From www.bloodjournal.org by guest on November 19, 2014. For personal use only. 1996 88: 1198-1205 Combination of quinine as a potential reversing agent with mitoxantrone and cytarabine for the treatment of acute leukemias: a randomized multicenter study E Solary, B Witz, D Caillot, P Moreau, B Desablens, JY Cahn, A Sadoun, B Pignon, C Berthou, F Maloisel, D Guyotat, P Casassus, N Ifrah, Y Lamy, B Audhuy, P Colombat and JL Harousseau Updated information and services can be found at: http://www.bloodjournal.org/content/88/4/1198.full.html Articles on similar topics can be found in the following Blood collections Information about reproducing this article in parts or in its entirety may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests Information about ordering reprints may be found online at: http://www.bloodjournal.org/site/misc/rights.xhtml#reprints Information about subscriptions and ASH membership may be found online at: http://www.bloodjournal.org/site/subscriptions/index.xhtml Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036. Copyright 2011 by The American Society of Hematology; all rights reserved. From www.bloodjournal.org by guest on November 19, 2014. For personal use only. Combination of Quinine as a Potential Reversing Agent With Mitoxantrone and Cytarabine for the Treatment of Acute Leukemias: A Randomized Multicenter Study By Eric Solary, Brigitte Witz, Denis Caillot, Philippe Moreau, Bernard Desablens, Jean-Yves Cahn, Alain Sadoun, Bernard Pignon, Christian Berthou, FranGois Maloisel, Denis Guyotat, Philippe Casassus, Norbert Ifrah, Yves Lamy, Bruno Audhuy, Philippe Colombat. and Jean-Luc Harousseau A phase 111 prospective randomized multicenter study was performed t o determine whether quinine could improve the response rate of poor-riskacute leukemias (ALs) t o standard chemotherapyincluding a multidrug resistance (MDR)-related cytotoxicagent. The rationale of the study was based on thenegative prognostic value of MDR phenotype in ALs and the ability of quinine t o reverse this phenotype both in vitro and ex vivo. Three hundred fifteen patients (median age, 49 years; range, 16 t o 65) with relapsed (n = 108) or refractory (n = 32) acute myeloblastic leukemia (AML), relapsed (n = 27) or refractory (n = 9) acute lymphoblastic leukemia (ALL), secondary AL (n = 22) or blastic transformation of myelodysplastic syndrome ([MDS] n = 74) or myeloproliferative syndrome ([MPS] n = 4 3 ) were randomly assigned t o receive mitoxantrone ([MXNI 12 mg/m2/d, days 2 t o 5) and cytarabine ([Ara-Cl 1 g/m2/12 h, days 1 t o 5) alone or in combination with quinine (30 mg/kg/d, days 1 t o 5; continuous intravenous infusion beginning 24 hours before MXN infusion). Side effects of quinine were observed in 56 of 161 quinine-treated patients and disappeared in all but four cases after one or t w o 20% dose decreases. Sera from quinine-treated patients showed increased MXN uptake in an MDR-positive cell line compared with matched sera obtained before quinineinfusion. Quinine induced a significant increase in the incidence of nausea, vomiting, mucositis, and cardiac toxicity. A complete response (CR) was observed in 85 of 161 patients (52.896) from the quinine-treated group versus 70 of 154 patients (45.5%) in the control group(P= .19). The most important differences between quinine and control group CR rates were observed in patients with refractory AMLs and blastic transformation of MDS andMPS. The CR rate was higher in P-glycoprotein-positive cases, although the difference was not significant. Failure of the regimen due t o blastic persistence or blast number increase was higher in the control group (61 of 154 patients) than in the quinine group (45 of 161, P = ,041. Early death was observed in eight cases (four in each arm) and death in aplasia in 27 cases (20 in quinine group Y seven in control group, P = .01). The significant increase of toxicity in thequinine arm could have masked the clinical benefit of MDR reversion in poor-risk ALs. 0 1996 by The American Society of Hematology. T ciently in vitro by a number of noncytotoxic agents.’”’ The in vivo use of most of these agents is precluded by serum protein binding or clinical toxicity,”,“ and circumvention of the MDR phenotype has not yet proven clinically useful. We previously showed that intravenous infusion of conventional doses of quinine allowed attainment of a sufficient concentrationin serumto reverse the anthracyclineresistance of rat colon cancer cells and MDR human leukemic cells.’J~15Then,we defined theconditionsfor the use of 1/11 quinineas a MDR modifier”andperformedaphase clinical trial to demonstrate that quinine could beused safely in combinationwithmitoxantrone (MXN) and cytarabine (Ara-C) for thetreatment of clinically resistant ALS.’‘ Here, we report the results of a phase I11 multicenter clinical study designed to answer the question of whether the response rate of poor-risk ALstoconventionalchemotherapy could be improved by quinine. In thistrial,patients with either relapsed, secondary, or refractory AL were treated with acombination of MXN and high-dose Ara-C. After randomization, half the patients also received quinine as a potential MDRreversing agent. HEMAINREASONforthefailure of conventional regimens combining an anthracycline or an aminoanthraquinone with cytarabine or etoposide for the treatment of acute leukemias (ALs) is the primary or secondary resistance of leukemic cells to cytotoxic drugs.According to the Goldie and Coldman’hypothesis, these regimens select drugresistant clones that develop by spontaneous mutation or tumor celladaptation. In vitro cell culture systems have identified several mechanisms of cellular resistance to cytotoxic drugs. One of these mechanisms is characterized by the multidrug resistance (MDR) phenotype that is associated with increased expression of the mdrl gene product,called Pglycoprotein, in the leukemic cell plasma membrane.’ This increased expression has been identified in clinical samples, mainly from relapsed and refractory ALs, and related to a lower response rate to conventional treatment and a shorter event-free survival and overall ~urvival.~” Thedecreased intracellular accumulation of a variety of cytotoxic agents, characteristic of the MDR phenotype, can be reversed effi- From the Clinical Hematology Unit, Centre Hospitalirr Univer,sitaire (CHU) Le Bocage, Dijon, France. Submitted February 26, 1996; accepted April 12, 1996. Supported by grants from the Direction de la Recherche Clinique and the Ligue Bourguignonne Contre le Cancer. Address reprint requests to Eric Solary,MD, Clinical Hematology Unit, CHU Le Bocage, BP1542, 21034 Dijon Cedex, France. The publication costs of this article were defrayedin part by page chargepayment. This article must thereforebeherebymarked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact. 0 1996 by The American Society of Hematology. 0006-4971/96/8804-0020$3.00/0 1198 PATIENTS AND METHODS Patients. The study was an open phase 111 prospective randomized trial with15 participating medical centers, performed from March 1992 to February 1995. The protocol received approval from the ethics board of the Dijon hospital. Before therapy, all patients provided informed consent after having been advised about the purpose and investigational nature of the study, as wellas potential risks. Patients eligible for the study were older than 14 and younger than 66 years, with a bone marrow diagnosis of acute nonlymphoblastic or acute lymphoblastic leukemia (ALL) as defined by the French-American-British classification system.” These patients eiBlood, Vol 88, NO 4 (August 15). 1996: pp 1198-1205 From www.bloodjournal.org by guest on November 19, 2014. For personal use only. 1199 MDR REVERSION IN ACUTELEUKEMIAS ther had relapsed from or were refractory to standard first-line chemotherapy. Refractory patients were defined as those with de novo AL failing to achieve complete remission (CR) after one or two cycles of chemotherapy. Relapsed patients included those with acute nonlymphoblastic leukemia or ALL who achieved a previous CR with a standard therapeutic regimen, which sometimes included autologous or allogeneic bone marrow transplantation. Patients with secondary leukemia or blastic transformation of myelodysplastic (MDS) or myeloproliferative (MPS) syndromes were also eligible. Other eligibility criteria included adequate kidney and hepatic function (creatinine <250 pmol/L, serum bilirubin < 14 m&, and transaminases <4N), adequate cardiac ejection fraction as measured by echocardiography (>30%) or radionuclide ejection (>50%), and cumulative dose of anthracycline less than 400 mg/m2 adriamycinequivalent. Patients with recent myocardial infarction or vascular cerebral infarction and those with known hypersensitivity to quinine were ineligible. Random permuted blocks of 10 were used to generate the randomization list. No stratification was used.All randomizations were performed by the Clinical Hematology Unit of the Dijon hospital. The data were collected centrally by this office and were subsequently reviewed by three clinicians. Regimen. The therapy regimen consisted ofAra-C 1 g/m' administered by 2-hour intravenous infusion twice daily on days 1 through 5 and MXN 12 mg/m' as a 30-minute infusion on days 2 through 5. After randomization, half the patients received quinine formiate (Quinimax; Labaz Laboratories, Paris, France) at a dosage of 30 mg/kg/d started 24 hours before the first dose of MXN and administered as a continuous intravenous infusion until 24 hours after the end of the last MXN infusion. Toxicity was assessed according to the World Health Organization (WHO) grading system. CR was defined by the presence of less than 5% leukemic blast cells in the bone marrow, their disappearance from the blood and possible extramedullary sites, including the cerebral fluid, and an increase of the peripheral granulocyte count to greater than 1,00O/pL. Partial response was defined by a percentage of bone marrow blast cells less than 25%, a percentage of peripheral blood blast cells less than 5%, and peripheral blood granulocyte count higher than l,OOO/pL. In patients in CR, the duration of critical cytopenia was evaluated by the time required for leukocyte (>500/pL), granulocyte (>500/ pL), and thrombocyte (> 100,OOO/pL) recoveries and the red blood cell transfusion requirement (hemoglobin <80 g/L) from the onset of treatment. Quantzjication of quinine concentrations in semm. Total quinine concentrations were determined as previously described.18 Patient serum was mixed with 0.5 mol/L borate buffer, pH 9.8, and hydroquinidine as internal standard. Extraction was performed with a mixture of dichloromethane and isoamylic alcohol (98:2 vol/vol). After centrifugation, the organic phasewas