We analyzed the effect of aspirin, salicylate, and other nonsteroidal antiinflammatory drugs (NSAIDs) on the viability of B-chronic lymphocytic leukemia (B-CLL) cells. Aspirin induced a decrease in cell viability in a dose- and time-dependent manner. The mean IC50 for cells from 5 patients was 5.9 ± 1.13 mmol/L (range, 4.4 to 7.3 mmol/L). In some cases, 2.5 mmol/L aspirin produced an important cytotoxic effect after 4 days of incubation. No effect was observed with other NSAIDs, at concentrations that inhibit cyclooxygenase, such as ketorolac (10 μmol/mL), NS-398 (100 μmol/mL), or indomethacin (20 μmol/mL), thus suggesting the involvement of cyclooxygenase-independent mechanisms in aspirin-induced cytotoxicity. Salicylate also produced dose-dependent cytotoxic effects on B-CLL cells and the mean IC50 for cells from 5 patients was 6.96 ± 1.13 mmol/L (range, 5 to 7.8 mmol/L). Both aspirin and salicylate induced DNA fragmentation and the proteolytic cleavage of poly(ADP(adenosine 5′-diphosphate)-ribose) polymerase (PARP), demonstrating that both compounds induce apoptosis of B-CLL cells. Finally, inhibition of caspases by Z-VAD.fmk blocked proteolytic cleavage of PARP, DNA fragmentation, and cytotoxicity induced by aspirin. Mononuclear cells from normal donors showed a lower sensitivity than cells from B-CLL patients to aspirin as determined by analysis of cell viability. B and T lymphocytes from normal donors and T lymphocytes from CLL patients are more resistant to aspirin-induced apoptosis, as determined by analysis of phosphatidylserine exposure. These results indicate that aspirin and salicylate induce apoptosis of B-CLL cells by activation of caspases and that this activation involves cyclooxygenase-independent mechanisms.

© 1998 by The American Society of Hematology.

B-CELL CHRONIC lymphocytic leukemia (B-CLL) is characterized by the accumulation of monoclonal CD5+ B lymphocytes.1,2 Most circulating cells appear to be arrested at the G0 phase of the cell cycle and it has been suggested that the clonal excess of B cells results from decreased apoptosis rather than increased proliferation.3 

Apoptosis is the physiological process whereby most cells, including B lymphocytes, are eliminated, which leads to homeostasis.4The evidence obtained in recent years indicates that many cancer chemotherapy agents induce apoptosis of tumor cells.5Glucocorticoids and other chemotherapeutic agents, such as chlorambucil, 2-chloro-2′-deoxyadenosine, fludarabine, camptothecin, and mitoxantrone,6-11 induce apoptosis in B-CLL lymphocytes, suggesting that apoptosis is one of the mechanisms of their therapeutic action.

Cysteine-proteases of the CED-3/ICE family, now named caspases, play an important role in apoptosis.12-14 Some of these proteases can cleave and inactivate poly(ADP-ribose) polymerase (PARP), and this cleavage can be used as a marker of activation of caspases and apoptosis. We recently reported that drug-induced apoptosis of B-CLL cells involves the activation of caspases.15 

Nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to induce apoptosis of v-src–transformed chicken embryo fibroblasts and different human cell lines.16-20 The mechanism responsible for this apoptotic effect of NSAIDs is not clear. Aspirin and other NSAIDs directly target cyclooxygenase (COX),21 a key enzyme in the production of prostaglandins, prostacyclins, and thromboxanes.22 There are 2 COX isoenzymes, COX-1 and COX-2, which differ in their expression regulation and tissue distribution23 and could be selectively inhibited. Although COX is the only molecular target known of most NSAIDs, both COX-dependent and COX-independent mechanisms in the apoptotic action of NSAIDs have been reported.17,24,25 

In the present study, we have analyzed the effect of aspirin, salicylate, and other NSAIDs on the viability of B-CLL cells and whether these drugs induce apoptosis of these leukemic cells.

Patients.

Seven patients (1 man and 6 women) with B-CLL who had not received treatment, with a median age of 75 years (range, 63 to 78 years), were studied. B-CLL was diagnosed according to standard clinical and laboratory criteria. The median peripheral blood leukocytosis was 85 × 109 leukocytes/L (range, 34 to 194 × 109). Leukemic cells were phenotyped for cell surface markers by flow cytometry and were positive in all cases for CD5 and CD19. According to Binet’s classification,26 at the time of inclusion, 5 patients were at stage A, 1 patient was at stage B, and 1 patient was at stage C.

Isolation of B-CLL cells.

Mononuclear cells from peripheral blood samples were isolated by centrifugation on a Ficoll/Hypaque (Seromed, Berlin, Germany) gradient and cryopreserved in liquid nitrogen in the presence of 10% dimethyl sulfoxide (DMSO).

Reagents.

Aspirin (acetylsalicylic acid), sodium salicylate, indomethacin, 3,(4,5-Dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide (MTT), and propidium iodide (PI) were obtained from Sigma Chemicals Co (St Louis, MO). N-benzyloxycarbonyl-Val-Ala-Asp-fluromethyl ketone (Z-VAD.fmk) was obtained from Enzyme Systems Products (Dublin, CA). Ketorolac was kindly provided by Almirall Laboratories (Barcelona, Spain). N-(2-ciclohexyloxy-4-nitrophenyl)-methanesulfonamide (NS-398) was obtained from BIOMOL Research Laboratories, Inc (Plymouth Meeting, PA). Stock solutions of aspirin and indomethacin in absolute ethanol, NS-398 in DMSO, and sodium salicylate and ketorolac in double-distilled water were freshly prepared for each experiment. The final concentrations of ethanol and DMSO did not affect cell viability (data not shown). Anti-PARP polyclonal antibody (Vi.5) raised against the recombinant human PARP overproduced in Sf9/baculovirus was kindly provided by Dr Gilbert de Murcia (Strasbourg, France).

Cell culture.

