Purpose: We examined the efficacy of flavopiridol, a cyclin-dependent kinase inhibitor that is undergoing clinical trials, on primary cancer cells isolated from the ascites or pleural fluids of patients with metastatic cancers.

Experimental Design: Metastasized cancer cells were isolated from the pleural fluids (n = 20) or ascites (n = 15) of patients, most of whom were refractory to chemotherapy. These primary cancer cells were used within 2 weeks of isolation without selecting for proliferative capacities. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide viability assay was used to characterize the response of these cancer cells to commonly used chemotherapeutic agents, and their response to flavopiridol was compared with rapidly dividing cultured cell lines.

Results: The primary cancer cells displayed phenotypes that were different from established cell lines; they had very low replication rates, dividing every 1 to 2 weeks, and underwent replicative senescence within five passages. These primary tumor cells retained their resistance to chemotherapeutic drugs exhibited by the respective patients but did not show cross-resistance to other agents. However, these cancer cells showed sensitivity to flavopiridol with an average LD50 of 50 nmol/L (range, 21.5-69 nmol/L), similar to the LD50 in established cell lines. Because senescent cells also showed similar sensitivity to flavopiridol, it suggests that the mechanism of action is not dependent on the activity of cyclin-dependent kinases that regulate the progression of the cell cycle.

Conclusion: Using cancer cells isolated from the ascites or pleural fluids, this study shows the potential of flavopiridol against cancer cells that have developed resistance to conventional chemotherapeutic agents.

Flavopiridol is a synthetic flavone that has shown potential as an antineoplastic agent and is currently undergoing clinical trials for the treatment of cancer (1). Biochemical studies have shown that it is a potent inhibitor of cyclin-dependent kinases (cdks), with a LD50 of ∼50 to 100 nmol/L, and can inhibit other kinases at higher concentrations (reviewed in ref. 2). Crystallography shows that flavopiridol binds to the ATP-binding domain of cdks. Because of its broad inhibition of cdks, flavopiridol can induce cell cycle arrest and affect other cellular functions, such as transcription and apoptosis (24).

Flavopiridol reduces transcriptional elongation through inhibition of cdk9 activity on RNA polymerase II, causing the reduction in expression of several genes, including those required for the cell cycle. Other effects of flavopiridol include inhibition of angiogenesis through the down-regulation of vascular endothelial growth factor and the induction of cellular differentiation (2). Because flavopiridol kills cells that are cycling as well as quiescent (57), the compound is most likely acting through a mechanism that does not involve the cdks that function to regulate cell cycle.

Experiments with flavopiridol on tumor xenografts in animal models showed activity against several solid tumors, and similar promising results were obtained in phase I clinical trials. However, in the phase II clinical trials that have been reported with flavopiridol thus far, the results have been mixed for its use as a single-agent treatment against solid tumors (810). Several in vitro experiments showed that flavopiridol may potentiate the effects of anticancer agents, and clinical trials are under way to determine its effectiveness in combination therapy (2, 10, 11).

To date, studies demonstrating the cytotoxicity of flavopiridol have mainly been done with established cell lines that divide rapidly. In this study, we report that primary cancer cells isolated from the ascites and pleural fluid of patients can be used to study sensitivity and resistance to antineoplastic agents. These primary cancer cells were isolated from patients with tumors of different origins, most of whom had developed resistance to the anticancer drugs used to treat the disease. The primary cancer cells showed sensitivity to flavopiridol with LD50 values of ∼50 nmol/L, comparable with that seen with the MCF-7 human breast cancer cell line.

Cell culture and medium. Cell lines used in the study were the human breast cancer MCF-7, osteosarcoma Saos-2, prostate cancer DU-145, cervical cancer HeLa, and normal human diploid fibroblast HSF-55. These cells were grown in DMEM containing 10% fetal bovine serum and antibiotics (100 units/mL penicillin, 100 μg/mL streptomycin, and 0.25 μg/mL amphotericin B) at 37°C in a 5% CO2 incubator. Primary cancer cells isolated from ascites or pleural fluid were cultured in the same medium, except that the dose of antibiotics was doubled.

Isolation of cancer cells. The process of obtaining consent from the patient was approved by the Thunder Bay Regional Health Sciences Centre Research Ethics Team (Thunder Bay, Ontario, Canada), and all required signed documents were obtained from patients before processing of patient samples. Tumor cells were isolated from the ascites of patients with ovarian cancer (n = 5), colon cancer (n = 2), breast cancer (n = 1), pancreatic cancer (n = 2), and one each of gastric cancer, mesothelioma, non-Hodgkin's lymphoma, cholangiocarcinoma, and adenocarcinoma. Tumor cells were also isolated from the pleural fluid of patients with lung cancer (n = 8), ovarian cancer (n = 3), breast cancer (n = 3), colon cancer (n = 2), and one each of mesothelioma, renal cancer, hepatocarcinoma, and non-Hodgkin's lymphoma. Table 1 lists all the cells that were isolated for use in the study. Ascites or pleural fluids drained from patients with metastatic cancers were transferred to 50 mL Falcon tubes and centrifuged at 2,500 rpm for 10 minutes to pellet the cells. The cells were resuspended in PBS, pooled, and recentrifuged to pellet the cells. Erythrocytes in the pellet were lysed by mixing each 5 mL of the ascites suspension with 40 mL erythrocyte lysis buffer (155 mmol/L NH4Cl, 10 mmol/L NH4HCO3, 100 μmol/L EDTA) and incubating at room temperature for 30 minutes. After centrifuging at 2,500 rpm for 10 minutes, the final cell pellets were washed once with PBS, pooled, resuspended in regular growth medium, transferred to 150 mm tissue culture dishes, and cultured at 37°C with 5% CO2. The growth medium was changed regularly every 2 or 3 days to remove cellular debris and any residual blood cells. After 2 weeks, the cells that remained adhered to the dish were trypsinized, counted with the Coulter cell counter (Mississauga, ON, Canada), and plated out for drug sensitivity studies. The yield of cancer cells from the ascites or pleural fluids varied between patients but typically ranged between 2 × 106 and 1 × 107 cells from 2 to 4 liters of fluid.

