LEADING ARTICLE Causality of myelodysplasia and acute ... - Nature

8 downloads 39 Views 218KB Size Report
... Larson RA, Le Beau MM, Skosey. C, Purl S, Hoffman PC, Wade J, Vardiman JW, Daly K, Rowley ..... Leukaemia in ben- zene workers. Lancet 1977; 2: 76–78.
Leukemia (2002) 16, 2177–2184  2002 Nature Publishing Group All rights reserved 0887-6924/02 $25.00 www.nature.com/leu

LEADING ARTICLE Causality of myelodysplasia and acute myeloid leukemia and their genetic abnormalities J Pedersen-Bjergaard, DH Christiansen, MK Andersen and F Skovby Cytogenetic Laboratory, Section of Hematology/Oncology, Department of Clinical Genetics, Juliane Marie Center, University Hospital, Rigshospitalet, Copenhagen, Denmark

New insights into causative factors for the development of myelodysplasia (MDS) and acute myeloid leukemia (AML), with associations to specific cytogenetic and genetic abnormalities have been obtained primarily from studies of patients with the therapy-related subsets of the two diseases. Current knowledge now makes it possible to distinguish between at least seven major genetic subgroups of MDS and AML, and has directed research towards more specific causative factors also for de novo MDS and AML. Leukemia (2002) 16, 2177–2184. doi:10.1038/sj.leu.2402764 Keywords: myelodysplasia; acute myeloid leukemia; therapy related leukemia; epidemiology; chromosome abnormalities; alkylating agents; topoisomerase II inhibitors

Introduction In most malignant diseases little is known of the etiological factors and the way they induce the genetic abnormalities resulting in neoplastic transformation. Examples of tumors with a specific etiology are lung cancer induced by smoking, pleural mesothelioma induced by inhalation of asbestos particles, and hepatocellular carcinoma following ingestion of aflatoxin. Myelodysplasia (MDS) and acute myeloid leukemia (AML) most often develop de novo without knowledge of previous mutagenic exposures. A subset of 10–15%, however, have a history of chemotherapy, of high voltage radiotherapy, or of combined modality therapy for another tumor. These therapyrelated cases of MDS (t-MDS) and AML (t-AML) are unique diseases, because in most cases they are induced by chemically well-defined drugs associated with specific genetic pathways of the disease. Because the pathology, the cytogenetics and the molecular biology of therapy-related and of de novo MDS and AML are almost indistinguishable, we propose hypotheses of the origin also of de novo disease. The risk of MDS and AML in patients treated intensively with chemotherapy has been shown to be increased 100 times or more as compared to the risk in the general population.1–7 This indicates that at least 99% of such cases of t-MDS or tAML must be considered as drug-induced. For this reason it is appropriate to relate their cytogenetic and genetic abnormalities to the mechanisms of action of the two types of leukemogenic drugs: alkylating agents and topoisomerase II inhibitors.

Correspondence: J Pedersen-Bjergaard, Section 4052, Rigshospitalet, Blegdamsvej 9, DK 2100 Copenhagen ⭋, Denmark; Fax: +45 3545 2577 Received 19 April 2002; accepted 26 July 2002