B-CLL lymphocytes were cultured immediately after the thawing of the cells at a concentration of 5 × 106 cells/mL in RPMI 1640 culture medium (GIBCO-BRL, Paisley, UK) supplemented with 10% heat-inactivated fetal calf serum (FCS), 2 mmol/L glutamine, and 0.04 mg/mL gentamicin at 37°C in a humidified atmosphere containing 5% carbon dioxide.27 Factors were added at the beginning of the culture.

Cell viability assay.

Cell viability was determined by the MTT assay.28 B lymphocytes (5 × 105 cells/well) were incubated in 96-well plates in the absence or in the presence of factors in a final volume of 100 μL. After 48 hours, 10 μL of MTT (5 mg/mL in phosphate-buffered saline) was added to each well for an additional 6 hours. The blue MTT formazan precipitated was dissolved in 100 μL of isopropanol:1 mol/L HCl (24:1) and the absorbance values at 550 nm were determined on a multiwell plate reader.

Western blot analysis of PARP cleavage.

Cells were lysed with Laemmli sample buffer29 and protein extracts were electrophoresed on 10% polyacrylamide gel and transferred to Immobilon-P (Millipore, Bedford, MA) membranes. After blocking for 1 hour with 5% dried skimmed milk in TBST (50 mmol/L Tris HCl, pH 8.0, 150 mmol/L NaCl, 0.5% Tween-20), the filters were incubated with Vi.5 PARP antibody diluted 1:1,000 in 5% dried skimmed milk in TBST, which recognizes both the native enzyme (116 kD) and the cleavage product (∼85 kD).15 Antibody binding was detected using a secondary antibody (swine antirabbit Ig; DAKO, Glostrup, Denmark) conjugated to horseradish peroxidase diluted 1:500 in 5% dried skimmed milk in TBST and an enhanced chemiluminescence (ECL) detection kit (Amersham, Buckinghamshire, UK). As an internal control of total cellular protein levels, blots were also probed with α-tubulin antibody (Oncogene Science, Inc, Cambridge, MA).

Analysis of DNA fragmentation.

Analysis of DNA fragmentation by agarose gel electrophoresis was performed as previously described.15 Five million cells were pelleted and lysed for 20 minutes at 4°C in 10 mmol/L Tris-HCl, pH 7.4, 0.2% Triton X-100, 1 mmol/L EDTA. After centrifugation at 14,000g for 15 minutes, the supernatant was treated with 0.2 mg/mL of proteinase K in a buffer containing 150 mmol/L NaCl, 10 mmol/L Tris HCl, pH 8.0, 40 mmol/L EDTA, and 1% sodium dodecyl sulfate (SDS) for 6 hours at 37°C. DNA was extracted twice with buffered phenol and precipitated with 140 mmol/L NaCl and 2 vol of ethanol at −20°C overnight. DNA precipitates were washed twice in 70% ethanol, dissolved in double-distilled water, and treated for 1 hour at 37°C with RNase (Boehringer Mannheim, Mannheim, Germany). Finally, DNA preparations were electrophoresed in 1% agarose gels. Gels were stained with ethidium bromide and visualized under UV light.

PI DNA staining.

Quantification of apoptosis by PI staining and fluorescence-activated cell sorting (FACS) analysis was performed as described previously.30 Briefly, cells were harvested and fixed in 70% ethanol. Cells were centrifuged, washed in phosphate-buffered saline (PBS), and resuspended in 0.5 mL PBS containing PI (5 μg/mL) and RNase (100 μg/mL). Tubes were incubated for 30 minutes at 37°C and placed at 4°C in the dark overnight before flow cytometry analysis to identify the sub-G0 peak corresponding to apoptosis.

Analysis of apoptosis by annexin binding.

Exposure of phosphatidylserine was quantified by surface annexin V staining as described previously.31 One million cells were incubated during 24 hours with the indicated factors. After that time, cells were washed with PBS and incubated with phycoerythrin-conjugated anti-CD19 (DAKO) and tri-color–conjugated anti-CD2 (Caltag Laboratories, Burlingame, CA) for 15 minutes in the dark. Cells were then washed, resuspended in 200 μL of binding buffer (10 mmol/L HEPES, pH 7.4 , 2.5 mmol/L CaCl2, 140 mmol/L NaCl), and incubated with 0.5 μg/mL of ApoAlert Enhanced Annexin V-FITC (Clontech Laboratories Inc, Palo Alto, CA) for 5 to 15 minutes in the dark, before analysis by flow cytometry (FACScan; Becton Dickinson, Mountain View, CA). Samples were acquired using Lysis-II software and data were analyzed with the Paint-a-gate Pro software (Becton Dickinson). To analyze a sufficient number of cells, a live-gate in side scatter (SSC) versus CD19 or SSC versus CD2 was drawn and at least 5,000 CD19+ cells or CD2+ cells were acquired.

Statistical analysis.

Levels of significance between samples were determined using thet-test for nonpaired samples and the analysis of variance (ANOVA), Fisher’s protected least significant difference (PLSD). To compare the effect of aspirin on normal PBMCs and B-CLL cells, we performed a multiple analysis of variance (MANOVA), Hottelling T2.

Cytotoxic effect of aspirin on B-CLL lymphocytes.

First, the effect of aspirin on the viability of B-CLL lymphocytes was studied. Cells from 5 patients were incubated for 48 hours with several doses of aspirin, ranging from 1 to 10 mmol/L, and cell viability was determined by the MTT assay. As seen in Fig 1A, a dose-dependent decrease in cell viability could be observed in all the cases studied, although cell sensitivity to aspirin differed from one patient to another. The mean IC50 of these dose-response studies was 5.9 ± 1.13 mmol/L (range, 4.4 to 7.3 mmol/L). In 3 patients (patients no. 1, 4, and 5), an effect was detected with 2.5 mmol/L aspirin.

Fig. 1.

Cytotoxic effect of aspirin on B-CLL cells. (A) Dose-response of the cytotoxic effect of aspirin on B-CLL cells. Cells from 5 patients were incubated for 48 hours with various concentrations of aspirin that ranged from 1 to 10 mmol/L. (B) Time course of aspirin-induced cytotoxicity. Cells from patients no. 1 and 2 were incubated with 2.5, 5, and 10 mmol/L aspirin (ASA) for the times indicated. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments.