Table 1.

Summary of the cancer cells that were tested for sensitivity to flavopiridol

AscitesTissueAge(y)SexLD50 (nM)
A-Ov1 Ovarian 59 57.5 
A-Ov3 Ovarian 58 44 
A-Ov16 Ovarian 67 40.7 
A-Ov40 Ovarian 50 21.5 
A-Ov55 Ovarian 71 25.3 
A-Col9 Colon 59 51 
A-Col37 Colon 79 22.3 
A-Br27 Breast 52 28.4 
A-Pan22 Pancreas 53 >100 
A-Pan45 Pancreas 60 24.5 
A-Gas34 Gastric 54 68.9 
A-Meso33 Mesothelioma 71 44.5 
A-NHL6 Non-Hodgkin's Lymphoma 57 52.5 
A-Chol48 Cholangiocarcinoma 69 45.8 
A-Aden23 Adenocarcinoma 49 41.5 
Pleural
 
Fluid
 
Tissue
 
Age(y)
 
Sex LD50 (nM)
 
P-Ov15 Ovarian 55 >100 
P-Ov21 Ovarian 57 40 
P-Ov30 Ovarian 51 55.2 
P-Col25 Colon 63 21.7 
P-Col58 Colon 66 25.9 
P-Br8 Breast 76 46.5 
P-Br51 Breast 59 24.2 
P-Br56 Breast 70 66.6 
P-Lu13 Lung 75 47.8 
P-Lu20 Lung 86 >100 
P-Lu26 Lung 59 43 
P-Lu38 Lung 65 37.8 
P-Lu39 Lung 70 33.4 
P-Lu52 Lung 67 59.5 
P-Lu53 Lung 81 50.3 
P-Lu54 Lung 74 28.5 
P-Meso41 Mesothelioma 82 23.7 
P-Ren11 Renal 69 35.7 
P-Hep35 Hepatocellular 49 37 
P-NHL50 Non-Hodgkin's Lymphoma 76 52.9 
AscitesTissueAge(y)SexLD50 (nM)
A-Ov1 Ovarian 59 57.5 
A-Ov3 Ovarian 58 44 
A-Ov16 Ovarian 67 40.7 
A-Ov40 Ovarian 50 21.5 
A-Ov55 Ovarian 71 25.3 
A-Col9 Colon 59 51 
A-Col37 Colon 79 22.3 
A-Br27 Breast 52 28.4 
A-Pan22 Pancreas 53 >100 
A-Pan45 Pancreas 60 24.5 
A-Gas34 Gastric 54 68.9 
A-Meso33 Mesothelioma 71 44.5 
A-NHL6 Non-Hodgkin's Lymphoma 57 52.5 
A-Chol48 Cholangiocarcinoma 69 45.8 
A-Aden23 Adenocarcinoma 49 41.5 
Pleural
 
Fluid
 
Tissue
 
Age(y)
 
Sex LD50 (nM)
 
P-Ov15 Ovarian 55 >100 
P-Ov21 Ovarian 57 40 
P-Ov30 Ovarian 51 55.2 
P-Col25 Colon 63 21.7 
P-Col58 Colon 66 25.9 
P-Br8 Breast 76 46.5 
P-Br51 Breast 59 24.2 
P-Br56 Breast 70 66.6 
P-Lu13 Lung 75 47.8 
P-Lu20 Lung 86 >100 
P-Lu26 Lung 59 43 
P-Lu38 Lung 65 37.8 
P-Lu39 Lung 70 33.4 
P-Lu52 Lung 67 59.5 
P-Lu53 Lung 81 50.3 
P-Lu54 Lung 74 28.5 
P-Meso41 Mesothelioma 82 23.7 
P-Ren11 Renal 69 35.7 
P-Hep35 Hepatocellular 49 37 
P-NHL50 Non-Hodgkin's Lymphoma 76 52.9 

NOTE: LD50 values were taken from the graphs of MTT assays showing sensitivity of cancer cells to flavopiridol.

