Promising Approaches in Acute Leukemia

26
Investigational New Drugs 18: 57–82, 2000. © 2000 Kluwer Academic Publishers. Printed in the Netherlands. 57 Invited review Promising approaches in acute leukemia Jorge Cortes and Hagop M. Kantarjian Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA Key words: Acute leukemia, therapy, monoclonal antibodies, immunomodulation Summary In the last few decades, there has been a significant improvement in the prognosis of patients with acute leukemias. Still, the majority of patients succumb to these diseases. In recent years there has been a great surge in the understanding of the molecular mechanisms of disease which have provided us with new targets for anti-leukemia therapy. These range from chemotherapeutic agents with novel mechanisms of action, such as topoisomerase I inhibitors or demethylating agents, to reversal of drug-resistance mechanisms, to monoclonal antibodies directed against specific antigens, and targeted therapy that inhibit the function of molecules such as tyrosine kinases or Ras. The research on many of these agents is still in the early phases, but these new approaches offer the promise of finding a cure for the majority of patients with leukemia in the near future. Here we describe some of the promising approaches that are currently being investigated in the treatment of acute leukemias. Introduction The prognosis of adult patients with acute leukemia has improved significantly over the past 3 decades. Still, most patients succumb to complications of dis- ease progression and therapy With standard chemo- therapy, 60% to 80% of patients with acute myelo- genous leukemia (AML) and 75% to 85% of those with acute lymphocytic leukemia (ALL) achieve a complete remission (CR). Long-term disease-free sur- vival (DFS) rates are 20% to 30%, and 30% to 40%, respectively, with prognosis depending on patient- and leukemia-associated characteristics including age, performance status, cytogenetics, and immunopheno- type [1–5]. This emphasizes our need to discover new strategies, and perhaps revisit old ones, to improve the long-term prognosis. Current investigations aim at discovering ap- proaches which might improve the prognosis of the 60% to 75% of patients in whom the disease ultimately progresses with established modalities. A variety of such avenues exist and are probably better investigated in pilot and later confirmatory trials. These include: 1) new agents with different mechanism of action from known active drugs; 2) exploring older drugs with new schedules or improved formulations which offer a pharmacologic advantage; 3) host immun- omodulation to enhance specific anti-leukemia effect (e.g interferons alpha or gamma, interleukin-2, li- nomide); 3) targeting leukemia-specific molecular or cellular components; and 4) selective enhancement of leukemia cell differentiation (Table 1). In this review, promising approaches for adult acute leukemia will be discussed with focus on 1) new or “rediscovered” chemotherapeutic agents, 2) mono- clonal antibodies, and 3) immune modulation. New and revisited chemotherapeutic agents There has been a recent surge in the interest in discov- ering new chemotherapeutic agents for the treatment of acute leukemia. This followed several success- ful discoveries of agents with significant activity in both solid tumors and hematologic malignancies (e.g taxanes, topo I inhibitors, navelbine, gemcitabine).

Transcript of Promising Approaches in Acute Leukemia

Investigational New Drugs18: 57–82, 2000.© 2000Kluwer Academic Publishers. Printed in the Netherlands.

57

Invited review

Promising approaches in acute leukemia

Jorge Cortes and Hagop M. KantarjianDepartment of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA

Key words:Acute leukemia, therapy, monoclonal antibodies, immunomodulation

Summary

In the last few decades, there has been a significant improvement in the prognosis of patients with acute leukemias.Still, the majority of patients succumb to these diseases. In recent years there has been a great surge in theunderstanding of the molecular mechanisms of disease which have provided us with new targets for anti-leukemiatherapy. These range from chemotherapeutic agents with novel mechanisms of action, such as topoisomerase Iinhibitors or demethylating agents, to reversal of drug-resistance mechanisms, to monoclonal antibodies directedagainst specific antigens, and targeted therapy that inhibit the function of molecules such as tyrosine kinases or Ras.The research on many of these agents is still in the early phases, but these new approaches offer the promise offinding a cure for the majority of patients with leukemia in the near future. Here we describe some of the promisingapproaches that are currently being investigated in the treatment of acute leukemias.

Introduction

The prognosis of adult patients with acute leukemiahas improved significantly over the past 3 decades.Still, most patients succumb to complications of dis-ease progression and therapy With standard chemo-therapy, 60% to 80% of patients with acute myelo-genous leukemia (AML) and 75% to 85% of thosewith acute lymphocytic leukemia (ALL) achieve acomplete remission (CR). Long-term disease-free sur-vival (DFS) rates are 20% to 30%, and 30% to 40%,respectively, with prognosis depending on patient-and leukemia-associated characteristics including age,performance status, cytogenetics, and immunopheno-type [1–5]. This emphasizes our need to discover newstrategies, and perhaps revisit old ones, to improve thelong-term prognosis.

Current investigations aim at discovering ap-proaches which might improve the prognosis of the60% to 75% of patients in whom the disease ultimatelyprogresses with established modalities. A variety ofsuch avenues exist and are probably better investigatedin pilot and later confirmatory trials. These include: 1)

new agents with different mechanism of actionfrom known active drugs; 2) exploring older drugswith new schedules or improved formulations whichoffer a pharmacologic advantage; 3) host immun-omodulation to enhance specific anti-leukemia effect(e.g interferons alpha or gamma, interleukin-2, li-nomide); 3) targeting leukemia-specific molecular orcellular components; and 4) selective enhancement ofleukemia cell differentiation (Table 1).

In this review, promising approaches for adultacute leukemia will be discussed with focus on 1) newor “rediscovered” chemotherapeutic agents, 2) mono-clonal antibodies, and 3) immune modulation.

New and revisited chemotherapeutic agents

There has been a recent surge in the interest in discov-ering new chemotherapeutic agents for the treatmentof acute leukemia. This followed several success-ful discoveries of agents with significant activity inboth solid tumors and hematologic malignancies (e.gtaxanes, topo I inhibitors, navelbine, gemcitabine).

58

Table 1. Promising approaches in acute leukemia therapy

1. Agents with novel antileukemic mechanisms, e.g., topoisomerase I inhibitors

2. New formulations of older drugs, e.g., liposomal agents, IV 6-MP

3. Dose intensive therapy, e.g. allogeneic and autologous stem cell transplantation

4. Leukemic cell sensitization e.g. G-CSF, GM-CSF, IL-3, other cytokines

5. Reversing leukemia drug-resistance e.g. MDR blocking agents

6. Immune modulation e.g. interferon alpha or gamma, interleukin 2, linomide

7. Cellular component therapy e.g. allogeneic or modified autologous lymphocyte re-infusion

8. Cellular targeted therapy e.g. CD33-monoclonal antibodies with or without immunotoxins

9. Selective enhancement of leukemia cell apoptosis e.g, anti-bcl-2, retinoids + cytokines

10. Leukemic cell differentiation e.g. retinoids

11. Gene-directed therapy, e.g. tyrosine kinase inhibitors, Ras inhibitors

I. Nucleoside analogues

The class of nucleoside analogues has been amongthe richest source of agents with active antileukemiceffects (Table 2).

A. Deoxyadenosine analogues

These include fludarabine and chlorodeoxyadenosineboth of which have demonstrated major efficacyin lymphoproliferative disorders including chroniclymphocytic leukemia (CLL), indolent lymphoma,hairy cell leukemia and Waldenström (macroglobu-linemic) lymphoma.

1. FludarabineThe phase I studies of fludarabine in acute leukemiashowed definite anti-AML activity, but at doses of 50to 100 mg/m2 daily for 5 days (i.e., total dose 250 to500 mg/m2 per course), a dose schedule associatedwith significant central and peripheral neurotoxicity(depressed consciousness, coma, blindness, paralysis)[6,7]. However, when used at doses shown to be effect-ive with acceptable toxicities in indolent lymphomaand CLL (25 to 30 mg/m2 daily for 5 days, total dose125 to 150 mg/m2 per course) fludarabine could in-crease phosphorylation of ara-C, increasing 3 to 5 foldthe intracellular levels of its active compound ara-CTPin leukemic cellsin vivo [8,9]. This led to a seriesof investigations of fludarabine and ara-C in differentschedules, either alone (FA), or combined with leuk-emic sensitizing agents such as G-CSF (FLAG), orother cytotoxic agents such as idarubicin (FLAG-IDA)in both AML and ALL [10–14]. In AML salvage, thefludarabine-ara-C combination was associated with atrend for higher CR rates compared with intermedi-

ate or high-doses of ara-C alone, particularly in themore sensitive leukemias, i.e. those associated with along first CR duration [10]. Fludarabine is currentlyused in many frontline and salvage trials in combina-tion with anthracyclines and ara-C [10–14]. However,comparative trials of standard induction-consolidationtherapy in AML with or without fludarabine in patientsubsets (e.g., good versus poor risk AML) are neededbefore fludarabine is established as beneficial in AMLtherapy.

2. 2-chlorodeoxyadenosine (2-CDA)2-CDA was reported to have significant activity inchildhood AML, with 50% of children treated in sal-vage achieving CR [15]. 2-CDA was then used innewly-diagnosed childhood AML before initiation ofstandard therapy (“therapeutic window”), producingCR rates of 27% after 2-CDA alone, and 72% aftermulti-agent chemotherapy [16]. In adult AML, twophase I studies using 2-CDA at doses up to 5 to 21mg/m2 daily for 5 to 7 days (cumulative doses of25 to 105 mg/m2 per course) produced three object-ive responses among 57 patients treated with onlyoccasional toxicity (neurologic, renal, prolonged my-elosuppression) [17,18]. The combination of 2-CDAand ara-C potentiates the antileukemic effect as a res-ult of the incorporation of 2-CDAMP and ara-CTP,inhibition of ribonucleotide reductase, and increasedaccumulation of ara-CTP, resulting in significant in-hibition of DNA synthesis [19]. Salvage therapy withconcomitant administration of 2-CDA and ara-C inAML resulted in 2 CR among 15 patients treated(13%) [18].

New deoxyadenosine analogues (e.g. chlorofluoroara-A) may also be effective and should be pursued[20].

59

Table 2. Nucleoside analogues of interest in acute leukemia

Deoxyadenosine analogues Deoxythimidine analogues

Fludarabine 5 fluorouracil

2′-chlorodeoxyadenosine Antiviral agents (e.g. AZT, DDI)

Deoxycoformycin

Chloro-fluoro adenosine arabinoside

Deoxyguanosine analogues Deoxycytidine analogues

6-thioguanine Cytosine arabinoside (ara-C)

6-mercaptopurine Gemcitabine

Guanosine arabinoside (ara-G) Decitabine

Compound 506 (GW506U78, ara-G prodrug) Azacytidine

Tiazofurin Troxacitabine (BCH-4556)

Fazarabine

B. Deoxyguanosine analogues

1. Guanosine arabinoside (ara-G) and compoundGW506U78Preclinical studies indicated the selective antileukemiceffect of ara-G in T cell leukemia [21]. However,ara-G is insoluble preventing its clinical development.Compound GW506U78 is a methoxy derivative ofara-G which upon administration is rapidly demeth-oxylated to ara-G, the active compound [22]. Ini-tial phase I clinical studies of compound 506U78 inchildhood and adult leukemia documented signific-ant responses in T-cell acute lymphoblastic leukemia,T-lymphoid blast phase chronic myeloid leukemia(CML), T-cell lymphomas and B-chronic lymphocyticleukemias (B-CLL) [22–24]. Among 13 evaluable pa-tients with these diseases, 5 (38%) achieved a CR and5 (38%) a partial remission (PR). Patients with B-ALL, B-lymphoma, AML or T-CLL did not respond[22,23]. Response correlated strongly with intracel-lular accumulation of ara-GTP [22]. The maximallytolerated dose was reported to be 40 to 60 mg/kg/dayfor 5 days and was age-dependent, with peripheralneuropathy (numbness, paresthesias, paresis) beingthe dose limiting toxicity [24]. Ongoing studies inboth childhood and adult hematologic malignanciesare investigating different schedules (i.e., day 1, 3,and 5 schedule) and using biochemical modulationwith fludarabine which can significantly increase ara-G phosphorylation and accumulation of ara-GTP [25].These schedules have produced remissions in patientswith refractory CLL [25].

Tiazofurin has also demonstrated activity in acuteleukemia and CML in blastic phase. The dose sched-

ule targeted the biologic endpoint of inhibiting inosine5′-phosphate dehydrogenase which, when combinedwith inhibition of the guanine-salvaging activity ofhypoxantine-guanine phosphoribosyltransferase, res-ults in suppression of guanosine triphosphate (GTP)pools [26]. However, this dose schedule was associ-ated with significant toxicity including neurotoxicity,pericarditis, renal dysfunction and others. Improvingthe therapeutic toxic ratio of Tiazofurin through dose-schedule modulations may render it a more attractiveinvestigational agent.

2. Intravenous 6-mercaptopurine (6-MP) and6-thioguanine (6-TG)

Both 6-TG and 6-MP have shown anti-leukemic ef-ficacy and have been used as part of induction (e.g.,6-thioguanine, daunorubicin, ara-C [TAD]) and main-tenance therapy in AML and ALL. Both drugs havebeen used in oral formulations because of ease of ad-ministration. However, oral 6-MP has erratic absorp-tion with bioavailability of 20% to 60%, with first-passrelated hepatic toxicity [27]. In childhood ALL, serumconcentrations of 6-MP are variable, and a directrelationship exists between 6-MP concentration-timecurve and relapse-free survival [28]. The intracellularlevels of the active metabolite of 6-MP, thioguan-ine nucleotides (6-TGN), are also variable, with lowlevels associated with a poor prognosis [29,30]. Anintravenous 6-MP formulation has been available forover 40 years, but has only recently been incorporatedmore extensively into the therapy of acute leukemias[27]. It offers 100% bioavailability, higher drug deliv-ery without dose-limiting hepatotoxicity, avoidance of

60

patient non-compliance with oral therapy, and cyto-toxic levels can be achieved in the CSF [27]. Asimilar dose-response relationship exists for metho-trexate (MTX) [31], and regimens using intravenous6-MP and MTX have reported improved results inchildhood [32–34] and adult ALL [35], but studiesdirectly comparing oral versus intravenous 6-MP areneeded. 6-MP has also demonstrated synergistic activ-ity with ara-C, increasing the intracellular levels ofara-CTP [36]. The combination of high-dose intraven-ous 6-MP and high-dose ara-C resulted in CR in 6 of13 children (46%) with refractory or relapsed AMLand in 1 of 10 (10%) with ALL [37]. This combin-ation was also used in post-induction chemotherapyfor children with newly diagnosed AML, with 7 of the14 children treated remaining in complete remission[38]. Late intensification with a combination of mer-captopurine, vincristine, methotrexate, and prednisone(POMP) prolonged survival in adult AML [39]. Theseresults indicate the need to revisit the role of intraven-ous 6-MP combinations in both childhood and adultAML and ALL.

6-TG has been used mostly in AML in the TADregimen and in maintenance (6-TG + ara-C). 6-TGis better tolerated than 6-MP, induces 4-fold higherintracellular levels of 6-TGN [40], and is probably amore potent cytotoxic drug than 6-MP [41]. A phaseI study in children showed that doses of 20 mg/m2/hadministered intravenously in a continuous infusionover 36 hours result in plasma concentrations wellabove cytotoxic concentrations with myelosupressionas the dose-limiting toxicity [42]. Further investigationof intravenous 6-TG in acute leukemias is warranted.

C. Deoxycytidine analogues

Based on the major activity of ara-C in acute leuk-emias and other hematologic disorders, other deoxy-citidine analogues have been explored. Interestingly, arelated structural analogue, gemcitabine, with signific-ant activity in solid tumors has shown little activity inleukemia. Despite the structural similarities of ara-Cand fazarabine, azacytidine and decitabine, fazara-bine was inactive while the latter two, through novelmechanisms of action, might prove complementary orsynergistic with ara-C in leukemia.

1. Azacytidine, decitabineHypermethylation is a general property of tumor pro-gression and resistance: it has been reported in 10to 50% of patients with newly diagnosed hematolo-

gic malignancies, and in 100% of recurrent ALL andCML in transformation [43]. Both azacytidine anddeoxyazacytidine are potent hypomethylating agents.Azacytidine has established efficacy in AML and isoffered on a compassionate basis for patients with re-fractory AML [44–46]. In myelodysplastic syndrome(MDS), azacytidine in a low-dose schedule has pro-duced CR in 10% to 15% of patients and objectiveresponses in 35% to 50% [47]. It is currently under-going phase III comparative trials with the standard ofcare in MDS.

Decitabine has been investigated mostly in Europe,with encouraging activity observed in AML, MDS andchronic myelomonocytic leukemia (CMML) (Table3). In AML salvage, decitabine has induced CR in5% to 30% of patients as a single agent, and in 25%to 50% in combination with anthracyclines [48–52].Schwartzman et al. reported 6 CR in 8 patients (75%)with newly-diagnosed AML treated with daunorubicin50 mg/m2 daily for 3 days and decitabine 90 mg/m2

daily for 5 days [52]. Lower dose schedules of de-citabine (15 mg/m2 over 4 hours every 8 hours×3 days; 135 mg/m2 per courses) have also producedobjective responses in 40% to 50% of patients withCMML and MDS. Wijermans et al. [53] treated 29elderly patients with high-risk MDS with decitabine40 to 50 mg/m2 daily for 3 days by continuous infu-sion. Eight achieved a CR (28%), 5 had a PR (17%),and 2 improved, for an overall response rate of 54%.Five patients (17%) died during induction. The me-dian remission duration was 8 months and the mediansurvival 11 months. Similar results were reported byPinto et al. [51] treated 46 patients (36 evaluable, me-dian age 69 years) with decitabine 405 mg/m2 percourse. In MDS, 8 of 35 patients achieved a CR and5 had PR. In CMML 3 of 11 patients achieved CRand 5 had PR. The CR duration was 10 months. Re-sponses required a median of 3 to 4 courses of therapyto develop fully.