The ‘alkylator types’ of MDS and AML The first cytostatic drugs shown to be leukemogenic were alkylating agents, administered as single agents1,2,4,5,8–10 or included in combination chemotherapy regimens,3,7,11 primarily for multiple myeloma1,3,10 and Hodgkin’s disease.6,12–14 Many of these studies showed that the risk of t-MDS and tAML increases with the cumulative dose of drug4,5,7,9,10,13,14 and with patient age.3,6,13,14 After the first reports of t-AML it was realized that in most of these patients the disease presents in a preleukemic stage as t-MDS with abrupt thrombocytopenia 2–5 years after start of therapy with alkylating agents. Characteristically, the karyotypes, even early in the course of the disease, show loss of whole chromosomes 5 or 7 or loss of various parts of the long arms of these two chromosomes, often with many additional chromosome abnormalities.15–20 Most patients with t-MDS die of refractory cytopenia before developing t-AML.16–20 Alkylating agents are known to induce interstrand crosslinking of DNA preventing transcription and replication, to form adducts with DNA bases resulting in gene mutations, and to induce various types of chromosome damage.21,22 These effects are not restricted to specific genes or chromosome regions, and the predominance of cases of t-MDS and t-AML with loss of various parts of the long arms of chromosomes 5 and 7 possibly reflects a selection of cells with these abnormalities due to a proliferative advantage. Chromosome damage which may lead to deletions or total chromosomal loss is frequent following chemotherapy with alkylating agents, and because two different deletions of the long arm of a chromosome 5 or a chromosome 7 are sometimes observed in cytogenetically unrelated clones in the same patient, the chromosome defects most likely are secondary and potentiate or activate the effects of other more important preclinical genetic changes23 (Figure 1a and c). Inactivated recessive genes on the remaining normal chromosome 5 or 7 have been searched for, so far without success,24–28 for which reason haploinsufficiency must also be considered. Two subgroups of the alkylator type of t-MDS and t-AML with deletion or loss of the long arms or complete loss of chromosomes 5 and 7 were recently identified (Figure 1).29,30 The first subgroup includes patients with abnormalities of chromosome 7 but normal chromosome 5. These patients generally have few other cytogenetic abnormalities, but often have mutations of the RAS genes and methylation of the p15 gene promotor. If loss of a whole chromosome 7 is the only cytogenetic abnormality observed, the patient may survive for long periods of time with a more indolent type of t-MDS. The second subgroup includes patients with t-MDS and tAML and deletion or loss of the long arm of chromosome 5,

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

2178

Figure 1 Genetic changes in different types of myelodysplasia and acute myeloid leukemia. (a) and (c) Putatitively inactivated recessive genes on the long arms of chromosome 5 and 7; (e) genetic abnormality specific for the 5q- syndrome; (f) genetic abnormality specific for juvenile myelomonocytic leukemia; (h–k) genetic abnormalities supplementing the dominant oncogenic effect of the transcripts of the recurrent balanced chromosome aberrations; (l) and (n) primary genetic changes in MDS with a normal karyotype or with +8; (b) (d) (g) (m) and (o) genetic changes involved in transformation from myelodysplasia to overt leukemia. Many of the genetic changes may be complex or identical for different types of MDS and AML.

with or without abnormalities of chromosome 7.29,30 These patients characteristically have a complex karyotype with many additional abnormalities and their prognosis is extremely poor.29 Mutations of p53 have been observed in 77% of such patients, often with loss of heterozygosity of the gene.29,31,32 The mutational spectrum seems to vary with the type of alkylating agent or cisplatin used, possibly due to different preferences for alkylation of the individual drugs at specific bases in the minor or in the major groove of DNA.29 These results support the notion that the p53 mutations are directly induced by the cytostatic agents. Many of the previously unidentified chromosome abnormalities of t-MDS and t-AML following therapy with alkylating agents have recently been identified by FISH as dicentric chroLeukemia

mosomes involving chromosomes 5 or 7,33 or as unbalanced translocations to their long arms, both type of abnormality resulting in loss of different but overlapping parts of 5q or 7q.34 In other cases, previously unidentified abnormalities have now been disclosed as duplications or amplifications of varying parts of the long arm of chromosome 11, including the 11q23 band and the unrearranged MLL gene, a phenomenon significantly associated with mutations of p53.35 Experimental research indicates that the entire scenario of multiple chromosome aberrations and amplifications could result directly from p53 mutations.36–38 As suggested for de novo AML,39 deleted genes at 5q and acquired mutations of p53 possibly cooperate in leukemogenesis, also in t-MDS and t-AML.29 In de novo MDS and AML similar deletions or loss of the