Fig. 1.

Cytotoxic effect of aspirin on B-CLL cells. (A) Dose-response of the cytotoxic effect of aspirin on B-CLL cells. Cells from 5 patients were incubated for 48 hours with various concentrations of aspirin that ranged from 1 to 10 mmol/L. (B) Time course of aspirin-induced cytotoxicity. Cells from patients no. 1 and 2 were incubated with 2.5, 5, and 10 mmol/L aspirin (ASA) for the times indicated. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments.

Close modal

The cytotoxicity of aspirin on B-CLL lymphocytes was further studied with time-course assays that were performed with cells from patients no. 1 and 2. Cells were incubated without or with aspirin (2.5, 5, or 10 mmol/L) for different periods of time, as indicated in Fig 1B. A significant decrease in cell viability was observed in both patients after only 24 hours of incubation with 10 mmol/L aspirin (P < .003). A dose of 5 mmol/L aspirin produced a significant decrease in cell viability of lymphocytes from patient no. 1 after 24 hours of incubation (P = .006) that increased with time of incubation. No significant effect was seen with this dose in cells from patient no. 2 after 24 hours (P = .29), and longer periods of incubation were needed to achieve an important cytotoxic effect. Incubation for 48 hours with 2.5 mmol/L aspirin induced cytotoxicity in cells from patient no. 1, but the viability of cells from patient no. 2 was not significantly decreased after 96 hours of incubation.

COX inhibition is not sufficient to induce apoptosis of B-CLL cells.

Because aspirin targets the enzyme COX, we tested whether other inhibitors of this enzyme could produce cytotoxic effects on B-CLL lymphocytes. An inhibitor of both COX-1 and COX-2 (indomethacin), a specific inhibitor of COX-1 (ketorolac), and a specific inhibitor of COX-2 (NS-398) were tested. Neither COX-1 nor COX-2 inhibitors produced any effect on cell viability with doses up to 10 μmol/L ketorolac, 20 μmol/L indomethacin, and 100 μmol/L NS-398, which have been previously described to inhibit COX32-36(Fig 2). Furthermore, we used ketorolac and NS-398 together, and no effect was observed either. All these assays were performed with cells from 5 patients and in all cases there were no effects. However, at the concentrations used in these experiments, these NSAIDs inhibited the production of prostaglandin E2(PGE2) by B-CLL cells, thus demonstrating that COX was being inhibited (data not shown). Taken together, these results indicate that inhibition of COX is not sufficient to induce the decrease in cell viability produced by aspirin on B-CLL lymphocytes.

Fig. 2.

Effect of COX inhibitors on the viability of B-CLL lymphocytes. Cells were incubated for 48 hours with 5 mmol/L aspirin, 10 μmol/L ketorolac, 100 μmol/L NS-398, or 20 μmol/L indomethacin. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments. Statistical significance of differences between control cells and cells treated with COX inhibitors was assayed by ANOVA, Fisher’s PLSD. **P < .0001.

Fig. 2.

Effect of COX inhibitors on the viability of B-CLL lymphocytes. Cells were incubated for 48 hours with 5 mmol/L aspirin, 10 μmol/L ketorolac, 100 μmol/L NS-398, or 20 μmol/L indomethacin. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments. Statistical significance of differences between control cells and cells treated with COX inhibitors was assayed by ANOVA, Fisher’s PLSD. **P < .0001.

Close modal
Cytotoxic effect of salicylate on B-CLL lymphocytes.

To test if salicylate, a structure-related compound of aspirin, produced any effect on cell viability, dose-response studies were performed with 1 to 20 mmol/L sodium salicylate. As seen in Fig 3, this substance produced cytotoxic effects on B-CLL cells when concentrations of 5 to 10 mmol/L were used. This effect was observed in cells from 5 patients studied. The mean IC50 for these 5 patients was 6.96 ± 1.13 mmol/L (range, 5 to 7.8 mmol/L).

Fig. 3.

Dose-response of the cytotoxic effect of salicylate on B-CLL cells. Cells from 5 patients were incubated for 48 hours with various concentrations of sodium salicylate as indicated. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments.

Fig. 3.

Dose-response of the cytotoxic effect of salicylate on B-CLL cells. Cells from 5 patients were incubated for 48 hours with various concentrations of sodium salicylate as indicated. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments.

Close modal
Aspirin and salicylate induce apoptosis and activation of caspases in B-CLL lymphocytes.

To determine whether this cytotoxic effect was due to induction of apoptosis, we analyzed whether incubation with aspirin or salicylate induced DNA fragmentation and proteolytic cleavage of PARP, a hallmark of apoptosis in most cells, including B-CLL cells.15 Cells from patients no. 1 and 2 were incubated with various doses of aspirin as described previously, ranging from 1 to 10 mmol/L. As seen in Fig 4, both DNA fragmentation (Fig 4A) and PARP cleavage (Fig 4B) were dose-dependent and a clear effect could be observed with 2.5 to 5 mmol/L aspirin. These doses corresponded to the doses that induced a cytotoxic effect as previously determined by the MTT assay. Similar results were obtained with cells from patients no. 3 and 4. The apparent decrease in the total amount of PARP in the Western blot analysis of patient no. 1 at the doses of 5 and 7.5 mmol/L is due to a decrease in the total amount of loaded protein, as demonstrated by immunoblotting of α-tubulin.

Fig. 4.

Induction of apoptosis by aspirin on B-CLL cells. (A) Effect of aspirin on DNA fragmentation in B-CLL cells. Cells from patients no. 1 and 2 were incubated for 24 hours with 1 to 10 mmol/L aspirin (ASA). DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of aspirin on PARP cleavage. Cells from patients no. 1 and 2 were incubated for 48 hours with 1 to 10 mmol/L aspirin (ASA). PARP cleavage was analyzed on protein extracts from these cells by Western blot as described in the Materials and Methods. The position of the native PARP (116 kD) and the proteolytic fragment (85 kD) is indicated. As an internal control, blots were also probed with -tubulin antibody. (C) Effect of aspirin on DNA content in a representative CLL patient. Cells from patient no. 1 were incubated for 48 hours with 1 to 10 mmol/L aspirin (ASA). DNA content was quantified by PI staining and flow cytometry analysis as described in the Materials and Methods. (D) Quantification of aspirin-induced apoptosis in cells from 4 patients by PI staining and FACS analysis after 48 hours of incubation with 1 to 10 mmol/L aspirin.

