Next Article in Journal
Physical Activity Associated with Public Transport Use—A Review and Modelling of Potential Benefits
Next Article in Special Issue
Using Bioinformatic Approaches to Identify Pathways Targeted by Human Leukemogens
Previous Article in Journal
First Report of a Toxic Nodularia spumigena (Nostocales/ Cyanobacteria) Bloom in Sub-Tropical Australia. II. Bioaccumulation of Nodularin in Isolated Populations of Mullet (Mugilidae)
Previous Article in Special Issue
Mechanism of Generation of Therapy Related Leukemia in Response to Anti-Topoisomerase II Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Secondary Leukemia Associated with the Anti-Cancer Agent, Etoposide, a Topoisomerase II Inhibitor

Hematology and Oncology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka Suita, Osaka 565-0871, Japan
Int. J. Environ. Res. Public Health 2012, 9(7), 2444-2453; https://doi.org/10.3390/ijerph9072444
Submission received: 16 May 2012 / Revised: 27 June 2012 / Accepted: 28 June 2012 / Published: 10 July 2012
(This article belongs to the Special Issue Leukemia Arising from Chemical Exposures and Chemotherapeutic Drugs)

Abstract

:
Etoposide is an anticancer agent, which is successfully and extensively used in treatments for various types of cancers in children and adults. However, due to the increases in survival and overall cure rate of cancer patients, interest has arisen on the potential risk of this agent for therapy-related secondary leukemia. Topoisomerase II inhibitors, including etoposide and teniposide, frequently cause rearrangements involving the mixed lineage leukemia (MLL) gene on chromosome 11q23, which is associated with secondary leukemia. The prognosis is extremely poor for leukemias associated with rearrangements in the MLL gene, including etoposide-related secondary leukemias. It is of great importance to gain precise knowledge of the clinical aspects of these diseases and the mechanism underlying the leukemogenesis induced by this agent to ensure correct assessments of current and future therapy strategies. Here, I will review current knowledge regarding the clinical aspects of etoposide-related secondary leukemia, some probable mechanisms, and strategies for treating etoposide-induced leukemia.

1. Introduction

Drugs that induce secondary leukemias comprise alkylating agents, like cyclophosphamide, topoisomerase II inhibitors, including epipodophylotoxins (etoposide, teniposide), and anthracyclinic agents, like doxorubicin.
Etoposide (VP-16, 4′-dimethylepipodophylloxin-9-[4,6-O-ethylidene-beta-D-glucopyranoside]) is an agent that targets DNA topoisomerase II. It has been widely used for the treatment of many types of cancers in children and adults [1]. However, etoposide has been associated with an increased risk of secondary leukemia. In 1987, Ratain et al. first propounded the relationship between etoposide and secondary leukemia when it was used as a therapy for advanced non-small-cell lung cancer [2]. Subsequently, several investigators reported secondary leukemias, particularly acute myelogenous leukemia (AML), associated with translocations of the mixed lineage leukemia (MLL) gene at human chromosomal band 11q23 when etoposide was used to treat lung cancer, non-Hodgkin lymphoma, neuroblastoma, acute lymphoid leukemia (ALL), Wilms tumor, and rhabdomyosarcoma, [3,4,5]. The mechanism that leads to MLL-specific rearrangement is largely unknown. However, recent studies have reported some speculative mechanisms based on in vitro experiments. In this paper, the findings from previous reports that described the clinical aspects, some probable mechanisms, and treatments for etoposide-related secondary leukemia will be summarized.

