Opioid effects on tumor growth have been a controversial topic of discussion. In the present study, morphine inhibited tumor cell proliferation at concentrations of ≥10 μm. This was primarily caused by inhibition of cell cycle progression from G1 to S phase. At higher concentrations (≥500 μm for 24 h), morphine also caused cell death. In nude mice, morphine significantly reduced the growth of MCF-7 and MDA-MB231 tumors but had no effect on HT-29 tumor growth. In these experiments, morphine plasma concentrations were similar to those found in cancer patients receiving chronic morphine treatment for pain relief (0.9–3.4 μm). In MCF-7 and MDA-MB231 cells, morphine caused a naloxone (Nx)- and pertussis toxin-sensitive, concentration-dependent increase of GTPase activity, indicating that morphine signals could be transduced by opioid receptors via a G protein. However, the antiproliferative effects of morphine were not antagonized by Nx, pertussis toxin, forskolin, and 8-bromo-cAMP, suggesting that the typical opioid receptor-coupled signaling cascade involving the Gi, adenylyl cyclase, and protein kinase A was not involved. Instead, morphine caused an NH2-terminal phosphorylation of p53 at Ser9 and/or Ser15 and a stabilization of p53 in MCF-7 cells that express wild-type p53. p53 phosphorylation was not antagonized by Nx and resulted in an increase of p53-dependent proteins including p21, Bax, and the death receptor Fas. Blockade of Fas by Fas-fusion protein or inhibition of caspase 8 resulted in a partial inhibition of morphine-induced apoptosis. In addition, Fas ligand only induced apoptosis when administered together with morphine. However, the sensitivity of the tumor cells toward Fas ligand remained low. HT-29 cells, which express dominant negative p53 and show no increase of GTPase activity when treated with morphine, were less sensitive in vitro and were not affected in vivo. Our results suggest that morphine, alone or in combination with Nx, may reduce the growth of certain tumors, apparently in part through activation of p53.

Most cancer patients suffer from severe pain. However, physicians often avoid the prescription of potent opioid analgesics, in part for fear of side effects. This attitude is strengthened by a recent study (1) that was published during the review process of the present article. This study (1) shows that morphine promotes the growth of breast cancer xenografts in nude mice by increasing angiogenesis. In addition, tumor-promoting effects of morphine have been found in mice that received an injection with leukemia or sarcoma cells (2). Because morphine caused suppression of concanavalin A-induced proliferation of lymphocytes in this study, the tumor-promoting effects were suggested to be due to an inhibition of the cellular immune response (2). On the other hand, some opioids [methadone, morphine, and buprenorphine (3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13)] and opioid peptides (14, 15, 16, 17, 18) were found to inhibit tumor cell proliferation and in vivo tumor growth in various models. The reasons for these conflicting results are unclear. It has been suggested that “atypical” opioid binding sites might be involved in tumor suppression because in some studies, the antiproliferative effects were not antagonized with Nx3(1, 5, 7, 10). However, this was not unequivocally confirmed in other studies (2, 6, 8). Hence, it is not known whether the observed tumor-promoting and/or tumor-suppressing effects of opioids are mediated through the well-characterized opioid-mediated intracellular signaling pathway that involves activation of a PTX-sensitive inhibitory G protein (Gi; Ref. 19), inhibition of adenylyl cyclase, and decrease of cAMP levels and thereby inhibition of PKA. In the present study, we primarily addressed the potential involvement of Gi and p53 for effects of morphine on tumor growth.

Cell Culture and Reagents

MCF-7 [wild-type p53, estrogen receptor positive (20)] and MDA-MB231 [p53 mutation (21), estrogen receptor negative (22)] human breast cancer cells were cultured in DMEM and RPMI 1640, respectively, supplemented with 10% FCS and 1% penicillin/streptomycin at 37°C in a 5% CO2 atmosphere. HT-29 colon cancer cells [dominant negative p53 (20)] were cultured in McCoy’s medium.

DAMGO, Nx, and actinomycin D were from Sigma (Steinheim, Germany). Morphine-sulfate trihydrate was from Merck (Darmstadt, Germany). The drugs were dissolved in water. Forskolin (adenylyl cyclase activator), recombinant human FasL, and Fas-fusion protein were from Alexis (Grünberg, Germany). The caspase 3, 8, and 9 inhibitors Z-DEVD-FMK, Z-IETD-FMK, and Z-LEHD-FMK, respectively, and 8-bromo-cAMP (PKA activator) were from Calbiochem (Schwalbach, Germany). Antibodies were from Santa Cruz Biotechnology (p21, Fas, FasL, Bax, Erk-2, PARP and secondary antibodies; Heidelberg, Germany), Gramsch Laboratories (μ- and δ-opioid receptor; Schwabhausen, Germany), and Cell Signaling (phospho-p53 and p53; Frankfurt, Germany).

Proliferation Assay

Cells (3 × 105) were treated for 24, 48, and 72 h; harvested by trypsinization; stained with trypan blue; and counted by a blinded observer.

Colony Forming Assay

Cells (500 cells/3-cm dish) were treated for 10 days, fixed with 100% methanol, and stained with 0.5% crystal violet, 5% Giemsa solution. Single cell clones were counted using QuantityOne (Bio-Rad, München, Germany). The IC50 was calculated using a standard sigmoidal Emax model.

Steady-state GTPase Activity Assay

Cell membranes were prepared as described previously (23). Membranes (100 ng of protein) were incubated for 10 min at room temperature in the presence or absence of morphine or morphine + Nx. The incubation (50 μl, final volume) also contained 50 mm Tris (pH 7.4), 100 μm EDTA, 1 mm DTT, 150 μm MgCl2, 1 mg/ml BSA, and a protease inhibitor mixture (5 mm benzamidine, 20 μg/ml lima bean trypsin inhibitor, 20 μg/ml leupeptin, and 20 μg/ml aprotinin). To the preincubated reaction mixture, 100 nm [γ-32P]GTP was added. Hydrolysis of [γ-32P]GTP was measured as described previously (24). Reactions were stopped after 20 min by transferring the assay mixture into tubes containing 0.75 ml of ice-cold 5% (w/v) Norit A in 50 mm NaH2PO4 (pH 8.0). After centrifugation, aliquots (500 μl) of the supernatant were decanted for determination of the [32P]phosphate content by scintillation counting.