evaporated to dryness, dissolved in the mobile phase (potassium dihydrogen phosphate 0.045 moUL, acetonitrile 4:1 vol/vol, pH 3.8), and injected into the highperformance liquid chromatography (HPLC) apparatus equipped with a U6K injector and a M45 pump (Waters Associates, SaintQuentin-en-Yvelines, France). The column was a Nova-pak C18 (150 X 3.9 mm, 5 pm). Detection was made with a FP 210 spectrofluorometer (Prolabo, Vaux-en-Velin, France) with excitation and emission wavelengths of 350 and 440 nm, respectively. Retention times were 2.4 and 3 minutes for quinine and hydroquinidine, respectively. Ex vivo assay. To determine the ability of serum from quininetreated patients to increase the MXN intracellular concentration by reversing the MDR phenotype, we designed an ex vivo flow-cytometry assay using the CEMNLB human leukemic cell line (Genne et al, manuscript submitted). This cell line overexpressed both mdrl mRNA and P-glycoprotein (not shown). CEMNLB cells (2 x IOs) were seeded in microtiter plates (24 wells/plate) and cultured for 24 hours, and then incubated for 2 hours at 37°C with 20 pmoUL MXN diluted in serum samples obtained from patients either before or 2 days after the beginning of quinine infusion. After a 2-hour incubation, cells were rinsed three times with ice-cold phosphate-buffered saline and resuspended in phosphate-buffered saline (PBS) for measurement of MXNintracellular accumulation, using a Becton Dickinson (Pont de Claix, France) FACScan flow cytometer at an excitation of 488 nm and an emission of 675 nm. Detection of P-glycoprotein. P-glycoprotein expression was determined using MRK16, a monoclonal antibody that recognizes the extracellular determinant of P-glycoprotein (Immunotech, Luminy, France), and a previously described indirect immunofluorescent assay." Briefly, cells werefixed in 1% paraformaldehyde for 30 minutes at 4°C and incubated inhuman AB serum before a 30minute incubation with MRK16 antibody (2 pg/mL), and then with fluorescein-conjugated F(ab'), fragments of goat anti-mouse IgG (37.5 pg/mL; Silenius Laboratories, Paris, France). The percentage of positive cells was determined by cytofluorimetry on gated blast cells using a Becton Dickinson FACScan flow cytometer. A nonrelesame goat vant IgG2a (Coulter, Raritan, NJ) wasusedwiththe antimouse second-step reagent to calibrate the assay on the FACScan and determine the positivity for individual cells. P-glycoproteinpositive K562/ADM cells were used as a positive control. Various cutoff values for positivity were tested. Statistical analysis. According to previous studies, the CR rate induced by the MXN-Ara-C regimen was approximately 50%.19-2' The study was designed to detect a 20% improvement intheCR rate (two-sided test) with a significance level of .05. A total of 137 patients per treatment group was calculated to be necessary to reach this goal, whichwas expected to require 3 years. At the time of statistical analysis, categorical data obtained from the patients before treatment and their response rates were compared by the x' test or Fisher's exact test in cases of small expected values. The MantelHaenszel test was used for adjustment ofthe results for thesex imbalance. The t test for nonpaired samples was used to compare quantitative parameters. A two-way ANOVA was performed to compare hepatic enzymes and bilirubinemia changes during treatment in both arms of the trial. A stepwise logistic regression model was used to assess the joint effect of prestudy variables on response. Validity of the model was evaluated by the Homer-Lameshow test. Statistical analyses were made with either Statview (Alsyd, Meylan, France) or BMDP software systems. RESULTS Characteristics of patients. From March 1992 to February 1995, 328 patients entered the study. Data were obtained from 318 patients. One patient was not eligible (diagnosis error), and two were not assessable (one did not receive the treatment, and the other died after randomization and before treatment onset). Three hundred fifteen patients were assessable: 161 received quinine, and 154 were treated without quinine. Despite the lack of stratification at initial randomization, the two arms of the study were well balanced in the 15 centers. Initial diagnoses of these assessable patients and their pretreatment characteristics are summarized in Table 1. MPSs included chronic myelogenous leukemias (n = 32), polycythemia vera (n = 6), essential thrombocytemia (n = 3), and myelofibrosis (n = 2). The only significant imbalance in the pretreatment variables was the higher number of women in the control arm ( P = .03). Analysis of response rates was adjusted for this imbalance. Serum quinine levels and MDR-reversing activity of sera. Serum samples from 40 patients (n = 20 per treatment arm) From www.bloodjournal.org by guest on November 19, 2014. For personal use only. SOLARY ET AL 1200 Table 1. Pretreatment Characteristics of Patients at Study Entry Characteristic Median age (yr) 46.0 Sex (n) Male Female Previous MDR-related drugs (n)" No Yes Diagnosis (n) Refractory AML Relapsed AML Refractory ALL Relapsed ALL Secondary AL M DS M PS Previous bone marrow transplantation (n) None Autologous Allogeneic Duration of first CR (mo) Median Range (n) Time between chronic and acute-phase MDS and MPS (mo) Median Range (n) Performance status, WHO grade (n) 0 1I2 314 Extramedullary site (n) No Yes