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

long arms of chromosomes 5 and 7 are observed, although less frequently than in therapy-related diseases.40–42 With some exceptions, most cases of de novo MDS or AML with the characteristic defects of chromosomes 5 or 7 show a striking similarity to cases of t-MDS and t-AML after the use of alkylating agents. In particular, cases with deletions or loss of 5q often present mutations of p53 and a complex karyotype,43– 45 and they all respond poorly to intensive antileukemic chemotherapy. De novo AML with abnormal chromosomes 5 and 7 has repeatedly been associated with previous exposure to chemicals, organic solvents and pesticides.46–48 Although referred to as a well-established fact, such an association has not been confirmed or rejected by larger prospective studies. The 5q- syndrome and juvenile myelomonocytic leukemia with monosomy 7 Cases of de novo MDS with interstitial deletion of the long arm of chromosome 5 as a sole abnormality, refractory anemia, and without an excess of blasts in the bone marrow represent a specific entity: ‘the 5q- syndrome’ (Figure 1).49,50 The commonly deleted region on 5q seems in these cases to be located more telomeric than in other cases of MDS and AML. These patients have normal platelet counts and an extremely good prognosis with a very low risk of leukemic transformation. They thereby differ completely, possibly also at the molecular level (Figure 1, genetic abnormality e), from other patients with MDS and an increased percentage of myeloblasts in the bone marrow or with t-AML and deletions of the long arm of chromosome 5. Monosomy 7 is frequently observed in children with MDS,51 and is characteristic of juvenile myelomonocytic leukemia.51,52 Although the specific genetic background of this disease entity remains to be identified, the molecular biology (Figure 1, genetic abnormality f) most likely differs from that of adult cases of MDS or AML with monosomy 7. The ‘topoisomerase II-related types’ of MDS and AML Anthracyclines, epipodophyllotoxins, mitoxantrone and the bisdioxopiperazine derivatives, all cytostatic drugs belonging to the group of topoisomerase II inhibitors, have recently been demonstrated as leukemogenic.53–63 Leukemic complications following therapy with these drugs characteristically present as overt t-AML rather than t-MDS, often with a latent period of only 1–2 years or less.11 Cytogenetic abnormalities include balanced reciprocal translocations, involving chromosome bands 11q23 or 21q22,53–55,58,59,64–67 more rarely inv(16)66,68 or t(15;17).61,63,66,69 These recurrent aberrations have been shown to generate fusions between the MLL, AML1, CBFB or RARA genes and various partner genes (Figure 1), and the new chimeric genes are supposed to act as dominant oncogenes.70–72 Although the molecular biology of the disease in this group of patients with t-MDS and t-AML is heterogeneous, they all present chimeric gene fusions and a common ethiology: therapy with topoisomerase II inhibitors. Indirect evidence now supports the notion that the balanced chromosome aberrations observed in t-MDS and t-AML are directly induced by cytostatic drugs.73 Topoisomerase II is a major component of the chromosomal scaffold,74 and as a part of its normal function the enzymes induce transient, 4 base pair staggered double strand breaks of DNA for strand passage and unwinding. Induction of double strand breaks of

DNA is a potent initiator of translocations,75 and topoisomerase II interacts preferentially with DNA crossovers.76 The enzyme has been associated with illegitimate gene recombinations at matrix-associated regions.77 Many cytostatic drugs, as an important mechanism of action, inhibit religation of the topoisomerase II-induced double strand breaks of DNA78,79 and this effect may enhance the risk of illegitimate chromosome recombinations. Etoposide has been shown to induce illegitimate gene recombinations,80 and etoposide and doxorubicin to induce site-specific cleavage within the MLL and the AML1 genes,81,82 a prerequisite for development of translocations and new fusion genes. A topoisomerase II cleavage site83 and a Dnase hypersensitive site have been shown to colocalize near exon 9 in the MLL breakpoint cluster region.84 Furthermore, a 4 base pair microduplication has been demonstrated at the translocation site between the NUP98 and the TOP1 genes in two cases of t-MDS with a t(11;20)(p15;q11) after treatment with topoisomerase II inhibitors.85 These results, if evaluated together, strongly support a direct role of topoisomerase II and its inhibition by cytostatic drugs in the development of balanced chromosome aberrations and fusion genes in therapy-related leukemia. Patients with de novo AML and recurrent balanced chromosome aberrations, like their therapy-related counterpart, are most often younger than patients with deletions or loss of 5q and 7q, and their initial response to chemotherapy is favorable.41,42 The long-term survival, however, of patients with translocations to chromosome band 11q23 is worse than the long-term survival of patients with other common balanced chromosome aberrations due to a high risk of relapse of the disease.86 Previously, no specific external exposures were suspected to play a role in the development of such de novo leukemias. Recently, a significantly increased maternal intake of agricultural products such as beans, fresh vegetables and fruit, all rich in bioflavonoids, has been demonstrated in cases of infant acute leukemia.87 These bioflavonoids are topoisomerase II inhibitors, pass the placental barrier and induce a cleavage of the MLL gene within the break point cluster region similarly to etoposide and doxorubicin.88 Thus, in one particular subgroup of de novo acute leukemias, an association between specific external exposures and definite cytogenetic abnormalities has now been established. In knock-in experiments in mice with the different fusion genes from the most common recurrent balanced chromosome aberrations, for instance the AML1-ETO gene of the t(8;21),89 it has been shown that additional cooperating mutations (Figure 1h–k) are required for development of overt leukemia. The fact that the AML1-ETO transcript can still be detected by the sensitive RT-PCR technique in many cases of AML with t(8;21) in long-term remission90 supports the idea that cooperating, additional mutations are also required for the development of human AML.