Western Blot Analysis

Cells were harvested by scraping them into 200 μl of lysis buffer [135 mm NaCl, 25 mm β-glycerophosphate, 20 mm Tris, 2 mm EDTA, 2 mm Na PPi, 2 mm DTT, 1 mm Na3VO4, 10% glycerol, 1% Triton X-100, 2 μg/ml aprotinin, and 5 μg/ml leupeptin (pH 7.5)]. The protein content was determined using the Bradford method. Proteins were separated electrophoretically by SDS-PAGE and transferred onto nitrocellulose membranes by semidry blotting. Membranes were incubated with primary and secondary antibodies according to standard procedures, and protein-antibody complexes were visualized with enhanced chemiluminescence (Amersham-Pharmacia, Freiburg, Germany).

RT-PCR

A one-tube RT-PCR system (Roche Diagnostics, Mannheim, Germany) was used according to the manufacturer’s instructions. PCR conditions were as follows: 30 min at 50°C; 35 cycles of 94°C for 1 min, 57°C (Fas) or 52°C (β-actin) for 1 min, and 72°C for 1.5 min; and 7 min at 72°C. Amplification products were separated by 1.2% agarose gel electrophoresis and visualized by ethidium bromide staining. Amplification of Fas mRNA was confirmed by DNA sequencing.

Primers

Primers were as follows: Fas, 5′-CTGTATGTGAACACTGTGACCC-3′ (forward) and 5′-AATTTATTGCCACTGTTTCAGG-3′ (reverse); and β-actin, 5′-CCGGATCCTCTTTGCTACTGAGACAGG-3′ (forward) and 5′-CCGAATTCGGGATCTGAATGCAATGTT-3′ (reverse).

Flow Cytometry

Cell Cycle Distribution.

Cells (5 × 105) were starved for 72 h and then treated in full medium for 24 or 48 h. Cells were harvested by trypsinization, fixed with ice-cold 80% ethanol, and kept at −20°C for 24 h. Cells were stained with PI, and the DNA content was determined using flow cytometry (Becton Dickinson). The cell cycle distribution was assessed with WinMDI 2.8.

Apoptosis.

MCF-7 cells (1 × 106) were treated for 24 h and harvested by trypsinization. Cells were stained with phycoerythrin-labeled annexin V and 7-AAD, and fluorescence was measured by flow cytometry. Ten thousand cells were counted.

Tumor Growth in Nude Mice

Tumor cells (5 × 106 MCF-7 cells, 1 × 107 MDA-MB231 cells, and 1 × 107 HT-29 cells in 150 μl of medium per mouse) were injected s.c. at the right dorsal flank of NMRI-nu/nu female mice (20–25 g; Harlan Winkelmann GmbH, Borchen, Germany). In case of MCF-7 cells, mice received 10 mg/kg β-estradiolvalerate i.m. at day 1, 7, and 14. Mice were treated with morphine, Nx, and morphine + Nx or vehicle (5% glucose/Ringer lactate) starting right after tumor cell implantation (8–15 mice/group). Because of a potential desensitization of opioid receptors, the dose of morphine and Nx was increased stepwise (10, 20, and 30 mg/kg i.p. for the first, second, and third week). For the drug combination, the Nx dose was one-tenth of the morphine dose because this ratio is generally considered to result in a complete antagonism of antinociceptive effects of morphine (25). The tumor volume was assessed three times a week [tumor volume = (long diameter × short diameter2)/2] for 2 or 3 weeks. The experiments were approved by the local ethics committee for animal research. Morphine plasma concentrations were analyzed by a liquid chromatography tandem mass spectrometry method as described previously (26).

Statistics

Data are presented as mean ± SE. For statistical comparisons, univariate ANOVA was performed. Subsequently, treatment groups were mutually compared using t tests with a Bonferroni α correction for multiple comparisons (α at 0.05). For time courses, the area under the curve was calculated, and areas under the curve were then submitted to one-way ANOVA.

Inhibition of Tumor Cell Proliferation and Survival.

Cell proliferation was significantly reduced at ≥10 μm morphine in MCF-7 cells (Fig. 1 A). Nx (250 μm) significantly reduced the proliferation in MCF-7 cells but had no significant effect in MDA-MB231 and HT-29 cells. The combination of morphine (250 μm) and Nx (250 μm) was as effective as either agent alone in MDA-MB231 cells and was more potent than either agent alone in MCF-7 and HT-29 cells. The effects of the selective μ-agonist DAMGO were indistinguishable from those of morphine.

In the colony forming assay (Fig. 1 B), the IC50 for morphine was 144.0 ± 23.3, 104.6 ± 18.7, and 182.9 ± 7.3 μm in MCF-7, MDA-MB231, and HT-29 cells, respectively. When morphine was combined 1:1 with Nx, the concentration effect curve was shifted to the left, indicating increased potency. The IC50 for the mixture of each drag of morphine and Nx was 30.7 ± 1.2, 10.6 ± 0.9, and 32.3 ± 10.6 μm in MCF-7, MDA-MB231, and HT-29 cells, respectively.

Inhibition of Cell Cycle Progression and Induction of Apoptosis.