2. Aspects of AML after Etoposide Treatments

Alkylating agents cause secondary AML characterized by antecedent myelodysplasia, and the typical latency period for the development of secondary leukemia is about 5–7 years. Often (60–90% of cases), complete or partial deletion of chromosome 5 or 7 occurs, and patients exhibit the M1 or M2 phenotype (French-American-British (FAB) classification; Table 1) [6,7]. The risk of causing secondary myelodysplastic syndrome (MDS) and leukemia associated with alkylating agents mainly depends on the cumulative dose of alkylating agents. Predisposing factors for the alkylating agent-associated secondary leukemias are germline NF-1 [8] and p53 gene mutations [9] and a null GSTT1 genotype [10], and Ras mutations are frequently accompanied with those leukemias [11,12]. On the other hand, epipodophyllotoxins and other DNA topoisomerase II inhibitors are linked to leukemias that bear MLL gene translocations at chromosome bands 11q23, t(8;21), t(3;21), inv(16), t(8;16), t(15;17), or t(9;22) [9]. Generally the leukemias following epipodophyllotoxins-treatments are likely to occur within 3 years after treatments, and the mean latency period from drug administration to the onset of secondary leukemia is about 2 years. Between 2 and 12% of patients that receive epipodophyllotoxin develop secondary AML. Most cases of epipodophyllotoxin-induced leukemia exhibit FAB M4 or M5 morphology, but observations have also included other FAB AML subtypes, MDS, ALL, and chronic myelogenous leukemia (CML). The prognosis of alkylating-agent-induced secondary leukemia appears to be worse than that for spontaneously occurring leukemias, and the prognosis of etoposide-related secondary leukemia is extremely poor. Pui et al. [13] reported that, out of 21 patients with etoposide-related secondary-leukemia, only two could maintain complete remission. Furthermore, Sandler et al. [14] reported that, even with therapy, including bone marrow transplantation, the 2-year disease free survival rate was 17.6% for children’s epipodophyllotoxin-induced secondary leukemia.
However, single-agent therapy may be rather uncommon in cancer treatments. Epipodophyllotoxins are generally used as part of alkylating agents- or cisplatin-based protocls in treatments of solid tumors [15,16], and are given with cytarabine in leukemia treatment [17]. In vitro study, combinations of etoposide with an alkylating agent or sisplatin produce synergistic cytocidal effects on leukemic cell. And In the Intergroup Rhabdomyosarcoma Study III, the risk of secondary AML was significantly higher for children receiving cyclophosphamide and etoposide compared to those receiving cyclophosphamide without etoposide [18]. Thus in assessments of the risk of secondary leukemia following etoposide therapy, we must consider the combination of other cytotoxic agents.
Table 1. Characteristics of secondary leukemia following epipodophyllotoxin or alkylating agent treatment.
Table 1. Characteristics of secondary leukemia following epipodophyllotoxin or alkylating agent treatment.
EpipodophyllotoxinAlkylating agent
Typical interval following therapy2–3 years5–7 years
FAB classificationM4/M5M1/M2
Karyotype abnormalities11q3 (MLL gene rearrangement)(–5)/del(5q), (–7)/del(7q)
Preceding myelodysplasiararefrequent

3. Schedule and Dose of the Drug

The risk of secondary AML appears to be dependent on both treatment schedule and dose. Typically, epipodophyllotoxin-induced AML occurs after multiple doses administered in brief intravenous infusions. The 4- to 5-year cumulative risk of the complication has ranged from 0% to 18.4% in patients treated with cumulative doses ranging from 5,200 mg/m2 to 19,200 mg/m2 [19]. Currently, low-dose, chronically administered regimens are more commonly given to patients with refractory or relapsed cancers; however, it is difficult to assess the risk of drug-induced AML associated with this regimen due to the short survival period. Caution must be exercised when speculating that leukemogenesis might occur less frequently with prolonged oral dosing; nevertheless, almost all cases of epipodophyllotoxin-induced AML have been reported after administration of short intravenous infusions [20]. Moreover, it is clear that factors other than cumulative dosage may predispose patients to developing this devastating adverse effect, like the administration schedule of epipodophyllotoxin (etoposide or teniposide) and other drugs that may be given concurrently.
Chen et al. [21] suggested, based on their in vitro site-specific DNA recombination analysis, that prolonged exposure to low-dose etoposide regimens did not produce less cytotoxicity than high-dose, short-term regimens; however, low-dose regimens induced less site specific, nonhomologous DNA recombination, which suggested that more prolonged dosage schedules might achieve adequate effects with lower risk of leukemogenesis.
Etoposide is used in many treatment regimens for various tumors, including several high-dose chemotherapy regimens. Pedersen-Bjergaard reported that the risk of developing secondary leukemia increased 336-fold with etoposide doses above 2.0 g/m2 compared to doses of 2.0 g/m2 or less [22]. Thereafter, several studies have investigated the possibility of a correlation between the risk of secondary AML and the administered cumulative dose of etoposide [20]. Based on some reports, the probability of secondary leukemia within 5 years after chemotherapy for testicular cancer was about 0.6% with cumulative etoposide doses up to 2.0 g/m2, but 3.4% in regimens with etoposide doses above 2.0 g/m2. This suggested that high-dose etoposide, especially higher than 2.0 g/m2 of cumulative dose, was associated with increased risk for developing secondary leukemia [23,24].
On the other hand, Pui et al. [13] reported that secondary AML was diagnosed in 21 out of 734 patients with newly diagnosed AML that had entered complete remission and were randomly assigned to receive continuation (maintenance) treatment; secondary AML was the initial adverse event in 17 of those patients. In that study, the probability that AML developed within six years was 3.8% for any patient in continuous complete remission; however, in patients that received teniposide or etoposide in weekly or twice-weekly doses for prolonged periods, the risk was approximately 12 times higher than that of patients treated according to other schedules. Nevertheless, they could not determine reliably whether the cumulative dose of epipodophyllotoxins played a major role in the development of AML. The recognition that etoposide is dependent on the cell cycle and proliferation [25] has led to the development of more prolonged dosage schedules. Doses given over a period of several days have shown improved antitumor effects compared to equivalent total doses given as a single, short intravenous infusion [26]. Sugita et al. reported that a B-8801 protocol for B-cell lymphoma, which contained a cumulative etoposide dose of 10,000 mg/m2, was not related to the occurrence of secondary leukemia; they suggested that the dose method was more important than the cumulative dose for leukemogenesis [27].
Furthermore, additional factors influence the risk of etoposide-related secondary leukemia, like concomitant radiotherapy or high doses of other agents, including platinum agents. In patients with refractory germ cell tumors, a treatment option of high-dose chemotherapy, including high-dose etoposide, was not related to a higher incidence of leukemogenesis than standard-dose chemotherapy regimens. However, a combination of high-dose platinum agents, like cisplatin or carboplatin, may promote etoposide-induced leukemogenesis [28].
Based on these data, some regimens have been developed that reduce the risk of secondary leukemia, including reduced doses of etoposide. For example, when reduced dose chemotherapy was given to patients with intermediate-risk neuroblastoma, the 3-year overall survival was greater than 90% and the cumulative incidence of secondary leukemia was 0.7% [29]. In the Children’s Oncology Group therapeutic trial (INT-0091), Ewing sarcoma was treated with Regimen A, which included doxorubicin (375 mg/m2) and cyclophosphamide (20.4 g/m2), and Regimen B, which included doxorubicin (375 mg/m2), cyclophosphamide (9.6 g/m2), ifosfamide (90 g/m2), and etoposide (5 g/m2); this regimen achieved a low rate (<1%) of secondary leukemia [30].