White blood cell count at diagnosis (per bL) Median Mean Range Mean hemoglobin (g/dL) Mean platelet count (x1.0001~L) Mean peripheral blood blast cell count (%) Mean marrow blast cell infiltration (%) Without Quinine (n = 154) With Quinine (n = 161) 49.5 Total ( n = 315) P 49.0 NS 84 70 107 54 191 124 .02 53 101 58 102 111 203 NS 15 55 5 13 12 36 18 17 53 4 14 10 38 25 32 NS 108 9 27 22 74 43 146 4 4 150 9 2 296 13 6 NS 11.0 0-288 91 8.0 0-70 88 9.0 0-288 179 NS 17.5 0-157 54 15.0 0- 180 63 16.0 0-180 117 NS 23 117 14 28 115 18 51 232 32 NS 119 35 127 33 246 68 NS 3.7 23.4 0.2-425 98.6 93.2 29.7 57.2 5.5 23.5 0.7-256 99.6 86.7 20.7 53.0 4.7 23.5 0.2-425 99.1 89.9 20.7 55.1 NS NS NS NS NS Abbreviation: NS, not significant. MDR-related drugs included anthracyclines, vinca alkaloids, and epipodophyllotoxins. were tested before and at day 2 of treatment. In quininetreated patients, sera obtained on day 2 showed increased MXN uptake in CEMNLB cells compared with matched sera obtained before treatment (mean k SD, 43% 2 30%; range, 13% to 117%).The mean concentration of quinine in these samples was 6.5 pg/mL (range, 1.5 to 13.0). When tested under the same conditions, sera from control patients did not demonstrate any significant effect on MXN accumulation (data not shown). When various concentrations of quinine were added to a control serum, a linear relationship was observed between quinine concentration and MXN uptake in CEMNLB cells incubated for 2 hours in this serum (R2 = .97, P = .0003; Fig 1A). The same experiment was per- formed with sera obtained before and 2 days after the beginning of treatment in 20 quinine-treated and 20 control patients. Although less significant (R2 = .67, P = .03; Fig lB), a linear correlation was again found between quinine serum level measured by HPLC and MXN uptake in CEMNLB cells. Toxicity of quinine. Adverse effects due to quinine infusion were observed in 56 of 161 quinine-treated patients (34.8%) and included tinnitus (45 of 161,28%),vertigo (24 of 161, 14.9%),and tachycardia or bradycardia (six of 161, 3.7%). Mild hearing loss was also reported in 10 patients (6.2%). In 48 patients, quinine-induced side effects decreased or disappeared after a unique 20% quinine dose From www.bloodjournal.org by guest on November 19, 2014. For personal use only. 1201 MDRREVERSIONINACUTELEUKEMIAS T e V 0 4 8 12 l 0 e 4 8 1 2 1 6 Quinine serum level (pglml) Fig 1. Effects of serum M X N uptake in MDR-positive CEMlVLB cells. (A) Effects of various concentrations of quinine added to a control serum (RZ= .97). (B) Relationships between serum quinine level determined by HPLC in serum samples from patients and M X N uptake in CEM/VLB cells (R2= ,671. decrease. In four patients, an additional 20% dose decrease was necessary. In the last four patients. quinine infusion was stopped after day 2 (one patient) or day 3 (three patients), due to an excessive QT interval increase or bradycardia. The mean age of patients who demonstrated hearing loss was significantly higher than the mean age of patients who did not report a hearing change (53.5 5 13 1’44.2 ? 14.2 years, P < .OS). No relationship was found between side effects and other pretreatment characteristics of patients. In quininetreated patients, the occurrence of quinine-related side effects was associated with a significant increase in the timeto discharge from hospital (37.8 ? 11.7 11 31.4 ? I I .0 days, P < .0008) and leukopenia duration (27.7 ? 1 1.4 1’ 22.3 ? 8.7, P = .02). By contrast, no difference was observed between quinine-treated patients who did not receive quinine and the control group. Nonhematopoietictoxicity. The nonhematopoietic toxicity associated with the regimen is reported in Fig 2. Quinine infusion significantly increased the incidence of nausea, vomiting, and .mucositis. The duration of these side effects was slightly increased in quinine-treated patients, but the differences did not reach significance. By contrast, quinine had no influence on renal and hepatic toxicity and did not induce a significant increase of bilirubinemia. Although cardiotoxicity was reported in only six patients during quinine perfusion, late toxicity (mainly cardiac failure) was observed in 10 additional patients from the quinine arm. compared with two patients from the control arm ( P = .03). Twelve of 16 patients who demonstrated cardiotoxicity were older than 50 years. In six patients, cardiotoxicity was reported in the absence of previous anthracycline treatment. Hrntatologictoxicity. Among responding patients, quinine slightly increased the median timeto leukocyte recovery over SOO/pL, granulocyte recovery over 500/pL, platelet recovery over 50.000/pL. and hemoglobin recovery over 8 g/ dL (Table 2). However, only the time to platelet count recovery in the relapsed AML group was significantly increased by quinine (mean, 35.6 2 12.5 v 29.4 ? 9.9 days. P = .037). In this group of patients, quinine also significantly increased the meantime to discharge fromhospital (38.1 ? 10.0 v 32.1 ? 10.0 days, P = .007). Seven responding patients from the control group and ninefrom the quinine-treated group never recovered a platelet count over 100 X I O 9 L Similarly. 