Bromodeoxyuridine incorporation. Incorporation of bromodeoxyuridine (BrdUrd) into DNA was used to correlate the proliferative capacity of the cells with drug sensitivity. The cells were seeded onto 18 × 18 mm cover glasses (Fisher, Ottawa, ON, Canada) and incubated with 30 μmol/L BrdUrd for 6 days before fixation with ice-cold methanol for 10 minutes. Immunostaining for BrdUrd was done according to the method described by MacDougall et al. (12). Briefly, following methanol fixation and one wash in PBS, the cells were incubated in 2 N HCl at 37°C for 20 minutes and neutralized in sodium borate/boric acid buffer. After a wash in PBS, mouse monoclonal anti-BrdUrd antibody (Sigma, Oakville, ON, Canada) was added at a dilution of 1:500 and incubated for 45 minutes at room temperature. This was followed by four 10-minute washes in PBS, and Cy3-conjugated anti-mouse antibody (Molecular Probes, Eugene, OR) was added at a dilution of 1:200 and incubated for a further 45 minutes. After four washes with PBS, the cells were counterstained with 1 μg/mL 4′,6-diamidino-2-phenylindole, mounted onto a microscope slide in mounting medium (90% glycerol, 1× PBS, containing 1 mg/mL p-phenylenediamine), and viewed with a Zeiss microscope (North York, ON, Canada) with epifluorescence. To score the percentage of replicating cells, two images of the cancer cells from different regions of each cover glass were taken using a CCD video camera (Sony, Toronto, ON, Canada), and the percentage of BrdUrd-labeled cells relative to the total number of 4′,6-diamidino-2-phenylindole–stained cells was determined for each image. Depending on whether the cells divided, each field of view typically contained between 80 and 100 cells, and the overall percentage of BrdUrd-labeled cells was averaged from the two fields.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. To measure sensitivity to flavopiridol and other antineoplastic agents, 1 × 104 cells were plated out into each well of a 96-well plate and allowed to adhere overnight. Flavopiridol (Aventis Pharmaceuticals, Inc., Bridgewater, NJ), cisplatin (Faulding Pharmaceuticals, Montréal, PQ, Canada), carboplatin (Bristol-Myers Squibb, Montréal, PQ, Canada), taxol (Bristol-Myers Squibb, Montréal, PQ, Canada), and Adriamycin (Pharmacia, Mississauga, ON, Canada) were then added at increasing concentrations and incubation continued for 3 days for cultured cells lines or 6 days for the primary cancer cells. Four wells were used for each treatment, and the average was used for LD50 determination. Measurement of viable cells remaining in each well was done using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method described by Carmichael et al. (13). MTT reagent was dissolved in PBS, added to each well to a concentration of 0.5 mg/mL, and incubated for 4 hours. The medium containing the MTT reagent was then aspirated, DMSO (100 μL) was added to each well, and the absorbance was measured in a microplate reader at a wavelength of 490 nm.

To measure the effect of exposure periods, the cells were seeded in the 96-well plates and incubated with varying concentrations of flavopiridol for up to 5 days for the MCF-7 cell line and up to 6 days for the A-Br27 breast cancer cells. For cells that were treated for less than the maximum number of days, the flavopiridol-containing medium was removed after incubation of the desired number of days and replaced with drug-free medium, and incubation was continued. MTT assays were done on the fifth day for the MCF-7 cells or the sixth day for the A-Br27 cells.

Western blot analyses. Cultured cell lines and primary cancer cells were incubated for 24 hours with 70 nmol/L flavopiridol and collected by trypsinization. Cells were lysed and extracts prepared for Western blot analysis as described in Derjuga et al. (14). Depending on the cells, total cellular protein (20-50 μg) was loaded in a 12% SDS-polyacrylamide gel and transferred onto a polyvinylidene difluoride membrane (Millipore, Billerica, MA) and immunoblotted for bcl-2 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA).

Growth characteristics of cells cultured from ascites and pleural fluids. The cancer cells isolated from the ascites or pleural fluids of patients with metastatic cancers grew very slowly, taking 1 to 2 weeks to double, and would typically undergo no more than three to five passages before entering a quiescent state. With such low replication rates, we used incorporation of BrdUrd into DNA as indication of cell division rates. With freshly isolated cancer cells that were cycling, incubation with BrdUrd for 3 days labeled <20% of the cells and a 6-day incubation period was required to get labeling in ∼80% of the cells (data not shown). With increased subcultures, the percentage of BrdUrd-positive cells decreased to <5% as they approached senescence.

Sensitivity of cancer cells to flavopiridol. Consistent with studies that have been done on established cell lines, we found that a 3-day treatment with 50 to 70 nmol/L flavopiridol killed ∼50% of the cells in several cell lines (Fig. 1, top). When we extended the study to >40 isolates of primary cancer cells prepared from the ascites or the pleural fluid, the concentration of flavopiridol required to kill 50% of the cells in 6 days was found to be within an order of magnitude of that seen with cultured cell lines, ranging from ∼20 nmol/L for a pancreatic cancer cell to ∼60 nmol/L for ovarian cancer cells isolated from the pleural fluid (Fig. 1, middle). With another isolate of ovarian cancer cells from the ascites of a different patient, the LD50 was found to be ∼20 nmol/L. These results show that the much reduced growth rates of the primary cancer cells did not affect their sensitivities to flavopiridol as was expected because of its specificity toward cdks. The results also showed that cancer cells isolated from the same disease site but from different patients could have a different LD50 for flavopiridol but remain within one-log value. As an example, we found the LD50 for eight ovarian cancer cells and eight lung cancer cells ranging between 20 and 60 nmol/L.