Flow cytometry (Fig. 2 A) revealed a dose-dependent relative increase of the number of cells in the G1 phase after morphine treatment. This indicates that morphine inhibited the progression from G1 to S phase and caused a G1 block. In MDA-MB231 cells, morphine also increased the number of sub-G1 cells, indicating apoptosis. Nx caused a G1 block in MCF-7 and HT-29 cells but had no obvious effect in MDA-MB231 cells. Again, morphine + Nx was more effective than either agent alone. The mixture caused a considerable increase of sub-G1 cells in all three cell lines. Nx was added 10 min before morphine in all combination experiments.

PARP cleavage was determined by Western blot analysis to further assess induction of apoptosis (Fig. 2 B). Both morphine and Nx caused a PARP cleavage at 2 or 3 mm for 24 h in MDA-MB231 and MCF-7 cells, respectively. In HT-29 cells, PARP cleavage occurred at 3 mm for 48 h. Again, effects of the mixture were somewhat stronger than those of either agent alone.

In MCF-7 cells, we additionally assessed the binding of annexin V to phosphatidylserine of the plasma membrane and 7-AAD DNA binding by means of flow cytometry (Fig. 2 C). Both morphine and Nx caused an increase of late apoptotic cells (stained with 7-AAD and annexin V) and necrotic cells (stained with 7-AAD) at concentrations of 3 mm. Effects of morphine were not antagonized by Nx or vice versa.

Inhibition of the effector caspase 3 caused a near complete inhibition of morphine-induced apoptosis in MDA-MB231 and HT-29 cells, as indicated by a disappearance of sub-G1 cells in cell cycle measurements (Fig. 2 D). Because MCF-7 cells lack caspase 3, inhibition of this effector caspase had no effect in these cells. Inhibition of caspase 8, which is primarily activated upon death receptor stimulation, caused a 50–70% inhibition of apoptosis in all three cell lines. Inhibition of caspase 9, which is activated in the course of the mitochondrial apoptosis cascade, caused a reduction of morphine-induced apoptosis by 30–50% in all three cell lines.

Inhibition of Tumor Growth in Nude Mice.

In mice treated with morphine, growth of MCF-7 and MDA-MB231 tumors was significantly reduced as compared with that seen in control mice (P = 0.001 for both cell lines; Fig. 3 A). Tumor growth was also inhibited by the combination of morphine + Nx in MCF-7 (P < 0.001) and MDA-MB231 tumors (P < 0.05), whereas Nx itself had no significant effect. HT-29 tumors were resistant to all treatments.

Plasma morphine concentrations were determined after administration of the last dose (Fig. 3 B). Peak morphine plasma concentrations ranged from 50–60 μm at 10–25 min after i.p. morphine injection. The elimination half-life was approximately 18 min. Concentrations were in the range of 0.9–3.4 μm between 1 and 2 h. These concentrations are similar to those found in cancer patients chronically receiving oral morphine for pain management (27, 28).

Potential Mechanisms: G Protein-dependent Pathway.

Morphine stimulated the steady-state GTPase activity in MDA-MB231 cell membranes with increasing concentrations (Fig. 4,A) and reached a maximum at 0.1–10 μm. Similar results were obtained in MCF-7 cells (data not shown). There was no increase of GTPase activity with HT-29 membranes (data not shown), although all three cell lines show immunoreactivity for at least one type of opioid receptor (Fig. 4 B). In MDA-MB231 cells pretreated with PTX (200 ng/ml for 24 h), the morphine-dependent increase in GTPase activity was significantly reduced. Addition of equimolar concentrations of Nx abolished morphine-stimulated steady-state GTPase activity.

To evaluate whether the typical opioid receptor-coupled signaling cascade was responsible for the growth-inhibitory effects of morphine, various antagonists were evaluated by flow cytometry. Neither PTX (200 ng/ml for 24 h), forskolin (5 μm for 24 h), nor 8-bromo-cAMP (10 μm for 24 h) was able to antagonize the morphine-induced cell cycle arrest (Fig. 4 C). The GTPase data suggest that morphine could signal through activation of a PTX-sensitive G protein in MDA-MB231 and MCF-7 cells as described for other cells expressing opioid receptors. However, because PTX and Nx abolished the morphine-induced Gi activation but not the morphine-induced growth inhibition, cell cycle arrest and apoptosis are obviously Gi independent.

Phosphorylation of p53 and Up-Regulation of p53-dependent Proteins.

Differences between MCF-7 (wild-type p53), MDA-MB231 (p53 mutation), and HT-29 (dominant negative p53) cells suggested that the p53 status might be important for the effects of morphine. Therefore, we assessed p53 and phospho-p53 (Ser15, Ser9) levels by means of Western blot analysis. Morphine and DAMGO caused an NH2-terminal phosphorylation of p53 at Ser15 in MCF-7 cells and at Ser9 in MDA-MB231 cells (Fig. 5,A, left panel and right panel, respectively). In MCF-7 cells, p53 phosphorylation was associated with an increase of total p53 protein levels (Fig. 5,B). Because p53 mRNA remained constant (data not shown), this was probably caused by a stabilization of p53 (29). The combination of morphine and Nx also caused a p53 stabilization, whereas Nx alone had no effect (Fig. 5 B, right panel). In MDA-MB231 cells, total p53 protein remained constant (data not shown).

Morphine-induced p53 phosphorylation was associated with an increase of the p53-dependent proteins p21 (cell cycle inhibitor) and Bax (proapoptotic mitochondrial protein; Fig. 5 C). Similar effects were observed with morphine + Nx. Nx alone had no effect.

Increase of Death Receptor Expression.

Because inhibition of caspase 8 was able to reduce morphine-induced apoptosis (Fig. 2 D), death receptors were likely to be involved. p53 regulates the transcription of Fas (30), DR5 [tumor necrosis factor-related apoptosis-inducing ligand receptor (31, 32)], the tumor necrosis factor α receptor (33), and Pidd, a new death domain-containing protein (34). Of these targets, we primarily addressed Fas because previous studies with lymphocytes and neurons have suggested that morphine might induce Fas expression (35, 36, 37).