13 responding patients from the control group and I I patients from the quinine-treated group required red blood cell transfusions for longer than 60 days. Quinine did not significantly increase the incidence of febrile episodes or aspergilloses (Table 2). Response to treatment. CR rates are listed in Table 3. Univariate analysis indicated that the overall CR rate was associated with younger age. lower white blood cell count, and lower percentages of peripheral blood and marrow blast cells at diagnosis (not shown). When these parameters were entered in the logistic regression in association with quinine and sex, a higher CR rate was related only to age less than 40, lower white blood cell count, and lower percentage of marrow blast cells. The CR rate was higher in the quinine120 100 Heart p = 0.03 51 1 0 1 2 3 4 0 1 WHO grade 2 3 4 Quinine w/o Quinine 0 1 2 3 4 WHO grade Fig 2. Effects of quinine infusion on the incidence and severom (according to the WHO grading system) of side effects induced by chemotherapy. NS, not statistically significant. From www.bloodjournal.org by guest on November 19, 2014. For personal use only. 1202 SOLARY ET AL Table 2. Hematologic Toxicity of the Two Arms of the Trial and Its Consequences on the Duration of Hospitalization and Febrile Episodes Without Quinine ToxicityIEffect Leukopenia (d) Mean ? SD Median Neutropenia (d) Mean i SD Median Thrombopenia (d) Mean ? SD Median Anemia (d) Mean f SD Median Time to discharge from hospital (d) Mean 2 SD Median No. of febrile episodes No. of CR/No. of Cases (%) Without Quinine P With Quinine 22.3 -t 7.9 22.0 23.9 f 9.1 .26 23.0 25.9 ? 7.8 25.0 28.2 i 9.2 .l0 26.0 P-GlycoproteinPositive Cells (%) 2 1 25 210 28.6 t 10.0 31.6 ? 11.5 .l1 32.0 27.0 29.0 t 8.8 28.0 32.1 t 12.0 .l0 30.0 35.5 f 17.9 35.8? 8.8 33.0 32.5 1 2 3 4 5 6 No. of aspergillosis episodes Table 4. Relationships Between P-Glycoprotein Expression and CR Rate 22 8 67 56 18 5 0 27 6 51 61 27 14 1 NS NS NS Abbreviation: NS, not significant. treated group (85 of 161 patients, 52.8%) than in the control group (70 of 154 patients, 45.5%), but the difference was not significant ( P = .19). The estimated relative odds ratio of CR (quininekontrol) adjusted for sex was 1.32 (confidence interval, 0.81 to 2.00). Quinine significantly improved the CR rate in previously treated patients who relapsed after a first CR shorter than 18 months (42 of 76 patients in the quinine arm v 26 of 67 in the control arm, P < .05). In MDS and MPS, the CR rate was significantly improved by quinine when the delay from chronic phase to blastic phase was less than 48 months (eight of 14 patients in the quinine arm v 26 of 67 in the control arm, P < .05). Failure of the regimen due to blastic persistence or blast number increase was higher in the control group (61 of 154 patients) than in the quinine group (45 of 161 patients, P = No. With Quinine % 40 14/35 7120 15/2554 44 4/13 % No. 28145 .05 62 60 9/17.28 53 P .09 .04). Eight patients died during treatment (four in each arm). Twenty-seven patients died in aplasia after day7:seven were inthe control group (refractory AML, one; relapsed AML, three; and blastic phase of MDS, three) and 20 in the quinine group (refractory AML, two; relapsed AML, six; relapsed ALL, four; secondary AL, two; blastic phase of MDS, three; and blastic phase of MPS, 3; P = .01). Postremission therapy was heterogeneous. The median duration of CR was 6.0 months and median survival was 11.5 months, without significant differences between control and quininetreated groups. Relationships between response rate and P-glycoprotein expression. P-glycoprotein expression was determined on blast cells from 102 patients (control arm, 47; quinine arm, 5 5 ) using MRK16 monoclonal antibody. Its expression ranged from 0% to 86% of positive cells. This expression was greater than 5% in 45 patients and greater than 10% in 30 patients, without a significant difference between treatment groups. The CR rate was higher in P-glycoproteinpositive cases at all tested cutoff values for P-glycoprotein positivity (Table 4). However, these differences never reached statistical significance. Similarly, no relationship was found between either CD34 expression on blast cells or other biologic or clinical pretreatment parameters and the influence of quinine on the response rate (not shown). DISCUSSION Although there is evidence that MDR phenotype is a prognostic factor in ALs, the issue of whether P-glycoprotein is a useful therapeutic target is still unresolved. Combined therapy with MDR-related drugs and P-glycoprotein modulators prolonged the life span in some animal and en- Table 3. Number of Patients Who Achieved CR Related to the Total Number of Patients in Each Group Without Quinine Group All patients Refractory AML Relapsed AML Refractory ALL Relapsed ALL Secondary AL Blastic phase of MDS Blastic phase of MPS % No. 851161 7011 54 4115 33/55 315 4113 511 2 16/36 10125 511 8 Abbreviation: NS, not significant. With Quinine 411 511 45.5 26.7 60.0 60.0 30.8 41.7 50.0 44.4 27.8 No. 1551315 12/32 8117 671108 34/53 619314 8/27 4 10/22 0 237/74 1/38 15/43 Overall % 52.8 47.1 64.2 75.0 28.6 50.0 55.3 40.0 No. % P 49.2 37.5 62.0 66.7 29.6 45.4 NS NS NS NS NS NS NS NS 34.9 From www.bloodjournal.org by guest on November 19, 2014. For personal use only. MDR REVERSION IN ACUTELEUKEMIAS 1203 couraging results have been obtained in individual cases23 the brain stem. Nausea and vomiting could also be the consequence of an increased drug exposure as an effect of quinine and phase VI1 pilot studies.'6324Here, we report the first infusion on