4. The Mechanism of Induction of Secondary Leukemia by Etoposide

Most chromosomal translocation breakpoints in 11q23 are located within an 8.3-kb breakpoint cluster region (BCR), which extends from exon 7 to exon 13 of the MLL gene [31,32]. However, it is largely unknown how etoposide induces 11q23 chromosome translocations in this region. The MLL gene spans 100 kb, encodes a 430-kDa protein homologous to the Drosophila trithorax gene, and has important functions in embryogenesis and hematopoiesis [33,34]. MLL fusion proteins induce high expression of MEIS1 and HOX genes through an epigenetic function, and thus, they inhibit differentiation and induce immortalization of immature hematopoietic cells [35]. Multiple lines of evidence have shown that etoposide-associated genomic alterations coincide with topoisomerase II cleavage activity, particularly within the MLL locus [36,37]. Etoposide addition to in vitro topoisomerase II cleavage assays enhanced DNA-topoisomerase II cleavage complexes within DNA substrates that contained MLL and partner gene sequences near translocation breakpoint sites identified in therapy-related leukemias [38,39]. Similarly, exposure of hematopoietic progenitor cells to etoposide led to detectable DNA-topoisomerase II cleavage complexes within the BCR, and some rearrangements were detectable after 16–24 h of continuous etoposide exposure in many hematopoietic cell lines [37,40] Sensitivity of the MLL locus may be a general consequence of chromatin architecture. MLL is an actively transcribed, open chromatin region throughout early hematopoietic development, and it contains a scaffold-associating region/matrix-associating region (SAR/MAR) that may be a site-specific target for topoisomerase II activity [41,42,43].
DNA damage activates the DNA damage response and repair pathways. Clinical data suggest that the MLL 11q23 locus is particularly susceptible to etoposide-mediated cleavage and subsequent illegitimate repair events that anneal heterologous chromosomes. Alternatively, etoposide may promote cleavage and illegitimate repair along all chromosomes with relatively equal frequency, but the specific oncogenic fusions with MLL lead to a growth advantage, and ultimately tumorigenesis, in a critical hematopoietic stem-cell subpopulation. Libura et al. [44] detected illegitimate repair events, including MLL tandem duplications and translocations (44%), in primary hematopoietic CD34+ cells treated with 20–50 mM etoposide ex vivo. Blanco et al. [45] reported that etoposide also induced MLL translocation in mouse ES cells. Bueno et al. [46] was the first study to use human embryonic stem cells (hESCs) as a model for testing the effects of etoposide in human embryonic development. Continuous exposure to etoposide induced MLL breaks and primed hESCs to acquire other major karyotype abnormalities. This linked embryonic genotoxic exposure to genomic instability. Recently, Vlasova et al. [47] reported that etoposide, which contains a hindered ring phenol, was oxidatively activated by myeloperoxidase (MPO) to a free radical species. These etoposide radicals resulted in redox cycling, which may play a role in enhanced etoposide genotoxicity. MPO expressed in CD34+ myeloid progenitor cells may make these cells particularly sensitive to the leukemogenic action of etoposide.
More recently, Cowell et al. explained the topoisomerase IIb dependent mechanism through which etoposide induces translocations [48]. For the effective repair of the etoposide-induced DNA double-strand breaks (DSBs) a specific machinery, Non-Homologous End-Joining (NHEJ), is required [49,50,51]. The hypersensitive sites for DNase I are also present in the BCRs of other transcription genes including AF9, AF4, RUNX1, ETO, and RARA. Cowell showed, using RNA-FISH analysis, that topoisomerase IIb is corecruited with components of the NHEJ machinery, which induces dynamic close proximity of MLL and t-AML-associated translocation partner genes, such as AF9 and AF4. This recruitment of genes to the shared machinery may contribute to the aberrant repairs in secondary AMLs.
The ataxia-telangiectasia mutation (ATM) was also reported to be involved in chromosome translocations involving 11q23. The ATM protein regulates the DNA damage response to DNA-double-strand breaks through its kinase activity [52]. Sun et al. [53] showed that ATM deficiency resulted in excessive binding of the DNA recombination protein, RAD51, at a translocation breakpoint hotspot on the 11q23 chromosome after etoposide exposure. This suggested that ATM modulated the loading of recombinational repair proteins onto translocational hotspots to avoid inappropriate recombination that could lead to chromosome translocation. Daurinol is a novel catalytic inhibitor of human topoisomerase IIa; its structure is quite similar to etoposide. However, unlike etoposide, it induces S-phase arrest through the ATM/Chk/Cdc25A pathway, without DNA cleavage or severe DNA damage [54]. This agent may represent an advantageous alternative to etoposide, because it may provide lower risk for developing secondary leukemias.