Fig. 4.

Induction of apoptosis by aspirin on B-CLL cells. (A) Effect of aspirin on DNA fragmentation in B-CLL cells. Cells from patients no. 1 and 2 were incubated for 24 hours with 1 to 10 mmol/L aspirin (ASA). DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of aspirin on PARP cleavage. Cells from patients no. 1 and 2 were incubated for 48 hours with 1 to 10 mmol/L aspirin (ASA). PARP cleavage was analyzed on protein extracts from these cells by Western blot as described in the Materials and Methods. The position of the native PARP (116 kD) and the proteolytic fragment (85 kD) is indicated. As an internal control, blots were also probed with -tubulin antibody. (C) Effect of aspirin on DNA content in a representative CLL patient. Cells from patient no. 1 were incubated for 48 hours with 1 to 10 mmol/L aspirin (ASA). DNA content was quantified by PI staining and flow cytometry analysis as described in the Materials and Methods. (D) Quantification of aspirin-induced apoptosis in cells from 4 patients by PI staining and FACS analysis after 48 hours of incubation with 1 to 10 mmol/L aspirin.

Close modal

Furthermore, we corroborated these results measuring apoptosis by PI staining and FACS analysis. As shown in Fig 4C and D, aspirin induced in a dose-dependent manner an increase in the percentage of subdiploid cells in 4 patients studied.

As seen in Fig 5, salicylate also induced DNA fragmentation and cleavage of PARP when cells were incubated with the doses that produced cytotoxic effects. Similar results to that shown in Fig 5 were obtained with cells from 2 additional patients (results not shown). These results demonstrate that the cytotoxicity of both aspirin and salicylate is due to apoptosis induction.

Fig. 5.

Induction of apoptosis by salicylate on B-CLL cells. (A) Effect of salicylate on DNA fragmentation in B-CLL cells. Cells from patients no. 3 and 4 were incubated for 24 hours with 5 and 10 mmol/L sodium salicylate (NaSal). DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of salicylate on PARP cleavage. Cells from patients no. 3 and 4 were incubated for 48 hours with 5 and 10 mmol/L sodium salicylate (NaSal). PARP cleavage was analyzed on protein extracts from these cells by Western blot as described in the Materials and Methods. The position of the native PARP (116 kD) and the proteolytic fragment (85 kD) is indicated.

Fig. 5.

Induction of apoptosis by salicylate on B-CLL cells. (A) Effect of salicylate on DNA fragmentation in B-CLL cells. Cells from patients no. 3 and 4 were incubated for 24 hours with 5 and 10 mmol/L sodium salicylate (NaSal). DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of salicylate on PARP cleavage. Cells from patients no. 3 and 4 were incubated for 48 hours with 5 and 10 mmol/L sodium salicylate (NaSal). PARP cleavage was analyzed on protein extracts from these cells by Western blot as described in the Materials and Methods. The position of the native PARP (116 kD) and the proteolytic fragment (85 kD) is indicated.

Close modal

To demonstrate the involvement of activation of caspases in the apoptotic effect of aspirin, we studied whether the caspase inhibitor Z-VAD.fmk prevented aspirin-induced apoptosis. We incubated B-CLL cells with aspirin in the presence or absence of 200 μmol/L Z-VAD.fmk, a concentration that inhibits apoptosis of B-CLL cells.15This concentration of Z-VAD.fmk had no cytotoxic effects (results not shown) and prevented the cytotoxic effect of both 5 and 10 mmol/L aspirin in the 3 cases studied, 2 of which are shown in Fig 6. Furthermore, Z-VAD.fmk completely blocked aspirin-induced DNA fragmentation (Fig 7A) and PARP cleavage (Fig 7B).

Fig. 6.

Effect of the caspase inhibitor Z-VAD.fmk on the viability of B-CLL cells. Cells from patients no. 2 and 6 were seeded in 96-microwell plates and incubated for 48 hours either alone or in the presence of aspirin (ASA; 5 or 10 mmol/L) or aspirin with 200 μmol/L Z-VAD.fmk. Z-VAD.fmk was added 1 hour before aspirin administration. Cytotoxicity was analyzed by the MTT method as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments. Statistical significance of differences between treatment with aspirin alone or in combination with Z-VAD.fmk was assayed ANOVA (Fisher’s PLSD). *P < .03; **P < .0001.

Fig. 6.

Effect of the caspase inhibitor Z-VAD.fmk on the viability of B-CLL cells. Cells from patients no. 2 and 6 were seeded in 96-microwell plates and incubated for 48 hours either alone or in the presence of aspirin (ASA; 5 or 10 mmol/L) or aspirin with 200 μmol/L Z-VAD.fmk. Z-VAD.fmk was added 1 hour before aspirin administration. Cytotoxicity was analyzed by the MTT method as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. Data are shown as the mean value ± SD of 3 independent experiments. Statistical significance of differences between treatment with aspirin alone or in combination with Z-VAD.fmk was assayed ANOVA (Fisher’s PLSD). *P < .03; **P < .0001.

Close modal
Fig. 7.

Effect of the caspase inhibitor Z-VAD.fmk on aspirin-induced apoptosis in B-CLL cells. (A) Effect of Z-VAD.fmk on aspirin-induced DNA fragmentation. B lymphocytes from 2 B-CLL patients were incubated for 24 hours with 10 mmol/L aspirin (ASA) in the presence or absence of 200 μmol/L Z-VAD.fmk. DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of Z-VAD.fmk on aspirin-induced PARP cleavage. Cells from patients no. 2 and 6 were incubated for 24 hours with 10 mmol/L aspirin (ASA) in the presence or absence of 200 μmol/L Z-VAD.fmk. Cells were lysed and analyzed by Western blot as described in the Materials and Methods. Z-VAD.fmk was added 1 hour before aspirin administration.