Decitabine is associated with an unusual pattern ofdelayed and prolonged myelosuppression, indicatingits potential value as part of dose-intensive regimensfollowed by stem cell support to improve antitumorefficacy, or as salvage therapy with stem cell support[54]. In CML transformation, decitabine has producedobjective responses in 25% of patients in blastic phase,and in 40% of patients with accelerated phase [55].Hypomethylating agents should be investigated fur-ther in hematologic malignancies and perhaps lymph-oproliferative disorders, and as part of regimens usingstem cell support. Correlating the degree of tumor-

61

Table 3. Decitabine in hematologic malignancies

Disease (reference) No. CR (%) Overall

response∗ (%)

AML salvage

Momparler [48] 6 2 (33)

Debusscher [49] 20 1 (5)

Willemze [50]

DAC+AMSA 30 8 (27)

DAC+Ida 33 15 (45)

Overall 63 23 (37)

Kantarjian [55] 8 1 (12)

Previously untreated AML

Schwartsmann [52] 8 6 (75)

MDS and CMML

Wijermans [53] 29 8 (28) 15 (54)

Pinto [51]

MDS 35 8 (23) 13 (37)

CMML 11 3 (27) 8 (73)

CML

Kantarjian [55]

Accelerated 17 5 (29) 8 (47)

Blastic 20 2 (10) 5 (25)

∗Overall response = Complete or partial remission, or hematologicimprovement (MDS, CMML), or complete or partial remission, orback top chronic phase (transformed CML).

specific DNA methylation patterns with response willhelp selecting patients who may benefit from decit-abine therapy. In AML, the value of decitabine couldbe evaluated in all patients as part of combinationchemotherapy or in high-risk patients who have com-pleted induction-consolidation, and who could then berandomized to no further therapy or to maintenancetherapy with decitabine. Based on thein vitro syn-ergism between decitabine and topo I reactive agents[56], such combinations should also be investigated inleukemia.

2. Other deoxycitidine analogues

Fazarabine combines properties of both ara-C and aza-cytidine. Despite encouraging preclinical data, thephase I-II study of fazarabine in acute leukemiashowed little activity. It also required about 15 dose es-calations from the phase II solid tumor dose limited bymyelosuppression (from 2 mg/m2 by continuous infu-sion daily× 5 up to 450 mg/m2 daily× 5). Among 72

patients entered on study, only one objective responsewas noted [57].

Gemcitabine (2′,2′-difluorodeoxycytidine; dFdC)is active in several solid tumors, andin vitro stud-ies showing activity against leukemia cell lines [58–60] stimulated interest in gemcitabine in acute leuk-emias. Pilot studies suggested that a dose rate of 10mg/m2/minute produce plasma concentrations greaterthan 20µmol/L and maximized the rate of dFdCTPaccumulation [61]. Hence phase I studies used a fixedincremental design of dose-per-unit time, where doseintensity was increased by prolonging the duration ofthe infusion at a fixed dose rate. A phase I study ofgemcitabine given weekly defined the MTD at 4800mg/m2 over 480 minutes, and was dose limited bymucositis and skin rashes [62]. No complete responseswere seen in this heavily pretreated group of patients[62]. A second phase I study of gemcitabine givenevery 3–4 weeks is ongoing with the current doseschedule at 12,500 mg/m2 over 1250 minutes (Es-tey E. – unpublished data). Another potential rolefor gemcitabine in acute leukemias is to increase theactivity of deoxycytidine kinase, which is the rate-limiting enzyme in the phosphorylation of ara-C intoits active metabolite [63]. This results in a 3-fold in-crease in intracellular accumulation of ara-CTP, andincreased cytotoxicity in leukemic cell lines [63,64].Gemcitabine could thus be used for modulation ofara-C activity in acute leukemia in a similar way asfludarabine. The higher intrinsic cytotoxic effect ofgemcitabine compared with fludarabine [64] makesthis modulation an interesting approach.

II. Topoisomerase I inhibitors

Uncoiling of DNA is through an excisional processcarried by two essential cellular enzymes, topoi-somerase II (topo II) for double-stranded DNA, andtopo I for single stranded DNA. Topo II inhibitors haveestablished anti-tumor activity and are part of currenttreatment schedules in several solid and hematologicmalignancies. Camptothecin (CPT) was investigatedin the 70’s and abandoned as being too toxic. The re-cent discovery of the topo I inhibitory effect of CPTrenewed interest in the drug and its analogues. TopoI levels were found to be significantly higher in tu-mor cells compared to their normal counterparts [65].Hence, inhibition of topo I enzyme may provide aselective tumor versus normal cell kill [65-67].

62

1. Topotecan

Topotecan is a semisynthetic analogue of the alkal-oid camptothecin which acts as a specific inhibitor oftopoisomerase I by stabilization of the topoisomeraseI-DNA complex leading to cell death. Two phase Istudies of topotecan as a continuous infusion scheduleover 5 days in acute leukemia defined the MTD at 2mg/m2 daily for 5 days, with mucositis as the dose-limiting toxicity. Objective response were observedin these trials with reduction in the number of circu-lating blasts in all patients included in one trial, andCR in 11% in the other [68,69]. A phase II studyof topotecan as a single agent in our institution didnot result in objective CRs among 14 patients withrefractory or relapsed AML (unpublished data). Thisfinding, together with the results obtained in MDS andCMML (discussed later), emphasize the risk of false-negative results even when using active agents, aspatients are treated with more effective frontline ther-apy and relapsing with more refractory disease. Usingan upfront “window” therapy approach has been sug-gested to explore activity of potentially effective drugsin a better-risk population but this is still controversial.

Topotecan was first investigated in MDS andCMML based on: 1) similarities between MDS andAML in terms of disease biology and treatment re-sponse; 2) less exposure of MDS than AML tochemotherapy thus providing a “therapeutic window”of opportunity to investigate topotecan in relativelychemotherapy-naive patients; and 3) the poor resultswith standard therapy. Sixty patients with high-riskMDS (refractory anemia with excess blasts [RAEB],or in transformation [RAEBT]) or CMML received to-potecan 2 mg/m2 as a continuous infusion for 5 days,every 3–4 weeks. The CR rate was 30% and was sim-ilar for MDS and CMML [70]. Patients with mutationsin the RAS-gene had a lower response rate, as didthose with monocytosis or thrombocytopenia.

Because of the positive results, combination ther-apy of topotecan with other agents, including ara-C, etoposide, anthracyclines, and cyclophosphamidewere initiated in different dose schedules in MDS andAML [71–73]. The combination of topotecan 1.25mg/m2 continuous infusion daily for 5 days and ara-C 1 g/m2 daily for 5 days was used to treat 86 patientswith MDS (n=59) or CMML (n=27), of which 57were previously untreated. CR was achieved in 48 pa-tients (56%), and it was higher in MDS (61%) thanCMML (44%) [73]. Preclinical studies indicated thattopoisomerase I inhibitory agents may potentiate DNA

damage induced by exposure of tumor cells to radi-ation or alkylating agents (e.g. cyclophosphamide) in aschedule-dependent way by preventing repair of DNAdamage [74,75]. Designed on the basis of this syn-ergism, the combination of cyclophosphamide, ara-Cand topotecan has been investigated in patients withrefractory or relapsed AML with a reported responserate of 23% [76], and is currently being evaluated infront-line therapy.

Topotecan is also available in an oral formulationwhich may offer practical, and perhaps more effect-ive longer-exposure schedules (21-day oral topotecanschedules) in indolent hematologic disorders (MDS,CMML, lymphoma, myeloma) [77].

2. Other topoisomerase I inhibitors

Camptothecin-11 (CPT-11) showed activity in the ini-tial phase I studies in acute leukemia [78] whichhas been confirmed in early phase II trials [79]. 9-aminocamptothecin (9-AC) has promising preclinicalactivity, and phase I studies of 3- to 21-day intra-venous and oral schedules are under investigation.9-nitrocamptothecin (9-NC) also offers the advantageof oral route delivery, and has produced objective re-sponses in solid tumors, and in MDS, CMML andPh-negative CML [80]. Correlative studies of topo I-DNA complex formation and response may help selectpatient who benefit specifically from therapy with topoI inhibitors.

III. Homoharringtonine (HHT)

HHT is a plant alkaloid derived from the bark ofCe-falotaxus fortuneii. Its antineoplastic mechanism ofaction is not clear, but it inhibits protein synthesis [81]resulting in cytotoxicity. HHT also induces differenti-ation of leukemic cell-line HL-60 into cells resemblingmonocytesin vitro [82], and of fresh leukemia cellswhen combined with cytarabine [83]. HHT has beenused in China for the treatment of acute and chronicleukemias, alone [84,85] and in combination withother agents [86,87]. Phase I studies in the UnitedStates with HHT bolus schedules, showed hypoten-sion, arrhythmias and myelosuppression to be dose-limiting [88,84], but hypotension and arrhythmiaswere reduced significantly when HHT was given ina continuous infusion schedule [89–91]. Several in-vestigators have used HHT alone or in combinationsfor the treatment of patients with acute leukemias,

63

mostly myeloid (Table 4). These trials used variabledose schedules and resulted in response rates of 0% to25% in patients with refractory or relapsed AML [92–96]. Warrel et al. reported 7 CRs among 28 patients(25%) treated with a total dose of 45 mg/m2 or higher,given as a seven-day continuous infusion of 7 mg/m2

daily or a nine-day continuous infusion of 5 mg/m2

daily [92]. The higher dose was associated with hy-potension in 63% of patients. Feldman et al. used asimilar schedule and reported CR in 16% of patients,with hypotension in 44% of patients [93]. Hypotensionwas also a significant problem in 40% of patients in anECOG study using a total dose of 52.5 mg/m2 over8 days, and no CR were achieved [94]. In contrast, aschedule of 2.5–3.0 mg/m2 daily continuous infusionover 15 to 21 days (total dose 37.5 to 52.5 mg/m2)resulted in a response rate of only 3%, but cardiovas-cular complications were minimal [95]. Feldman etal. used HHT 5 mg/m2 daily continuous infusion for9 days in patients with newly diagnosed myelodys-plastic syndrome (MDS, n=16) or AML evolving afteran MDS (n=12). Seven patients (25%) achieved a CR,and hypotension was reported only in 3 patients [96].HHT has also been used in combination with ara-Cin 22 patients with relapsed or refractory AML whoreceived ara-C 100 mg/m2 continuous infusion dailyfor 7 days and HHT 1.5 to 5 mg/m2 CI daily for 7days [97]. Five achieved CR (23%), and four of theresponders received doses of 4 mg/m2 which was thedose recommended for further studies. Interestingly,4 of the 5 responders in this study had acute promy-elocytic leukemia (APL). Responses in patients withAPL have been reported in other small series [98–100] ranging from 46% to 100%. In one study of 10patients with APL (nine were previously untreated),CR was achieved in 7 (70%), and treatment producedbone marrow aplasia in only one, while 3 had mildhypoplasia, and six maintained a hypercellular bonemarrow [100]. Feldman et al. reported a 60% remis-sion rate in patients with relapsed APL [98]. Theseresults, together with the differentiating properties ofHHT [82,83], and the sensitivity of the HL-60 cellline to HHT compared to other leukemia or lymph-oma derived cell-lines, suggest that HHT could be aninteresting agent in patients with relapsed APL [112].Only a few patients with ALL have been treated withHHT, and no responses were observed [92,93].

Therefore, there is clear evidence that HHT hasantileukemic effect, and trials of combinations includ-ing HHT are warranted. Particularly intriguing is itspotential role in patients with APL.

IV. Alkylating agents

Alkylating agents have been used for a long time inthe treatment of hematologic malignancies and solidtumors. The availability of new alkylating agents, andnew formulations and schedules for old alkylators,have renewed the interest in this group of drugs.

1. Tallimustine

Tallimustine, a distamycin A derivative, is a novel al-kylating benzoyl mustard which alkylates N3 adeninein the minor groove of DNA with very stringent se-quence specificity [101]. This mechanism of actionis different from other alkylating agents and thereforeresults in different cell cycle perturbation compared tothe more conventional alkylators [102], and differentmechanism of resistance [103]. Although significantactivity was found in preclinical studies against avariety of malignancies, initial trials in solid tumorshave yielded modest results [104,105]. In phase I andII trials the dose limiting toxicity was neutropenia,which prompted the interest of tallimustine in leuk-emia [104–105]. In a human leukemia model usingSCID mice, tallimustine at 0.86 to 3.0 mg/kg dailyfor 3 days resulted in the cure of most mice [107].Based on this model, a phase I trial was conductedin patients with refractory or relapsed acute leukemia.The MTD was 900 mcg/m2 daily for 3 days, every 3to 4 weeks, which is 3 times higher than the MTDin solid tumors, with the dose limiting toxicity be-ing mucositis. Magnesium and potasium wasting werealso observed. Two of nineteen patients (11%) withAML or AUL achieved CR, and two others had a mar-row CR but with persistent thrombocytopenia [107].Based on the SCID mouse model demonstrating thesafety and efficacy of combining tallimustine with ara-C, studies with combinations including tallimustineare needed, as well as additional trials of tallimustinein other hematologic malignancies (lymphomas, my-eloma, Waldenström’s macroglobulinemia).

2. Platinum analogues

Despite the low degree of activity of cisplatin inhematologic malignancies, a synergistic effect of cis-platin and ara-C was demonstrated bothin vitro andin clinical trials in lymphoma. Combination studiesof cisplatin plus amsacrine, or cisplatin with VP16,ara-C, and steroids (ESHAP) have resulted in highCR rates (Kantarjian H. – unpublished data). Phar-macologic modulation of cisplatin with fludarabine

64

Table 4. Results with homoharringtonine (HHT) in AML

Author (reference) No. CR (%) Comments

Warrell [92] 31 7 (23) – 7/28 CR when cumulative dose≥ 45 mg/m2

– Hypotension in 63% using 7 mg/m2 ×7 days

Feldman [93] 57 7 (12) – All CR among patients with relapse AML; no CR among 14 patients withprimary refractory AML

Stewart [94] 20 0 (0)

Kantarjian [95] 31 1 (3) – Low-dose, continuous infusion for 15 to 21 days

Feldman [96] 22 5 (23) – HHT + ara-C

– No CR among 8 patients with primary refractory disease

–4/5 CR among patients with APL

results in increased cytotoxicity and inhibition of re-pair of cisplatin-induced cross links [108]. Other plat-inum analogues have also been investigated. The mostpromising results were achieved with carboplatin,which was associated with CR rates of 5% to 20%in patients with refractory or relapsed AML or ALL[109,110], and of 40% to 60% in newly-diagnosedhigh-risk AML. Salvage therapy with carboplatin-based combination regimens have resulted in CR ratesof 8% to 50% [110–116].

CI-973 is a new platinum analogue with signific-ant anti-leukemic effect against a variety of leukemiacell lines at concentrations 10-fold lower than the peakplasma levels achieved in phase I trials for solid tu-mors [117]. In a phase I study of CI-973 in acute leuk-emia, no responses were achieved among 22 patientswith refractory or relapsed acute leukemia (18 AML,4 ALL) [118]. There might be a role for platinumanalogues, particularly carboplatin, in combination forsalvage therapy, or as frontline/consolidation therapyin AML, but further studies are required.

3. Other alkylating agents

Alkylating agents have been used for the treatment ofacute leukemias mostly in the context of bone marrowtransplantation, but may have a role in the setting ofnon-myeloablative therapy. The self-renewal capacityof AML progenitors, rather than terminal divisionsmight be differentially affected by alkylating agents[119,120]. Some alkylating agents may be more ef-fective inhibitors of self-renewal capacity than ara-C[120]. Therefore, drugs such as melphalan, busulfanand cyclophosphamide should be re-evaluated, par-ticularly considering that intravenous formulations ofmelphalan and busulfan could make drug deliverymore reliable, accurate, and adjustable to effective

blood levels. Alkylating agents may be used in syner-gistic combinations. Topo I inhibitors and nucleosideanalogues inhibit DNA repair thus enhancing DNAdamage by alkylating agents [74,75]. These combin-ations are under investigation.

V. Liposomal agents

Liposomal encapsulation of chemotherapeutic agentsand other drugs changes the pharmacologic propertiesof the active compound and has been advocated to re-duce toxicity, therefore potentially allowing for high-dose therapy and potentially improve the therapeuticbenefit of these drugs [121]. Dose-intensive anthra-cycline therapy is associated with improved prognosisin acute promyelocytic leukemia, AML, and ALL butdose escalation is limited by mucositis and, in thelong term, dose-dependent cardiac toxicity. Liposomaldaunorubicin (DNX) has been used in phase I trialsin solid tumors with no extramedullary toxicity, andlimited only by myelosuppression at doses of between100 mg/m2 and 120 mg/m2 repeated every three weeks[122] DNX has demonstrated significant activity inAIDS-associated Kaposi’s sarcoma, and no cardiactoxicity has been reported among patients receivingcumulative daunorubicin doses of>600 mg/m2 (range600 to 3159 mg/m2) [123]. In a phase I study inpatients with refractory or relapsed acute leukemia,mucositis was the dose-limiting toxicity at a doseof 150 mg/m2 [124]. No significant cardiac toxicitywas reported and 2 patients (1 with CML in blasticphase and 1 with acute promyelocytic leukemia insecond relapse) achieved a complete remission [124].Preliminary results of a study using DNX in combina-tion with high-dose ara-C (1 g/m2 continuous infusiondaily for 4 days) reported a CR rate of 21% with an

65

additional 11% with hematologic improvement (i.e.,met all criteria for CR except platelet count improvedbut<100× 109/L) [125]. The MTD for DNX was 125mg/m2. Preliminary results of a combination chemo-therapy regimen for salvage ALL have produced aCR rate of 33% [126]. Dose escalation of liposomaldoxorubicin has been limited by hand-foot syndrome[127], although this could be possibly ameliorated bypyridoxine [128]. Whether these anthracyclines willbe safer in the long term and therefore valuable inhigh-risk populations (e.g., elderly) and the role ofhigh-dose anthracyclines using these formulations iscurrently being investigated.

A liposomal formulation of vincristine has demon-strated increased efficacy against leukemia cell lines[129,130]. The MTD in a phase I study in solid tu-mors was defined as 2.4 mg/m2 [131] and studiesin hematologic malignancies are currently underway.Liposomal formulations of topoisomerase I inhibit-ors, including topotecan [132] and its analogue GL147211C [133] are available with increased activity inpreclinical models.

Monoclonal antibodies

Monoclonal antibodies (MoAb) may target cytotoxictherapy (e.g., radioisotopes, toxins) specifically to ma-lignant cells. MoAb may also induce on their ownimmune-mediated killing of leukemic cells. Severalconditions need to be met for MoAb to become ef-fective therapy, including: 1) identification of tumor-specific antigens, 2) development of humanized MoAbto avoid antibodies against murine MoAb (HAMA),and 3) use of toxins with tight binding to MoAb andproper targeting to tumor cells to avoid normal tis-sue toxicities, or else, antibodies that are capable ofinducing immune destruction of tumor cells withoutthe need for toxin complexes. Some MoAb have beendeveloped and used with promising results in acuteleukemia.