5. Treatments for Etoposide-Induced Leukemia

As pointed out above, patients with etoposide-related secondary leukemias with MLL-rearrangements have a significantly worse prognosis compared to patients with leukemia cells that possess a normal MLL. Currently, intense strategies are being developed, including stem-cell transplantation, to treat MLL-rearranged acute leukemias, but the outcome has remained poor. More recently, the National Cancer Institute/National Institute of Health Developmental Therapeutics Program (NCI/NIH) proposed testing an approved set of drugs designed to combat MLL-rearranged pediatric leukemia cell lines [55]. Results from that study showed that 42 of the 89 agents tested had measurable cytotoxicity against leukemia cells. Among those, 12 agents were effective against all five MLL-rearranged cell lines; these included cladribine, dactinomycin, daunorubicin, docetazel, etoposide, gemcitabine, mitomycin C, mitoxantrone, teniposide, topotecan, triethylnemelamine, and vinblastine. These data are extremely informative, and may facilitate further clinical trials for testing new treatment strategies for leukemia with MLL abnormalities.

6. Conclusions

In conclusion, it is necessary to gain more exact knowledge of secondary leukemia, identify the optimal therapy administration method, including the application schedule and dosage, and perform long-term follow-up studies to advance the prevention and treatment of secondary leukemias.