Fig. 1.

Sensitivity of cultured cell lines and cancer cells to flavopiridol. Cytotoxic effects of flavopiridol on several cultured cell lines as measured by the MTT cell viability test (top). At concentrations >10 nmol/L flavopiridol (20 nmol/L for DU-145), the survival in all cells was significantly decreased compared with untreated controls using a one-tailed Student's t test (P < 0.05). Effects of flavopiridol on several of the cancer cells isolated from patients with metastatic cancers (middle). These cancer cells were isolated from the ascites of patients with pancreatic cancer (A-Pan45) or ovarian cancer (A-Ov40) or from the pleural fluid of a patient with ovarian cancer (P-Ov30). The decrease in survival in all treated cells was statistically significant (P < 0.05). Among the cancer cells studied, three isolates were found to be resistant to flavopiridol (bottom). These cancer cells were isolated from the ascites of patients with ovarian cancer (A-Ov15) or pancreatic cancer (A-Pan22) or from the pleural fluid of a patient with lung cancer (P-Lu20). Although there were significant declines in survival of A-Pan22 cells at all flavopiridol concentrations and of P-Lu20 cells when treated with higher concentrations of flavopiridol (P < 0.05), >50% of the cells were not killed and were resistant to the treatment.

Fig. 1.

Sensitivity of cultured cell lines and cancer cells to flavopiridol. Cytotoxic effects of flavopiridol on several cultured cell lines as measured by the MTT cell viability test (top). At concentrations >10 nmol/L flavopiridol (20 nmol/L for DU-145), the survival in all cells was significantly decreased compared with untreated controls using a one-tailed Student's t test (P < 0.05). Effects of flavopiridol on several of the cancer cells isolated from patients with metastatic cancers (middle). These cancer cells were isolated from the ascites of patients with pancreatic cancer (A-Pan45) or ovarian cancer (A-Ov40) or from the pleural fluid of a patient with ovarian cancer (P-Ov30). The decrease in survival in all treated cells was statistically significant (P < 0.05). Among the cancer cells studied, three isolates were found to be resistant to flavopiridol (bottom). These cancer cells were isolated from the ascites of patients with ovarian cancer (A-Ov15) or pancreatic cancer (A-Pan22) or from the pleural fluid of a patient with lung cancer (P-Lu20). Although there were significant declines in survival of A-Pan22 cells at all flavopiridol concentrations and of P-Lu20 cells when treated with higher concentrations of flavopiridol (P < 0.05), >50% of the cells were not killed and were resistant to the treatment.

Close modal

In our study of >40 isolates of cancer cells, three were found resistant to flavopiridol, displaying LD50 values that were >100 nmol/L (Fig. 1, bottom). One such cancer cell (A-Pan22) was from a patient with pancreatic cancer, with ∼60% of the cells remaining viable after 6-day continuous incubation with 100 nmol/L flavopiridol. A similar level of resistance was seen with P-Lu20 cells isolated from a patient with lung cancer. A great level of resistance was seen with A-Ov15 ovarian cancer cells, where >90% of the cells remained viable after 6-day exposure to 100 nmol/L flavopiridol. When tested for cross-resistance, these cells also had increased tolerance to cisplatin and taxol, showing 3- to 5-fold increase in LD50 (data not shown).

Cell cycle–independent chemosensitivity of flavopiridol. Because flavopiridol has a high specificity toward cdks, we compared its cytotoxicity with agents that are dependent on cell division, such as taxol and cisplatin, and correlated their respective effectiveness against growth rates as measured by BrdUrd uptake. As shown in Fig. 2, when ovarian cancer cells were cycling, as indicated by 76% of cells having incorporated BrdUrd, taxol and cisplatin were highly effective, killing the cells with LD50 of ∼2 μmol/L. With subsequent passages, when the percentage of BrdUrd-labeled cells declined to ∼50%, the LD50 increased to ∼7 μmol/L for cisplatin and 9 μmol/L for taxol. When the cells reached senescence and did not take up BrdUrd, the ovarian cancer cells were resistant to both agents.

Fig. 2.

Cell division and chemosensitivity of cancer cells. Ovarian cancer cells of three different passages were treated with cisplatin (top), taxol (middle), or flavopiridol (bottom), and cell survival was measured by MTT cell viability test. Relative division rates of the cells at each of these passages were measured as a percentage of cells that incorporated BrdUrd into the DNA. Treatment with all three drugs at each concentration tested significantly decreased survival in the 76% and 50% BrdUrd-incorporated cells compared with corresponding untreated controls using a one-tailed Student's t test (P < 0.05). Survival of the noncycling (0% BrdUrd-incorporated) cells treated with the highest concentration of cisplatin (17.6 μmol/L) and with all concentrations of flavopiridol was statistically significantly different from corresponding control. *, P < 0.05, significant difference in cell survival between 0% and 50% BrdUrd-incorporated cells using a two-tailed Student's t test. +, P < 0.05, significant difference in cell survival between 0% and 76% BrdUrd-incorporated cells.