I. Anti-CD33

CD33 is a surface glycoprotein expressed in commit-ted normal myelomonocytic and erythroid progenitorcells but not on stem cells [134]. It is also found onmost myeloid leukemic blasts and leukemic progen-itors [135]. Monoclonal anti-CD33 antibodies wereinitially IgG2 derived from mice, and were able to

bind avidly their target and kill cells in the presenceof rabbit but not human complement [136]. In phaseI trials, there was saturation of binding sites at doses≥5 mg/m2, with uptake in the bone marrow within 1hour after administration [137]. Murine anti-CD33 an-tibodies have no intrinsic cytotoxic activity, but whenlabeled with131I-significant selective irradiation wasdelivered to the bone marrow [137]. Using this131I-labeled anti-CD33, Schwartz et al. [138] treated 24patients with myeloid malignancies, including 16 withrefractory or relapsed AML, 5 with “blastic” MDS,2 with secondary leukemias, and 1 with CML inblastic phase. Ninety six percent of patients developedpancytopenia, and 89% had significant decreases inthe percentage of marrow blasts. Eight patients hadsufficient cytoreduction to proceed to bone marrowtransplantation (BMT). However, 37% of patients de-veloped human anti-mouse antibodies which resultedin loss of activity of the antibody when patients werere-treated [138].

Recently, a humanized anti-CD33 antibody hasbeen constructed, which retained its specificity, hada higher binding avidity than the murine antibody and,unlike the murine counterpart, mediated antibody-dependent cell-mediated cytotoxicity [139]. In a phaseI study, 13 patients with refractory or relapsed AMLwere treated at dose levels from 0.5 to 10 mg/m2/dosefor 6 doses over 18 days [140]. Total doses as high as216 mg could be safely administered, with the optimalbiodistribution and saturation of binding sites occur-ring at 3 mg/m2. The antibody was rapidly internal-ized, and no patient developed antihuman antibodies[140]. In a recent trial, 7 patients with acute promy-elocytic leukemia received ATRA followed by131I-labeled anti-CD33. Two of six patients with detectablePML/RARα mRNA after treatment with ATRA hadnegative determinations after receiving the antibody[141]. The treatment was well tolerated.

These antibodies have also been used in the settingof BMT. When used in conjunction with conventionalconditioning for allogeneic bone marrow transplant-ation, 131I-labeled anti-CD33 antibodies was admin-istered safely, delivered significantly higher doses ofradiotherapy to the bone marrow and spleen, but thehalf-life of the radioactive component in the bone mar-row was relatively short (9 to 41 hours) which maylimit its use in this setting [142]. However, these anti-bodies may be of value for bone marrowin vitro pur-ging. The sequential treatment of bone marrow withVP16 plus ara-C followed by complement-mediatedlysis using an anti-CD33 antibody synergistically

66

killed CFU-L, sparing early normal hematopoieticprogenitor cells [143].

MoAb have recently been conjugated with othermolecules in an attempt to increase specific cytotox-icity. An anti-CD33 MoAb conjugated to the potentcytotoxic agent colicheamicin was used to treat 23patients with AML in first relapse; three patientsachieved a CR and 7 others met all criteria for CRexcept for platelets< 100×109/L (but> 50×109/L)[143a]. Other combinations include anti-CD33 anti-body conjugated to gelonin, a toxin which inhibitsribosomal protein synthesis. This conjugate resultedin 4500 increased activity in inhibiting myeloid celllines while conserving the specificity of the antibody[144] and significantly suppressed leukemia growthin a mouse model [145]. Anti-CD33 antibodies havebeen conjugated to another toxin, ricin, to increase thespecific cytotoxicity [146]. Incubation of normal bonemarrow with this conjugate resulted in enrichment oflong-term culture initiating cells [146]. Bispecific an-tibodies to CD33 and CD 16 have been effective inaugmenting the killing of resistant leukemic cellsinvitro by peripheral blood lymphocytes and NK cells[147].

Further studies are underway using these and newantibodies in the treatment of patients with AML, butthe early results offer the potential of a useful approachfor the therapy of AML.

II. Anti-CD52 (CAMPATH-1)

CD52 is a 21 to 28 kDa phosphatidylinositolglycan(PIG)-anchoredglycoprotein expressed at high densityon normal lymphocytes, monocytes and macrophages[148]. Although its function is unknown, it may parti-cipate in the activation of T-lymphocytes [149]. CD52is expressed in the majority of lymphoid malignan-cies including non-Hodgkin’s lymphomas, CLL, hairycell leukemia, prolymphocytic leukemia, Sezary’ssyndrome, and ALL, although only a few myeloidmalignancies express CD52 [150]. CAMPATH-1 is amonoclonal antibody which binds to most circulatinglymphocytes and has been used successfully to purgebone marrow from T-lymphocytes prior to bone mar-row transplantation [151]. Several subtypes are avail-able with distinctive properties. The IgM rat antibodyfixes human complement and has been very effectivefor bone marrow purging [151,152]. The IgG vari-ety can be cytolytic with human complement, and theIgG2b isotype can mediate antibody-dependent cell-

mediated cytotoxicity [153]. A humanized version ofCAMPATH-1 (CAMPATH-1H) was engineered to re-duce the immunogenecity of the antibody [154]. Haleet al. first treated two patients with non-Hodgkin’slymphoma with CAMPATH-1H and noted clearing oftumor in the blood and bone marrow and resolutionof splenomegaly [154]. Interestingly, administrationof the IgM and IgG2a isotypes have resulted in tran-sient depletion of peripheral blood lymphocytes andconsumption of complement with little effect on tu-mor masses or bone marrow. In contrast, the IgG2bisotype has produced long-lasting lymphocyte deple-tion from peripheral blood and bone marrow, as wellas improvement in splenomegaly [155].

Information is starting to accumulate on the ef-fect of CAMPATH-1H in leukemias. A response rateof 89% was observed in a small series of patientswith CLL treated with CAMPATH-1H for front-linetherapy [156]. Among 29 patients with refractory orrelapsed CLL 11 (38%) achieved a PR and 1 (4%)a CR after treatment with CAMPATH-1H [157]. Al-though most ALL and a large proportion of AMLexpress CD52 [150] only a handful of patients withacute leukemia treated with this antibody have beenreported. Dyer et al. [155] included 5 patients withALL in their trial with CAMPATH-1H: 2 with B-cellALL, 2 with common ALL, and one with Philadelphiachromosome-positive ALL. The first four patients hadfailed conventional chemotherapy, whereas the latterreceived CAMPATH-1H as primary therapy. In allcases the peripheral blood cleared from leukemic cellsand the bone marrow showed a reduction in leukemiccells, complete in 3 patients and partial in 2 (includingthe patient treated with CAMPATH-1H as front-linetherapy, in whom the percentage of blasts in the bonemarrow decreased from 97% to 21%) [155]. There-fore, CAMPATH-1H deserves further investigation assalvage therapy in patients with CLL and ALL, or astherapy for minimal residual disease for patients infirst CR.

III. Anti CD19 monoclonal antibodies

The CD19 antigen is expressed in most normal andneoplastic B cells, and can be detected early in theirontogeny [158]. Although CD19 MoAb can recognizemalignant cells of many B-cell malignancies, it hasvery weak direct antitumor effect [159,160]. Whenconjugated to the plant toxin ricin, with potent ribo-some inactivating activity, CD19 has shown specific

67

cytotoxicity in vitro against B-cells [161]. Anti-CD19antibodies conjugated with blocked ricin (to preventnonspecific binding of ricin, anti-B4-bR) have shownantitumor activity in SCID mouse models of sev-eral CD19-positive human neoplasms, including ALL[162]. Phase I/II trials have been conducted in pa-tients with B-cell neoplasms, documenting the safetyof these antibodies, with the dose limiting toxicity be-ing elevation of liver enzymes [163,164]. Thirty five to70% of patients treated in phase I trials developed an-timouse antibodies and/or anti-ricin antibodies whichcould potentially limit the long-term treatment effic-acy [163,164]. The effect of anti-B4-bR on minimalresidual disease in patients with B-lineage ALL wasrecently reported [165]. Forty six adults with CD19-positive ALL who were in CR after one courseof consolidation were treated with anti-B4-bR at 30µg/kg/day as IV continuous infusion on days 1 to 7and 15 to 21. Of 5 patients with BCR-ABL positivedisease at diagnosis, 3 were positive prior to receivingthe immunotoxin and all remained positive, whereas1 of 4 who had become negative after chemotherapyconverted to positive. Of 7 patients with T-cell receptoror Ig heavy chain rearrangement at diagnosis 4 re-mained positive, 1 converted from positive to negative,and 2 converted from negative to positive after anti-B4-bR [165]. Although these results are not encour-aging, other antibodies and different strategies mayprove more useful in the setting of ALL. One possibil-ity is to combine the immunotoxin with chemotherapy.In vitro studies have shown synergistic activity of anti-B4-bR with chemotherapy in lymphoma cell lines,including some multidrug-resistant [166]. This effectmay be related to downregulation of P-glycoproteinexpression in resistant cells after exposure to theimmunotoxin [166]. This approach has resulted inimproved survival of mice treated with the combina-tion of anti-B4-bR and doxorubicin or etoposide [166]or with combination chemotherapy [167]. Anti-B4-bR has also been used effectively for purging bonemarrow grafts of leukemia or lymphoma cells [168].A reduction of more than 3 logs of malignant cellshas been reported with minimal effect against normalhematopoietic cells [168].

IV. Anti CD20 monoclonal antibodies

CD20, a 32 kD non-glycosylated phosphorylated pro-tein, is expressed on the surface of the majority ofnormal B cells but not early pre-B cells [169]. A vari-

ety of lymphoid malignancies express CD20, includ-ing non-Hodgkin’s lymphoma, ALL and CLL [170].Radiolabeled anti-CD20 antibodies were investigatedin the treatment for non-Hodgkin’s lymphomas withpromising results, but responses were observed withonly trace doses of the labeled antibody, suggest-ing activity of the unconjugated antibody [171,172].A humanized, unconjugated version of anti-CD20MoAb showed significant anti-B cell activityin vitroand in vivo in animal models through ill antibody-dependent cell-mediated cytotoxicity [173]. Severaltrials have been performed in B-cell lymphomas [174–176]. A 48% response rate was reported among 166patients with relapsed low-grade or follicular lymph-oma treated with anti-CD20 as a single agent [176].Toxicity was minimal and no immune response to theinfused antibody was reported [174–176]. In combin-ation with CHOP chemotherapy, 95% of 40 patientstreated responded (55% CR, 40% PR) [177]. Re-sponses have also been observed in CLL [178] and ag-gressive lymphomas [179]. The activity of anti-CD20MoAb therapy alone or in combination with chemo-therapy is currently under investigation in patientswith ALL who express CD20 on leukemic blasts.

Multidrug resistance modulation

The effectiveness of chemotherapeutic agents in acuteleukemia is limited by mechanisms of leukemic cellsresistance. Resistance can be induced by exposure ofleukemic cells to these agents or may be intrinsic to theneoplastic cells. Several mechanisms leading to multi-drug resistance (MDR) have been identified in acuteleukemia and their pharmacologic manipulation is anactive area of research [180]. Probably the best knownis the MDR1 gene, which translates into a membraneglycoprotein, P-glycoprotein (PgP) that can functionas a drug efflux pump [181]. The overexpression ofPgP has been linked to resistance to anthracyclines,vinca alkaloids, taxanes, epipodophyllotoxins, andsome alkylating agents. Overexpression of MDR1 andPgP have been reported with variable frequencies inAML, and has been associated with a poor response tochemotherapy and shortened survival [180,182,183].Measures of PgP function such as retention of rhodam-ine [184] and intracellular daunorubicin accumulation[185] also correlate with outcome in some reports.

Several agents have been used to inhibit PgP func-tion as a means to prevent development of resistanceor to restore leukemic cell sensitivity. These include

68

quinine, verapamil and dexverapamil, tamoxifen, cyc-losporin A and its non-immunosuppressive derivativePSC 833. These agents have shown efficacyin vitro,and clinical trials have produced some interestingresults.

The clinical applications of verapamil in MDRmodulation are limited by cardiovascular toxicitywhen used at doses that produce plasma levels withPgP inhibitory activity [186]. R-verapamil has demon-strated similar potency as verapamil in reversing MDRbut has a better toxicity profile [187,188] and is cur-rently being evaluated in acute leukemias [189].

Quinine therapy was well tolerated and plasmalevels within vitro inhibitory activity against PgP wereachieved in patients [190]. A large randomized trialincluding 315 patients with a variety of acute leuk-emias, most of them refractory or relapsed, found asimilar response rate for patients treated with (53%)and without (46%) quinine together with chemother-apy [191]. There was a trend for better CR rates amongMDR-positive patients who received quinine (60% vs35%; p=0.06) [191].

Cyclosporine-A (CsA) is a potent inhibitor of PgPat concentrations which can be achievedin vivo (500to 2,000 ng/ml). When used with chemotherapy forAML, CsA prolonged the clearance of anthracyclines[192]. Initial trials in AML have reported CR rates in82% of patients with relapsed AML and in 25% ofthose with refractory disease [193]. The response ratedid not correlate with MDR phenotype but correlatedwith the development of hyperbilirubinemia, whichoccurred in over 60% of patients [193]. In a recentmulticenter trial, patients with refractory or relapsedAML, secondary AML or RAEB-T were randomizedto receive daunorubicin and ara-C alone or in combin-ation with CsA. There was no difference in the CRrate between the two groups, but patients treated withCsA had a superior relapse-free survival and overallsurvival [194]. PSC 833 is a non immunosuppress-ive analog of CsA with a 10- to 30-fold more potentinhibiting activity against PgP [195,196]. PSC 833also caused hyperbilirrubinemia in most patients, andat doses of 12 mg/kg/d ataxia has been observed inphase I trials [197]. Studies in acute leukemia arecurrently underway [180], but preliminary results aredisappointing.

Some caution notes should be considered regard-ing approaches aimed at MDR modulation Thesedrugs may increase the toxicity of chemotherapy byaltering the clearance of the antineoplastic agents. Pre-clinical data with PSC 833 suggest that both acute

and delayed toxicity can be potentiated, and increasedconcentrations of doxorubicin can be found in theheart and other organs [198,199]. Increased acutetoxicity has been reported in some trials in acuteleukemia [191], and the effect on long-term toxicityremains to be determined (i.e., cardiotoxicity, second-ary malignancies). Another concern is the appearanceof alternative mechanisms of resistance. List et al.[193] reported 5 patients who achieved CR after ther-apy with cytarabine and daunorubicine plus CsA whoeventually relapsed. In 4 of these patients the diseasewas MDR-negative at relapse suggesting that a clonewith alternative mechanisms of resistance had beenselected [193]. Thus, even when MDR expression iseffectively inhibited in MDR-positive leukemia, othermechanisms of resistance may persist or evolve andmake cells unresponsive to therapyin vitro or in vivoin animals [200,201]. At least two other mechanismsof resistance mediated by non-PgP transport proteinshave been described. MDR-related protein (MRP) isexpressed in normal hematopoietic cells as well as inacute leukemias particularly resistant AML [202,203].The MRP gene is located in chromosome 16p13.1[204] and, interestingly, in patients with AML andinv(16)(p13q22), deletion of the MRP gene is associ-ated with prolonged failure-free survival [205]. Lungresistance protein (LRP) has been demonstrated in avariety of cell lines and has been detected in 37% ofpatients with AML, including patients with de novo(33%) and relapsed (38%) disease [206]. In contrastto MDR, LRP expression had an independent adverseprognostic effect for response [206]. The LRP is alsolocalized on the short arm of chromosome 16, closeto the MRP gene [207]. Unfortunately, these mechan-isms seem to be only weakly sensitive to known PgPinhibitors [208,209]. Additional mechanisms of drugresistance exist, for which no drug manipulation hasbeen designed and which are not related to transportproteins. Therefore, MDR manipulation as an area ofresearch remains in its early phase of development.

Immune modulation and other biologic therapies

Several biologic agents and manipulations are cur-rently being investigated in the management of acuteleukemias. In many cases, these biologic agents in-duce immune mediated killing of the leukemic cells,whereas in others the mechanism may be more com-plex.

69

I. Interferon alpha (IFN-A)

IFN-A has potent inhibitory activity against AMLcells and cell lines [210–212]. Inhibition may bethrough blocking IL-1 and IL-6 mediated growth[212]. IFN-A can also increase the lysing activity ofheterologous natural killer (NK) cells against AMLcells [213]. In somein vitro conditions, IFN-A hassynergistic cytotoxic activity against AML cell lines[214]. IFN-A has been used as maintenance therapyin adult AML after induction of CR with TAD: noimprovement in CR duration or survival was seencompared to no maintenance or maintenance withthioguanine and cytarabine [214]. Maintenance withIFN-A in patients with Ph-positive ALL in CR alsodid not result in improved CR duration (KantarjianH; unpublished data). In contrast, an induction re-gimen combining IFN-A and ara-C showed remark-able activity in patients with refractory or relapsedAML (27/35 CR, 77%) andde novoAML (15/15CR, 100%) [214]. Similar activity has been reportedin patients with refractory or relapsed non-Hodgkin’slymphoma [214,215]. A combination of IFN-A, ara-C, and thymopentin also had significant activity inpatients with myelodysplastic syndromes (overall re-sponse rate 75%) [216]. Further studies are neededto determine the role of IFN-A and IFN-A-basedcombinations in acute leukemias.