References

  1. Hande, K.R. Etoposide: Four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 1998, 34, 1514–1521. [Google Scholar]
  2. Ratain, M.J.; Kaminer, L.S.; Bitran, J.D.; Larson, R.A.; Le Beau, M.M.; Skosey, C.; Purl, S.; Hoffman, P.C.; Wade, J.; Vardiman, J.W.; et al. Acute nonlymphocytic leukemia following etoposide and cisplatin combination chemotherapy for advanced non-small-cell carcinoma of the lung. Blood 1987, 70, 1412–1417. [Google Scholar]
  3. Potzsch, C.; Fetscher, S.; Mertelsmann, R.; Lubbert, M. Acute myelomonocytic leukemia secondary to synchronous carcinomas of the breast and lung, and to metachronous renal cell carcinoma. J. Cancer Res. Clin. Oncol. 1997, 123, 678–680. [Google Scholar]
  4. Shearer, P.; Kapoor, G.; Beckwith, J.B.; Takashima, J.; Breslow, N.; Green, D.M. Secondary acute myelogenous leukemia in patients previously treated for childhood renal tumors: A report from the National Wilms Tumor Study Group. J. Pediatr. Hematol. Oncol. 2001, 23, 109–111. [Google Scholar]
  5. Domer, P.H.; Head, D.R.; Renganathan, N.; Raimondi, S.C.; Yang, E.; Atlas, M. Molecular analysis of 13 cases of MLL/11q23 secondary acute leukemia and identification of topoisomerase II consensus-binding sequences near the chromosomal breakpoint of a secondary leukemia with the t(4;11). Leukemia 1995, 9, 1305–1312. [Google Scholar]
  6. Levine, E.G.; Bloomfield, C.D. Leukemias and myelodysplastic syndromes secondary to drug, radiation, and environmental exposure. Semin. Oncol. 1992, 19, 47–84. [Google Scholar]
  7. Kollmannsberger, C.; Hartmann, J.T.; Kanz, L.; Bokemeyer, C. Risk of secondary myeloid leukemia and myelodysplastic syndrome following standard-dose chemotherapy or high-dose chemotherapy with stem cell support in patients with potentially curable malignancies. J. Cancer Res. Clin. Oncol. 1998, 124, 207–214. [Google Scholar]
  8. Kaneko, Y.; Maseki, N.; Sakurai, M.; Shibuya, A.; Shinohara, T.; Fujimoto, T.; Kannno, H.; Nishikawa, A. Chromosome pattern in juvenile chronic myelogenous leukemia, myelodysplastic syndrome, and acute leukemia associated with neurofibromatosis. Leukemia 1989, 3, 36–41. [Google Scholar]
  9. Felix, C.A. Secondary leukemias induced by topoisomerase-targeted drugs. Biochim. Biophys. Acta 1998, 1400, 233–255. [Google Scholar]
  10. Hayes, J.D.; Pulford, D.J. The glutathione S-transferase supergene family: Regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Crit. Rev. Biochem. Mol. Biol. 1995, 30, 445–600. [Google Scholar]
  11. Willman, C.L.; Sever, C.E.; Pallacicini, M.G.; Harada, H.; Tanaka, N.; Slovak, M.L.; Yamamoto, H.; Harada, K.; Meeker, T.C. Deletion of IRF-1, mapping to chromosome 5q31.1, in human leukemia and preleukemic myelodysplasia. Science 1993, 259, 968–971. [Google Scholar]
  12. Le Beau, M.M.; Espinosa, R., 3rd.; Neuman, W.L.; Stock, W.; Roulston, D.; Larson, R.A.; Keinanen, M.; Westbrook, C.A. Cytogenetic and molecular delineation of the smallest commonly deleted region of chromosome 5 in malignant myeloid diseases. Proc. Natl. Acad. Sci. USA 1993, 90, 5484–5488. [Google Scholar]
  13. Pui, C.H. Epipodophyllotoxin-related acute myeloid leukaemia. Lancet 1991, 338. [Google Scholar]
  14. Sandler, E.S.; Friedman, D.J.; Mustafa, M.M.; Winick, N.J.; Bowman, W.P.; Buchanan, G.R. Treatment of children with epipodophyllotoxin-induced secondary acute myeloid leukemia. Cancer 1997, 79, 1049–1054. [Google Scholar]
  15. Rimmer, Y.; Chester, J.; Joffe, J.; Stark, D.; Shamash, J.; Powles, T.; White, J.; Wason, J.; Parashar, D.; Armstrong, G.; Mazhar, D.; Williams, M.V. Accelerated BEP: A phase I trial of dose-dense BEP for intermediate and poor prognosis metastatic germ cell tumour. Br. J. Cancer 2011, 105, 766–772. [Google Scholar]