CD95/Fas protein expression was found in untreated MDA-MB231 and HT-29 cells but was minimal in untreated MCF-7 cells (Western blot analysis; Fig. 6,A), suggesting that MCF-7 cells in particular down-regulate Fas, which is a common feature of tumor cells (38, 39). Therefore, we used this cell line to assess the effects of opioid treatment. DAMGO (500 μm) caused a rapid increase of Fas mRNA (RT-PCR; Fig. 6,B). Its effects were stronger than those of 1 μg/ml actinomycin D (positive control) and were not antagonized by Nx. Fas protein levels were increased at 6 h after DAMGO treatment (Fig. 6 C).

If cells were treated with morphine together with Fas-fusion protein (200 ng/ml), which inhibits the activation of the Fas receptor, morphine-induced apoptosis was partly antagonized (Fig. 6,D). However, Fas-fusion protein was less antagonistic than the caspase 8 inhibitor (Fig. 2,D). On the other hand, treatment with recombinant human FasL (60 ng/ml) caused apoptosis only in the presence of morphine at morphine concentrations that per se did not cause apoptosis (Fig. 6 D). However, the sensitivity of the tumor cells toward FasL remained low even in the presence of morphine.

Previously, morphine was found to cause a delay of normal cell death in neurons (40), to protect astrocytes from apoptosis-promoting agents (41), and to increase the proliferation of endothelial and tumor cells (1, 2, 42, 43). On the other hand, opioids were also found to promote cell death in lymphocytes (44, 45, 46), tumor cells (3, 4, 5, 6, 8, 10, 47, 48, 49, 50), and neurons (51, 52, 53). Effects of Nx were highly controversial [antagonism (8, 42, 43) or no antagonism (1, 5, 7)]. The protective effects of morphine have been linked to a Gβ/γ-mediated activation of phosphatidylinositol 3′-kinase (54), PKB/Akt (54), and Erks [Erk-1 and Erk-2 (1)]. On the other hand, the proapoptotic effects of morphine have been suggested to involve activation of p38 mitogen-activated protein kinase (37), nuclear factor κB inhibition (3), or up-regulation of death receptors (35, 36). In the present study, antiproliferative effects of morphine were associated with p53 activation and up-regulation of p53-dependent genes, including CD95/Fas. Although morphine facilitated FasL-evoked apoptosis, the sensitivity toward this death receptor ligand remained low even in the presence of morphine, suggesting that up-regulation of the Fas receptor might contribute in part to the growth-inhibitory effects of morphine but probably does not represent the primary mechanism. This is probably due to the variety of adaptations that allow tumor cells to escape from Fas-induced killing, of which down-regulation of the receptor is only one mechanism (39). In addition to the role of p53, the lack of in vivo effects of morphine toward HT-29 tumors in mice that did not respond with an increase of GTPase activity upon morphine exposure suggests that the antitumoral effects of morphine also depend on the abundance of opioid receptors. At first sight, this appears illogical because the growth-inhibitory effects of morphine occurred primarily at concentrations where Gi was no longer active. This suggests that possibly the uncoupling of Gi, rather than its activation, may be the initiating event that ultimately leads to p53 phosphorylation. This idea is supported by the finding that neuronal apoptosis induced by morphine was associated with morphine tolerance (52), which is known to be caused by opioid receptor desensitization and uncoupling of the Gi protein (55, 56, 57). On the other hand, increase of tumor growth with opioids occurred at very low doses (1, 43) and/or rapidly discontinued administration (1–3 doses; Ref. 42), i.e., with a dosing regimen avoiding tolerance. Because treatment of cancer pain usually requires long-term morphine use with increasing doses, opioid tolerance generally develops, possibly along with proapoptotic effects of morphine. However, morphine-induced apoptosis is not restricted to tumor cells. In particular, effects on immunocytes may have a negative impact on the antitumoral immune response (2), which may result in a faster progression of tumor growth or metastasis despite direct proapoptotic effects of morphine on tumor cells. In the present study, the growth-inhibitory effects of morphine were increased by Nx, suggesting that the combination of morphine and Nx might be useful to supplement cancer therapy, provided that tumors express opioid receptors and have no loss of function mutation of p53.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

Supported by a grant from the Klinikum der Johann Wolfgang Goethe-University (to I. T.).

3

The abbreviations used are: Nx, naloxone; PTX, pertussis toxin; PK, protein kinase; FasL, Fas ligand; DAMGO, (d-Ala2,N-methyl-Phe4,Gly-ol5)-enkephalin; RT-PCR, reverse transcription-PCR; 7-AAD, 7-amino-actinomycin D; PARP, poly(ADP-ribose) polymerase; Erk. extracellular signal-regulated kinase; PI, propidium iodide.

Fig. 1.

Proliferation and survival of MCF-7, MDA-MB231, and HT-29 cells. A, proliferation. Cells were treated as indicated, stained with trypan blue, and counted by a blinded observer. Data are means ± SE of four to six experiments. The asterisks beside the curves indicate a statistically significant difference versus control cells (P < 0.05). B, survival of MCF-7, MDA-MB231, and HT-29 cells in the colony forming assay. The relative survival rate was calculated in relation to that of untreated control cells, which was set at 100%. Data are the mean ± SE of four experiments. IC50 data are given in “Results.”.

Fig. 1.

Proliferation and survival of MCF-7, MDA-MB231, and HT-29 cells. A, proliferation. Cells were treated as indicated, stained with trypan blue, and counted by a blinded observer. Data are means ± SE of four to six experiments. The asterisks beside the curves indicate a statistically significant difference versus control cells (P < 0.05). B, survival of MCF-7, MDA-MB231, and HT-29 cells in the colony forming assay. The relative survival rate was calculated in relation to that of untreated control cells, which was set at 100%. Data are the mean ± SE of four experiments. IC50 data are given in “Results.”.

Close modal
Fig. 2.