II. Interleukin 2 (IL-2)

Interleukin-2 is a pleomorphic cytokine capable ofstimulating the activity of cytotoxic T-cells and NKcells, and can induce the generation of lymphokine-activated killer (LAK) cells [217]. IL-2 with orwithout LAK cells can inhibit the growth of murineleukemia cellsin vivo, and NK-resistant myeloid orlymphoid leukemic cells can be lysedin vitro by LAKcells [218–221]. This effect seems to be mediatedby the Fas/APO-1 pathway [222]. IL-2 was extens-ively used after BMT, mostly autologous [223]. IL-2has also been used in treating acute leukemias aloneor in combination with chemotherapy or LAK cells(Table 5). Several trials have documented CRs in upto 57% of patients with AML and low percentage ofbone marrow blasts at relapse (e.g., less than 20% to25%); little effect was seen in patients with higherpercentages [224–226]. IL-2 may have prolonged CRduration when used as maintenance [227] or consolid-ation [228] therapy in patients in second CR. A pilotstudy using a 12 week continuous infusion of IL-2with weekly bolus in patients in first CR suggested

an improved CR duration compared to historical con-trols [229]. IL-2 has been used as maintenance therapyin combination with histamine with promising results[230]. IL-2 may also have a role in the management ofmyelodysplastic syndromes [231], and anecdotal re-sponses have been reported using low-dose IL-2 [232].There is little if any information on the use of IL-2in ALL, but ALL blasts may be less susceptible toLAK cell-mediated lysis than AML cells [233]. Trans-duction of IL-2 into acute leukemia cells reduces theoncogenic potential of these cells in animal modelsand may be a model for manipulation of this suscept-ibility [234]. The exact role of IL-2 in the managementof acute leukemia needs to be better defined, includingthe optimal dose, schedule and timing. However, IL-2 may be an valuable addition to the therapy of acuteleukemia [235].

III. Linomide

Linomide (roquinimex) is a quinoline derivative withtumor suppressing activity in some melanoma animalmodels [236]. Linomide has a variety of immun-omodulatory activities including stimulation of NKcell [237] and lymphokine-activated killer (LAK) cell[238] activity, enhancement of delayed-type hyper-sensitivity (DTH) reactions [239] and proliferativeT-cell responses [240], and increased IL-6 production[241]. Low concentrations of linomide are synergisticwith IL-2 in stimulating lymphocyte and NK-cell pro-liferation, but at higher concentrations the prolifera-tion decreases [242]. A pilot study in patients withsolid tumors used linomide at doses of 0.05 to 0.6mg/kg orally once a week. An increase number ofCD16+ NK cells and CD14+ monocytes was docu-mented, with increase NK activity at a dose of 0.2mg/kg, but not at 0.6 mg/kg [243]. In hematologicmalignancies, linomide has been considered for themanagement of AML and CML after autologous bonemarrow transplantation in an effort to produce a graft-versus-leukemia effect after this procedure [241,244].In several cases, an acute graft-versus-host reactionhas been induced after autologous bone marrow trans-plantation followed by linomide therapy in patientswith acute or chronic myeloid leukemias [244,245].Five patients with AML were treated with linomide0.3 mg/kg/week orally in cycles of three weeks fol-lowed by three weeks of rest for up to six months afteran autologous BMT [246]. During treatment periodsthere was an increase in the number and cytotoxicactivity of circulating NK cells, as well as increased

70

Table 5. Interleukin 2 in AML

Author No Disease status (No.) CR (%) Outcome Comments

(reference)

Foa [224] 12 Refractory (2) 4 (33) 1 CCR (30+ months) All patients who responded

Relapse (10) 3 relapsed (20 days, 4 mo, 9 had<20% BM blasts

mo)

Meloni [226] 14 Refractory (3) 8 (57) 5 CCR (14+ to 68+ months) All pts had<25% BM blasts

Relapse (11) 3 relapsed (2,5,9 months) No patient with refractory

AML responded

Lim [225] 13 Refractory (1) 1 (17) 1 relapsed (6 months)

Relapse (5)

1st CR (3) 1st CR in CCR (13+,13+,22+

2nd CR (4) months)

2nd CR: 2 relapsed (5,6

months), 2 CCR (7+,12+

months)

Bergmann [228] 21 2nd CR 7 CCR at 7+ to 49+ months

5 pts CR2 duration> CR1

duration

Wiernick [227] 7 2nd CR CR duration 4–49 months PEG-IL-2

2 CCR (43+,49+ months)

3 pts CR2 duration> CR1

duration

Brune [230] 28 1st CR (17) Median CR1 duration 12+ Ara-C + 6-TG between IL-2

2nd CR (8) months (range 7–51 months) courses

3rd CR (2) (12/17 remain in CR)

4th CR (1) Median CR> 1 duration 13

months (range 6–49) (5/11

remain in CR)

8/10 evaluable CR> 1 longer

than previous CR

Cortes [229] 18 1st CR 6 CCR after 4 years

TNFα, IFNγ , and IL-1 secretion [246]. All patientsengrafted, but one died of intracerebral hemorrhage 13weeks after the transplant, and one relapsed 4 monthsafter transplant [246]. Linomide has also been usedafter autologous BMT for patients with CML [244].Six patients (one in second chronic phase, one in ac-celerated phase, four in first chronic phase) receivedLinomide after engraftment and continued for twoyears. The preliminary analysis had little informationon cytogenetic responses, but interestingly 3 patientsdeveloped graft-versus-host disease [244].

The experience on linomide outside the transplantsetting is limited. One study reported on 17 patientswith myelodysplastic syndrome (6 refractory anemia,1 refractory anemia with ringed sideroblasts, 10 re-fractory anemia with excess blasts) treated with Li-

nomide [247]. There was an increase in CD8+ andCD56+/CD3-cells, but no increase in NK or LAKcell activity [247]. After therapy, neutrophil countsimproved in 4 patients, platelet counts in two, andhemoglobin in one patient who became transfusionindependent [247]. Linomide, and other immunomod-ulatory drugs (e.g., histamine) are therefore attractivealternatives in the management of acute leukemiaswhich deserve further investigation.

IV. Specific immunotherapy

Although there is little clinical data for specific im-munotherapy, preclinical results make this an excitingopportunity to explore. One approach has been theuse of AML cells transducted with B7.1, a criticalco-stimulatory molecule in the generation of an ef-

71

fective immune response [248]. Mice treated withirradiated B7.1-transfected cells 2 weeks prior to in-oculation of untreated leukemia cells are protectedfrom developing leukemia, and the immunity is long-lasting (up to 5 to 6 months) upon rechallenge [249].Furthermore, mice vaccinated early (i.e., one week)after inoculation of untreated leukemia cells reject theleukemia, although later vaccination (i.e., 2 weeksafter AML inoculation) was ineffective [249]. How-ever, the combination of early chemotherapy and latevaccination cured 100% of the mice, suggesting thatsuch an approach could be used in patients withAML [250]. Using a similar strategy, the same au-thors used AML cells transfected with B7.2 or oneof several cytokines with dendritic cell-stimulatingproperties (GM-CSF, IL-4, or TNF-α) [251]. Micevaccinated with GM-CSF-transfected AML cells de-veloped anti-leukemia immunity that protected themwhen challenged with untreated AML cells. Further-more, GM-CSF-transfected AML cells rescued 100%of mice when vaccinated one week prior to injec-tion with untreated AML and up to 80% when givenlate (i.e., 2 weeks after AML inoculation) [251].Therefore, GM-CSF gene immunotherapy might beeven more effective than B7.1-based cell vaccines andconstitute a promising approach to take to the clinic.

Another approach is to develop “vaccines” againstleukemia-specific antigens. One approach has beento use peptides derived from chromosomal transloca-tions resulting in ocogenic fusion proteins. Althoughmany of these proteins are intracellular, their abilityto bind HLA class I molecule and therefore their po-tential to stimulate a specific immunologic responsehas been reported [252]. This approach would belimited to leukemias where a specific fusion proteinis known (e.g., BCR/ABL, PML/RARα, etc.) but itmight prove beneficial in such cases. An alternativeto this strategy is the use of proteins expressed in amore uniform fashion. Proteinase 3 is overexpressedin a variety of myeloid leukemias [253] and it mightbe involved in the process of leukemotrenesis or main-tenance of the leukemic phemotype [254]. PR-1, apeptide contained in proteinase 3, can bind HLA-A2.1and stimulate the generation of cytotoxic T lympho-cytes which preferentially lyse myeloid leukemia cells[255–256]. Clinical studies using these “vaccines” arecurrently underway.

Dendritic cells are potent antigen-presenting cellsresponsible for eliciting T-cell responses and are cur-rently being investigated to generate effective can-cer immunotherapy [257]. Dendritic cells generated

from leukemic cells could present the necessary an-tigens to elicit a specific immune response. Dendriticcells have been generated ex vive from myelomono-cytic precursors from chronic mylogenous leukemia[258] and acute myelogenous leukemia [259]. Auto-logous lymphocytes co-cultured with these dendriticcells were able to lyse autologous leukemia cells, butnot normal cells, raising the possibility of specificimmunotherapy.

Other approaches

Multiple other approaches are currently under devel-opment and might represent the future of the treatmentof acute leukemia. Many of them are in early phases ofdevelopment and the data available is still preliminary.Although not all inclusive, some of the most promisingand challenging new drugs are:

I. Retinoids. The activity of all-trans retinoic acid(ATRA) in acute promyelocytic leukemia has beenwell established and is beyond the scope of this re-view. However, retinoids are being investigated inother acute leukemias with intriguing results. Retin-oids increase the sensitivity of leukemia cells to ara-C[260] and anthracyclines [261]. This effect might bemediated by downregulation of bcl2 [261,262] and ad-dition of G-CSF might augment this phenomenon in asynergistic way [263].

Based on these data, a recent trial investigated theeffect of adding ATRA to chemotherapy on a ran-domized trial of fludarabine, ara-C and idarrubicin±G-CSF± ATRA in patients with poor-prognosis AML(excluding APL) [264]. Patients who received ATRAhad an early survival advantage but it was lost withmore follow-up [264]. ATRA alone has been reportedto induce a complete remission in a patient with AML[265]. In a small series of 13 patients with refractoryor resistant AML, ATRA in combination with low-dose ara-C (20 mg/m2 subcutaneous, twice daily for10 days every month) induced remission in 8 patients.Therefore, the role of retinoids in acute leukemiasneeds to be further explored, looking at schedules thatmight be more appropriate [260,261], new formula-tions such as liposomal ATRA, and other retinoidssuch as 9-cis retinoic acid which have a more potentanti-leukemia activityin vitro against AML [267].

II. Arsenic Trioxide. Arsenic trioxide (As2O3) wasinitially reported to have significant activity in APL,

72

with 9 of 10 patients who had relapsed after treatmentwith ATRA achieved a remission when treated withAs2O3 alone [268]. No myelosuppression was seenon these patients. These results have been expanded,including the achievement of molecular remissions in8 of 11 patients who achieved a CR [269]. As2O3 in-duces differentiation [269] and apoptosis [269–271] ofthe leukemic cells, including downregulation of bcl-2[270] and increased caspase expression and activation[269]. Induction of apoptosis may occur without in-duction of differentiation and is associated with theloss of the PML/RAR alpha fusion protein [271]. In-terestingly, As2O3 has been reported to inhibit growthand induce apoptosis independent of PML/RAR ex-pression in a variety of myeloid leukemia cell lines[272], and significant cytotoxicity against other tumorcell-lines, including breats and prostate has been re-ported [273–274]. The activity of As2O3 in patientswith other leukemias is currently being investigated.

II. Targeted Therapy. CGP 57148 (STI 571) is anovel agent that inhibits the tyrosine kinase activity ofABL and the platelet-derived growth factor receptorand other activated ABL tyrosine kinases [275–277],and suppresses the growth of cells expressing theseproteins [275–278]. Early clinical results suggest avery potent anti-leukemia activity with minimal tox-icity in patients with Interferon-resistant Ph-positiveCML [279] and studies in ALL and transformed CMLare ongoing. Another approach is the use of Ras in-hibitors. Ras mutations occur frequently in leukemias,the highest frequency being reported in chronic my-elomonocytic leukemia where as many as 65% ofpatients have been shown to have mutations in Ras.Mutations in AML have been reported in 25% to 44%of patients. Ras may be activated by other mechan-isms in the absence of mutations (e.g., BCR-ABL)[280,281]. Recently, a new class of drugs has beendescribed which targets the posttranslational prenyla-tion of Ras critical for its membrane localization andactivity. These drugs target the enzyme FTase and arecalled farnesyl transferase inhibitors [282].

Conclusions

Several promising strategies exist which will hope-fully find a role and lead to improved prognosis inacute leukemia. Some of them (new chemotherapyagents, MoAb, MDR modulation) have been dis-cussed in this review. One important lesson from

some of these trials is the importance of molecularpharmacology in the rational design of chemother-apy combinations to obtain the most benefit of certaindrugs, and investigate potential synergism (and avoidantagonisms). Appropriate schedules of combinationchemotherapy and even single agents should be de-signed based on their mechanisms of action and phar-macologic properties. Other approaches, such as doseintensive chemotherapy and stem cell transplantationand leukemia sensitization (i.e., using growth factorssuch as G-CSF, GM-CSF, IL-3, and others) have beencovered by excellent recent reviews [283,284]. Thenewer approaches directed to specific targets, such asleukemic cells (e.g., monoclonal antibodies), specificgenes (e.g., tyrosine kinase inhibitors, Ras inhibit-ors), immune modulators (e.g., n-2, dendritic cells),or mechanisms of resistance (e.g., MDR blockers, in-hibitors of DNA repair) offer a refreshing promiseto the treatment of leukemias and might change ourmanagement of these diseases in the coming years.

References

1. Estey EH: Treatment of acute myelogenous leukemia andmyelodysplastic syndromes. Semin Hematol 32: 132–151,1995

2. Rowe JM, Liesveld JL: Treatment and prognostic factors inacute myeloid leukemia. Baillieres Clin Hematol 9: 87–105,1996

3. Cortes J, Kantarjian HM: Acute lymphoblastic leukemia. Acomprehensive review with emphasis on biology and therapy.Cancer 76: 2393–2417, 1995

4. Copelan EA, McGuire EA: The biology and treatment ofacute lymphoblastic leukemia in adults. Blood 85: 1151-1168, 1995

5. Russell NH: Biology of acute leukemia. Lancet 349: 118–122, 1997

6. Spriggs DR, Stopa E, Mayer RJ, Schoene W, Kufe DW:Fludarabine phosphate (NSC 312878) infusions for the treat-ment of acute leukemia: phase I and neuropathological study.Cancer Res 46: 5953–5958, 1986

7. Warrell RP Jr, Berman E: Phase I and II study of fludarabinephosphate in leukemia: therapeutic efficacy with delayedcentral nervous system toxicity. J Clin Oncol 4: 74–79, 1986

8. Gandhi V, Kemena A, Keating, MJ, Plunkett W: Fludara-bine infusion potentiates arabinosylcytosine metabolism inlymphocytes of patients with chronic lymphocytic leukemia.Cancer Res 52: 897–903, 1992

9. Gandhi V, Estey E, Keating MJ, Plunkett W: Fludarabinepotentiates metabolism of cytarabine in patients with acutemyelogenous leukemia during therapy. J Clin Oncol 11:116–124, 1993

10. Estey E, Plunkett W, Gandhi V, Rios MB, Kantarjian H,Keating MJ: Fludarabine and arabinasylcytosine therapy ofrefractory and relapsed acute myelogenous leukemia. Leuk-emia Lymphoma 9: 343–350, 1993

73

11. Estey E, Thall P, AndreeffM, Beran M, Kantarjian H,O’Brien S, Escudier S, Robertson LE, Koller C, KornblauS, Pierce S, Freireich EJ, Deisseroth A, Keating M: Useof granulocyte colony-stimulating factor before, during, andafter fludarabine plus cytarabine induction therapy of newlydiagnosed acute myelogenous leukemia or myelndysplasticsyndromes: comparison with fludarabine plus cytarabinewithout granulocyte colony-stimulating factor. J Clin Oncol12: 671–678, 1994

12. Visani G, Tosi P, Zinzani PL, Manfroi S, Ottaviani E, TestoniN, Clavio M, Cenacchi A, Gamberi B, Carrara P: FLAG(fludarabine + high-dose cytarabine + G-CSF): an effectiveand tolerable protocol for the treatment of “poor risk” acutemyeloid leukemias. Leukemia 8: 1842–1846, 1994

13. Huhmann IM, Watzke HH, Geissler K, Gisslinger H, LagerU, Knobl P, Pabinger I, Korninger L, Mannhalter C, Mit-terbauer G, Schwarzinger I, Kalhs P, Haas OA, Lechner K:FLAG (fludarabine, cytosine arabinoside, G-CSF) for re-fractory and relapsed acute myeloid leukemia. Ann Hematol73: 265–271, 1996

14. Parker JE, Cullis JO, Mijovic A, Pagliuca A, Mufti GJ: Idaru-bicin, fludarabine, cytarabine and G-CSF (FLAG-IDA) forthe treatment of high risk myeloid malignancies. Blood 88(Suppl 1): 454a (Abstract # 1803), 1996

15. Santana VM, Mirro J, Kearns C, Schell MJ, Crom W,Blakley RL: 2-chlorodeoxyadenosine produces a high rate ofcomplete hematologic remission in relapsed acute myeloidleukemia. J Clin Oncol 10: 364–370, 1992

16. Santana VM, Hunvitz CA, Blakley RL, Crom WR, Luo X,Roberts WM, Ribeiro R, Mahmoud H, Krance RA: Completehematologic remissions induced by 2-chlorodeoxyadenosinein children with newly diagnosed acute myeloid leukemia.Blood 84: 1237–1242, 1994

17. Vahdat L, Wong ET, Wile MJ, Rosemblum M, Foley KM,Warrell RP Jr: Therapeutic and neurotoxic effects of 2-chlorodeoxyadenosine in adults with acute myeloid leuk-emia. Blood 84: 3429–3434, 1994

18. Kornblau SM, Gandhi V, Andreeff HM, Beran M, KantarjianHM, Koller CA, O’Brien S, Plunkett W, Estey E: Clinicaland laboratory studies of 2-chlorodeoxyadenosine± cytosinearabinoside for relapsed or refractory acute myelogenousleukemia in adults. Leukemia 10: 1563–1569, 1996

19. Gandhi V, Estey E, Keating MJ, Chucrallah A, PlunkettW: Chlorodeoxyadenosine and arabinosylcytosine in pa-tients with acute myelogenous leukemia: pharmacokinetic,pharmacodynamic, and molecular interactions. Blood 87:256–264, 1996

20. Xie C, Plunkett W: Metabolism and actions of 2-chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-adenine in humanlymphoblastoid cells. Cancer Res 55: 2847–2852, 1995

21. Shewach DS, Daddona PE, Ashcraft E, Mitchell BS: Me-tabolism and selective cytotoxicity of 9-beta-D-arabinofuranosylguanine in human lymphoblasts. CancerRes 45: 1008–1014, 1985