Fig. 7.

Effect of the caspase inhibitor Z-VAD.fmk on aspirin-induced apoptosis in B-CLL cells. (A) Effect of Z-VAD.fmk on aspirin-induced DNA fragmentation. B lymphocytes from 2 B-CLL patients were incubated for 24 hours with 10 mmol/L aspirin (ASA) in the presence or absence of 200 μmol/L Z-VAD.fmk. DNA was extracted and subjected to agarose gel electrophoresis as described in the Materials and Methods. (B) Effect of Z-VAD.fmk on aspirin-induced PARP cleavage. Cells from patients no. 2 and 6 were incubated for 24 hours with 10 mmol/L aspirin (ASA) in the presence or absence of 200 μmol/L Z-VAD.fmk. Cells were lysed and analyzed by Western blot as described in the Materials and Methods. Z-VAD.fmk was added 1 hour before aspirin administration.

Close modal
Aspirin effect on normal peripheral blood mononuclear cells (PBMCs).

Finally, we studied the cytotoxic effects of aspirin in normal PBMCs. Incubation of PBMCs with aspirin produced no significant cytotoxic effect (P > .12) with doses up to 7.5 mmol/L (Fig 8A). The mean aspirin IC50value for normal cells was 15.8 ± 6.2 mmol/L (range, 9.8 to 26.3 mmol/L). The comparison between the dose-responses of the cytotoxic effect of aspirin on B-CLL cells and normal PBMCs indicates that normal PBMCs are more resistant to aspirin (P = .003). A similar dose-dependency was found when DNA fragmentation and PARP cleavage were analyzed. The concentration of aspirin or salicylate necessary to induce detectable DNA fragmentation and PARP cleavage was 7.5 mmol/L (results not shown).

Fig. 8.

Effect of aspirin on normal PBMCs and B cells. (A) Dose-response of the cytotoxic effect of aspirin on normal PBMCs. Cells from 6 normal donors were incubated with various concentrations of aspirin as indicated for 48 hours. Data are presented as the means ± SD of 3 experiments each for all 6 normal donors and compared with the means of the 5 B-CLL patients represented in Fig 1A. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. (B) Comparison between the induction of apoptosis in B cells and T cells from B-CLL patients and normal donors. Cells were incubated with 1 to 10 mmol/L aspirin for 24 hours and phosphatidylserine exposure was measured by binding of annexin V to CD19+ or CD2+ cells as described in the Materials and Methods. Statistical significance was determined using the t-test for nonpaired samples. *P < .05; **P < .01; ***P < .001.

Fig. 8.

Effect of aspirin on normal PBMCs and B cells. (A) Dose-response of the cytotoxic effect of aspirin on normal PBMCs. Cells from 6 normal donors were incubated with various concentrations of aspirin as indicated for 48 hours. Data are presented as the means ± SD of 3 experiments each for all 6 normal donors and compared with the means of the 5 B-CLL patients represented in Fig 1A. Cell viability was determined by the MTT assay as described in the Materials and Methods and is expressed as a percentage of the viability of control cells at the beginning of the culture. (B) Comparison between the induction of apoptosis in B cells and T cells from B-CLL patients and normal donors. Cells were incubated with 1 to 10 mmol/L aspirin for 24 hours and phosphatidylserine exposure was measured by binding of annexin V to CD19+ or CD2+ cells as described in the Materials and Methods. Statistical significance was determined using the t-test for nonpaired samples. *P < .05; **P < .01; ***P < .001.

Close modal

Because most normal PBMCs are T lymphocytes, it was interesting to distinguish the effects of aspirin on B and T cells. To specifically analyze the induction of apoptosis in normal B cells by aspirin, we quantified phosphatidylserine exposure in response to aspirin in CD19+ cells. As seen in Fig 8B, aspirin did not induce a significant increase in the apoptosis of normal B cells with doses up to 2.5 mmol/L (P > .13); in contrast, aspirin induced apoptosis of B-CLL cells at 2.5 mmol/L, indicating that B cells from normal donors have a lower degree of sensitivity to aspirin than that of B-CLL cells. Furthermore, T cells from both normal donors and B-CLL patients appeared to be more resistant to aspirin-induced apoptosis than B-CLL cells (Fig 8B).

The results shown in this report demonstrate that aspirin and its metabolite salicylate induce apoptosis of B-CLL cells. This is the first report showing apoptotic action of NSAIDs in human primary tumor cells.

NSAIDs have been shown to induce apoptosis of human colon carcinoma and in vitro transformed colon adenoma cell lines.17-20Furthermore, different NSAIDs cause apoptosis in v-src–transformed chicken embryo fibroblasts16 and sulindac sulfide induces apoptosis of promyelocytic leukemia cell line HL-60.18 It has been reported that aspirin, in contrast to other NSAIDs, does not induce apoptosis in either HT-29 colon carcinoma cells or v-src–transformed chicken fibroblasts.16,20 The discrepancy between these results and our results could be explained by the fact that the doses used in previous studies were too low (150 μmol/L and 1.5 mmol/L). Very recently, it has been reported that aspirin induces apoptosis of HT-29 cells at concentrations higher that 1 mmol/L.37 

The mechanisms by which NSAIDs induce apoptosis are not clear. In agreement with the hypothesis that the apoptotic action of NSAIDs is mediated by the inhibition of COX, overexpression of COX-2 in rat epithelial intestinal cells inhibits butyrate-induced apoptosis and this inhibition was reversed by the NSAID sulindac sulfide.24 However, the sulfone metabolite of sulindac, which does not inhibit cyclooxygenases,38 also induces apoptosis of HT-29 cells,17 and treatment with different prostaglandins failed to reverse the effects of NSAIDs on apoptosis.25 Considering the results herein reported, two different arguments indicate the involvement of cyclooxygenase-independent mechanisms in the apoptotic action of aspirin and salicylate on B-CLL cells. (1) Neither COX-1 nor COX-2 inhibitors induced cytotoxicity in B-CLL cells. (2) Although aspirin is a much more potent inhibitor of COX than salicylate, both drugs show similar potency as inducers of apoptosis. Taken together, these results suggest the existence of COX-dependent and COX-independent mechanisms in the apoptotic action of NSAIDs. Interestingly, we recently demonstrated that aspirin inhibits DNA synthesis in Swiss 3T3 fibroblasts by both COX-dependent and COX-independent mechanisms, depending on the dose used.39 