Cell cycle distribution and apoptosis in MCF-7, MDA-MB231, and HT-29 cells treated with morphine, Nx, or the combination of morphine and Nx. A, cell cycle distribution. Cells were treated for 24 h as indicated. PI-stained cells were analyzed by flow cytometry. B, Western blot analysis of PARP cleavage. Cells were treated as indicated (C, controls; MOR, morphine; Mix, morphine + Nx). C, flow cytometric analysis of annexin V and 7-AAD binding in MCF-7 cells (10,000 cells were counted). D, inhibition of apoptosis with caspase inhibitors. Cells were treated for 24 h with morphine (3 mm for MCF-7, 2 mm for MDA-MB231, and 4 mm for HT-29 cells) in the presence or absence of specific inhibitors for caspase 3, 8, and 9. Cells were stained with PI, and cell cycle distribution was analyzed by flow cytometry. The sub-G1 peak represents the apoptotic fraction. Representative result of three experiments is shown.

Fig. 2.

Cell cycle distribution and apoptosis in MCF-7, MDA-MB231, and HT-29 cells treated with morphine, Nx, or the combination of morphine and Nx. A, cell cycle distribution. Cells were treated for 24 h as indicated. PI-stained cells were analyzed by flow cytometry. B, Western blot analysis of PARP cleavage. Cells were treated as indicated (C, controls; MOR, morphine; Mix, morphine + Nx). C, flow cytometric analysis of annexin V and 7-AAD binding in MCF-7 cells (10,000 cells were counted). D, inhibition of apoptosis with caspase inhibitors. Cells were treated for 24 h with morphine (3 mm for MCF-7, 2 mm for MDA-MB231, and 4 mm for HT-29 cells) in the presence or absence of specific inhibitors for caspase 3, 8, and 9. Cells were stained with PI, and cell cycle distribution was analyzed by flow cytometry. The sub-G1 peak represents the apoptotic fraction. Representative result of three experiments is shown.

Close modal
Fig. 3.

Tumor growth in nude mice. A, cells were injected s.c. at the dorsal flank of NMRI-nu/nu female mice. Drugs were administered daily by an i.p. injection starting at the day of tumor cell implantation. The doses of morphine and Nx were 10, 20, and 30 mg/kg in the first, second, and third week, respectively. In mice treated with the drug combination, the Nx dose was one-tenth of the morphine dose. A total of 8–15 mice were treated in each group. The tumor volume was determined 3×/week [tumor volume = (long diameter × short diameter2)/2]. Data are means ± SE. The asterisk beside the curve indicates a statistically significant difference versus control mice with P < 0.05. B, plasma concentrations of morphine and Nx in individual mice. Blood was sampled after injection of the last dose.

Fig. 3.

Tumor growth in nude mice. A, cells were injected s.c. at the dorsal flank of NMRI-nu/nu female mice. Drugs were administered daily by an i.p. injection starting at the day of tumor cell implantation. The doses of morphine and Nx were 10, 20, and 30 mg/kg in the first, second, and third week, respectively. In mice treated with the drug combination, the Nx dose was one-tenth of the morphine dose. A total of 8–15 mice were treated in each group. The tumor volume was determined 3×/week [tumor volume = (long diameter × short diameter2)/2]. Data are means ± SE. The asterisk beside the curve indicates a statistically significant difference versus control mice with P < 0.05. B, plasma concentrations of morphine and Nx in individual mice. Blood was sampled after injection of the last dose.

Close modal
Fig. 4.

Typical opioid receptor-dependent signaling pathways are not involved in morphine-induced antiproliferative effects. A, steady-state GTPase activity in membrane preparations of MDA-MB231 cells (100 ng) treated as indicated. Where indicated, cells were pretreated for 24 h with PTX before membrane preparation. The mean ± SE of at least three experiments, each done in duplicates, is represented. B, Western blot analysis of μ- and δ-opioid receptor expression (Mor and Dor, respectively) in untreated MCF-7, MDA-MB231, and HT-29 cells. C, flow cytometry shows that the morphine-induced cell cycle arrest was not abolished by concomitant treatment with PTX, forskolin, and 8-bromo-cGMP at the indicated concentrations. PTX, forskolin, and 8-bromo-cGMP were added 10 min before morphine. Cells were treated for 24 h. Doses of morphine were 1 mm in MCF-7 and MDA-MB231 cells and 2 mm in HT-29 cells.

Fig. 4.

Typical opioid receptor-dependent signaling pathways are not involved in morphine-induced antiproliferative effects. A, steady-state GTPase activity in membrane preparations of MDA-MB231 cells (100 ng) treated as indicated. Where indicated, cells were pretreated for 24 h with PTX before membrane preparation. The mean ± SE of at least three experiments, each done in duplicates, is represented. B, Western blot analysis of μ- and δ-opioid receptor expression (Mor and Dor, respectively) in untreated MCF-7, MDA-MB231, and HT-29 cells. C, flow cytometry shows that the morphine-induced cell cycle arrest was not abolished by concomitant treatment with PTX, forskolin, and 8-bromo-cGMP at the indicated concentrations. PTX, forskolin, and 8-bromo-cGMP were added 10 min before morphine. Cells were treated for 24 h. Doses of morphine were 1 mm in MCF-7 and MDA-MB231 cells and 2 mm in HT-29 cells.

Close modal
Fig. 5.

Phosphorylation and stabilization of p53 and up-regulation of p53-dependent genes. A, time course of phospo-p53 in MCF-7 (Ser15, left panel) and MDA-MB231 cells (Ser9, right panel) treated as indicated. Erk-2 served as loading control. Representative result of four experiments is shown. B, time course of total p53 protein expression in MCF-7 cells treated with morphine (left panels) or morphine + Nx (right panel) for the indicated times. Representative result of four experiments is shown. Erk-2 was used as loading control. C, Western blot analysis of total p53, p21, and Bax at 3 or 6 h. Treatments were as follows: 1, control; 2, 1 μg/ml actinomycin D; 3, 500 μm DAMGO; 4, 100 μm Nx; and 5, 500 μm DAMGO +100 μm Nx. Representative result of three experiments is shown.