22. Gandhi V, Plunkett W, Rodriguez CO, Nowak BJ, Du M,Ayres M, Kisor DF, Mitchell BS, Kurtzberg J, Keating MJ:Compound GW506U78 in refractory hematologic malig-nancies: relationship between cellular pharmacokinetics andclinical response. J Clin Oncol 16: 3607–3615, 1998

23. Kurtzberg J, Keating M, Moore JO, Gandhi V, Blaney S,Gold S, Ernst T, Henslee-Downey J, Chang A, Kisor D, Plun-kett W, Mitchell B: 2-amino-9-B-D-arabinosyl-6-methoxy-9H-guanine (GW 506U; compound 506U) is highly activein patients with T-cell malignancies: results of a phase I trial

in pediatric and adult patients with refractory hematologicalmalignancies. Blood 88 (Suppl 1): 669a (Abstract #2666),1996

24. Aguayo A, Cortes JE, Kantarjian HM, Beran M, Gandhi V,Plunkett W, Kurtzberg J, Keating MJ: Complete hematologicand cytogenetic response to 2-amino-9-β-D-arabinosyl-6-methoxy-9H-guanine in a patient with chronic myelogenousleukemia in T-cell blastic phase. Cancer 85: 58–64, 1999

25. O’Brien S, Thomas D, Kantarjian H, Freireich E, Koller C,Cortes J, Giles F, Bivens C, Lerner S, Keating M, Hodge J,Spector N: Compound 506 has activity in mature lymphoidleukemia. Blood 92 (Suppl 1): 490a, 1998

26. Tricot G, Weber G: Biochemically targeted therapy ofrefractory leukemia and myeloid blast crisis of chronicgranulocytic leukemia with Tiazofurin, a selective blocker ofinosine 5′-phosphate dehydrogenase activity. Anticancer Res16: 3341–3347, 1996

27. Pinkel D: Intravenous mercaptopurine: life begins at 40. JClin Oncol 11: 1826-1831, 1993

28. Koren G, Ferrazini G, Sulh H, Langevin AM, KapelushnikJ, Klein J, Giesbrecht E, Soldin S, Greenberg M: Systemicexposure to mercaptopurine as a prognostic factor in acutelymphocytic leukemia in children. N Engl J Med 323: 17–21,1990

29. Lennard L, Lilleyman JS: Variable mercaptopurine meta-bolism and treatment outcome in childhood lymphoblasticleukemia. J Clin Oncol 7: 1816–1823, 1989

30. Schmiegelow K, Schrøder H, Gustafsson G, Kristinsson J,Glomstein A, Salmi T, Wranne L: Risk of relapse in child-hood acute lymphoblastic leukemia is related to RBC metho-trexate and mercaptopurine metabolites during maintenancechemotherapy. J Clin Oncol 13: 345–351, 1995

31. Evans WE, Crom WR, Abromowitch M, Dodge R, Look AT,Bowmen WP, George SL, Pui CH: Clinical pharmacodynam-ics of high-dose methotrexate in acute lymphocytic leukemia.Identification of a relation between concentration and effect.N Engl J Med 314: 471–477, 1986

32. Camitta B, Leventhal B, Lauer S, Shuster JJ, Adair S, CasperJ, Civin C: Intermediate-dose intravenous methotrexate andmercaptopurine theraphy for non-T, non-B acute lympho-cytic leukemia of childhood: a pediatric oncology groupstudy. J Clin Oncol 10: 1539–1544, 1989

33. Camitta B, Mahoney D, Leventhal B, Lauer SJ, Shuster JJ,Adair S, Civin C, Munoz L, Steuber P, Strother D: Intensiveintravenous methotrexate and mercaptopurine treatment ofhigher-risk non-T, non-B acute lymphocytic leukemia: a Pe-diatric Oncology Group study. J Clin Oncol 12: 1383–1389,1994

34. Mahoney DH, Shuster J, Nitschke R, Lauer SJ, Winick N,Steuber CP, Camitta B: Intermediate-dose intravenous me-thotrexate with intravenous mercaptopurine is superior torepetitive low-dose oral methotrexate with intravenous mer-captopurine for children with lower-risk B-lineage acutelymphoblastic leukemia: a Pediatric Oncology Group PhaseIII Trial. J Clin Oncol 16: 246–254, 1998

35. Kantarjian H, O’Brien S, Keating MJ, Cortes J, Giles FJ, Be-ran M, Koller C, Andreeff M, Konblau S, Pierce S, MurphyS, Freireich EJ: Update of the Hyper-CVAD dose-intensiveregimen in adult acute lymphoblastic leukemia. Blood 92(Suppl 1): 313a (Abstract 1285), 1998

36. Ramilo-Torno LV, Avramis VI: Intracellular pharmaco-dynamic studies of the synergistic combination of 6-mercaptopurine and cytosine arabinoside in human leukemiacell lines. Cancer Chemother Pharmacol 35: 191–199, 1995

74

37. Lockhart S, Plunkett W, Jeha S, Ramirez I, Zipf T,Cork A, Pinkel D: High-dose mercaptopurine followed byintermediate-dose cytarabine in relapsed acute leukemia. JClin Oncol 12: 587–595, 1994

38. Canpolat C, Jeha S, Lockhart S, Ramirez I, Zipf T, Pinkel D:High-dose mercaptopurine and intermediate-dose cytarabineduring first remission of acute myeloid leukemia. CancerInvest 15: 121–126, 1997

39. Morrison FS, Kopecky KJ, Head DR, Athens JW, BalcerzakSP, Gumbart C, Dabich L, Costanzi JJ, Coltman CA, SaikiJH: Late intensification with POMP chemotherapy prolongssurvival in acute myelogenous leukemia. Results of a South-west Oncology Group study of rubidazone versus adriamycinfor remission induction, prophylactic intrathecal therapy, lateintensification, and levamisole maintenance. Leukemia 6:708–714, 1992

40. Lennard L, Lilleyman JS, Von Loon J, Weinshilboum RM:Genetic variation in response to 6-mercaptopurine for child-hood acute lymphoblastic leukemia. Lancet 336: 225-229,1990

41. Adamson PC, Poplack DG, Balis FM: The cytotoxicity ofthioguanine versus mercaptopurine in acute lymphoblasticleukemia. Leuk Res 18: 805–810, 1994

42. Kitchen BJ, Balls FM, Poplack DG, O’Brien M, Craig CE,Adamson PC: A pediatric phase I trial and pharmacokineticstudy of thioguanine administered by continuous IV infusion.Clin Cancer Res 3: 713–717, 1997

43. Issa J-PJ, Zehnbauer BA, Kaufmann SH, Biel MA, BaylinSE: HIC1 hypermethylation is a late event in hematopoieticneoplasms. Cancer Res 57: 1678–1681, 1997

44. Steuber CP, Krischer J, Holbrook T, Camitta B, Land V,Sexauer C, Mahoney D, Weinstein H: Therapy of refractoryor recurrent childhood acute myeloid leukemia using amsac-rine and etoposide with or without azacitidine: a PediatricOncology Group randomized phase II study. J Clin Oncol14: 1521–1525, 1996

45. Wang C, McCulloch EA: Sensitivity to 5-azacytidine of blastprogenitors in acute myeloblastic leukemia. Blood 69: 553–559, 1987

46. Case DC Jr: 5-azacytidine in refractory acute leukemia.Oncology 39: 218–221, 1982

47. Silverman LR, Holland JF, Weinberg RS, Alter BP, DavisRB, Ellison RR, Demakos EP, Cornell CJ Jr, Carey RW,Schiffer C, Frei E, McIntyre OR: Effects of treatment with5-azacytidine on thein vivo andin vitro hematopoiesis in pa-tients with myelodysplastic syndromes. Leukemia 7 (Suppl1): 21–29, 1993

48. Momparler RL, Gyger M, Rivard GE: Clinical trial on 5-aza-2′-deoxycytidine therapy in patients with acute leukemia.Pharmacol Ther 30: 277–286, 1985

49. Debusscher L, Marie JP, Dodion P, Blanc GM, ArrigoC, Zittoun R, Stryckmans P: Phase I–II trial of 5-aza-2′ -deoxycytidine in adult patients with acute leukemia. In:Momparler RL, De Vos D (eds): 5-aza-2′ -deoxycytidine: pre-clinical and clinical studies. PCH, Haarlem, Netherlands, pp:131–142, 1990

50. Willemze R, Suciu S, Archimbaud E, Muus P, Stryckmans P,Louwagie EA, Berneman Z, Tjean M, Wijermans P, DohnerH, Jehn U, Labar B, Jaksic B, Dardenne M, Zittoun R:A randomized phase II study on the effects of 5-aza-2′ -deoxycytidine combined with either amsacrine or idarubicinin patients with relapsed acute leukemia: an EORTC Leuk-emia Cooperative Group phase II study (06893). Leukemia11 (Suppl 1): S24–S27, 1997

51. Pinto A, Zagonel V: 5-aza-2′-deoxycytidine (decitabine) and5-azacytidine in the treatment of acute myeloid leukemiasand myelodysplastic syndromes: past, present, and futuretrends. Leukemia 7 (Suppl 1): 51–60, 1993

52. Schwartsmann G, Fernandes MS, Schaan MD, Moschen M,Gerhardt LM, Di Leone L, Loitzembauer B, Kalakun L: De-citabine (5-aza-2′ -deoxycytidine; DAC) plus daunorubicin asa first line treatment in patients with acute myeloid leukemia:preliminary observations. Leukemia 11(Suppl 1): S28–S31,1997

53. Wijermans PW, Krulder JW, Huijgens PC, Neve P: Con-tinuous infusion low-dose 5-Aza-2′-deoxycytidine in elderlypatients with high-risk myelodysplastic syndrome. Leukemia11: 1–5, 1997

54. Giralt S, Davis M, O’Brien S, van Besien K, Champlin R, deVos D, Kantarjian H: Studies of decitabine with allogeneicprogenitor cell transplantation. Leukemia 11 (Suppl 1): S32–S34, 1997

55. Kantajian HM, O’Brien SM, Keating M, Beran M, Estey E,Giralt S, Kornblau S, Rios MB, de Vos D, Talpaz M: Resultsof decitabine therapy in the accelerated and blastic phases ofchronic myelogenous leukemia. Leukemia 11: 1617–1620,1997

56. Anzai H, Frost P, Abbruzzese JL: Synergistic cytotoxicitywith 2′-deoxy-5-azacytidine and topotecanin vitro and invivo. Cancer Res 52: 2180–2185, 1992

57. Surbone A, Ford H Jr, Kelley JA, Ben-Baruch N, Thomas RV,Fine R, Cowan KH: Phase I and pharmacokinetic study ofarabinofuranosyl-5-azacytosine (fazarabine, NSC 281272).Cancer Res 50: 1220–1225, 1990

58. Hertel LW, Boder GB, Kroin JS, Rinzel SM, Poore GA,Todd GC, Grindey GB: Evaluation of the antitumor activityof gemcitabine (2′,2′-difluoro-2′-deoxycytidine). Cancer Res50: 4417–4422, 1990

59. Cory AH, Hertel LW, Kroin J S, C ory JG: Effects of2′,2′-difluoro deoxycytidine (Gemcitabine) on wild type andvariant mouse leukemia L1210 cells. Oncol Res 5: 59–63,1993

60. Huang P, Plunkett W: Induction of apoptosis by gemcitabine.Semin Oncol 22 (4 Suppl 11): 19–25, 1995

61. Grunewald R, Kantarjian H, Keating MJ, Abbruzzese J,Tarassoff P, Plunkett W: Pharmacologically directed designof the dose rate and schedule of 2′,2′difluorodeoxycytidine(Gemcitabine) administration in leukemia. Cancer Res 50:6823–6826, 1990

62. Grunewald R Kantarjian H, Du M, Faucher K, TarassoffP,Plunkett W: Gemcitabine and leukemia: a phase I clinical,plasma, and cellular pharmacology study. J Clin Oncol 10:406–413, 1992

63. Gandhi V, Plunkett W: Modulatory activity of 2′,2′-difluorodeoxycytidine on the phosphorylation and cytotox-icity of arabinosyl nucleosides. Cancer Res 50: 3675–3680,1990

64. Santini V, D’Ippolito G, Bernabei PA, Zoccolante A, ErminiA, Possi-Ferrini P: Effects of fludarabine and gemcitabineon human acute myeloid leukemia cell line HL 60: dir-ect comparison of cytotoxicity and cellular Ara-C uptakeenhancement. Leukemia Res 20: 37–45, 1996

65. Husain I, Mohler JL, Seigler HF, Besterman JM: Elevation oftopoisomerase I messenger RNA, protein, and catalytic activ-ity in human tumors: demonstration of tumor-type specificityand implications for cancer chemotherapy. Cancer Res 54:539–546, 1994

75

66. Liu LF: DNA topoisomerase poisons as antitumor drugs. AnnRev Biochem 58: 351–375, 1989

67. Giovanella BC, Stehlin JS, Wall ME, Wani MC, NicholasAW, Liu LF, Silber R, Potmesil M: DNA topoisomerase I-targeted chemotherapy of human colon cancer xenografts.Science 246: 1046–1048, 1989

68. Kantarjian HM, Beran M, Ellis A, Zwelling L, O’Brien S,Cazenave L, Koller C, Rios MB, Plunkett W, Keating JM,Estey EH: Phase I study oftopotecan, a new topoisomeraseI inhibitor, in patients with refractory or relapsed acuteleukemia. Blood 81: 1146–1151, 1993

69. Rowinsky EK, Adjei A, Donehower RC, Gore SD, Jones RJ,Burke PJ, Cheng Y-C: Gorchow LB, Kaufman SH: Phase Iand pharmacodynamic study of the topoisomerase I inhibitortopotecan in patients with refractory acute leukemia. J ClinOncol 12: 2193–2203, 1994

70. Beran M, Kantarjian H, O’Brien S, Koller C, Albitar M,Arbuck S, Pierce S, Moore M, Abbruzzese JL, Andreeff M,Keating M, Estey E: Topotecan, a topoisomerase I inhibitor,is active in the treatment of myelodysplastic syndrome andchronic myelomonocytic leukemia. Blood 88: 2473–2479,1996

71. Crump M, Lipton J, Hedley D, Stewart AK, Sutton D, Keat-ing A, Minden M, Messner H, Eisenhauer E: A phase I trialof sequential topotecan and etoposide in adult acute myeloidleukemia. Blood 88 (Suppl 1): 220a (Abstract #867), 1996

72. Seiter K, Feldman EJ, Halicka HD, Traganos F, Dar-zynkiewicz Z, Lake D, Ahmed T: Phase I clinical and labor-atory evaluation of topotecan and cytarabine in patients withacute leukemia. J Clin Oncol 15: 44–51, 1997

73. Beran M, Estey EH, O’Brien S, Cortes J, Koller CA, GilesFJ, Kornblau S, AndreeffM, Vey N, Pierce SR, Hayes K,Wong GC, Keating M, Kantarjian H: Topotecan and cyta-rabine is an active combination regimen in myelodysplasticsyndromes and chronic myelomonocytic leukemia. J ClinOncol (in press), 1999

74. Boothman DA, Wang M, Schea RA, Burrows HL, Strick-faden S, Owens JK: Posttreatment exposure to camptothecinenhances the lethal effects of x-rays on radioresistant hu-man malignant melanoma cells. Int J Radiat Oncol Biol Phys24(5): 939–948, 1992

75. Kano Y, Suzuki K, Akutsu M, Suda K, Inoue Y, Yoshida M, etal.: Effects of CPT-11 in combination with other anti-canceragents in culture. Int J Cancer 50(4): 604–610, 1992

76. Cortes J, Estey E, Beran M, O’Brien S, Giles F, Koller C,Keating M, Kantarjian: Cyclophosphamide, ara-C, and to-potecan (CAT) for patients with refractory or relapsed acuteleukemia. Leukemia Lymphoma (in press), 1999

77. Gerrits CJH, Schellens JHM, Burris H, Eckardt JR, Plant-ing AST, van der Burg MEL, Rodriguez GI, Loos WJ, vanBeurden V, Hudson I, von Hoff DD, Venveij J: A comparisonof clinical pharmacodynamics of different administrationschedules of oral topotecan (Hycamtin). Clin Cancer Res 5:69–75, 1999

78. Armand JP: CPT-11: clinical experience in phase I studies.Semin Oncol 23 (Suppl 1): 27–33, 1996

79. Ohno R, Okada K, Masaoka T, Kuramoto A, Arima T,Yoshida Y, Ariyoshi H, Ichimaru M, Sakai Y, Oguro M: Anearly phase II study of CPT-11: a new derivative of camp-tothecin, for the treatment of leukemia and lymphoma. J ClinOncol 8: 1907–1912, 1990

80. Kantarjian H, Cortes J, O’Brien S, Verschraegen CF, Giovan-ella BC, Cao Z, Stehlin JS: 9-nitro-20-(S)-camptothecin (9-NC, RFS2000): an effective agent for treatment of chronic

myelomonocytic leukemia and Philadelphia chromosome-negative chronic myelogenous leukemia. Proc ASCO 1999

81. Huang MT: Harringtonine, an inhibitor of initiation of pro-tein biosynthesis. Mol Pharmacol 11: 511–519, 1975

82. Boyd AW, Sullivan JR: Leukemic cell differentiationin vivoandin vitro: arrest of proliferation parallels the differentiationinduced by the antileukemic drug Harringtonine. Blood 63:384–392, 1984

83. Changliang H, Mingliang D, Renjie G, Mingtang W, Feng-zhu L, Yingming L, Qingda Q: A study of the inductionof differentiation of human leukemic cells by harringtoninecombined with cytarabine. Leukemia 2: 518–522, 1988

84. Chinese People’s Liberation Army 187th Hospital: Harring-tonine in acute leukemia: Clinical analysis of 31 cases. ChinMed J 3: 319–324, 1977

85. Chinese People’s Liberation Army 187th Hospital: Harring-tonine in the treatment of acute leukemia: Clinical analysesof 72 cases. Chin Med J 3: 163, 1978

86. Hematology Research Division and Hematology Sectionof the Children’s Hospital, Suzhou Medical College. Highremission-induction (traditional sine-Western) HOAP) regi-men for acute nonlympholeukemia. Chin Med J 93: 565–568,1980