COX-independent mechanisms have been proposed to explain the inhibition of the transcription factor NF-κB by aspirin and salicylate.40-43 It is noteworthy that inhibition of NF-κB causes apoptosis of B lymphocytes44,45 and that NF-κB has an essential role in preventing tumor necrosis factor-α (TNF-α)– and cancer therapy-induced apoptosis.46-49Another COX-independent mechanism induced by aspirin and salicylate is the inhibition of AP-1 activity, which may involve the elevation of H+ concentration.50 In addition, it was very recently reported that salicylate induces apoptosis of fibroblasts via p38 mitogen-activated protein kinase,51 and this kinase has been involved in mIgM-induced apoptosis of human B lymphocytes.52 The results presented in this report demonstrate that aspirin-induced apoptosis of B-CLL cells involves the activation of caspases, as previously described for glucocorticoid-, fludarabine-, and mitoxantrone-induced apoptosis.11,15 The elucidation of the mechanisms involved in the apoptotic action of aspirin and salicylate in B-CLL cells needs further investigation.

The finding that salicylate also induces apoptosis of B-CLL cells is important, because aspirin is substantially hydrolyzed to salicylate on first pass through portal circulation. Consequently, plasma concentrations of salicylate rapidly exceed plasma concentrations of aspirin after oral administration of aspirin, and the half-life of salicylate in the plasma is considerably longer than that of aspirin.53 Although we only observe effects of aspirin and salicylate on apoptosis of B-CLL cells at relatively high concentrations (1.5 to 10 mmol/L), the lower range of such concentrations is achieved and even maintained in the treatment of chronic inflammatory states such as rheumatoid arthritis, in which the therapeutic plasma concentration is 150 to 300 mg/L (0.94 to 1.88 mmol/L).54-57 Unfortunately, not all patients are able to tolerate the adequate dosage to maintain these plasma levels for a long time.57 On the other hand, normal peripheral blood mononuclear cells are more resistant to aspirin than B-CLL cells, thus suggesting that clinical use of aspirin at concentrations that induce apoptosis of B-CLL cells could have only minor effects on normal lymphocytes in certain patients.

Several epidemiological studies have demonstrated an association between the long-term consumption of aspirin and a reduced risk of colon cancer.58-61 Although no association with reduced risk of other types of cancer has been clearly demonstrated,61 the incidence of B-CLL in cohorts of individuals taking aspirin on a regular basis has not been analyzed. This issue, as well as whether there is a potential role for aspirin in B-CLL management, deserves investigation. Finally, the elucidation of the mechanisms underlying the induction of the apoptosis of B-CLL cells by aspirin and salicylate would provide important information to understand the biology of B-CLL and to design new strategies for its therapy.

The authors are indebted to Dr G. de Murcia for the kind gift of PARP antibodies. We thank Drs V. Jiménez and J. Ros for the determinations of PGE2 levels. We thank Drs R. Bartrons, C. Pastor, and P. Feliz for their help and suggestions and R. Rycroft for language assistance.

B.B., M.P., M.B., and E.C. are recipients of a research fellowship from Fundación de la Asociación Española Contra el Cáncer, CIRIT, Fundació August Pi i Sunyer, and Universitat de Barcelona, respectively. This work was supported by “Fondo de Investigaciones Sanitarias de la Seguridad Social” (FIS 95/0873), by Quı́mica Farmacéutica Bayer S.A. (Division Consumer Care), and by the “Generalitat de Catalunya” (1995 SGR 00427).

Address reprint requests to Joan Gil, PhD, Departament de Ciències Fisiològiques II, Campus de Bellvitge, Universitat de Barcelona, Pavelló de Govern, 4a planta, 08907 L’Hospitalet, Spain; e-mail: joangil@bellvitge.bvg.ub.es.