Fig. 5.

Phosphorylation and stabilization of p53 and up-regulation of p53-dependent genes. A, time course of phospo-p53 in MCF-7 (Ser15, left panel) and MDA-MB231 cells (Ser9, right panel) treated as indicated. Erk-2 served as loading control. Representative result of four experiments is shown. B, time course of total p53 protein expression in MCF-7 cells treated with morphine (left panels) or morphine + Nx (right panel) for the indicated times. Representative result of four experiments is shown. Erk-2 was used as loading control. C, Western blot analysis of total p53, p21, and Bax at 3 or 6 h. Treatments were as follows: 1, control; 2, 1 μg/ml actinomycin D; 3, 500 μm DAMGO; 4, 100 μm Nx; and 5, 500 μm DAMGO +100 μm Nx. Representative result of three experiments is shown.

Close modal
Fig. 6.

Up-regulation of Fas (A) protein expression in untreated MCF-7, MDA-MB231, and HT-29 cells. B, Fas mRNA levels as assessed by RT-PCR. β-Actin is used a control. Representative result of three experiments is shown. C, Western blot analysis of CD95/Fas protein expression. Treatments were as follows: 1, control; 2, 1 μg/ml actinomycin D; 3, 500 μm DAMGO; 4, 100 μm Nx; and 5, 500 μm DAMGO +100 μm Nx. Erk-2 was used as loading control. D, fluorescence-activated cell-sorting analysis showing the effects of Fas-fusion protein on morphine-induced apoptosis. Cells were treated for 48 h as indicated. Fas-fusion protein was added 10 min before morphine. The sub-G1 peak represents the apoptotic fraction. Representative result of two experiments is shown.

Fig. 6.

Up-regulation of Fas (A) protein expression in untreated MCF-7, MDA-MB231, and HT-29 cells. B, Fas mRNA levels as assessed by RT-PCR. β-Actin is used a control. Representative result of three experiments is shown. C, Western blot analysis of CD95/Fas protein expression. Treatments were as follows: 1, control; 2, 1 μg/ml actinomycin D; 3, 500 μm DAMGO; 4, 100 μm Nx; and 5, 500 μm DAMGO +100 μm Nx. Erk-2 was used as loading control. D, fluorescence-activated cell-sorting analysis showing the effects of Fas-fusion protein on morphine-induced apoptosis. Cells were treated for 48 h as indicated. Fas-fusion protein was added 10 min before morphine. The sub-G1 peak represents the apoptotic fraction. Representative result of two experiments is shown.