87. Zhang ZY, Hou CH, Zhu YF, You XY, Li JL, Fu Z: Curat-ive effect of harringtonine semisynthetic harringtonine andHOAP on nonlymphocytic leukemias. Analysis of 304 cases.Chin Med J 100: 565–568, 1987

88. Legha SS, Keating M, Picket S, Ajani JA, Ewer M, BodeyGP: Phase I clinical investigation ofhomoharringtonine. Can-cer Treat Rep 68: 1085–1091, 1984

89. Stewart JA, Krakoff IH: Homoharringtonine: a phase I eval-uation. Invest New Drugs 3: 279–286, 1985

90. Coonley CJ, Warrell RP, Young CW: Phase I trial of homo-harringtonine administered as a 5-day continuous infusion.Cancer Treat Rep 67: 693–696, 1983

91. Neidhart JA, Young DC, Kraut E, Howinstein B, MetzEN: Phase I trial of homoharringtonine administered byprolonged continuous infusion. Cancer Res 46: 967–969,1986

92. Warrell RP, Coonley CJ, Gee TS: Homoharringtonine: aneffective new drug for remission induction in refractorynonlymphoblastic leukemia. J Clin Oncol 3: 617–621, 1985

93. Feldman E, Arlin Z, Ahmed T, Mittelman A, Puccio C, ChunY Cook P, Baskind P: Homoharringtonine is safe and effect-ive for patients with acute myelogenous leukemia. Leukemia6: 1185–1188, 1992

94. Stewart JA, Cassileth PA, Bennett JM, O’Connell MJ: Con-tinuous infusion homoharringtonine (NSC 141633) in re-fractory acute nonlymphacytic leukemia. Am J Clin Oncol11: 627–629, 1988

95. Kantarjian HM, Keating MJ, Walters RS, Koller CA, Mc-Credie KB, Freireich EJ: Phase II study of low-dose continu-ous infusion homoharringtonine in refractory acute myelo-genous leukemia. Cancer 63: 813–817, 1989

96. Feldman EJ, Seiter KP, Ahmed T, Baskind P, Arlin ZA:Homoharringtonine in patients with myelodysplastic syn-drome (MDS) and MDS evolcing to acute myeloid leukemia.Leukemia 10: 40–42, 1996

97. Feldman E, Arlin Z, Ahmed T, Mittelman A, Puccio C,Chun H, Cook P, Baskind P: Homoharringtonine in com-bination with cytarabine for patients with acute myelogenousleukemia. Leukemia 6: 1189–1191, 1992

98. Feldman E, Arlin Z, Ahmed T, Mittelman A, Ascensao JL,Puccio C, Coombe N, Baskind P: Acute promyelocytic leuk-

76

emia: a 5-year experience with new antileukemic agentsand a new approach at preventing fatal hemorrhage. ActaHematol 82: 117–121, 1989

99. Jing-song Y, Xiao-hong W, Guang-hui F, Gui-ren L, Zhi-pingL: Small-dose harringtonine induces complete remission inpatients with acute promyelocytic leukemia. Leukemia 2:427–429, 1988

100. Takemura Y, Ohnuma T, Chou TC, Okano T, Holland IF:Biologic and pharmacologic effects of harringtonine on hu-man leukemia-lymphoma cells. Cancer Chemother Phar-macol 14: 206–210, 1985

101. Broggini M, Coley HM, Mongelli N, Pesenti E, Wyatt MD,Hartley JA, D’Incalci: DNA sequence-specific adenine al-kylation by the novel antitumor drug tallimustine (FCE24517), a benzoyl nitrogen mustard derivative of distamycin.Nucleic Acids Res 23: 81–87, 1995

102. Erba E, Mascellani E, Pifferi A, D’Incalci M: Comparison ofcell-cycle phase perturbations induced by the DNA-minor-groove alkylator tallimustine and by melphalan in the SW626cell line. Int J Cancer 62: 170–175, 1995

103. Ciomei M, Pastori W, Capolongo L, Geroni C, MelegaroG, Penella G, Grandi M: Decreased tyrosine phosphorilationin tumor cells resistant to FCE 24517 (Tallimustine). Br JCancer 72: 1504–1508, 1995

104. Punt CJ, Humblet Y, Roca E, Dirix LY, Wainstein R, Polli A,Corradino I: Tallimustine in advanced previously untreatedcolorectal cancer, a phase II study. Br J Cancer 73: 803–804,1996

105. Viallet J, Stewart D, Shepherd F, Ayoub J, Cormier Y, DiPi-etro N, Steward W: Tallimustine is inactive in patients withpreviously treated small cell lung cancer. A phase II trialof the National Cancer Institute of Canada Clinical TrialsGroup. Lung Cancer 15: 367–373, 1996

106. Sessa C, Pagani O, Zurlo MG, de Jong J, Hofmann C, LassusM, Marrari P, Benedetti SM, Cavalli F: Phase I study of thenovel distamycin derivative tallimustine (FCE 24517). AnnOncol 5: 901–907, 1994

107. Beran M, Jeha S, O’Brien S, Estey E, Vitek L, Zurlo M, RiosMB, Keating M, Kantarjian H: Tallimustine, an effectiveantileukemic agent in a SCID mouse model of adult myelo-genous leukemia induces remissions in a phase I study. Blood88 (Suppl 1): 220a (Abs # 868), 1996

108. Yang L-Y, Li L, Keating MJ, Plunkett W: Arabinosyl-2-fluoroadenine augments cisplatin cytotoxicity and inhibitscisplatin-DNA cross-link repair. Mol Pharmacol 47: 1072–1079, 1995

109. Vogler WR, Harrington DP, Winton EF, Lazarus HM, Ben-nett JM, Cassileth PA, Oken NM: Phase II clinical trial ofcarboplatin in relapsed and refractory leukemia. Leukemia 6:1072–1075, 1992

110. Welborn JL, Kopecky KJ, Meyers FJ, Veith R, Shurafa M,Doroshow JH, Balcerzak SP, Appelbaum FR: Carboplatininfusion in relapsed and refractory acute myeloid leukemia.A Southwest Oncology Group trial. Leukemia 9: 1126–1129,1995

111. Kornblau SM, Andreeff M, Betari M, Estey E, Keat-ing M, Koller C, O’Brien S, Kantajian H: CECA-Cyclophosphamide, etoposide, carboplatin, and cytosine ar-abinoside. A new salvage regimen for relapsed or refractoryacute myelogenous leukemia. Blood 86 (Suppl 1): 514a (Abs# 2045), 1995

112. Fantl D, Nucitfora E, Viñuales S, Penchasky D, ArbelbideJ, Iastrebner M, Dibar E, Makiva M, Tartas N: Infusioncarboplatin and mitoxantrone treatment of relapsed and re-

fractory acute leukemia. Blood 86 (Suppl 1): 758a(Abs #3021), 1995

113. Spiers A, Saba H, Balducci L, Moscinski L, Richard J,Huegel V, Daniel P, Cosgrove D: The Cadet regimen(Carboplatin/ara-C/Daunorubicin/Etoposide/Thioguanine) inacute myeloid leukemia: results in standard risk patients.Blood 86 (Suppl 1): 781a (Abs # 3111), 1995

114. Larrea L, Martin G, Sanz GF, Martinez JA, Arnao M, JarqueI, Jimenez C, Sanz MA: Carboplatin plus cytarabine in thetreatment of high-risk acute myeloblastic leukemia. Blood 88(Suppl 1): 217a (Abs # 856), 1996

115. Lioure B, Moreau P, Witz F, Witz B, Vilque JP, Francois S,Brion A, Delain M, Casassus P, Ojeda M, Cahn JY, OberlingF, Harousseau JL: New association of high-dose mitoxan-trone, carboplatin and VP16 as treatment for relapsed orrefractory acute myelogenous leukemia. Blood 88 (Suppl 1):215a (Abs # 848), 1996

116. Amadori S, Picardi A, Fazi P, Testi AM, Petti MC, Monte-fusco E, Mandelli F: A phase II study of VP-16, intermediate-dose ara-C and carboplatin (VAC) in advanced acute myelo-genous leukemia and blastic chronic myelogenous leukemia.Leukemia 10: 766–768, 1996

117. O’Brien S, Kantarjian H, Freireich E, Johnston D, Nguyen K,Beran M: CI-973, a new platinum derivative with potentialantileukemic activity. Cancer Res 52: 4130–4134, 1992

118. Kantarjian HM, Beran M, O’Brien S, Robertson L, SiddikK, Yoshida M, Yang LY, Rios MB, Keating MJ, Meyer M:Evaluation of CI-973, a platinum analogue, in refractory orrelapsed acute leukemia. Leukemia 10: 396–401, 1996

119. Demur C, Chiron M, Saivin S, Attal M, Dastugue N,Bousquet C, Galinier JL, Colombies P, Laurent L: Effect ofmelphalan against self-renewal capacity of leukemic progen-itors in acute myeloblastic leukemia. Leukemia 6: 204–208,1992

120. Tohda S, Nagata K, Suzuki T, Nara N: Comparative effectsof busulfan, cytosine arabinoside and adriamycin on differentmaturation stages of normal human bone marrow cells. ActaHaematologica 83: 16–21, 1990

121. Kim S: Liposomes as carriers of cancer chemotherapy. Cur-rent status and future prospects. Drugs 46: 618–638, 1993

122. Guaglianone P, Chan K, DelaFlor-Weiss E, et al.: Phase Iand pharmacologic study of liposomal daunorubicin (Daun-oXome). Invest New Drugs 12: 103–110, 1994

123. Cohen P, Gill PS, Wernz J, et al.: Absence of cardiac toxicityin patients who received 600 mg/m2 of liposomal encap-sulated daunorubicin (DaunoXome). Proc ASCO 16: 217a(Abs # 761), 1997

124. Cortes J, O’Brien S, Estey E, Giles F, Keating M, KantarjianH: Phase I study of liposomal daunorubicin in patients withacute leukemia. Invest New Drugs 1999 (in press)

125. Cortes J, Kantarjian H, O’Brien S, Giles FJ, Koller C, BeranM, Keating M, Estey E: High-dose liposomal Daunorubicinand ara-C for refractory or relapsed acute leukemias: a dosesearching study. Blood 92 (Suppl 1): 234a (Abs # 957), 1998

126. Koller C, Cortes J, O’Brien S, Giles F, Keating M, KantajianH. A pilot study of dose-intensive anthracyclines for acutelymphoblastic leukemia (ALL) using liposomal daunorubi-cin (LD) with hyperfractionated cyclophosphamide (CTX),vincristine (VCR), and prednisone (PDN) (HyperCVXD).Proc ASCO 17: 28a (Abs # 110), 1998

127. Gordon KB, Tajuddin A, Guitart J, Kuzel TM, Eramo LR,VonRoenn J: Hand-foot syndrome associated with liposome-encapsulated doxorubicin therapy. Cancer 75: 2169–2173,1995

77

128. Vail DM, Chun R, Thamm DH, Garrett LD, Cooley AJ,Obradovich JE: Efficacy of pyridoxine to ameloriate thecutaneous toxicity associated with doxorubicin containingpegylated (Stealth) liposomes: a randomized, double-blindclinical trial using a canine model. Clin Cancer Res 4:1567–1571, 1998

129. Mayer LD, Bally MB, Loughrey H, Masin D, Cullis PR:Liposomal vincristine preparations which exhibit decreaseddrug toxicity and increased activity against murine L1210and P388 tumors. Cancer Res 50: 575–579, 1990

130. Mayer LD, Masin D, Nayar R, Boman NL, Bally MB: Phar-macology ofliposomal vincristine in mice bearing L1210ascitic and B16/BL6 solid tumours. Br J Cancer 71: 482–488,1995

131. Gelmon KA, Tolcher A, Diab AR, Bally MB, Embree L,Hudon N, Dedhar C, Ayers D, Eisen A, Melosky B, Burge C,Logan P, Mayer LD: Phase I study of liposomal vincristine. JClin Oncol 17: 697–705, 1999

132. Subramanian D, Muller MT: Liposomal encapsulation in-creases the activity of the topoisomerase I inhibitor topo-tecan. Oncology Res 7: 461–469, 1995

133. Colbern GT, Dykes DJ, Engbers C, Musterer R, Hiller A,Pegg E, Saville R, Weng S, Luzzio M, Uster P, Amantea M,Working PK: Encapsulation of the topoisomerase I inhibitorGL147211C in pegylated (Stealth) liposomes: pharmacokin-etics and antitumor activity in HT29 colon tumor xenografts.Clin Cancer Res 4: 3077–3082, 1998

134. Verfaillie CM, Miller JS: CD34+/CD33- cells reselectedfrom macrophage inflammatory protein 1 alpha+interleukin-3-supplemented “stroma noncontact” cultures are highly en-riched for long-term bone marrow culture initiating cells.Blood 84: 1442–1449, 1994

135. Scheinberg DA, Tanimoto M, McKenzie S, Strife A, OldLJ, Clarkson BD: Monoclonal antibody M195: a diagnosticmarker for acute myelogenous leukemia. Leukemia 3: 440–445, 1989

136. Tanimoto M, Scheinberg DA, Cordon-Cardo C, Huie D,Clarkson ED, Old LJ: Restricted expression of an earlymyeloid and monocytoid cell surface antigen defined bymonoclonal antibody M195. Leukemia 3: 339–348, 1989

137. Scheinberg DA, Lovett D, Divgi CR, Graham MC, Ber-man E, Pentlow K, Feirt N, Finn RD, Clarkson BD, GeeTS: A phase I trial of monoclonal antibody M195 in acutemyelogenous leukemia: specific bone marrow targeting andinternalization of radionuclide. J Clin Oncol 9: 478–490,1991

138. Schwartz MA, Lovett DR, Redner A, Finn RD, GrahamMC, Divgi CR, Dantis L, Gee TS, Andreeff M, Old LJ:Dose escalation trial of M195 labeled with iodine 131 frocytoreduction and marrow ablation in relapsed or refractorymyeloid leukemias. J Clin Oncol 11: 294–303, 1993

139. Caron PC, Co MS, Bull MK, Avdalovic NM, Queen C,Scheinberg DA: Biological and immunological features ofhumanized M195 (anti-CD33) monoclonal antibodies. Can-cer Res 52: 6761–6767, 1992

140. Caron PC, Jurcic JG, Scott AM, Finn RD, Divgi CR, Gra-ham MC, Jureidini IM, Sgouros G, Tyson D, Old LJ: Aphase 1B trial of humanized monoclonal antibody M195(anti-CD33) in myeloid leukemia: specific targeting withoutimmunogenicity. Blood 83: 1760–1768, 1994

141. Jurcic JG, Caron PC, Miller WH Jr, Yao TJ, Maslak P, FinnRD, Larson SM, Warrell RP Jr, Scheinberg DA: Sequen-tial targeted therapy for relapsed acute promyelocytic leuk-

emia with all-trans retinoic acid and anti-CD33 monoclonalantibody M195. Leukemia 9: 244–248, 1995

142. Appelbaum FR, Matthews DC, Eary JF, Badger CC, Kel-logg M, Press OW, Martin PJ, Fisher DR, Nelp WB, ThomasED: The use of radiolabeled anti-CD33 antibody to augmentmarrow irradiation prior to marrow transplantation for acutemyelogenous leukemia. Transplantation 54: 829–833, 1992

143. Stiff PJ, Schulz WC, Bishop M, Marks L: Anti-CD33 mono-clonal antibody and etoposide/cytosine arabinoside combin-ations for theex vivepurification of bone marrow in acutenonlymphocytic leukemia. Blood 77: 355–362, 1991

144. Sievers EL, Larson RA, Estey E, Stadtmauer E, Berger M,Eten C, Bernstein E, Appelbaum F: Interim analysis of theefficacy and safety of CMA-676 in patients with AML in firstrelapse. Blood 92 (Suppl 1): 613a (Abs # 2527), 1998

145. McGraw KJ, Rosemblum MJ, Cheung L, Scheinberg DA:Characterization of murine and humanized anti-CD33,gelonin immunotoxins reactive against myeloid leukemias.Cancer Immunol Immunother 39: 367–374, 1994

146. Xu Y, Xu Q, Rosemblum MJ, Scheinberg DA: Antileukemicactivity of recombinant humanized M195-gelonin immuno-toxin in nude mice. Leukemia 10: 321–326, 1996

147. Silla LM, Chen J, Zhong RK, Whiteside TL, Ball ED: Po-tentiation of lysis of leukemia cells by a bispecific antibodyto CD33 and CD16 (Fc gamma RIII) expressed by humannatural killer (NK) cells. Br J Hematol 89: 712–718, 1995

148. Hale G. Xia MQ, Tighe HP, Dyer MJS, Waldmann H:The CAMPATH-1 antigen (Cdw52). Tissue Antigens 35:118–127, 1990

149. Rowan WC, Hale G, Tite JP, Brett SJ: Cross-linking ofthe CAMPATH-1 antigen (CD52) triggers activation of nor-mal human T lymphocytes. International Immunol 7: 69–77,1995

150. Hale G, Swirsky D, Walsmann H, Chan LC: Reactivity ofrat monoclonal antibody CAMPATH-1 with human leukemiacells and its possible application for autologous bone marrowtransplantation. Br J Hematol 60: 41–48, 1985

151. Hale G, Bright S, Chumbley G, Hoang T, Metcalf D, MunroAJ, Waldmann H: Removal of T cells from bone marrow fortransplantation: a monoclonal antilymphocyte antibody thatfixes human complement. Blood 62: 873–872, 1983

152. Waldmann H, Polliak A, Hale G, Or R Cividalli G, Weiss L,Weshler Z, Samuel S, Manor D, Brautbar C: Elimination ofgraft-versus-host disease byin vitro depletion of alloreactivelymphocytes with a monoclonal rat anti-human lymphocyteantibody (CAMPATH-1). Lancet 2: 483–486, 1984

153. Hale G, Clark M, Waldmann H: Therapeutic potential ofrat monoclonal antibodies: isotype specificity of antibody-dependent cell-mediated cytotoxicity with human lympho-cytes. J Immunol 134: 3056, 1985

154. Hale G, Dyer MJ, Clark MR, Phillips JM, Marcus R, Riech-mann L, Winter G, Waldmann H: Remission induction innon-Hodgkin lymphoma with reshaped human monoclonalantibody CAMPATH-1H. Lancet 2: 1394–1399, 1988