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

1
Foon
KA
Rai
KR
Gale
RP
Chronic lymphocytic leukemia: New insights into biology and therapy.
Ann Intern Med
113
1990
525
2
Rozman
C
Montserrat
E
Chronic lymphocytic leukemia.
N Engl J Med
333
1995
1052
3
Gale
RP
Caligaris-Cappio
F
Dighiero
G
Keating
M
Montserrat
E
Rai
K
Recent progress in chronic lymphocytic leukemia.
Leukemia
8
1994
1610
4
Krammer
PH
Behrmann
I
Daniel
P
Dhein
J
Debatin
KM
Regulation of apoptosis in the immune system.
Curr Opin Immunol
6
1994
279
5
Hannun
YA
Apoptosis and the dilemma of cancer chemotherapy.
Blood
89
1997
1845
6
McConkey
DJ
Aguilar-Santelises
M
Hartzell
P
Eriksson
I
Mellstedt
H
Orrenius
S
Jondal
M
Induction of DNA fragmentation in chronic B-lymphocytic leukemia cells.
J Immunol
146
1991
1072
7
Carrera CJ, Piro LD, Saven A, Beutler E, Terai C, Carson DA: 2-Chlorodeoxyadenosine chemotherapy triggers programmed cell death in normal and malignant lymphocytes. Adv Exp Med Biol 309A:15, 1991
8
Robertson
LE
Chubb
S
Meyn
RE
Story
M
Ford
R
Hittelman
WN
Plunkett
W
Induction of apoptotic cell death in chronic lymphocytic leukemia by 2-chloro-2′-deoxyadenosine and 9-β-D-arabinosyl-2-fluoroadenine.
Blood
81
1993
143
9
(suppl 1)
Begleiter
A
Lee
K
Israels
LG
Mowat
MRA
Johnston
JB
Chlorambucil induced apoptosis in chronic lymphocytic leukemia (CLL) and its relationship to clinical efficacy.
Leukemia
8
1994
S103
10
Thomas
A
El Rouby
S
Reed
JC
Krajewski
S
Silber
R
Potmesil
M
Newcomb
EW
Drug-induced apoptosis in B-cell chronic lymphocytic leukemia: Relationship between p53 gene mutation and bcl-2/bax proteins in drug resistance.
Oncogene
12
1996
1055
11
Bellosillo
B
Colomer
D
Pons
G
Gil
J
Mitoxantrone, a topoisomerase II inhibitor, induces apoptosis of B-chronic lymphocytic leukaemia cells.
Br J Haematol
100
1998
142
12
Henkart
PA
ICE family of proteases: Mediators of all apoptotic cell death?
Immunity
4
1996
195
13
Yang
E
Korsmeyer
SJ
Molecular thanatopsis: A discourse on the BCL2 family and cell death.
Blood
88
1996
386
14
Nagata
S
Apoptosis by death factor.
Cell
88
1997
355
15
Bellosillo
B
Dalmau
M
Colomer
D
Gil
J
Involvement of CED-3/ICE proteases in the apoptosis of B-chronic lymphocytic leukemia cells.
Blood
89
1997
3378
16
Lu
X
Xie
W
Reed
D
Bradshaw
WS
Simmons
DL
Nonsteroidal antiinflammatory drugs cause apoptosis and induce cyclooxygenases in chicken embryo fibroblasts.
Proc Natl Acad Sci USA
92
1995
7961
17
Piazza
GA
Rahm
AL
Krutzsch
M
Sperl
G
Paranka
NS
Gross
PH
Brendel
K
Burt
RW
Alberts
DS
Pamukcu
R
Ahnen
DJ
Antineoplastic drugs sulindac sulfide and sulfone inhibit cell growth by inducing apoptosis.
Cancer Res
55
1995
3110
18
Shiff
SJ
Qiao
L
Tsai
LL
Rigas
B
Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells.
J Clin Invest
96
1995
491
19
Elder
DJE
Hague
A
Hicks
DJ
Paraskeva
C
Differential growth inhibition by the aspirin metabolite salicylate in human colorectal tumor cell lines: Enhanced apoptosis in carcinoma and in vitro-transformed adenoma relative to adenoma cell lines.
Cancer Res
56
1996
2273
20
Shiff
SJ
Koutsos
MI
Qiao
L
Rigas
B
Nonsteroidal antiinflammatory drugs inhibit the proliferation of colon adenocarcinoma cells: Effects on cell cycle and apoptosis.
Exp Cell Res
222
1996
179
21
Vane
JR
Inhibition of prostaglandin synthesis as mechanism of action for aspirin-like drugs.
Nat New Biol
231
1971
232
22
Smith
WL
The eicosanoids and their biochemical mechanism of action.
Biochem J
259
1989
315
23
Smith
WL
Garavito
RM
DeWitt
DL
Prostaglandin endoperoxide H syntases (cyclooxygenases)-1 and -2.
J Biol Chem
271
1996
33157
24
Tsujii
M
Dubois
RN
Alterations in cellular adhesion and apoptosis in epithelial cells overexpressing prostaglandin endoperoxide synthase-2.
Cell
83
1995
493
25
Hanif
R
Pittas
A
Feng
Y
Koutsos
MI
Qiao
L
Staiano-Coico
L
Shiff
SI
Rigas
B
Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway.
Biochem Pharmacol
52
1996
237
26
Binet
JL
Auquier
A
Dighiero
G
Chastang
C
Piguet
H
Goasguen
J
Vaugier
G
Potron
G
Colona
P
Oberling
F
Thomas
M
Tchernia
G
Jacquillat
C
Boivin
P
Lesty
C
Duault
MT
Monconduit
M
Belabbes
S
Gremy
F
A new prognostic classification of chronic lymphocytic leukemia derived from a multivariate survival analysis.
Cancer
48
1981
198
27
Carballo
E
Colomer
D
Vives Corrons
JL
Blackshear
PJ
Gil
J
Characterization and purification of a protein kinase C substrate in human B cells.
J Immunol
156
1996
1709
28
Mosmann
T
Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays.
J Immunol Methods
65
1983
55
29
Laemmli
UK
Cleavage of structural proteins during the assembly of the head of bacteriophage T4.
Nature
227
1970
680
30
Nicoletti
I
Migliorati
G
Pagliacci
MC
Grignani
F
Riccardi
C
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry.
J Immunol Methods
139
1991
271
31
Koopman
G
Reutelingsperger
CP
Kuijten
GA
Keehnen
RM
Pals
ST
van Oers
MH
Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis.
Blood
84
1994
1415
32
Gierse
JK
Hauser
SD
Creely
DP
Koboldt
C
Rangwala
SH
Isakson
PC
Seibert
K
Expression and selective inhibition of the constitutive and inducible forms of human cyclo-oxygenase.
Biochem J
305
1995
479
33
Barnett
J
Chow
J
Ives
D
Chiou
M
Mackenzie
R
Osen
E