MXN pharmacokinetics in humans.29 Bothveralarge randomized trial that evaluates the clinical relevance pamil and cyclosporin were reported to increase the plasma of MDR reversion in ALs. Although this study failed to area under the curve of several MDR-related drugs in anidemonstrate a significant increase in the response rate for the mals Cinchonine, a diastereoisomer of quinine arm, several arguments suggest that MDR reversion and quinine and a potent MDR-reversing agent in vitro and in could improve the efficacy of conventional treatments invivo, did not modify doxorubicin pharmacokinetics in rat cluding an MDR-related agent. The most important effects but modified the distribution of the cytotoxic drug in several of quinine on the response rate were observed in refractory tissues.33 Such a pharmacokinetic effect could explain the AML and blastic transformation of MDS and MPS, which increased incidence of mucositis and could be involved in are also the most resistant ALs to conventional therapy. the increased cardiotoxicity of the regimen observed mainly Moreover, quinine induced a significant decrease in the failin older patients.34Whatever its mechanism, this significant ure rate due to blastic persistence or blastic growth during increase in toxicity of the regimen could have masked the treatment. Lastly, there was a trend for quinine to improve clinical benefit of MDR reversion, and better management the response rate in P-glycoprotein-positive ALs. Therefore, of this toxicity could reveal the clinical advantage of the quinine could be useful in a selected group of poor-risk combination. leukemias that overexpress P-glycoprotein. P-glycoprotein is expressed only in a subset of patients, The lack of significant improvement in the overall reand leukemic cells without P-glycoprotein should not response rate could be related to insufficient potency of quinine spond toattempted modulation. Previous studies have shown as MDR-reversing agent. Quinine andmany other agents that the number of P-glycoprotein-expressing cases was tested so far are first-generation chemosensitizers that have higher in poor-risk ALs than in de novo ALs.3-7.35 However, been developed for pharmacologic effects other than MDR results are discordant from one study to another, depending circumvention. All these agents increase accumulation of on the disease and the technique chosen to identify MDR MDR-related drugs in MDR-positive cells in Quinine p h e n ~ t y p e Using . ~ ~ MRK16 monoclonal antibody to detect also allows subcellular redistribution of doxorubicin in a P-glycoprotein in 102 assessable patients, MDR phenotype MDR-positive leukemic cell line.26Many of the potent rewas identified in a limited number of cases, suggesting that versing agents in vitro are too toxic in vivo at doses that only a minority of the quinine-treated patients could have mimic the conditions in vitro.13 Consequently, a number of been sensitive to MDR-reversing activity in our series of studies actually used suboptimal concentrations. Even when patients. Moreover, some P-glycoprotein-positive leukemic the chemosensitizer plasma level is within the range of concentrations active in vitro, protein binding could limit its cells could not respond to quinine because of a lack of functional P-glycoprotein. The extent to which mutations and ability to reverse MDR in V ~ V O . ' * * ' ~ The * ~ ~ MDR-reversing polymorphism of P-glycoprotein may confer altered binding activity of quinine is retained in serum despite serum protein specificity in leukemic cells is notknown.37Functional MDR binding. The ability of serum to increase MXN accumulation phenotype was reported to be predominant in CD34+ leukein a MDR-positive leukemic cell line was linearly related mic cells, whereas P-glycoprotein could not be functional in to quinine level in the serum of quinine-treated patients. CD34- cells.38Nevertheless, no significant relationship was Conversely, the ability of serum samples from control pafound between CD34 expression and the response to treattients to reverse MDR phenotype did not increase during ment in 136 patients in whom its expression was studied cytotoxic treatment (data not shown). However, quinine se(data not shown). rum levels remained sometimes as low as 1.5 kg/mL at day 2 of continuous intravenous infusion. Tinnitus and vertigo Two other reasons could account for the lack of clinical are well-known complications of quinine administration benefit from quinine in the present study. EvenwhenPwhose appearance was previously related to the highest quiglycoprotein is present and functional, its inhibition could nine serum levels.27A significant relationship was observed not be sufficient to render the leukemic cells sensitive between the occurrence of these side effects and the inenough to a MDR-related cytotoxic drug, due to thepresence creased duration of neutropenia in responding patients. This of other mechanisms of resistance in poor-risk A L s . ~Ac~ later effect could be explained by either the reversion of Pcordingly, quinine and other reversing agents could be more glycoprotein function inbonemarrow precursors28or the potent in MDR-positive de novo ALs inwhich other mechaalteration of MXN and Arg-C metabolism or excretion by nisms of resistance might be less present. Lastly, high doses normal tissues that express the MDR phen~type.'