Fig. 2.

Cell division and chemosensitivity of cancer cells. Ovarian cancer cells of three different passages were treated with cisplatin (top), taxol (middle), or flavopiridol (bottom), and cell survival was measured by MTT cell viability test. Relative division rates of the cells at each of these passages were measured as a percentage of cells that incorporated BrdUrd into the DNA. Treatment with all three drugs at each concentration tested significantly decreased survival in the 76% and 50% BrdUrd-incorporated cells compared with corresponding untreated controls using a one-tailed Student's t test (P < 0.05). Survival of the noncycling (0% BrdUrd-incorporated) cells treated with the highest concentration of cisplatin (17.6 μmol/L) and with all concentrations of flavopiridol was statistically significantly different from corresponding control. *, P < 0.05, significant difference in cell survival between 0% and 50% BrdUrd-incorporated cells using a two-tailed Student's t test. +, P < 0.05, significant difference in cell survival between 0% and 76% BrdUrd-incorporated cells.

Close modal

Unlike taxol and cisplatin, the MTT assays showed that noncycling senescent cancer cells were just as sensitive to flavopiridol as those that were cycling. As shown in Fig. 2 (bottom), flavopiridol was as effective in killing the dividing ovarian cancer cells that took up 76% BrdUrd as the noncycling ones that did not incorporate BrdUrd. This result suggests that flavopiridol is capable of inducing cell death through an alternate pathway that does not depend on the cell division and cdk activities.

Flavopiridol sensitivity and exposure period. To determine the extent of exposure that is required to kill cells, we incubated MCF-7 cells for periods of up to 5 days in varying concentrations of flavopiridol. As seen in Fig. 3 (top), a 1-day treatment of the rapidly cycling MCF-7 cells with the higher concentrations of 75 or 100 nmol/L flavopiridol was sufficient to kill >80% of the cells. At 50 nmol/L, a 5-day incubation killed ∼30% of the cells, and the lowest concentration of 25 nmol/L did not result in significant killing. When we compared this with the A-Br27 breast cancer cells isolated from ascites fluid, these slower cycling primary cancer cells showed a wider variation in sensitivity to the exposure period of up to 6 days (bottom). At the lower concentrations of flavopiridol, these cells showed considerable sensitivity, with ∼40% to 50% cell death at 25 nmol/L. At the higher concentration of 100 nmol/L, a 1-day treatment was not as effective as that seen with MCF-7 cells, killing ∼70% of the cells. At least a 2-day exposure was required to kill ∼90%. At the 50 and 75 nmol/L concentrations, increasing the number of days of exposure resulted in correspondingly greater percentages of cell death. Although the results showed that continuous exposure was not required to induce cell death, the extent of incubation period required is dependent on the cells and the concentrations used.

Fig. 3.

Flavopiridol exposure and sensitivity of cancer cells. Human breast cancer MCF-7 cells (top) were treated with flavopiridol for 1- to 5-day periods and assayed for survival on the fifth day. For the cells that were treated for <5 days, the flavopiridol-containing medium was replaced with drug-free medium for the remaining incubation period before the MTT assay was done. Compared with the corresponding untreated control, the survival in MCF-7 cells was statistically significantly decreased at 75 and 100 nmol/L flavopiridol for all treatment times and at 50 nmol/L flavopiridol for 5-day treatment (P < 0.05). A-Br27 breast cancer cells isolated from ascites fluid (bottom)were similarly treated with flavopiridol for a period of up to 6 days. Survival in all treated A-Br27 cells was statistically significantly decreased compared with corresponding untreated control using a one-tailed Student's t test (P < 0.05).

Fig. 3.

Flavopiridol exposure and sensitivity of cancer cells. Human breast cancer MCF-7 cells (top) were treated with flavopiridol for 1- to 5-day periods and assayed for survival on the fifth day. For the cells that were treated for <5 days, the flavopiridol-containing medium was replaced with drug-free medium for the remaining incubation period before the MTT assay was done. Compared with the corresponding untreated control, the survival in MCF-7 cells was statistically significantly decreased at 75 and 100 nmol/L flavopiridol for all treatment times and at 50 nmol/L flavopiridol for 5-day treatment (P < 0.05). A-Br27 breast cancer cells isolated from ascites fluid (bottom)were similarly treated with flavopiridol for a period of up to 6 days. Survival in all treated A-Br27 cells was statistically significantly decreased compared with corresponding untreated control using a one-tailed Student's t test (P < 0.05).