Nguyen
B
Tsing
S
Bach
C
Freire
J
Chan
H
Sigal
E
Ramesha
C
Purification, characterization and selective inhibition of human prostaglandin G/H synthase 1 and 2 expressed in the baculovirus system.
Biochim Biophys Acta
1209
1994
130
34
Chulada
PC
Langenbach
R
Differential inhibition of murine prostaglandin synthase-1 and -2 by non-steroidal anti-inflammatory drugs using exogenous and endogenous sources of arachidonic acid.
J Pharmacol Exp Ther
280
1997
606
35
Kargman
S
Wong
E
Greig
GM
Falgueyret
JP
Cromlish
W
Ethier
D
Yergey
JA
Riendeau
D
Evans
JF
Kennedy
B
Tagari
P
Francis
DA
O’Neill
GP
Mechanism of selective inhibition of human prostaglandin G/H synthase-1 and -2 in intact cells.
Biochem Pharmacol
52
1996
1113
36
Miralpeix
M
Camacho
M
Lopez-Belmonte
J
Canalias
F
Beleta
J
Palacios
JM
Vila
L
Selective induction of cyclo-oxygenase-2 activity in the permanent human endothelial cell line HUV-EC-C: Biochemical and pharmacological characterization.
Br J Pharmacol
121
1997
171
37
Qiao
L
Hanif
R
Sphicas
E
Shiff
SJ
Rigas
B
Effect of aspirin on induction of apoptosis in HT-29 human colon adenocarcinoma cells.
Biochem Pharmacol
55
1998
53
38
Thompson
HJ
Briggs
S
Paranka
NS
Piazza
GA
Brendel
K
Gross
PH
Sperl
GH
Pamukcu
R
Ahnen
DJ
Inhibition of mammary carcinogenesis in rats by sulfone metabolite of sulindac.
J Natl Cancer Inst
87
1995
1259
39
Castaño
E
Dalmau
M
Martı́
M
Berrocal
F
Bartrons
R
Gil
J
Inhibition of DNA synthesis by aspirin in Swiss 3T3 fibroblasts.
J Pharmacol Exp Ther
280
1997
366
40
Kopp
E
Ghosh
S
Inhibition of NF-κB by sodium salicylate and aspirin.
Science
265
1994
956
41
Weber
C
Erl
W
Pietsch
A
Weber
PC
Aspirin inhibits nuclear factor-κB mobilization and monocyte adhesion in stimulated human endothelial cells.
Circulation
91
1995
1914
42
Pierce
JW
Read
MA
Ding
H
Luscinskas
FW
Collins
T
Salicylates inhibit IκB-α phosphorylation, endothelial-leukocyte adhesion molecule expression and neutrophil transmigration.
J Immunol
156
1996
3961
43
Grilli
M
Pizzi
M
Memo
M
Spano
P
Neuroprotection by aspirin and sodium salicylate through blockade of NF-κB activation.
Science
274
1996
1383
44
Wu
M
Lee
H
Bellas
RE
Schauer
SL
Arsura
M
Katz
D
Fitzgerald
MJ
Rothstein
TL
Sherr
DH
Sonenshein
GE
Inhibition of NF-κB/Rel induces apoptosis of murine B cells.
EMBO J
15
1996
4682
45
Arsura
M
Wu
M
Sonenshein
GE
TGFβ-1 inhibits NF-κB/Rel activity inducing apoptosis of B cells: Transcriptional activation of IκBα.
Immunity
5
1996
31
46
Beg
AA
Baltimore
D
An essential role for NF-κB in preventing TNF-α-induced cell death.
Science
274
1996
782
47
Wang
CY
Mayo
MW
Baldwin
AS
TNF- and cancer therapy-induced apoptosis: Potentiation by inhibition of NF-κB.
Science
274
1996
784
48
Van Antwerp
DJ
Martin
SJ
Kafri
T
Green
DR
Verma
IM
Suppression of TNF-α-induced apoptosis by NF-κB.
Science
274
1996
787
49
Liu
ZG
Hsu
H
Goeddel
DV
Karin
M
Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-κB activation prevents cell death.
Cell
87
1996
565
50
Dong
Z
Huang
C
Brown
RE
Ma
WY
Inhibition of activator protein 1 activity and neoplastic transformation by aspirin.
J Biol Chem
272
1997
9962
51
Schwenger
P
Bellosta
P
Vietor
I
Basilico
C
Skolnik
EY
Vilcek
J
Sodium salicylate induces apoptosis via p38 mitogen-activated protein kinase but inhibits tumor necrosis factor-induced c-jun N-terminal kinase/stress-activated protein kinase activation.
Proc Natl Acad Sci USA
94
1997
2869
52
Graves
JD
Draves
KE
Craxton
A
Saklatvala
J
Krebs
EG
Clark
EA
Involvement of stress-activated protein kinase and p38 mitogen-activated protein kinase in mIgM-induced apoptosis of human B lymphocytes.
Proc Natl Acad Sci USA
93
1996
13814
53
Higgs
GA
Salmon
JA
Henderson
B
Vane
JR
Pharmacokinetics of aspirin and salicylate in relation to inhibition of arachidonate cyclooxygenase and antiinflammatory activity.
Proc Natl Acad Sci USA
84
1987
1417
54
Insel
PA
Analgesic-antipyretic and antiinflammatory agents and drugs employed in the treatment of gout
Hardman
JG
Limbird
LE
Molinoff
PB
Ruddon
RW
Goodman Gilman
A
Goodman & Gilman’s The Pharmacological Basis of Therapeutics.
1996
617
McGraw Hill
New York, NY
55
USP Dispending Information—Vol I.
Drug Information for the Health Care Professional. The United States Pharmacopeial Convention, Inc.
1996
2592
Rand McNally
Taunton, MA
56
Hart
FD
Huskisson
EC
Non-steroidal anti-inflammatory drugs. Current status and rational therapeutic use.
Drugs
27
1984
232
57
Rosenberg
L
Palmer
JR
Zauber
AG
Warshauer
ME
Stolley
PD
Shapiro
S
A hypothesis: Nonsteroidal anti-inflammatory drugs reduce the incidence of large-bowel cancer.
J Natl Cancer Inst
83
1991
355
58
Thun
MJ
Namboodiri
MM
Heath
CW
Aspirin use and reduced risk of fatal colon cancer.
N Engl J Med
325
1991
1593
59
Giovannucci
E
Egan
KM
Hunter
DJ
Stampfer
MJ
Colditz
GA
Willett
WC
Speizer
FE
Aspirin and the risk of colorectal cancer in women.
N Engl J Med
333
1995
609
60
(suppl 220)
Weiss
HA
Forman
D
Aspirin, non-steroidal anti-inflammatory drugs and protection from colorectal cancer: A review of the epidemiological evidence.
Scand J Gastroenterol
31
1996
137
61
Rosenberg
L
Nonsteroidal anti-inflammatory drugs and cancer.
Prev Med
24
1995
107
Sign in via your Institution