~ Only one of Ara-C were demonstrated to induce high response rates third of the patients reported quinine-related side effects, in poor-risk ALs.@Ara-C is not involved in MDR, and idensuggesting that the quinine serum level could have been too tification of both MDR phenotype and resistance to Ara-C low to efficiently inhibit P-glycoprotein in a majorityof was shown recently to better identify the more resistant patients. ALs.~'Therefore, the clinical benefit of quinine could have P-glycoprotein is expressed in endothelial cells of the cenbeen masked by the efficacy of Ara-C. tral nervous system and is thought to contribute to the bloodSeveral recent randomized studies also failed to show any brain barrier.30 Therefore, the increase of nausea and benefit of MDR-reversing agents, including quinidine and vomiting in quinine-treated patients could be related to the verapamil, in terms of response to chemotherapy or length increased distribution of MXN to the vomiting centers of of survival in patients with multiple myeloma, small-cell From www.bloodjournal.org by guest on November 19, 2014. For personal use only. 1204 SOLARY ET AL lung cancer, and breast ~ a n c e r . ~These ~ - ~ ’ negative results should not prevent further trials that testthe potential interest of these and more potent MDR-reversing agents in ALs. Better management of the regimen toxicityor the use of second-generation chemosensitizers that have been designed in this respectzs could improve either the response rate or the survival. While waiting for the possibility of testing new compounds in randomized controlled trials, the Groupe Est Ouest LeucCmies Aigues MyCloblastiques has initiated a phase 111 study in de novo AML testing the effect of quininemediated MDR reversion on survival. ACKNOWLEDGMENT The authors gratefully acknowledge Bruno Chauffert and Philippe Genne for a critical reading of the manuscript, and Serge Aho for advice about statistical analysis. This study was performed on behalf of the GOELAMS group. REFERENCES 1. Goldie JH, Coldman AJ: Quantitative model for multiple levels of drug resistance in clinical tumors. Cancer Treat Rep 67:923, 1983 2. Gottesman MM, Pastan I: Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem 62:385, 1993 3. Marie JP, Zittoun R, Sikic BI: Multidrug resistance (mdrl) gene expression in adult acute leukemias: Correlations with treatment outcome and in vitro drug sensitivity. Blood 78586, 1991 4. Campos L, Guyotat D, Archimbaud E, Calmard-Oriol P, TSUNOT, Troncy J, Treille D, Fiere D: Clinical significance of multidrug resistance P-glycoprotein expression on acute nonlymphoblastic leukemia cells at diagnosis. Blood 79:473, 1992 S. Pirker R, Wallner J, Geissler K, Linkesch W, Haas OA, Bettelheim P, Hopfner M, Scherrer R, Valent P, Havelec L, Ludwig H, Lechner K: MDRl gene expression and treatment outcome in acute myeloid leukemia. J Natl Cancer Inst 83:708, 1991 6. Goasguen JE, Dossot JM, Fardel 0, Le Mee F, Le Gall E, Leblay R, LePrise PY, Chaperon J, Fauchet R: Expression of the multidrug resistance-associated P-glycoprotein (P-170) in S9 cases of de novo acute lymphoblastic leukemia: Prognostic implications. Blood 81:2394, 1993 7. Holmes J, Jacobs A, Carter G, Janowska WA, Padua RA: Multidrug resistance in haematopoietic cell lines, myelodysplastic syndromes and acute myeloblastic leukemia. Br J Haematol 72:40, 1989 8. Tsuruo T, Iida H, Tsukagoshi S, Sakurai Y: Overcoming of vincristine resistance in P388 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil. Cancer Res 41:1697, 1981 9. Chauffert B, Martin M, Hammann A, Michel MF, Martin F: Amiodarone-induced enhancement of doxorubicin and 4”deoxydoxorubicin cytotoxicity to rat colon cancer cells in vitro andin vivo. Cancer Res 465325, 1986 IO. Ford JM, Hait WN: Pharmacology of drugs that alter multidrug resistance in cancer. Pharmacol Rev 42:155, 1990 1 1. Zamora JM, Pearse HL, Beck WT: Physical properties shared by compounds that modulate multidrug resistance in human leukemic cells. Mol Pharmacol 33:454, 1988 12. Genne P, Dimanche-Boitrel MT, Mauvernay RY, Gutierrez G, Duchamp 0, Petit JM, Martin F, Chauffert B: Cinchonine, a potent efflux inhibitor to circumvent anthracycline resistance in vitro and in vivo. Cancer Res 52:2797, 1992 13. Pennock GD, Dalton WS, Roeske WR, Appleton CP, Mosley K, Plezia P, Miller TP, Salmon SE: Systemic toxic effects associated with high-dose verapamil infusion and chemotherapy administration. J Natl Cancer Inst 83:105, 1991 14. Chauffert B, Pelletier H, Corda C, Solary E, Bedenne L. Caillot D, Martin F: Potential usefulness of quinine for the circumvention of the anthracycline resistance in clinical practice. Br J Cancer 62:39S, 1990 1.5. Solary E, Velay I, Chauffert B, Bidan JM, Caillot D, Dumas M, Guy H: Sufficient levels of quinine in the serum circumvent the multidrug resistance of the human leukemic cell line K562/ADM. Cancer 68: 17 14, l99 I 16. Solary E, Caillot D, Chauffert B, Casasnovas RO, Dumas M, Maynadie M, Guy H: Feasibility of using quinine, a potential multidrug resistance-reversing agent, in combination with mitoxantrone and cytarabine for the treatment of acute leukemia. J Clin Oncol 10: 1730, I992 17. Benett JM, Catovsky D, Daniet MT, Flandrin G, Galton DAG, Gralnick HR, Sultan C: Proposals for the classification of the acute leukemias. Br J Haematol 33:451, 1976 18. Solary E, Bidan JM, Calvo F, Chauffert B, Caillot D, Mugneret