Close modal

Effect of flavopiridol on chemoresistant cancer cells. The cancer cells used in this study were isolated from the ascites or the pleural fluids that were obtained from patients with tumors that were resistant to standard chemotherapy. The chemotherapy agents employed in this study were used only for some types of cancers, and we examined the records of the patients to determine the agents to which they had developed resistance. When compared with the results of the MTT analyses, it was found that the cancer cells did not have broad-ranged resistance to all the antineoplastic agents but were resistant only to the drugs that the patients had become refractory to. An example of this selective resistance is shown in the experiment described in Fig. 4. The cells (P-Hep35) were isolated from the pleural fluid of a patient with hepatocellular carcinoma who was treated with, and eventually became refractory to, Adriamycin (Fig. 4, top). The pleural fluid was drained a total of eight times and the cancer cells from earlier isolations showed sensitivity to Adriamycin. However, with subsequent removal of fluids from the patient, the hepatocarcinoma cells showed a gradual increase in resistance to Adriamycin. When tested against other agents, these cells were sensitive to the cytotoxic effects of carboplatin (middle) regardless of when the isolates were prepared. Similar sensitivity was also seen with flavopiridol (bottom).

Fig. 4.

Development of chemoresistance in cancer cells. Hepatocarcinoma cells (P-Hep35) were isolated from the pleural fluids of a patient who was drained up to eight times within a 9-week time period. Numbers indicate the time that the fluids were removed from the patient. Adriamycin was one of the chemotherapy agents used to treat the cancer. Cancer cells were tested for sensitivity to Adriamycin by the MTT assay (top). *, P < 0.05, significant decrease in cell survival between treated and corresponding untreated control cells using a one-tailed Student's t test. Sensitivity of P-Hep35 cells to carboplatin (middle) and flavopiridol (bottom). Compared with the corresponding untreated control, survival of cells obtained at all times indicated was statistically significantly decreased with all carboplatin and flavopiridol concentrations tested, except for P-Hep35 cells obtained for the eighth time at 9 μmol/L carboplatin.

Fig. 4.

Development of chemoresistance in cancer cells. Hepatocarcinoma cells (P-Hep35) were isolated from the pleural fluids of a patient who was drained up to eight times within a 9-week time period. Numbers indicate the time that the fluids were removed from the patient. Adriamycin was one of the chemotherapy agents used to treat the cancer. Cancer cells were tested for sensitivity to Adriamycin by the MTT assay (top). *, P < 0.05, significant decrease in cell survival between treated and corresponding untreated control cells using a one-tailed Student's t test. Sensitivity of P-Hep35 cells to carboplatin (middle) and flavopiridol (bottom). Compared with the corresponding untreated control, survival of cells obtained at all times indicated was statistically significantly decreased with all carboplatin and flavopiridol concentrations tested, except for P-Hep35 cells obtained for the eighth time at 9 μmol/L carboplatin.

Close modal

Expression of bcl-2. The expression of antiapoptotic bcl-2 has been reported to be down-regulated by flavopiridol in several cell lines (1518), but this was not consistently seen in other cells (7). In examining the effects of flavopiridol on proteins that affect apoptosis, we saw a down-regulation of bcl-2 in several cell lines (Fig. 5, top). However, with the primary cancer cells, we did not see any correlation between sensitivity to flavopiridol and cellular levels of bcl-2. As shown in Fig. 5 (bottom), the pancreatic cancer cells A-Pan22 and A-Pan45 expressed different levels of bcl-2 that were not affected by the presence of flavopiridol. The MTT viability test showed that A-Pan22 was resistant to flavopiridol and A-Pan45 was sensitive (see Fig. 1). With the ovarian cancer P-Ov30, the expression level of bcl-2 was unaffected by flavopiridol, although these cells were sensitive (see Fig. 1), whereas the resistant ovarian cancer cell A-Ov15 down-regulated their expression of bcl-2 when incubated with flavopiridol.

Fig. 5.

Expression of bcl-2 in cells treated with flavopiridol as determined by Western blot analysis. Cell lines or cancer cells were incubated with 50 or 70 nmol/L flavopiridol (Fp), respectively, for 24 hours and protein extracts were prepared and immunoblotted for expression levels of bcl-2. Untreated, protein-standardized extracts of the same cells were used as controls (Con).

Fig. 5.

Expression of bcl-2 in cells treated with flavopiridol as determined by Western blot analysis. Cell lines or cancer cells were incubated with 50 or 70 nmol/L flavopiridol (Fp), respectively, for 24 hours and protein extracts were prepared and immunoblotted for expression levels of bcl-2. Untreated, protein-standardized extracts of the same cells were used as controls (Con).

Close modal

We describe the use of cancer cells isolated from ascites or pleural fluids for studying responses to anticancer agents that are currently in clinical use and to novel anticancer agents that are undergoing clinical trials. Cancer cells isolated from ascites and pleural fluid have been used to establish cell lines that retain the genotype and phenotype of the cancers of interest. These cells were of clonal origins and were selected for their increased proliferative capacity. In our study, we used the cancer cells directly, without selection for any growth advantages or clonal expansion of any particular subpopulations, and these relatively mixed population of cells should reflect more accurately the clinical situation. There are obvious differences between cultured cell lines and the tumors in a human body, although the biochemical pathways and responses to chemotherapy may be similar. One major difference between the tumor cell lines and the primary cancer cells isolated from ascites or pleural fluid that may influence response to cancer treatment is the growth rate. Cultured cell lines have much shorter doubling times, typically dividing every 18 to 24 hours. The higher metabolic activities would drive the cells to respond faster to anticancer agents. Furthermore, because every cell would traverse every phase of the cell cycle within a day, they would be susceptible to agents that affect S and G2-M phases. With the reduced growth rates and lower metabolism of the primary cancer cells, incubation periods have to be extended for cytotoxic effect of chemotherapeutic agents to be seen. In the body where drugs are constantly cleared, the periods of exposure would have to be considered for effectiveness of anticancer agents. These primary cancer cells would be a more accurate model system for initial drug trials, as they reflect the situation seen in the body where cancer cells have a much slower growth rate than established cell lines.