2179

The ‘non-mutagenic type’ of MDS and AML A normal karyotype has been observed consistently in 40– 50% of patients with de novo MDS and AML.40–42 New techniques such as RT-PCR and Southern blotting for detection of the rearranged genes of the recurrent balanced translocations, and multicolor fluorescence in situ hybrization (M-FISH) for detection of cryptic chromosome rearrangements, have disclosed abnormalities in only a smaller fraction of patients with a normal karyotype. Other genetic changes (Figure 1l and m) Leukemia

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

2180

are supposed to play a role, and recently internal tandem duplications of the MLL and the FLT3 genes have been demonstrated in 6–9% and 28–36%, respectively, of such patients (Figure 1).45,91–97 Classification of cases of AML de novo with a normal karyotype as a single entity is now supported by a characteristic gene expression profile of such cases,98 different from the profile of normal CD34-positive cells and different from the profile of AML cells with trisomy 8. In patients with t-MDS and t-AML only 10–15% have a normal karyotype.16–21 It has not been possible in these cases to relate the disease to any specific type of chemotherapy or radiotherapy. Recently, internal tandem duplications of the MLL and the FLT3 genes were also observed in patients with tAML and a normal karyotype.99 Even these duplications were unrelated to any type of therapy. As the junction points for duplications of the MLL gene consistently occur within Alurepeats,96,97,99 these duplications have been supposed to arise as spontaneous homologous recombinations. As the genetic changes of t-MDS and t-AML with a normal karyotype seem to arise unrelated to exposure to cytostatic drugs or irradiation, the genetic changes of de novo MDS and AML with a normal karyotype could perhaps likewise arise without any external leukemogenic exposures. Other general types of MDS and AML The three major types of MDS and AML discussed above include more than 80% of patients with therapy-related and de novo MDS and AML. At least two other types must be considered. The first type includes patients with de novo or therapy-related MDS or AML and trisomy 8, the second comprises patients with t-MDS and t-AML after radiotherapy only. Trisomy 8 with or without other chromosome aberrations is present in 5% and 9% of patients with de novo MDS and AML, respectively,40–42 and is associated with an intermediate or poor prognosis depending on the number of additional aberrations. In de novo AML trisomy 8 has been related to previous exposure to organic solvents.100 In t-MDS and t-AML trisomy 8 is less common than in de novo disease,19,20 and is often observed in only a subclone of cells or as an evolutionary event during the course of the disease,101 unrelated to any specific type of previous therapy. For these reasons trisomy 8 has been considered a secondary phenomenon.20,30 Recently, however, a distinct gene expression profile was demonstrated in patients with de novo AML and trisomy 8 as the sole abnormality.98 Leukemic cells with trisomy 8 showed down-regulation of several apoptosis-related genes and a moderate overexpression of many genes located to chromosome 8 as compared to leukemic cells with a normal karyotype and to normal cells. The results suggest a simple gene-dosage effect of the supernumerary chromosome 8 and support trisomy 8 as well as a normal karyotype to represent specific genetic entities of leukemia (Figure 1). Cytogenetic investigations of t-MDS and t-AML following radiotherapy only have been published for single cases or small series of patients.16–20 Karyotypes have not differed markedly from those of de novo MDS and AML, taking the often advanced age of patients receiving radiotherapy into consideration. Other very extensive studies have consistently reported a much lower risk of t-AML following high voltage radiotherapy only than following intensive chemotherapy. Irradiation confers relative risks of AML in the order of 1.3– 2.4 as compared to the risk of AML in the general popu-