Close modal
1
Gupta K., Kshirsagar S., Chang L., Schwartz R., Law P. Y., Yee D., Hebbel R. P. Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth.
Cancer Res.
,
62
:
4491
-4498,  
2002
.
2
Ishikawa M., Tanno K., Kamo A., Takayanagi Y., Sasaki K. Enhancement of tumor growth by morphine and its possible mechanism in mice.
Biol. Pharm. Bull.
,
16
:
762
-766,  
1993
.
3
Sueoka E., Sueoka N., Kai Y., Okabe S., Suganuma M., Kanematsu K., Yamamoto T., Fujiki H. Anticancer activity of morphine and its synthetic derivative, KT-90, mediated through apoptosis and inhibition of NF-κB activation.
Biochem. Biophys. Res. Commun.
,
252
:
566
-570,  
1998
.
4
Yeager M. P., Colacchio T. A. Effect of morphine on growth of metastatic colon cancer in vivo.
Arch. Surg.
,
126
:
454
-456,  
1991
.
5
Maneckjee R., Minna J. D. Nonconventional opioid binding sites mediate growth inhibitory effects of methadone on human lung cancer cells.
Proc. Natl. Acad. Sci. USA
,
89
:
1169
-1173,  
1992
.
6
Maneckjee R., Minna J. D. Opioids induce while nicotine suppresses apoptosis in human lung cancer cells.
Cell Growth Differ.
,
5
:
1033
-1040,  
1994
.
7
Kugawa F., Arae K., Ueno A., Aoki M. Buprenorphine hydrochloride induces apoptosis in NG108-15 nerve cells.
Eur. J. Pharmacol.
,
347
:
105
-112,  
1998
.
8
Maneckjee R., Biswas R., Vonderhaar B. K. Binding of opioids to human MCF-7 breast cancer cells and their effects on growth.
Cancer Res.
,
50
:
2234
-2238,  
1990
.
9
Kampa M., Bakogeorgou E., Hatzoglou A., Damianaki A., Martin P. M., Castanas E. Opioid alkaloids and casomorphin peptides decrease the proliferation of prostatic cancer cell lines (LNCaP, PC3 and DU145) through a partial interaction with opioid receptors.
Eur. J. Pharmacol.
,
335
:
255
-265,  
1997
.
10
Hatzoglou A., Bakogeorgou E., Castanas E. The antiproliferative effect of opioid receptor agonists on the T47D human breast cancer cell line is partially mediated through opioid receptors.
Eur. J. Pharmacol.
,
296
:
199
-207,  
1996
.
11
Panagiotou S., Bakogeorgou E., Papakonstanti E., Hatzoglou A., Wallet F., Dussert C., Stournaras C., Martin P. M., Castanas E. Opioid agonists modify breast cancer cell proliferation by blocking cells to the G2/M phase of the cycle: involvement of cytoskeletal elements.
J. Cell. Biochem.
,
73
:
204
-211,  
1999
.
12
Harimaya Y., Koizumi K., Andoh T., Nojima H., Kuraishi Y., Saiki I. Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells.
Cancer Lett.
,
187
:
121
2002
.
13
Sasamura T., Nakamura S., Iida Y., Fujii H., Murata J., Saiki I., Nojima H., 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
.
14
Scholar E. M., Violi L., Hexum T. D. The antimetastatic activity of enkephalin-like peptides.
Cancer Lett.
,
35
:
133
-138,  
1987
.
15
Zagon I. S., Hytrek S. D., Lang C. M., Smith J. P., McGarrity T. J., Wu Y., McLaughlin P. J. Opioid growth factor ([Met5]enkephalin) prevents the incidence and retards the growth of human colon cancer.
Am. J. Physiol.
,
271
:
R780
-R786,  
1996
.
16
Srisuchart B., Fuchs B. A., Sikorski E. E., Munson A. E., Loveless S. E. Antitumor activity of enkephalin analogues in inhibiting PYB6 tumor growth in mice and immunological effects of methionine enkephalinamide.
Int. J. Immunopharmacol.
,
11
:
487
-500,  
1989
.
17
McLaughlin P. J., Zagon I. S., Skitzki J. Human neuroblastoma cell growth in tissue culture is regulated by opioid growth factor.
Int. J. Oncol.
,
14
:
373
-380,  
1999
.
18
Zagon I. S., Roesener C. D., Verderame M. F., Ohlsson-Wilhelm B. M., Levin R. J., McLaughlin P. J. Opioid growth factor regulates the cell cycle of human neoplasias.
Int. J. Oncol.
,
17
:
1053
-1061,  
2000
.
19
Standifer K. M., Pasternak G. W. G proteins and opioid receptor-mediated signalling.
Cell Signalling
,
9
:
237
-248,  
1997
.
20
Nagasawa H., Li C. Y., Maki C. G., Imrich A. C., Little J. B. Relationship between radiation-induced G1 phase arrest and p53 function in human tumor cells.
Cancer Res.
,
55
:
1842
-1846,  
1995
.
21
Watson N. C., Di Y. M., Orr M. S., Fornari F. A., Jr., Randolph J. K., Magnet K. J., Jain P. T., Gewirtz D. A. Influence of ionizing radiation on proliferation, c-myc expression and the induction of apoptotic cell death in two breast tumour cell lines differing in p53 status.
Int. J. Radiat. Biol.
,
72
:
547
-559,  
1997
.
22
Dickson R. B., Bates S. E., McManaway M. E., Lippman M. E. Characterization of estrogen responsive transforming activity in human breast cancer cell lines.
Cancer Res.
,
46
:
1707
-1713,  
1986
.
23
Kassis S., Fishman P. H. Different mechanisms of desensitization of adenylate cyclase by isoproterenol and prostaglandin E1 in human fibroblasts. Role of regulatory components in desensitization.
J. Biol. Chem.
,
257
:
5312
-5318,  
1982
.
24
Sun H., Seyer J. M., Patel T. B. A region in the cytosolic domain of the epidermal growth factor receptor antithetically regulates the stimulatory and inhibitory guanine nucleotide-binding regulatory proteins of adenylyl cyclase.
Proc. Natl. Acad. Sci. USA
,
92
:
2229
-2233,  
1995
.
25
Eisenberg E., Vos B. P., Strassman A. M. The peripheral antinociceptive effect of morphine in a rat model of facial pain.
Neuroscience
,
72
:
519
-525,  
1996
.
26
Lotsch J., Skarke C., Schmidt H., Grosch S., Geisslinger G. The transfer half-life of morphine-6-glucuronide from plasma to effect site assessed by pupil size measurement in healthy volunteers.
Anesthesiology
,
95
:
1329
-1338,  
2001
.
27
Faura C. C., Olaso M. J., Garcia Cabanes C., Horga J. F. Lack of morphine-6-glucuronide antinociception after morphine treatment. Is morphine-3-glucuronide involved?.
Pain
,
65
:
25
-30,  
1996
.
28
Aherne G. W., Piall E. M., Twycross R. G. Serum morphine concentration after oral administration of diamorphine hydrochloride and morphine sulphate.
Br. J. Clin. Pharmacol.
,
8
:
577
-580,  
1979
.
29