In our studies of the response of these cancer cells to chemotherapy, we have made several observations. Because not all the patients were treated with the drugs that were used in this study, we examined the clinical records of those who were treated with taxol, cisplatin, carboplatin, or Adriamycin and compared the response of their cancer cells to incubation with these agents. We found that the cancer cells retained resistance to the chemotherapeutic agents reflective of the patient's history but were sensitive to other drugs that had not been used for treatment. This shows that the cancer cells in the body were directly resistant to the drugs, and the resistances to treatment were not due to reduced exposures from enhanced clearance or elevated metabolism of the drugs, although these factors cannot be excluded from the development of chemoresistance. Because the cancer cells showed little cross-resistance to other agents, it suggests that the resistance is due to altered drug metabolism in the cell that conferred resistance and not a result of enhanced efflux due to an overexpression of multidrug-resistant proteins. The exception may be the three isolates of cancer cells that were resistant to the cytotoxic effects of flavopiridol. These cells also showed increased resistance to other anticancer agents, suggesting the involvement of a multidrug resistant protein. Because overexpression of MDR-1 has modest effects on flavopiridol resistance (19), the most likely candidate may be ABCG2, a member of the ATP-binding cassette transporter family that was found to enhance flavopiridol resistance in MCF-7 breast cancer cell line (2022). Increased efflux has also been suggested as the cause of flavopiridol resistance in an ovarian cancer cell line although the protein responsible has not been identified (23).

To date, studies on biochemical and cytotoxic properties of flavopiridol have been done on established cell lines. Flavopiridol was the first specific inhibitor of cdks to undergo clinical trials, and in vitro testing showed antineoplastic activity. However, clinical trials have yielded mixed results. Using cancer cells isolated from the fluids that were drained from patients, we show here that flavopiridol was cytotoxic to the cells that had become resistant to standard anticancer drugs. Flavopiridol was equally effective against actively cycling and nonproliferating cells, suggesting that the compound's cytotoxic effects were not solely dependent on the inhibition of cdks that regulate the cell cycle but might have affected cdks that function in other cellular process, including transcription (24). This observation is consistent with published results with other cell lines and indicates that flavopiridol can induce cell death by other pathways (reviewed in ref. 2). However, in clinical trials, flavopiridol treatments have not consistently resulted in antitumor effects measured by complete or partial response in cancer patients (1, 10). It is possible that rapid clearance from the body can limit flavopiridol from inducing any apoptotic pathways. In a recent study, it has been found that the clearance of flavopiridol can vary by up to 10-fold between patients (24). Our studies with the MCF-7 breast cancer cell line show that a 1-day exposure to 100 nmol/L flavopiridol was sufficient to kill more that 90% the cells, and the absence of variance between the treatments could reflect the genetic homogeneity of the cultured cell line (Fig. 3). The result was more varied with the breast cancer cells taken from a patient, likely because these cancer cells were genetically heterogenous as they had not undergone clonal selection and would show some variations in sensitivity to drug treatments. The results shown in Fig. 3 (bottom) indicate that a subpopulation of these cancer cells had greater sensitivity and were eliminated by 25 nmol/L flavopiridol, with the remaining cells displaying varied sensitivities that corresponded to exposure periods. This may have implications in clinical applications of chemotherapy, indicating that an inadequate treatment period with flavopiridol may kill the more sensitive cancer cells in the patient, selecting for the more resistant subpopulation of cancer cells. The results show that, even at the higher concentration of 100 nmol/L, a 1-day treatment would leave ∼30% of cells and at least a 2-day treatment was required to kill >90% of the cells. Hence, the variable rate at which patients clear the drug from the body would affect the exposure of the cancer cells to flavopiridol and would affect the efficacy of treatment.

In our study, we were able to analyze cancer cells that were repeatedly drained from a patient with hepatic cancer who was treated with Adriamycin. In the early isolates, the cells were sensitive to Adriamycin. However, with subsequent isolates, the cancer cells showed a steady increase in resistance to Adriamycin. This result suggests that at the early stages of the disease most of the cancer cells had normal sensitivity to Adriamycin, and there may be a very small percentage that was resistant. This resistance may have arisen as a result of random mutations, and this random development of resistance may be a common occurrence during the growth of tumors in the body. With repeated exposures, the sensitive cells were gradually eliminated, allowing for the selection of cells that were resistant and finally leaving the drug-resistant cancers.

In summary, we have shown that cancer cells cultured from the ascites or pleural fluids of patients with metastatic cancers are useful tools for the study of cellular responses to anticancer agents and would complement the work done with established cell lines.

Grant support: Canadian Institute of Health Research (J.P.H. Th'ng and W. Duivenvoorden) and Prostate Cancer Research Foundation of Canada (W. Duivenvoorden).