F, Gauville C, Tsuruo T, CarliPM,GuyH: P-glycoprotein expression and in vitro reversion of doxorubicin resistance by verapamilin clinical specimens from acute leukaemia and myeloma. Leukemia 5:592, 1991 19. Hiddemann W, Kreutzmann H, Sraif K, Ludwig WD, Mertelsmann R, Donhuijsen-Ant R, Lengfelder E, Arlin Z, Biichner T: High-dose cytosine arabinoside and mitoxantrone: A highly effective regimen in refractory acute myeloid leukemia. Blood 69:744, 1987 20. Bezwoda WR, Bernasconi C, Hutchinson RM, Winfield DA, de Bock R, Mandelli F: Mitoxantrone for refractory and relapsed acute leukemia. Cancer 66:418, 1990 21. O‘Brien S, Kantarjian H,Estey E, Koller C, BeranM, McCredie K, Keating M: Mitoxantrone and high-dose etoposide for patients with relapsed or refractory acute leukemia. Cancer 68:691, 1991 22.Loor F, BoeschD, Gaveriaux C, Jachez B, Pourtier-Manzanedo A, Emmer G: SDZ 280-446, a novel semisynthetic cyclopeptide: In vitro and in vivo circumvention of P-glycoprotein mediated tumor cell multidrug resistance. Br J Cancer 65: 11, 1992 23. Sonneveld P, Nooter K: Reversal ofdrug resistance by cyclosporin-A in a patient with acute myelocytic leukaemia. Br J Haematol 75:208, 1990 24. List AF, Spier C, Greer J, Wolff S, Hutter J, Dorr R, Salmon S, Futscher B, Baier M, Dalton W: Phase I/II trial of cyclosporine as a chemotherapy resistance modifier in acute leukemia. J Clin Oncol 11:1652, 1993 25. Lehnert M: Reversal of P-glycoprotein-associated multidrug resistance: The challenge continues. Eur J Cancer 29:636, 1993 26. Bennis S, lchas F, Robert J: Differential effects of verapamil and quinine on the reversal of doxorubicin resistance in a human leukemia cell line. Int J Cancer 62283, 1995 27. Boland ME, Brennand Roper SM, Henry JA: Complications of quinine poisoning. Lancet 1:384, 1985 28. Chaudhary PM, Roninson IB: Expression and activity ofPglycoprotein, a multidrug efflux pump, in humanhematopoietic stem cells. Cell 6 6 3 5 , 1991 29. Lum BL, Fisher GA, Brophy NA, Yahanda AM, Adler KM, Kaubisch S, Halsey J, Sikic BI: Clinical trials of modulation of multidrug resistance. Pharmacokinetic and pharmacodynamic considerations. Cancer 72:3502, 1993 30. Cordon-Cardo C, O’Brien JP, Casals D, Rittman-Grauer L, Biedler JL, MelamedMR, Bertino JR: Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci USA 86:695, 1989 31. Nooter K, Oostrum R, Deurloo J: Effects of verapamilon From www.bloodjournal.org by guest on November 19, 2014. For personal use only. MDR REVERSION IN ACUTELEUKEMIAS the pharmacokinetics of daunorubicin in the rat. Cancer Chemother Pharmacol 20:176, 1987 32. Marie J P , Bastie J N , Coloma F, Faussat Suberville AM, Delmer A, Rio B, Delmas-Marsalet B, Leroux G, Casassus P, Baumelou E, Catalin J, Zittoun R: Cyclosporin A as a modifier agent in salvage treatment of acute leukemia (AL). Leukemia 7:821, 1993 33. Genne P, Duchamp P, Solary E, Magnette J, Belon JP, Chauffert B: Cinchonine per os: Efficient circumvention of P-glycoprotein-mediated multidrug resistance. Anticancer Drug Design 10:103, 1995 34. Bright JM, Buss DD: Effects of verapamil on chronic doxorubicin-induced cardiotoxicity in dogs. J Natl Cancer Inst 82:963, 1990 35. Arceci RI: Clinical significance of P-glycoprotein in multidrug resistance malignancies. Blood 81:2215, 1993 36. Haber DA: Multidrug resistance (MDR 1) in leukemia: Is it time to test? Blood 79:295, 1992 37. Holmes JA, Whittaker JA, Padua R: Effect of position 185 mutations of the MDRl gene on drug resistance in leukemia. Leukemia 6:484, 1992 38. Bailly JD, Muller C, Jaffrezou JP, Demur C, Cassar G, Bordier C, Laurent G: Lack of correlation between expression and function of P-glycoprotein in acute myeloid leukemia cell lines. Leukemia 9:799, 1995 39. Harker WG, Slade DL, Dalton WS, Meltzer PS, Trent JM: Multidrug resistance in mitoxantrone-selected HL-60 leukemia cells in the absence of P-glycoprotein overexpression. Cancer Res 49:4542, 1989 1205 40. Herzig RH, Wolff SN, Lazarus HM, Phillips GL, Karanes C, Herzig GP: High-dose cytosine arabinoside therapy for refractory leukemia. Blood 62:361, 1983 41. Schuurhuis GJ, Broxterman HJ, Ossenkoppele GJ, Baak JPA, Eekman CA, Kuiper CM, Feller N, van Heijningen TI", Klumper E, Pieters R, Lankelma J, Pinedo HM: Functional multidrug resistance phenotype associated with combined overexpression of Pgpl senMDRl and MRP together with l-0-D-arabinofuranosykytosine sitivity may predict clinical response in acute myeloid leukemia. Clin Cancer Res 1:81, 1995 42. Milroy R: A randomised clinical study of verapamil in addition to combination chemotherapy in small cell lung cancer. Br J Cancer 68:813, 1993 43. Millward MJ, Cantwell BMJ, MUNONC, Robinson A, Conis PA, Harris AL: Oral verapamil with chemotherapy for advanced non-small cell lung cancer: A randomized study. Br J Cancer 67:1031, 1993 44. Wishart GC, Bisett D, Paul J, Jodrell D, Harnett A, Habeshaw T, KenDJ, Macham MA, Soukop M, Leonard RCF, Knepil J, Kaye SB: Quinidine as a resistance modulator of epirubicin in advanced breast cancer: Mature results of a placebo-controlled randomized trial. J Clin Oncol 12:1771, 1994 45. Dalton WS, Crowley JJ, Salmon SS, Grogan TM, Laufman LR, Weiss GR, Bonnet JD: A phase I11 randomized study of oral verapamil as a chemosensitizer to reverse drug resistance in patients with refractory myeloma: A Southwest Oncology Group study. Cancer 75:815, 1995
© Copyright 2024