  16. Bajetta, E.; di Bartolomeo, M.; Carnaghi, C.; Buzzoni, R.; Mariani, L.; Gebbia, V.; Comella, G.; Pinotti, G.; Ianniello, G.; Schieppati, G.; Bochicchio, A.M.; Maiorino, L. FEP regimen (epidoxorubicin, etoposide and cisplatin) in advanced gastric cancer, with or without low-dose GM-CSF: An Italian Trial in Medical Oncology (ITMO) study. Br. J. Cancer 1998, 77, 1149–1154. [Google Scholar] [CrossRef]
  17. Pui, C.H.; Ribeiro, R.C.; Hancock, M.L.; Rivera, G.K.; Evans, W.E.; Raimondi, S.C.; Head, D.R.; Behm, F.G.; Mahmoud, M.H.; Sandlund, J.T. et al. Acute myeloid leukemia in children treated with epipodophyllotoxins for acute lymphoblastic leukemia. N. Engl. J. Med. 1991, 325, 1682–1687. [Google Scholar]
  18. Heyn, R.; Khan, F.; Ensign, L.G.; Donaldson, S.S.; Ruymann, F.; Smith, M.A.; Vietti, T.; Maurer, H.M. Acute myeloid leukemia in patients treated for rhabdomyosarcoma with cyclophosphamide and low-dose etoposide on Intergroup Rhabdomyosarcoma Study III: An interim report. Med. Pediatr. Oncol. 1994, 23, 99–106. [Google Scholar]
  19. Smith, M.A.; Rubinstein, L.; Ungerleider, R.S. Therapy-related acute myeloid leukemia following treatment with epipodophyllotoxins: Estimating the risks. Med. Pediatr. Oncol. 1994, 23, 86–98. [Google Scholar]
  20. Kollmannsberger, C.; Beyer, J.; Droz, J.P.; Harstrick, A.; Hartmann, J.T.; Biron, P.; Flechon, A.; Schoffski, P.; Kuczyk, M.; Schmoll, H.J.; et al. Secondary leukemia following high cumulative doses of etoposide in patients treated for advanced germ cell tumors. J. Clin. Oncol. 1998, 16, 3386–3391. [Google Scholar]
  21. Chen, C.L.; Fuscoe, J.C.; Liu, Q.; Pui, C.H.; Mahmoud, H.H.; Relling, M.V. Relationship between cytotoxicity and site-specific DNA recombination after in vitro exposure of leukemia cells to etoposide. J. Natl. Cancer Inst. 1996, 88, 1840–1847. [Google Scholar]
  22. Pedersen-Bjergaard, J.; Daugaard, G.; Hansen, S.W.; Philip, P.; Larsen, S.O.; Rorth, M. Increased risk of myelodysplasia and leukaemia after etoposide, cisplatin, and bleomycin for germ-cell tumours. Lancet 1991, 338, 359–363. [Google Scholar]
  23. Bokemeyer, C.; Schmoll, H.J.; Kuczyk, M.A.; Beyer, J.; Siegert, W. Risk of secondary leukemia following high cumulative doses of etoposide during chemotherapy for testicular cancer. J. Natl. Cancer Inst. 1995, 87, 58–60. [Google Scholar]
  24. Boshoff, C.; Begent, R.H.; Oliver, R.T.; Rustin, G.J.; Newlands, E.S.; Andrews, R.; Skelton, M.; Holden, L.; Ong, J. Secondary tumours following etoposide containing therapy for germ cell cancer. Ann. Oncol. 1995, 6, 35–40. [Google Scholar]
  25. Sullivan, D.M.; Glisson, B.S.; Hodges, P.K.; Smallwood-Kentro, S.; Ross, W.E. Proliferation dependence of topoisomerase II mediated drug action. Biochemistry 1986, 25, 2248–2256. [Google Scholar]
  26. Clark, P.I. Clinical pharmacology and schedule dependency of the podophyllotoxin derivatives. Semin. Oncol. 1992, 19, 20–27. [Google Scholar]
  27. Sugita, K.; Furukawa, T.; Tsuchida, M.; Okawa, Y.; Nakazawa, S.; Akatsuka, J.; Ohira, M.; Nishimura, K. High frequency of etoposide (VP-16)-related secondary leukemia in children with non-Hodgkin’s lymphoma. Am. J. Pediatr. Hematol. Oncol. 1993, 15, 99–104. [Google Scholar]
  28. Houck, W.; Abonour, R.; Vance, G.; Einhorn, L.H. Secondary leukemias in refractory germ cell tumor patients undergoing autologous stem-cell transplantation using high-dose etoposide. J. Clin. Oncol. 2004, 22, 2155–2158. [Google Scholar]
  29. Baker, D.L.; Schmidt, M.L.; Cohn, S.L.; Maris, J.M.; London, W.B.; Buxton, A.; Stram, D.; Castleberry, R.P.; Shimada, H.; Sandler, A.; et al. Outcome after reduced chemotherapy for intermediate-risk neuroblastoma. N. Engl. J. Med. 2010, 363, 1313–1323. [Google Scholar]