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
Zhai S, Senderowicz A, Sausville E, Figg W. Flavopiridol, a novel cyclin-dependent kinase inhibitor, in clinical development.
Ann Pharmacother
2002
;
36
:
905
–11.
2
Senderowicz AM. The cell cycle as a target for cancer therapy: basic and clinical findings with the small molecule inhibitors flavopiridol and UCN-01.
Oncologist
2002
;
7
:
12
–9.
3
Sausville EA. Complexities in the development of cyclin-dependent kinase inhibitor drugs.
Trends Mol Med
2002
;
8
:
S32
–7.
4
Sausville EA. Cyclin-dependent kinase modulators studied at the NCI: pre-clinical and clinical studies.
Curr Med Chem Anti-Canc Agents
2003
;
3
:
47
–56.
5
Brusselbach S, Nettelbeck DM, Sedlacek HH, Muller R. Cell cycle-independent induction of apoptosis by the anti-tumor drug flavopiridol in endothelial cells.
Int J Cancer
1998
;
77
:
146
–52.
6
Bible KC, Kaufmann SH. Flavopiridol: a cytotoxic flavone that induces cell death in noncycling A549 human lung carcinoma cells.
Cancer Res
1996
;
56
:
4856
–61.
7
Sedlacek HH. Mechanisms of action of flavopiridol.
Crit Rev Oncol Hematol
2001
;
38
:
139
–70.
8
Elsayed YA, Sausville EA. Selected novel anticancer treatments targeting cell signaling proteins.
Oncologist
2001
;
6
:
517
–37.
9
Liu G, Gandara DR, Lara PN Jr, et al. A phase II trial of flavopiridol (NSC #649890) in patients with previously untreated metastatic androgen-independent prostate cancer.
Clin Cancer Res
2004
;
10
:
924
–8.
10
Shapiro GI. Preclinical and clinical development of the cyclin-dependent kinase inhibitor flavopiridol.
Clin Cancer Res
2004
;
10
:
4270
–5S.
11
Tan AR, Yang X, Berman A, et al. Phase I trial of the cyclin-dependent kinase inhibitor flavopiridol in combination with docetaxel in patients with metastatic breast cancer.
Clin Cancer Res
2004
;
10
:
5038
–47.
12
MacDougall JR, Matrisian LM. Targets of extinction: identification of genes whose expression is repressed as a consequence of somatic fusion between cells representing basal and luminal mammary epithelial phenotypes.
J Cell Sci
2000
;
113
:
409
–23.
13
Carmichael J, Degraff WG, Gazdar AF, et al. Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing.
Cancer Res
1987
;
47
:
936
–42.
14
Derjuga A, Richard C, Crosato M, et al. Expression of p21Waf1/Cip1 and cyclin D1 is increased in butyrate-resistant HeLa cells.
J Biol Chem
2001
;
276
:
37815
–20.
15
Li Y, Bhuiyan M, Alhasan S, et al. Induction of apoptosis and inhibition of c-erbB-2 in breast cancer cells by flavopiridol.
Clin Cancer Res
2000
;
6
:
223
–9.
16
Li Y, Chinni SR, Senderowicz AM, Sarkar FH. Induction of growth inhibition and apoptosis in prostate cancer cells by flavopiridol.
Int J Oncol
2000
;
17
:
755
–9.
17
Konig A, Schwartz GK, Mohammad RM, et al. The novel cyclin-dependent kinase inhibitor flavopiridol downregulates bcl-2 and induces growth arrest and apoptosis in chronic B-cell leukemia lines.
Blood
1997
;
90
:
4307
–12.
18
Newcomb EW, Tamasdan C, Entzminger Y, et al. Flavopiridol induces mitochondrial-mediated apoptosis in murine glioma GL261 cells via release of cytochrome c and apoptosis inducing factor.
Cell Cycle
2003
;
2
:
243
–50.
19
Boerner SA, Tourne ME, Kaufmann SH, Bible KC. Effect of P-glycoprotein on flavopiridol sensitivity.
Br J Cancer
2001
;
84
:
1391
–6.
20
Nakanishi T, Karp JE, Tan M, et al. Quantitative analysis of breast cancer resistance protein and cellular resistance to flavopiridol in acute leukemia patients.
Clin Cancer Res
2003
;
9
:
3320
–8.
21
Robey RW, Medina-Perez WY, Nishiyama K, et al. Overexpression of the ATP-binding cassette half-transporter, ABCG2 (MXR/BCRP/ABCP1), in flavopiridol-resistant human breast cancer cells.
Clin Cancer Res
2001
;
7
:
145
–52.
22
Doyle LA, Ross DD. Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2).
Oncogene
2003
;
22
:
7340
–58.
23
Bible KC, Boerner SA, Kirkland K, et al. Characterization of an ovarian carcinoma cell line resistant to cisplatin and flavopiridol.
Clin Cancer Res
2000
;
6
:
661
–70.
24
Rudek MA, Bauer KS Jr, Lush RM III, et al. Clinical pharmacology of flavopiridol following a 72-hour continuous infusion.
Ann Pharmacother
2003
;
37
:
1369
–74.