Stewart Z. A., Tang L. J., Pietenpol J. A. Increased p53 phosphorylation after microtubule disruption is mediated in a microtubule inhibitor- and cell-specific manner.
Oncogene
,
20
:
113
-124,  
2001
.
30
Gutierrez del Arroyo A., Gil-Lamagniere C., Lazaro I., de Marco M. C., Layunta I., Silva A. Involvement of p53 and interleukin 3 in the up-regulation of CD95 (APO-1/Fas) by X-ray irradiation.
Oncogene
,
19
:
3647
-3655,  
2000
.
31
Wen J., Ramadevi N., Nguyen D., Perkins C., Worthington E., Bhalla K. Antileukemic drugs increase death receptor 5 levels and enhance Apo-2L-induced apoptosis of human acute leukemia cells.
Blood
,
96
:
3900
-3906,  
2000
.
32
Chinnaiyan A. M., Prasad U., Shankar S., Hamstra D. A., Shanaiah M., Chenevert T. L., Ross B. D., Rehemtulla A. Combined effect of tumor necrosis factor-related apoptosis-inducing ligand and ionizing radiation in breast cancer therapy.
Proc. Natl. Acad. Sci. USA
,
97
:
1754
-1759,  
2000
.
33
Kannan K., Amariglio N., Rechavi G., Jakob-Hirsch J., Kela I., Kaminski N., Getz G., Domany E., Givol D. DNA microarrays identification of primary and secondary target genes regulated by p53.
Oncogene
,
20
:
2225
-2234,  
2001
.
34
Lin Y., Ma W., Benchimol S. Pidd, a new death-domain-containing protein, is induced by p53 and promotes apoptosis.
Nat. Genet.
,
26
:
122
-127,  
2000
.
35
Yin D., Mufson R. A., Wang R., Shi Y. Fas-mediated cell death promoted by opioids.
Nature (Lond.)
,
397
:
218
1999
.
36
Boronat M. A., Garcia-Fuster M. J., Garcia-Sevilla J. A. Chronic morphine induces up-regulation of the pro-apoptotic Fas receptor and down-regulation of the anti-apoptotic Bcl-2 oncoprotein in rat brain.
Br. J. Pharmacol.
,
134
:
1263
-1270,  
2001
.
37
Singhal P. C., Bhaskaran M., Patel J., Patel K., Kasinath B. S., Duraisamy S., Franki N., Reddy K., Kapasi A. A. Role of p38 mitogen-activated protein kinase phosphorylation and Fas-Fas ligand interaction in morphine-induced macrophage apoptosis.
J. Immunol.
,
168
:
4025
-4033,  
2002
.
38
Hahne M., Rimoldi D., Schroter M., Romero P., Schreier M., French L. E., Schneider P., Bornand T., Fontana A., Lienard D., Cerottini J., Tschopp J. Melanoma cell expression of Fas(Apo-1/CD95) ligand: implications for tumor immune escape.
Science (Wash. DC)
,
274
:
1363
-1366,  
1996
.
39
O’Connell J., O’Sullivan G. C., Collins J. K., Shanahan F. The Fas counterattack: Fas-mediated T cell killing by colon cancer cells expressing Fas ligand.
J. Exp. Med.
,
184
:
1075
-1082,  
1996
.
40
Meriney S. D., Gray D. B., Pilar G. Morphine-induced delay of normal cell death in the avian ciliary ganglion.
Science (Wash. DC)
,
228
:
1451
-1453,  
1985
.
41
Kim M. S., Cheong Y. P., So H. S., Lee K. M., Kim T. Y., Oh J., Chung Y. T., Son Y., Kim B. R., Park R. Protective effects of morphine in peroxynitrite-induced apoptosis of primary rat neonatal astrocytes: potential involvement of G protein and phosphatidylinositol 3-kinase (PI3 kinase).
Biochem. Pharmacol.
,
61
:
779
-786,  
2001
.
42
Simon R. H., Arbo T. E. Morphine increases metastatic tumor growth.
Brain Res. Bull.
,
16
:
363
-367,  
1986
.
43
Moon T. D. The effect of opiates upon prostatic carcinoma cell growth.
Biochem. Biophys. Res. Commun.
,
153
:
722
-727,  
1988
.
44
Singhal P. C., Reddy K., Franki N., Sanwal V., Gibbons N. Morphine induces splenocyte apoptosis and enhanced mRNA expression of cathepsin-B.
Inflammation
,
21
:
609
-617,  
1997
.
45
Singhal P. C., Sharma P., Kapasi A. A., Reddy K., Franki N., Gibbons N. Morphine enhances macrophage apoptosis.
J. Immunol.
,
160
:
1886
-1893,  
1998
.
46
Singhal P., Kapasi A., Reddy K., Franki N. Opiates promote T cell apoptosis through JNK and caspase pathway.
Adv. Exp. Med. Biol.
,
493
:
127
-135,  
2001
.
47
Sueoka N., Sueoka E., Okabe S., Fujiki H. Anti-cancer effects of morphine through inhibition of tumour necrosis factor-α release and mRNA expression.
Carcinogenesis (Lond.)
,
17
:
2337
-2341,  
1996
.
48
Kawase M., Sakagami H., Furuya K., Kikuchi H., Nishikawa H., Motohashi N., Morimoto Y., Varga A., Molnar J. Cell death-inducing activity of opiates in human oral tumor cell lines.
Anticancer Res.
,
22
:
211
-214,  
2002
.
49
Kugawa F., Ueno A., Aoki M. Apoptosis of NG108-15 cells induced by buprenorphine hydrochloride occurs via the caspase-3 pathway.
Biol. Pharm. Bull.
,
23
:
930
-935,  
2000
.
50
Page G. G., Ben-Eliyahu S., Yirmiya R., Liebeskind J. C. Morphine attenuates surgery-induced enhancement of metastatic colonization in rats.
Pain
,
54
:
21
-28,  
1993
.
51
Hu S., Sheng W. S., Lokensgard J. R., Peterson P. K. Morphine induces apoptosis of human microglia and neurons.
Neuropharmacology
,
42
:
829
-836,  
2002
.
52
Mao J., Sung B., Ji R. R., Lim G. Neuronal apoptosis associated with morphine tolerance: evidence for an opioid-induced neurotoxic mechanism.
J. Neurosci.
,
22
:
7650
-7661,  
2002
.
53
Yin D. L., Ren X. H., Zheng Z. L., Pu L., Jiang L. Z., Ma L., Pei G. Etorphine inhibits cell growth and induces apoptosis in SK-N-SH cells: involvement of pertussis toxin-sensitive G proteins.
Neurosci. Res.
,
29
:
121
-127,  
1997
.
54
Polakiewicz R. D., Schieferl S. M., Gingras A. C., Sonenberg N., Comb M. J. μ-Opioid receptor activates signaling pathways implicated in cell survival and translational control.
J. Biol. Chem.
,
273
:
23534
-23541,  
1998
.
55
Whistler J. L., von Zastrow M. Morphine-activated opioid receptors elude desensitization by β-arrestin.
Proc. Natl. Acad. Sci. USA
,
95
:
9914
-9919,  
1998
.
56
Pei G., Kieffer B. L., Lefkowitz R. J., Freedman N. J. Agonist-dependent phosphorylation of the mouse δ-opioid receptor: involvement of G protein-coupled receptor kinases but not protein kinase C.
Mol. Pharmacol.
,
48
:
173
-177,  
1995
.
57
Bohn L. M., Lefkowitz R. J., Gainetdinov R. R., Peppel K., Caron M. G., Lin F. T. Enhanced morphine analgesia in mice lacking β-arrestin 2.
Science (Wash. DC)
,
286
:
2495
-2498,  
1999
.