  30. Bhatia, S.; Krailo, M.D.; Chen, Z.; Burden, L.; Askin, F.B.; Dickman, P.S.; Grier, H.E.; Link, M.P.; Meyers, P.A.; Perlman, E.J.; et al. Therapy-related myelodysplasia and acute myeloid leukemia after Ewing sarcoma and primitive neuroectodermal tumor of bone: A report from the Children’s Oncology Group. Blood 2007, 109, 46–51. [Google Scholar]
  31. Broeker, P.L.; Super, H.G.; Thirman, M.J.; Pomykala, H.; Yonebayashi, Y.; Tanabe, S.; Zeleznik-Le, N.; Rowley, J.D. Distribution of 11q23 breakpoints within the MLL breakpoint cluster region in de novo acute leukemia and in treatment-related acute myeloid leukemia: Correlation with scaffold attachment regions and topoisomerase II consensus binding sites. Blood 1996, 87, 1912–1922. [Google Scholar]
  32. Hess, J.L. MLL: A histone methyltransferase disrupted in leukemia. Trends Mol. Med. 2004, 10, 500–507. [Google Scholar]
  33. Ernst, P.; Fisher, J.K.; Avery, W.; Wade, S.; Foy, D.; Korsmeyer, S.J. Definitive hematopoiesis requires the mixed-lineage leukemia gene. Dev. Cell 2004, 6, 437–443. [Google Scholar]
  34. Hess, J.L.; Yu, B.D.; Li, B.; Hanson, R.; Korsmeyer, S.J. Defects in yolk sac hematopoiesis in Mll-null embryos. Blood 1997, 90, 1799–1806. [Google Scholar]
  35. Yokoyama, A.; Somervaille, T.C.; Smith, K.S.; Rozenblatt-Rosen, O.; Meyerson, M.; Cleary, M.L. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005, 123, 207–218. [Google Scholar]
  36. Strissel, P.L.; Strick, R.; Rowley, J.D.; Zeleznik-Le, N.J. An in vivo topoisomerase II cleavage site and a DNase I hypersensitive site colocalize near exon 9 in the MLL breakpoint cluster region. Blood 1998, 92, 3793–3803. [Google Scholar]
  37. Aplan, P.D.; Chervinsky, D.S.; Stanulla, M.; Burhans, W.C. Site-specific DNA cleavage within the MLL breakpoint cluster region induced by topoisomerase II inhibitors. Blood 1996, 87, 2649–2658. [Google Scholar]
  38. Lovett, B.D.; Lo Nigro, L.; Rappaport, E.F.; Blair, I.A.; Osheroff, N.; Zheng, N.; Megonigal, M.D.; Williams, W.R.; Nowell, P.C.; Felix, C.A. Near-precise interchromosomal recombination and functional DNA topoisomerase II cleavage sites at MLL and AF-4 genomic breakpoints in treatment-related acute lymphoblastic leukemia with t(4;11) translocation. Proc. Natl. Acad. Sci. USA 2001, 98, 9802–9807. [Google Scholar]
  39. Lovett, B.D.; Strumberg, D.; Blair, I.A.; Pang, S.; Burden, D.A.; Megonigal, M.D.; Rappaport, E.F.; Rebbeck, T.R.; Osheroff, N.; Pommier, Y.G.; et al. Etoposide metabolites enhance DNA topoisomerase II cleavage near leukemia-associated MLL translocation breakpoints. Biochemistry 2001, 40, 1159–1170. [Google Scholar]
  40. Stanulla, M.; Wang, J.; Chervinsky, D.S.; Thandla, S.; Aplan, P.D. DNA cleavage within the MLL breakpoint cluster region is a specific event which occurs as part of higher-order chromatin fragmentation during the initial stages of apoptosis. Mol. Cell. Biol. 1997, 17, 4070–4079. [Google Scholar]
  41. Stanulla, M.; Chhalliyil, P.; Wang, J.; Jani-Sait, S.N.; Aplan, P.D. Mechanisms of MLL gene rearrangement: Site-specific DNA cleavage within the breakpoint cluster region is independent of chromosomal context. Hum. Mol. Genet. 2001, 10, 2481–2491. [Google Scholar]
  42. Betti, C.J.; Villalobos, M.J.; Diaz, M.O.; Vaughan, A.T. Apoptotic triggers initiate translocations within the MLL gene involving the nonhomologous end joining repair system. Cancer Res. 2001, 61, 4550–4555. [Google Scholar]
  43. Betti, C.J.; Villalobos, M.J.; Diaz, M.O.; Vaughan, A.T. Apoptotic stimuli initiate MLL-AF9 translocations that are transcribed in cells capable of division. Cancer Res. 2003, 63, 1377–1381. [Google Scholar]