155. Dyer MJS, Hale G, Hayhoe FGJ, Walsmann H: Effects ofCAMPATH-1 antibodiesin vivo in patients with lymph-oid malignancies: influence of antibody isotype. Blood 73:1431–1439, 1989

156. Osterborg A, Fassas AS, Anagnostopoulos A, Dyer MJS,Catovsky D, Mellstedt H: Humanized CD52 monoclonal an-tibody Campath-1H as first-line treatment in chronic lymph-ocytic leukemia. Br J Hematol 93: 151–153, 1996

157. Osterborg A, Dyer MJS, Bunjes D, Pangalis GA, Bastion Y,Catovsky D, Mellstedt H: Phase II multicenter study of hu-

78

man CD52 antibody in previosly treated chronic lymphocyticleukemia. J Clin Oncol 15: 1567–1574, 1997

158. Nadler LM, Anderson KC, Marti G, Bates MP, Park E,Daley JF, Schlossman SF: B4, a human B lymphocyte asso-ciated antigen expressed on normal, mitogen-activated, andmalignant B lymphocytes. J Immunol 131: 244, 1983

159. Hooijberg E, Sein JJ, van den Berk PC, Hart AA, van derValk MA, Kast WM, Melief CJ, Hekman A: Erradication oflarge human B cell tumors in nude mice with unconjugatedCD20 monoclonal antibodies and interleukin 2. Cancer Res55: 2627–2634, 1995

160. Hooijberg E, van den Berk PC, Sein JJ, Wijdenes J, Hart AA,de Boer RW, Melief CJ, Hekman A: Enhanced antitumor ef-fects of CD20 over CD19 monoclonal antibodies in a nudemouse xenograft model. Cancer Res 55: 840–846, 1995

161. Lambert JM, Goldmacher VS, Collinson AR, Nadler LM,Blattler WA: An immunotoxin prepared with blocked ricin:a natural plant toxin adapted for therapeutic use. Cancer Res51: 6236–6242, 1991

162. Shah SA, Halloran PM, Ferris CA, Levine BA, Bourret LA,Goldmacher VS, Blattler WA: Anti-B4-blocked ricin im-munotoxin shows therapeutic efficacy in four different SCIDmouse tumor models. Cancer Res 53: 1360–1367, 1993

163. Grossbard ML, Lambert JM, Goldmacher VS, Spector NL,Kinsella J, Eliseo L, Coral F, Taylor JA, Blattler WA, EpsteinCL, Nadler LM: Anti-B4-blocked ricin: a phase I trial of 7-day continuous infusion in patients with B-cell neoplasms. JClin Oncol 11: 726–737, 1993

164. Grossbard ML, Freedman AS, Ritz J, Coral F, GoldmacherVS, Eliseo L, Spector N, Dear K, Lambert JM, Blattler WA,Taylor JA, Nadler LM: Serotherapy of B-cell neoplasms withanti-B4-blocked ricin: a phase I trial of daily bolus infusion.Blood 79: 576–585, 1992

165. Szatrowski TP, Larson RA, Dodge R, Sklar J, Reynolds C,Westbrook CA, Hurd D, Kolitz J, Velez-Garcia E, FrankelSR, Stewart C, Bloomfield CD, Schiffer CA: The effect ofanti-B4-blocked ricin (anti-B4-BR) on minimal residual dis-ease in adults with B-lineage acute lymphoblastic leukemia.Blood 88 (Suppl 1): 669a (Abs # 2665), 1996

166. O’Connor R, Liu C, Ferris CA, Guild BC, Teicher BA, CorviC, Liu Y, Arceci RJ, Goldmacher VS, Lambert JM, BlattlerWA: Anti-B4-blocked ricin synergizes with doxorubicin andetoposide on multidrug-resistant and drug-sensitive tumors.Blood 86: 4286–4294, 1995

167. Liu C, Lambert JM, Teicher BA, Blattler WA, O’ConnorR: Cure of multidrug-resistant human B-cell lymphomaxenografts by combinations of anti-B4-blocked ricin andchemotherapeutic drugs. Blood 87: 3892–3898, 1996

168. Roy DC, Perreault C, Belanger R, Gyger M, Le Houillier C,Blattler WA, Lambert JM, Ritz J: Elimination of B-lineageleukemia and lymphoma cells from bone marrow grafts usinganti-B4-blocked-ricin immunotoxin. J Clin Immunol 15: 51–57, 1995

169. Stashenko P, Nadler LM, Hardy R, Schlossman SF: Char-acterization of a human B lymphocyte-specific antigen. JImmunol 125: 1678–1685, 1980

170. Anderson KC, Bates MP, Slaughenhoupt BL, Pinkus GS,Schlossman SF, Nadler LM: Expression of human B cell-associated antigens on leukemias and lymphomas: a modelof human B cell differentiation. Blood 63: 1424–1433, 1984

171. Kaminski MS, Zasadny KR, Francis IR, Milik AW, Ross CW,Moon SD, Crawford SM, Burgess JM, Petry NA, ButchkoGM, Glenn SD: Radioimmunotherapy of B-cell lymphoma

with 131I-anti-B1 (anti-CD20) antibody. N Engl J Med 329:459–465, 1993

172. Press OW, Eary JF, Appelbaum FR, Martin PJ, Badger CC,Nelp WB, Glenn S, Butchko G, Fisher D, Porter B, MatthewsDC, Fisher LD, Bernstein ID: Radiolabeled-antibody therapyof B-cell lymphoma with autologous bone marrow support.N Engl J Med 329: 1219–1224, 1993

173. Reff ME, Carner K, Chambers KS, Chinn PC, Leonard JE,Raab R, Newman RA, Hanna N, Anderson DR: Depletionof B cells in vivo by a chimeric mouse human monoclonalantibody to CD20. Blood 83: 435–445, 1994

174. Maloney DG, Liles TM, Czerwinski DK, Waldichuk C,Rosenberg J, Grillo-Lopez A, Levy R: Phase I clinicaltrial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody 9IDEC-C2B8) in patients withrecurrent B-cell lymphoma. Blood 84: 2457–2466, 1994

175. Maloney DG, Grillo-Lopez AJ, Bodkin D, White C, Foon K,Schilder RJ, Neidhart J, Janakiraman N, Waldichuk C, DavisT, Dallaire BK, Royston I, Levy R: IDEC-C2B8 anti-CD20antibody: results of long-term follow-up of relapsed NHLphase II trial patients. Blood 86 (Suppl 1): 54a (Abs # 205),1995

176. McLaughlin P, Grillo-Lopez AJ, Link BK, Levy R, Czucz-man M, Heyman MR, Williams M, Heyman MR, Bence-Bruckler I, White CA, Cabanillas F, Jain V, Ho AD, Lister J,Wey K, Shen D, Dallaire BK: Rituximab chimeric anti-CD20monoclonal. J Clin Oncol 16: 2825–2833, 1998

177. Czuczman MS, Grillo-Lopez AJ, White CA, Saleh M, Gor-don L, LoBuglio AF, Jonas C, Klippenstein D, Dallaire B,Varns C: Treatment of patients with low-grade B-cell lymph-oma with the combination of chimeric anti-CD20 mono-clonal antibody and CHOP chemotherapy. J Clin Oncol 17:268–276, 1999

178. O’Brien S, Freireich E, Andreeff M, Lerner S, Keating M:Phase I/II study of Rituxan in chronic lymphocytic leukemia.Blood 92 (Suppl 1): 105a (Abs # 431), 1998

179. Coiffier B, Haioun C, Ketterer N, Engert A, Tilly H, Ma D,Johnson P, Lister A, Feuring-Buske M, Radford JA, Capdev-ille R, Diehl V, Reyes F: Rituximab (Anti-CD20 monoclonalantibody) for the treatment of patients with relapsing or re-fractory aggressive lymphoma: a multicenter phase II study.Blood 92: 1927–1932, 1998

180. List AF: Role of multidrug resistance and its pharmacolo-gical modulation in acute myeloid leukemia. Leukemia 10:937–942, 1996

181. Chen CJ, Chin JE, Ueda K, Clark DP, Pastan I, GottesmanMM, Roninson IB: Internal duplication and homology withbacterial transport proteins in themdr-1 (P-glycoprotein)gene from multidrug-ressistant human cells. Cell 47: 381,1986

182. Campos L, Guyotat D, Archibaud E, Calmard-Oriol P,Tsuruo T, Troncy J, Treille D, Fiere D: Clinical signific-ance of multidrug resistance P-glycoprotein expression onacute nonlymphoblastic leukemia cells at diagnosis. Blood79: 473–476, 1992

183. Pirker R, Wallner J, Geissler K, Linkesch W, Haas OA,Bettelheim P, Hopfner M, Scherrer R, Valent P, Haveloc L,Ludwig H, Lechner K: MDR1 gene expression and treatmentoutcome in acute myeloid leukemia. J Natl Cancer Inst 83:708–712, 1991

184. Ivy SP, Olshefski RS, Taylos BJ, Patel KM, Reaman GH:Correlation of P-glycoprotein expression and function inchildhood acute leukemia: a children’s cancer group study.Blood 88: 309–318, 1996

79

185. Guerci A, Merlin JL, Missoum N, Felmann L, Marchal S,Witz F, Rose C, Guerci O: Predictive value for treatmentoutcome in acute myeloid leukemia of cellular daunorubicinaccumulation and P-glycoprotein expresion simultaneouslydetermined by flow cytometry. Blood 85: 2147–2153, 1995

186. Miller TP, Grogan TM, Dalton WS, Spier CM, Scheper RJ,Salmon SE: P-glycoprotein expression in malignant lymph-oma and reversal of clinical drug resistance with chemo-therapy plus high-dose verapamil. J Clin Oncol 9: 17–24,1991

187. Haussermann K, Benz B, Gekeler V, Schumacher K, Eichel-baum M: Effects of verapamil enantiomers and major meta-bolites on the cytotoxicity of vincristine and daunomycin inhuman lymphoma cell lines. Eur J Clin Pharmacol 40: 53–59,1991

188. Ahmed JH, Godden J, Meredith PA, Elliott HL: R-verapamil:pharmacokinetics and effects of PR interval, blood pressureand heart rate. Br J Clin Pharmacol 36: 93–98, 1993

189. Pirker R, Zochbauer S, Kupper H, Lassmann A, Gsur A,Frass M, Knobl P, Lechner K: Dexverapamil as resistancemodifier in acute myeloid leukemia. Leukemia 9: 539, 1995

190. Solary E, Caillot D, Chauffert B, Casasnovas RO, Dumas M,Maynadie M: Feasibility of using quinine, a potential mul-tidrug resistance-reversing agent, in combination with mitox-antrone and cytarabine for the treatment of acute leukemia. JClin Oncol 10: 1730–1736, 1992

191. Solary E, Witz B, Caillot D, Moreau P, Desablens B, CahnJY, Maloisel F, Sadoun A, Pignon B, Guyotat D, Casas-sus P, Ifrah N, Audhuy B, Berthou C, Delain M, Lamy T,Harousseau JL: Prospectic multicentric randomized study ofquinine as a potential MDR-reversing agent for the treatmentof acute leukemias. Blood 86 (Suppl 1): 780a (Abs # 3109),1995

192. Marie JP, Bastie JN, Coloma F, Suberville AMF, Delmer A,Rio B, Delmas-Marsalet B, Leroux G, Casassus P, BaumelouE, Catalin J, Zittoun R: Cyclosporin A, as a modifier agentin the salvage treatment of cute leukemia. Leukemia 7: 821–824, 1993

193. List AF, Spier C, Greer J, Wolff S, Hutter J, Dorr R, Sal-mon S, Futscher B, Baier M, Dalton W: Phase I/II trial ofcyclosporine as a chemotherapy-resistance modifier in acuteleukemia. J Clin Oncol 11: 1652–1660, 1993

194. List AF, Kopecky KJ, Willman CL, Spier C, Dorr R, Fla-herty L, Hynes H, Appelbaum F: Benefit of cyclosporinemodulation of anthracycline resistance in high-risk AML: aSouthwest Oncology Group study. Blood 92 (Suppl 1): 312a(Abs #1281), 1998

195. Twentyman PR, Bleehen NM: Resistance modification byPSC-833, a novel non-immunosuppressive cyclosporin A.Eur J Cancer 27: 1639–1642, 1991

196. Jiang XR, Kelsey SM, Wu YL, Newland AC: Circumven-tion of P-glycoprotein-mediated drug resistance in humanleukemic cells by non-immunosupressive cyclosporin D ana-logue, SDZ PSC 833. Br J Hematol 90: 375–383, 1995

197. Boote DJ, Dennis IF, Twentyman PR, Osborne RJ, LaburteC, Hensel S, Smyth JF, Brampton MH, Bleehen NM: PhaseI study of etoposide with SDZ PSC 833 as a modulator ofmultidrug resistance in patients with cancer. J Clin Oncol:610–618, 1996

198. Gonzalez O, Colombo T, DeFusco M, Imperatori I, Zuc-chetti M, D’Incalci M: Changes in doxorubicin distributionand toxicity in mice pretreated with the cyclosporin analogueSDZ PSC 833. Cancer Chemother Pharmacol 36: 335–340,1995

199. Keller RP, Altermatt HJ, Nooter K, Poschmann G, Lais-sue JA, Bollinger P, Hiestand PC: SDZ PSC 833, a non-immunosuppresive cyclosporine: its potency in overcom-ing P-glycoprotein-mediated multidrug resistance of murineleukemia. Int J Cancer 50: 593–597, 1992

200. Bailly JD, Muller C, Jaffrezou JP, Demur C, Gassar G,Bordier C, Laurent G: Lack of correlation between ex-pression and function of P-glycoprotein in acute myeloidleukemia cell lines. Leukemia 9: 799–807, 1995

201. Colombo T, Gonzalez Paz O, D’Incalci M: Distribution andactivity of doxorubicin combined with SDZ PSC 833 in micewith P388 and P388/DOX leukemia. Br J Cancer 73: 866–871, 1996

202. Abbaszadegan MR, Futscher BW, Klimecki WT, List A,Dalton WS: Analysis of multidrug resistance-associated pro-tein (MRP) messenger RNA in normal and malignant hema-topoietic cells. Cancer Res 54: 4676–4679, 1994

203. Schneider E, Cowan KH, Bader H, Toomey S, Scwartz GN,Karp JE, Kaufmann SH: Increased expression of the mul-tidmg resistance-associated protein gene in relapsed acuteleukemia. Blood 85: 186–193, 1995

204. Slovak ML, Ho JP, Bhardwaj G, Kurz EU, Deeley RG, ColeSPC: Localization of a novel multidrug resistance-associatedgene in the HT1080/DR4 and H69AR human tumor celllines. Cancer Res 53: 3221–3225, 1993

205. Kuss BJ, Deeley RG, Cole SP, Willman CL, Kopecky KJ,Wolman SR, Eyre HJ, Lane SA, Nancarrow JK, WhitmoreSA, Callen DF: Deletion of gene for multidrug resistance inacute myeloid leukemia with inversion in chromosome 16:prognostic implications. Lancet 343: 1531–1534, 1994

206. List AF, Spier CS, Grogan TM, Johnson C, Roe DJ, Greer JP,Wolff SN, Broxterman HJ, Scheffer GL, Scheper RJ, DaltonWS: Overexpression of the major vault transporter proteinlung-resistance protein predicts treatment outcome in acutemyelogenous leukemia. Blood 87: 2464–2469, 1996

207. Scheffer GL, Wijnagaard PLJ, Flens MJ, Izquierdo MA,Slovak ML, Pinedo HM, Meijer CJLM, Clevers HC, ScheperRJ: The drug resistance related protein LRP is a major vaultprotein. Nat Med 1: 578–582, 1995

208. Leier I, Jedlitschky G, Buchholz U, Cole SP, Deeley RG,Keppler D: The MRP gene encodes an ATP-dependent exportpump for leukotriene C4 and structurally related conjugates.J Biol Chem 269: 27807–27810, 1994

209. Sceper RJ, Broxterman HJ, Scheffer GL, Kaaijk P, DaltonWS, van Heijningen TH, van Kalken CK, Slovak ML, deVries EG, van der Valk P: Overexpression of a M(r) 110,000vesicular protein in non-P-glycoprotein-mediated multidrugresistance. Cancer Res 53: 1475–1479, 1993

210. Carter A, Silvian-Draxler I, Tatarsky I: Effect of interleukin-1, tumor necrosis factor-alpha, and interferon-alpha on theblast cells of acute myeloblastic leukemia. Am J Hematol 40:245–251, 1992

211. Gallagher RE, Lurie KJ, Leavitt RD, Wiernik PH: Effectsof interferon and retinoic acid on the growth and differen-tiation of clonogenic leukemic cells from acute myelogenousleukemia patients treated with recombinant leukocyte-αAinterferon. Leukemia Res 11: 609–619, 1987

212. Sissolak G, Hoffbrand AV, Mehta AB, Ganeshaguru K:Effects of interferon-alpha (IFN) on the expression of in-terleukin 1-beta (IL-1), interleukin 6 (IL-6), granulocyte-macrophage colony-stimulating factor (GM-CSF) and tu-mor necrosis factor-alpha (TNF) in acute myeloid leukemia(AML) blasts. Leukemia 6: 1155–1160, 1992

80

213. Brune M, Hansson M, Mellqvist UH, Hermodsson S, Hell-strand K: NK cell-mediated killing of AML blasts: roleof histamine, monocytes and reactive oxygen metabolites.European J Hematol 57: 312–319, 1996

214. Bezwoda WR, Seymour L, Mansoor N, Dansey R, Davidge-Pitts M, Dreosti L: Interferon alpha enhances the cytotoxicityof cytosine arabinoside in HL60 cells by increasing apop-tosis and is synergistic with cytosine arabinoside in clinicalcombination treatment regimens. Proc ASCO 14: 338 (Abs#1013), 1995

215. Palmieri G, Morabito A, Rea A, Caraglia M, Tagliaferri P, Bi-anco AR: Cytosine arabinoside (ara-C) plus alpha-interferon(alpha IFN) determine prolonged complete remissions inpatients with agressive non-Hodgkin’s lymphoma partiallyresponsive to first-line doxorubicin-containing regimens. BrJ Hematol 88: 421–423, 1994

216. Venditti A, Scimo MT, del Poeta G, Buccisano F, Stasi R,Mastino A, Grelli S, Favalli C, Garaci E, Papa G: Recom-binant interferon alpha 2a, thymopentin and low doses ofcytosine arabinoside for the treatment of myelodysplasti syn-dromes: a pilot study. Leukemia Lymphoma 16: 335–342,1995

217. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA:Lymphokine-activated killer cell phenomenon. Lysis of nat-ural killer-resistant fresh solid tumor cells by interleukin2-activated autologous human peripheral blood lymphocytes.J Exp Med 155: 1823–1841, 1982

218. Thompson JA, Peace DJ, Klamet JP, Kern DE, GreenbergPD, Cheever MA: Eradication of disseminated murine leuk-emia by treatment with high-dose interleukin-2. J Immunol137: 3675–3680, 1986

219. Johnson CS, Thurlow SM, Furmanski P: Lymphokine-activated killer cell plus recombinant interleukin-2 therapyof erythroleukemia in mice. Leukemia 3: 91–96, 1989

220. Oshimi K, Oshimi Y, Akutsu M, Takei Y, Saito H, Okada M,Mizoguchi H: Cytotoxicity of interleukin 2 activated lymph-ocytes for leukemia and lymphoma cells. Blood 68: 938–948,1986

221. Fierro MT, Xin-Sheng L, Lusso P, Bonferroni M, Matera L,Cesano A, Lista P, Arione R, Forni G, Foa R:In vitro andin vivo susceptibility of human leukemic cells to lymphokineactivated killer activity. Leukemia 2: 50–54, 1988

222. Komada Y, Zhou Y-W, Zhang X-L, Chen T-X, Tanaka S,Azuma E, Sakurai M: Fas/APO-1 (CD95)-mediated cytotox-icity is responsible for the apoptotic cell death of leukemiccells induced by interleukin-2-activated T cells. Br J Hematol96: 147–157, 1997

223. Klingemann HG, Phillips GL: Is there a place for immun-otherapy with interleukin-2 to prevent relapse after autolog-ous stem cell transplantation for acute leukemia? LeukemiaLymphoma 16: 397–405, 1995

224. Foa R, Meloni G, Tosti S, Novarino A, Fenu S, Gavosto F,Mandelli F: Treatment of acute myeloid leukemia patientswith recombinant interleukin 2: a pilot study. Br J Hematol77: 491–496, 1991

225. Lim SH, Newland AC, Kelsey S, Bell A, Offerman E,Rist C, Gozzard D, Bareford D, Smith MP, Goldstone AH:Continuous intravenous infusion of high-dose recombinantinterleukin-2 for acute myeloid leukemia: a phase II study.Cancer Immunol Immunother 34: 337–342, 1992

226. Meloni G, Foa R, Vignetti M, Guarini A, Fenu S, Tosti S, TosAG, Mandelli F: Interleukin-2 may induce prolonged remis-sions in advanced acute myelogenous leukemia. Blood 84:2158–2163, 1994

227. Wiernik PH, Dutcher JP, Todd M, Caliendo G, Benson L:Polyethylene glycolated interleukin-2 as maintenance ther-apy for acute myelogenous leukemia in second remission.Am J Hematol 47: 41–44, 1994

228. Bergmann L, Heil G, Kolbe K, Lengfelder E, Puzicha E,Martin H, Lohmeyer J, Mitrou PS, Hoelzer D: Interleukin-2bolus infusion as late consolidation therapy in second remis-sion of acute myeloblastic leukemia. Leukemia Lymphoma16: 271–279, 1995

229. Cortes JE, Kantarjian HM, O’Brien S, Giles F, Keating MJ,Freireich EJ, Estey EH: A pilot study of interleukin-2 forpatients with acute myelogenous leukemia in first completeremission. Cancer 85: 1506–1513, 1999

230. Brune M, Hellstrand K: Remission maintenance therapy withhistamine and interleukin-2 in acute myelogenous leukemia.Br J Hematol 92: 620–626, 1996

231. Ogata K, Yokose N, Nomura T: Interleukin-2 therapy for my-elodysplastic syndrome: dose it work? Leukemia Lymphoma17: 411–415, 1995

232. Toze CL, Barnett MJ, Klingemann HG: Response of therapy-related myelodysplasia to low-dose interleukin-2. Leukemia7: 463–465, 1993

233. Lauria F, Raspadori D, Rondelli D, Ventura MA, Foa R:Invitro susceptibility of acute leukemia cells to the cytotoxicactivity of allogeneic and autologous lymphokine activatedkiller (LAK) effecters: correlation with the rate and durationof complete remission and with survival. Leukemia 8: 724–728, 1994

234. Cignetti A, Guarini A, Carbone A, Forni M, Cronin K, ForniG, Gansbacher B, Foa R: Transduction of the IL2 geneinto human acute leukemia cells: induction of tumor rejec-tion without modifying cell proliferation and IL2 receptorexpression. J Natl Cancer Inst 86: 785–791, 1994

235. Foa R: Interleukin 2 in the management of acute leukemia.Br J Hematol 92: 1–8, 1996

236. Kalland T: Effects of the immunomodulator LS 2616 ongrowth and metastasis of the murine B16-F10 melanoma.Cancer Res 46: 3018–3022, 1986

237. Kalland T: Regulation of natural killer progenitors. Studieswith a novel immunomodulator with distinct effects at theprecursor level. J Immunol 144: 4472–4476, 1990

238. Vaz F, Silva MR, Ascensao JL: Enhanced lymphokine-activated killer cell activity by an immunomodulator,Roquinimex. Br J Cancer 72: 1498–1503, 1995

239. Stalhandske T, Kalland T: Effect of the novel immunomodu-lator LS 2616 on the delayed-type hypersensitivity reactionto Bordetella pertussis in the rat. Immunopharmacol 11:87–92, 1986

240. Larson EL, Joki A, Stalhandske T: Mechanism of action ofthe immunomodulator LS 2616 on T cell response. Int JImmunopharmacol 9: 425–431, 1987

241. Rowe JM, Nilsson BI, Simonsson B: Treatment of min-imal residual disease in myeloid leukemia – The immun-otherapeutic options with emphasis on linomide. LeukemiaLymphoma 11: 321–329, 1993

242. Candiotti R, Slavin S, Barak V, Nagler A: The novel im-munomodulator, linomide, stimulates interleukin-2-inducedhuman natural killer (NK) cell and PHA-stimulated T cellproliferation from normal donors. Leukemia Res 20: 57–63,1996

243. Bergh JCS, Totterman TH, Termander BC, StrandgardenKAMP, Gunnarsson POG, Nilsson BI: The first clinical pilotstudy of roquinimex (Linomider ) in cancer patients with

81

special focus on immunological effects. Cancer Invest 15:204–211, 1997

244. Rowe J, Ryan D, Dipersio J, Gaspari A, Nilsson B, LarssonL, Liesveld J, Kouides P, Simonsson B: Autografting inchronic myelogenous leukemia followed by immunotherapy.Stem Cell 11 (Suppl 3): 34–42, 1993

245. Gaspari AA, Cheng SF, DiPersio JF, Rowe JM; Roquinimex-induced graft-versus-host reaction after autologous bonemarrow transplantation. J Am Acad Derm 33: 711–717, 1995

246. Bengtsson M, Simonsson B, Carlsson K, Nilsson B,Smedmyr B, Termander B, Oberg G, Totterman TH: Stimula-tion of NK cell, T cell, and monocyte functions by the novelimmunomodulator Linomide after autologous bone marrowtransplantation. Transplantation 53: 882–888, 1992

247. Rosenfeld CS, Zeigler ZR, Shadduck RK, Nilsson BO: PhaseII study of Roquinimex in myelodysplastic syndrome. Am JClin Oncol 20: 189–192, 1997

248. Guinan EC, Gribben JG, Boussiotis VA, Freeman GJ, NadlerLM: Pivotal role of the B7:CD28 pathway in transplantationtolerance and tumor immunity. Blood 84: 3261–3282, 1994

249. Dunussi-Joannopoulos K, Weinstein HJ, Nickerson PW,Strom TB, Burakoff SJ, Croop JM, Arceci RJ: Irradiated B7-1 transduced primary acute myelogenous leukemia (AML)Gell can be used as therapeutic vaccines in murine AML.Blood 87: 2938–2946, 1996

250. Dunussi-Joannopoulos K, Krenger W, Weinstein HJ, FerraraJL, Croop JM: CD8+ T cells activated during the courseof murine acute myelogenous leukemia elicit therapeutic re-sponses to late B7 vaccines after cytoreductive treatment.Blood 89: 2915–2924, 1997

251. Dunussi-Joannopoulos K, Dranoff G, Weinstein HJ, Fer-rara JLM, Bierer BE, Croop JM: Gene immunotherapy inmurine acute myeloid leukemia: granulocyte-macrophagecolonystimulating factor tumor cell vaccines elicit more po-tent anti-tumor immunity compared with B7 family and othercytokine vaccines. Blood 91: 222–230, 1998

252. Bocchia M, Wentworth PA, Southwood S, Sidney J, McGrawK, Scheinberg DA, Sette A: Specific binding of leukemiaoncogene fusion protein peptides to HLA class 1 molecules.Blood 85: 2680–2684, 1995

253. Dengler R, Munstermann U, Al-Batran S, Hausner I, FaderlS, Nerl C, Emmercih B: Immunocytochemical and flow cyto-metric detection of proteinase 3 (myeloblastin) in normal andleukemic myeloid cells. Br J Haematol 89: 250, 1995

254. Bories D, Raynal MC, Solomon DH, Darzynkiewicz Z,Cayre YE: Down-regulation of a serine protease, myelo-blastin, causes growth arrest and differentiation of promye-locytic leukemia cells. Cell 59: 959, 1989

255. Molldrem J, Dermime S, Parker K, Jiang YZ, Mavroudis D,Hensel N, Fukushima P, Barrett AJ: Targeted T-cell therapyfor human leukemia: cytotoxic T lymphocytes specific forpeptide derived from proteinase 3 preferentially lyse humanmyeloid leukemia cells. Blood 88: 2450–2457, 1996

256. Molldrem JJ, Clave E, Jiang YZ, Mavroudis D, Raptis A,Hensel N, Agarwala V, Barrett AJ: Cytotoxic T lymphocytesspecific for a nonpolymorphic proteinase 3 peptide preferen-tially inhibit chronic myeloid leukemia colony-forming units.Blood 90: 2529–2534, 1997

257. Banchereau J, Steinman RM: Dendritic cells and the controlof immunity. Nature 392: 245–252, 1998

258. Choudhury A, Gajewski JL, Liang JC, Popat U, Claxton DF,Kliche KO, Andreeff M, Champlin RE: Use of leukemicdendritic cell for the generation of antileukemic cellular cyto-

toxicity against Philadelphia chromosome-positive chronicmyelogenous leukemia. Blood 89: 1133–1142, 1997

259. Choudhury A, Liang JC, Thomas EK, Flores-Romo L, XieQS, Agusala K, Sutaria S, Sinha I, Champlin RE, ClaxtonDF: Dendritic cells derivedin vitro from acute myelogen-ous leukemia cells stimulate autologous, antileukemic T-cellresponses. Blood 93: 780–786, 1999

260. Lishner M, Curtis JE, Minkin S, McCulloch EA: Interactionbetween retinoic acid and cytosine arabinoside affecting theblast cells of acute myeloblastic leukemia. Leukemia 3: 78,1989

261. Yang GS, Minden MD, McCulloch EA: Regulation by retin-oic acid and hydrocortisone of the anthracycline sensitivityof blast cells of acute myeloblastic leukemia. Leukemia 8:2065–2075, 1994

262. Hu Z-B, Minden MD, McCulloch EA: Direct evidence forthe participation of bcl-2 in the regulation by retinoic acidof the ara-C sensitivity of leukemic stem cells. Leukemia 9:1667, 1995

263. Bradbury DA, Aldington S, Zhu YM, Russell NH: Down-regulation of bcl-2 in AML blasts by all-trans retinoicacid and its relationship to CD34 antigen expression. Br JHaematol 94: 671–675, 1996

264. Hu Z-B, Minden MD, McCulloch EA: Regulation of the syn-thesis of bcl-2 protein by growth factors. Leukemia 10: 1925,1996

265. Estey EH, Thall PF, Pierce S, Cortes J, Beran M, KantarjianH, Keating MJ, Andreeff M, Freireich E: Randomized phaseII study of fludarabine + cytosine arabinoside + idarubicin± all-trans retinoic acid± granulocyte colony-stimulatingFactor in poor prognosis newly diagnosed acute myelo-genous leukemia and myelodysplastic syndrome. Blood 93:2478–2484, 1999

266. Griggs JJ, Henley SE, Rowe JM: Treatment of refractoryundifferentiated acute myelogenous leukemia with all-transretinoic acid. Am J Hematol 45: 177–180, 1994

267. Venditti A, Stasi R, Del Poeta G, Buccisano F, Aronica G,Bruno A, Pisani F, Caravita T, Masi M, Tribalto M: All-transretinoic acid and low-dose cytosine arabinoside for the treat-ment of “poor prognosis” acute myeloid leukemia. Leukemia9: 1121–1125, 1995

268. Sakashita A, Kizaki M, Pakkala S, Schiller G, Tsuruoka N,Tomosaki R, Cameron JF, Dawson MI, Koeffler HP: 9-cis-retinoic acid: effects on normal and leukemic hematopoiesisin vitro. Blood 81: 1009–1016, 1993

269. Shen Z-X, Chen G-Q, Ni J-H, Li X-S, Xiong S-M, Qiu Q-Y,Zhu J, Tang W, Sun G-L, Yang K-Q, Chen Y, Zhou L, FangZ-W, Wang Y-T, Ma J, Zhang P, Zhang T-D, Chen S-J, ChenS-J, Chen Z, Wang Z-Y: Use of arsenic trioxide (As2O3)in the treatment of acute promyelocytic leukemia (APL): II.Clinical efficacy and pharmacokinetics in relapsed patients.Blood 89: 3354–3360, 1997

270. Soignet SL, Maslak P, Wang Z-G, Jhanwar S, Calleja E,Dardashti LJ, Corso D, DeBlasio A, Gabrilove J, ScheinbergDA, Pandolfi PP, Warrell RP: Complete remission after treat-ment of acute promyelocytic leukemia with arsenic trioxide.N Engl J Med 339: 1341–1348, 1998

271. Chen G-Q, Zhu J, Shi X-G, Ni J-H, Zhong H-J, Si G-Y, JinX-L, Tang W, Li X-S, Xong S-M, Shen Z-X, Sun G-L, Ma J,Zhang P, Zhang T-D, Gazin C, Naoe T, Chen S-J, Wang Z-Y,Chen Z:In vitro studies on cellular and molecular mechan-isms of arsenic trioxide (As2O3) in the treatment of acutepromyelocytic leukemia: As2O3 induces NB4 cell apoptosis

82

with downregulation of Bcl-2 expression and modulation ofPML-RARα/PML proteins. Blood 88: 1052–1061, 1996

272. Shao W, Fanelli M, Ferrara F, Riccioni R, Rosenauer A,Davison K, Lamph WW, Waxman S, Pelicci PG, LoCocoF, Avvisati G, Testa U, Peschle C, Gambacorti-PasseriniC, Nervi C, Miller WH Jr: Arsenic trioxide as an inducerof apoptosis and loss of PML/RAR alpha protein in acutepromyelocytic leukemia cells. J Natl Cancer Inst 90: 86–88,1998

273. Wang Z-G, Rivi R, Delva L, Konig A, Scheinberg DA,Gambacorti-Passerini C, Gabrilove JL, Warrell RP, PandolfiPP: Aresenic trioxide and melarsoprol induce programmedcell death in myeloid leukemia cell lines and function ina PML and PML-RARα independent manner. Blood 92:1497–1504, 1998

274. Calleja EM, Gabrilove JL, Warrell RP: Arsenic trioxide in-hibits cell survival in a variety of cancer cell types. Proc AmSoc Clin Oncol 17: 218a (Abs #844), 1998

275. Yang CH, Wang TY, Chen YC: Cytotoxicity of arsenic triox-ide in cancer cell lines. Proc Am Assoc Cancer Res 39: 227(Abs), 1998

276. Carroll M, Ohno-Jones S, Tamura S, Buchdunger E, Zimmer-mann J, Lydon NB, Gilliland DG, Druker BJ: CGP 57148, atyrosine kinase inhibitor, inhibits the growth of cells express-ing BCR-ABL, TEL-ABL, and TEL-PDGFR fusion proteins.Blood 90: 4947–4952, 1997

277. Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM,Fanning S, Zimmermann J, Lydon NB: Effects of a selectiveinhibitor of the Abl tyrosine kinase on the growth of Bcr-Ablpositive cells. Nature Medicine 2: 561–566, 1996

278. Beran M, Cao X, Estrov Z, Jeha S, Jin G, O’Brien S, TalpazM, Arlinghaus RB, Lydon NB, Kantarjian H: Selective in-hibition of cell proliferation and BCR-ABL phosphoryla-tion in acute lymphoblastic leukemia cells expressing Mr190,000 BCR-ABL protein by a tyrosine kinase inhibitor(CGP-57148). Clinical Cancer Research 4: 1661–1672, 1998

279. Druker BJ, Sawyers CL, Talpaz M, Resta D, Peng B, FordJ: Phase I trial of a specific ABL tyrosine kinase inhibitor,CGP 57148, in interferon refractory chronic myelogenousleukemia patients. Proc ASCO 18: 7a (Abs), 1999

280. Ahuja HG, Foti A, Bareli M, et al.: The pattern of mutationalinvolvement of ras genes in human hematologic malignan-cies determined by DNA amplification and direct sequen-cing. Blood 75: 1684–1690, 1990

281. Janssen JWG, Steenvoorden ACM, Lyons J, et al.: Rasgene mutations in acute and chronic myelocytic leukemias,chronic myeloproliferative disorders, and myelodysplasticsyndromes. Proc Natl Acad Sci USA 84: 9228–9232, 1987

282. Tamanoi F: Inhibitors of ras farnesyltransferase. Trends Bio-chem Sci 18: 349-353, 1993

283. Geller RB: Use of cytokines in the treatment of acutemyelocytic leukemia: A critical review. J Clin Oncol 14:1371–1382, 1996

284. Armitage JO: Emerging applications of recombinant humangranulocyte-macrophage colony-stimulating factor. Blood92: 4491–4508, 1998

Address for offprints:Jorge Cortes, Leukemia Department, Box 61,M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX77030; Fax: 215 728 3639