Leukemia

lation.102–104 Consequently, only about half of the radiotherapy-related cases can be considered as radiation-induced, whereas the other half represents incidental cases of de novo MDS or AML. The fact that some of these leukemias have been observed only a few months after radiotherapy, and others after more than 10 years, supports the assumption that far from all radiotherapy-related cases are radiation-induced. In our own series of 19 consecutive cases of t-MDS or tAML after radiotherapy only,105 eight patients presented deletion or loss of 7q whereas six patients had a normal karyotype. These findings are in agreement with data from Felix Mitelman’s Catalog of Chromosome Aberrations in Cancer,106 according to which deletions or loss of the long arm of chromosome 5 are also common in radiotherapy-related leukemias. It is still an open question whether radiotherapy-induced cases of t-MDS and t-AML show characteristic chromosome aberrations, or whether the aberrations observed merely relate to an advanced age of most of these patients. If characteristic abnormalities exist, these include deletions or loss of 5q and 7q or loss of the whole chromosomes.

Epidemiologic studies of de novo MDS and AML Many studies have searched for etiologic factors in malignancy including MDS and AML as previously reviewed.107 Apart from chemotherapy or radiotherapy, only few other exposures have consistently been verified as leukemogenic. These include inhalation of benzene vapor at high concentrations108–111 and smoking.112–114 There may be several reasons for this lack of success. First, many cases of MDS and AML may develop as the result of spontaneous gene mutations or recombinations, unrelated to external mutagenic exposures, as suggested for the ‘non-mutagenic type’ of MDS and AML with a normal karyotype. Second, many environmental exposures are difficult to quantify or even to verify, and the low incidence of MDS and AML makes it difficult to gather large numbers of patients for study. Third, only a few out of numerous potential gene mutations or rearrangements are present in each case. If only some of these are associated with a specific external exposure, it may be impossible to identify such an exposure as leukemogenic by epidemiologic methods.

t-MDS and t-AML as a model of leukemic transformation The induction by chemically well-defined substances with a known mechanism of action, administered in exact doses and at a known time, make t-MDS and t-AML following chemotherapy exceptional diseases. Further studies may be able to relate chemical structure and reactivity to specific genetic changes more precisely. In the closely followed cohorts of patients treated intensively it may be possible to study a stepwise accumulation, alternatively a ‘one hit’ development, of the genetic changes required for leukemic transformation. Experience from chemotherapy-related MDS and AML may also help in identifying new important chemical exposures, as exemplified by recent studies suggesting that consumption of natural products and medicines,115 and absorption of phenol and hydroquinone from the gastrointestinal tract,116 may be responsible for some cases of de novo MDS and AML.

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

Acknowledgements Supported by grants from the Danish Cancer Society and HS Forskningspulje 1997.