  44. Libura, J.; Slater, D.J.; Felix, C.A.; Richardson, C. Therapy-related acute myeloid leukemia-like MLL rearrangements are induced by etoposide in primary human CD34+ cells and remain stable after clonal expansion. Blood 2005, 105, 2124–2131. [Google Scholar]
  45. Blanco, J.G.; Edick, M.J.; Relling, M.V. Etoposide induces chimeric Mll gene fusions. FASEB J. 2004, 18, 173–175. [Google Scholar]
  46. Bueno, C.; Catalina, P.; Melen, G.J.; Montes, R.; Sanchez, L.; Ligero, G.; Garcia-Perez, J.L.; Menendez, P. Etoposide induces MLL rearrangements and other chromosomal abnormalities in human embryonic stem cells. Carcinogenesis 2009, 30, 1628–1637. [Google Scholar]
  47. Vlasova, I.I.; Feng, W.H.; Goff, J.P.; Giorgianni, A.; Do, D.; Gollin, S.M.; Lewis, D.W.; Kagan, V.E.; Yalowich, J.C. Myeloperoxidase-dependent oxidation of etoposide in human myeloid progenitor CD34+ cells. Mol. Pharmacol. 2011, 79, 479–487. [Google Scholar]
  48. Cowell, I.G.; Sondka, Z.; Smith, K.; Lee, K.C.; Manville, C.M.; Sidorczuk-Lesthuruge, M.; Rance, H.A.; Padget, K.; Jackson, G.H.; Adachi, N.; et al. Model for MLL translocations in therapy-related leukemia involving topoisomerase IIbeta-mediated DNA strand breaks and gene proximity. Proc. Natl. Acad. Sci. USA 2012, 109, 8989–8994. [Google Scholar]
  49. Willmore, E.; de Caux, S.; Sunter, N.J.; Tilby, M.J.; Jackson, G.H.; Austin, C.A.; Durkacz, B.W. A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood 2004, 103, 4659–4665. [Google Scholar]
  50. Toyoda, E.; Kagaya, S.; Cowell, I.G.; Kurosawa, A.; Kamoshita, K.; Nishikawa, K.; Iiizumi, S.; Koyama, H.; Austin, C.A.; Adachi, N. NK314, a topoisomerase II inhibitor that specifically targets the alpha isoform. J. Biol. Chem. 2008, 283, 23711–23720. [Google Scholar]
  51. Adachi, N.; Suzuki, H.; Iiizumi, S.; Koyama, H. Hypersensitivity of nonhomologous DNA end-joining mutants to VP-16 and ICRF-193: Implications for the repair of topoisomerase II-mediated DNA damage. J. Biol. Chem. 2003, 278, 35897–35902. [Google Scholar]
  52. Shiloh, Y. ATM and related protein kinases: Safeguarding genome integrity. Nat. Rev. Cancer 2003, 3, 155–168. [Google Scholar]
  53. Sun, J.; Oma, Y.; Harata, M.; Kono, K.; Shima, H.; Kinomura, A.; Ikura, T.; Suzuki, H.; Mizutani, S.; Kanaar, R.; Tashiro, S. ATM modulates the loading of recombination proteins onto a chromosomal translocation breakpoint hotspot. PLoS One 2010, 5. [Google Scholar]
  54. Kang, K.; Oh, S.H.; Yun, J.H.; Jho, E.H.; Kang, J.H.; Batsuren, D.; Tunsag, J.; Park, K.H.; Kim, M.; Nho, C.W. A novel topoisomerase inhibitor, daurinol, suppresses growth of HCT116 cells with low hematological toxicity compared to etoposide. Neoplasia 2011, 13, 1043–1057. [Google Scholar]
  55. Hoeksema, K.A.; Jayanthan, A.; Cooper, T.; Gore, L.; Trippett, T.; Boklan, J.; Arceci, R.J.; Narendran, A. Systematic in-vitro evaluation of the NCI/NIH Developmental Therapeutics Program Approved Oncology Drug Set for the identification of a candidate drug repertoire for MLL-rearranged leukemia. Onco Targets Ther. 2011, 4, 149–168. [Google Scholar]

Share and Cite

MDPI and ACS Style

Ezoe, S. Secondary Leukemia Associated with the Anti-Cancer Agent, Etoposide, a Topoisomerase II Inhibitor. Int. J. Environ. Res. Public Health 2012, 9, 2444-2453. https://doi.org/10.3390/ijerph9072444

AMA Style

Ezoe S. Secondary Leukemia Associated with the Anti-Cancer Agent, Etoposide, a Topoisomerase II Inhibitor. International Journal of Environmental Research and Public Health. 2012; 9(7):2444-2453. https://doi.org/10.3390/ijerph9072444

Chicago/Turabian Style

Ezoe, Sachiko. 2012. "Secondary Leukemia Associated with the Anti-Cancer Agent, Etoposide, a Topoisomerase II Inhibitor" International Journal of Environmental Research and Public Health 9, no. 7: 2444-2453. https://doi.org/10.3390/ijerph9072444

Article Metrics

Back to TopTop