zitat Deegan, C. A., Murray, D., Doran, P., Ecimovic, P., Moriarty, D. C., & Buggy, D. J. (2009). Effect of anaesthetic technique on oestrogen receptor-negative breast cancer cell function in vitro. British Journal of Anaesthesia, 103(5), 685–690.PubMed Deegan, C. A., Murray, D., Doran, P., Ecimovic, P., Moriarty, D. C., & Buggy, D. J. (2009). Effect of anaesthetic technique on oestrogen receptor-negative breast cancer cell function in vitro. British Journal of Anaesthesia, 103(5), 685–690.PubMed
97.
Zurück zum Zitat Singleton, P. A., & Moss, J. (2010). Effect of perioperative opioids on cancer recurrence: A hypothesis. Future Oncology, 6(8), 1237–1242.PubMed Singleton, P. A., & Moss, J. (2010). Effect of perioperative opioids on cancer recurrence: A hypothesis. Future Oncology, 6(8), 1237–1242.PubMed
98.
Zurück zum Zitat Ishikawa, M., Tanno, K., Kamo, A., Takayanagi, Y., & Sasaki, K. (1993). Enhancement of tumor growth by morphine and its possible mechanism in mice. Biological & Pharmaceutical Bulletin, 16(8), 762–766. Ishikawa, M., Tanno, K., Kamo, A., Takayanagi, Y., & Sasaki, K. (1993). Enhancement of tumor growth by morphine and its possible mechanism in mice. Biological & Pharmaceutical Bulletin, 16(8), 762–766.
99.
Zurück zum Zitat Maneckjee, R., & Minna, J. D. (1994). Opioids induce while nicotine suppresses apoptosis in human lung cancer cells. Cell Growth & Differentiation, 5(10), 1033–1040. Maneckjee, R., & Minna, J. D. (1994). Opioids induce while nicotine suppresses apoptosis in human lung cancer cells. Cell Growth & Differentiation, 5(10), 1033–1040.
100.
Zurück zum Zitat Kawase, M., Sakagami, H., Furuya, K., Kikuchi, H., Nishikawa, H., & Motohashi, N. (2002). Cell death-inducing activity of opiates in human oral tumor cell lines. Anticancer Research, 22(1A), 211–214.PubMed Kawase, M., Sakagami, H., Furuya, K., Kikuchi, H., Nishikawa, H., & Motohashi, N. (2002). Cell death-inducing activity of opiates in human oral tumor cell lines. Anticancer Research, 22(1A), 211–214.PubMed
101.
Zurück zum Zitat Yeager, M. P., & Colacchio, T. A. (1991). Effect of morphine on growth of metastatic colon cancer in vivo. Archives of Surgery, 126(4), 454–456.PubMed Yeager, M. P., & Colacchio, T. A. (1991). Effect of morphine on growth of metastatic colon cancer in vivo. Archives of Surgery, 126(4), 454–456.PubMed
102.
Zurück zum Zitat Page, G. G., Ben-Eliyahu, S., Yirmiya, R., & Liebeskind, J. C. (1993). Morphine attenuates surgery-induced enhancement of metastatic colonization in rats. Pain, 54(1), 21–28.PubMed Page, G. G., Ben-Eliyahu, S., Yirmiya, R., & Liebeskind, J. C. (1993). Morphine attenuates surgery-induced enhancement of metastatic colonization in rats. Pain, 54(1), 21–28.PubMed
103.
Zurück zum Zitat Page, G. G., Ben-Eliyahu, S., & Liebeskind, J. C. (1994). The role of LGL/NK cells in surgery-induced promotion of metastasis and its attenuation by morphine. Brain, Behavior, and Immunity, 8(3), 241–250.PubMed Page, G. G., Ben-Eliyahu, S., & Liebeskind, J. C. (1994). The role of LGL/NK cells in surgery-induced promotion of metastasis and its attenuation by morphine. Brain, Behavior, and Immunity, 8(3), 241–250.PubMed
104.
Zurück zum Zitat Page, G. G., McDonald, J. S., & Ben-Eliyahu, S. (1998). Pre-operative versus postoperative administration of morphine: Impact on the neuroendocrine, behavioural, and metastatic-enhancing effects of surgery. British Journal of Anaesthesia, 81(2), 216–223.PubMed Page, G. G., McDonald, J. S., & Ben-Eliyahu, S. (1998). Pre-operative versus postoperative administration of morphine: Impact on the neuroendocrine, behavioural, and metastatic-enhancing effects of surgery. British Journal of Anaesthesia, 81(2), 216–223.PubMed
105.
Zurück zum Zitat Fuggetta, M. P., Di, F. P., Falchetti, R., Cottarelli, A., Rossi, L., Tricarico, M., et al. (2005). Effect of morphine on cell-mediated immune responses of human lymphocytes against allogeneic malignant cells. Journal of Experimental & Clinical Cancer Research, 24(2), 255–263. Fuggetta, M. P., Di, F. P., Falchetti, R., Cottarelli, A., Rossi, L., Tricarico, M., et al. (2005). Effect of morphine on cell-mediated immune responses of human lymphocytes against allogeneic malignant cells. Journal of Experimental & Clinical Cancer Research, 24(2), 255–263.
106.
Zurück zum Zitat Sasamura, T., Nakamura, S., Iida, Y., Fujii, H., Murata, J., Saiki, I., et al. (2002). Morphine analgesia suppresses tumor growth and metastasis in a mouse model of cancer pain produced by orthotopic tumor inoculation. European Journal of Pharmacology, 441(3), 185–191.PubMed Sasamura, T., Nakamura, S., Iida, Y., Fujii, H., Murata, J., Saiki, I., et al. (2002). Morphine analgesia suppresses tumor growth and metastasis in a mouse model of cancer pain produced by orthotopic tumor inoculation. European Journal of Pharmacology, 441(3), 185–191.PubMed
107.
Zurück zum Zitat Dai, X., Cui, S. G., Wang, T., Liu, Q., Song, H. J., & Wang, R. (2008). Endogenous opioid peptides, endomorphin-1 and -2 and deltorphin I, stimulate angiogenesis in the CAM assay. European Journal of Pharmacology, 579(1–3), 269–275.PubMed Dai, X., Cui, S. G., Wang, T., Liu, Q., Song, H. J., & Wang, R. (2008). Endogenous opioid peptides, endomorphin-1 and -2 and deltorphin I, stimulate angiogenesis in the CAM assay. European Journal of Pharmacology, 579(1–3), 269–275.PubMed
Metadaten
Titel
Morphine and tumor growth and metastasis
verfasst von
Banafsheh Afsharimani
Peter Cabot
Marie-Odile Parat
Publikationsdatum
01.06.2011
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 2/2011
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-011-9285-0

Weitere Artikel der Ausgabe 2/2011

Cancer and Metastasis Reviews 2/2011 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

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