18

References 1 Kyle RA, Pierre RV, Bayrd ED. Multiple myeloma and acute myelomonocytic leukemia. Report of four cases possibly related to melphalan. N Engl J Med 1970; 283: 1121–1125. 2 Reimer RR, Hoover R, Fraumeni JF Jr, Young RC. Acute leukemia after alkylating-agent therapy of ovarian cancer. N Engl J Med 1977; 297: 177–181. 3 Bergsagel DE, Bailey AJ, Langley GR, MacDonald RN, White DF, Miller AB. The chemotherapy of plasma-cell myeloma and the incidence of acute leukemia. N Engl J Med 1979; 301: 743–748. 4 Pedersen-Bjergaard J, Nissen NI, Sørensen HM, Hou-Jensen K, Larsen, MS, Ernst P, Ersbol J, Knudtzon S, Rose C. Acute nonlymphocytic leukemia in patients with ovarian carcinoma following long-term treatment with treosulfan (=dihydroxybusulfan). Cancer 1980; 45: 19–29. 5 Greene MH, Boice JD Jr, Greer BE, Blessing JA, Dembo AJ. Acute nonlymphocytic leukemia after therapy with alkylating agents for ovarian cancer. N Engl J Med 1982; 307: 1416–1421. 6 Pedersen-Bjergaard J, Larsen SO. Incidence of acute nonlymphocytic leukemia, preleukemia, and acute myeloproliferative syndrome up to 10 years after treatment of Hodgkin’s disease. N Engl J Med 1982; 307: 965–971. 7 Greene MH, Young RC, Merrill JM, De Vita VT. Evidence of a treatment dose response in acute nonlymphocytic leukemias which occur after therapy of non-Hodgkin’s lymphoma. Cancer Res 1983; 43: 1891–1898. 8 Berk PD, Goldberg JD, Silverstein MN, Weinfeld A, Donovan PB, Ellis JT, Landaw SA, Laszlo J, Najean Y, Pisciotta AV, Wasserman LR. Increased incidence of acute leukemia in polycythemia vera associated with chlorambucil therapy. N Engl J Med 1981; 304: 441–447. 9 Boice JD Jr, Greene MH, Killen JY Jr, Ellenberg SS, Keehn RJ, McFadden E, Chen TT, Fraumeni JF Jr. Leukemia and preleukemia after adjuvant treatment of gastrointestinal cancer with semustine (methyl-CCNU). N Engl J Med 1983; 309: 1079–1084. 10 Cuzick J, Erskine S, Edelman D, Galton DA. A comparison of the incidence of the myelodysplastic syndrome and acute myeloid leukaemia following melphalan and cyclophosphamide treatment for myelomatosis. Br J Cancer 1987; 55: 523–529. 11 Pedersen-Bjergaard J, Philip P, Larsen SO, Andersson M, Daugaard G, Ersboll J, Hansen SW, Hou-Jensen K, Nielsen D, Sigsgaard TC, Specht L, ⭋sterlind K. Therapy-related myelodysplasia and acute myeloid leukemia. Cytogenetic characteristics of 115 consecutive cases and risk in seven cohorts of patients treated intensively for malignant diseases in the Copenhagen series. Leukemia 1993; 7: 1975–1986. 12 Coleman CN, Williams CJ, Flint A, Glatstein EJ, Rosenberg SA, Kaplan HS. Hematologic neoplasia in patients treated for Hodgkin disease. N Engl J Med 1977; 297: 1249–1252. 13 Pedersen-Bjergaard J, Specht L, Larsen SO, Ersboll J, Struck J, Hansen MM, Hansen HH, Nissen NI. Risk of therapy-related leukaemia and preleukaemia after Hodgkin’s disease. Lancet 1987; II: 83–88. 14 Tucker MA, Coleman CN, Cox RS, Varghese A, Rosenberg SA. Risk of second cancers after treatment for Hodgkin’s disease. N Engl J Med 1988; 318: 76–81. 15 Rowley JD, Golomb HM, Vardiman J. Nonrandom chromosomal abnormalities in acute nonlymphocytic leukemia in patients treated for Hodgkin disease and non-Hodgkin lymphomas. Blood 1977; 50: 759–770. 16 Pedersen-Bjergaard J, Philip P, Mortensen BT, Ersboll J, Jensen G, Panduro J, Thomsen M. Acute nonlymphocytic leukemia, preleukemia, and acute myeloproliferative syndrome secondary to treatment of other malignant diseases. Clinical and cytogenetic characteristics and results of in vitro culture of bone marrow and HLA typing. Blood 1981; 57: 712–723. 17 Rowley JD, Golomb HM, Vardiman JW. Nonrandom chromosome

19 20

21 22 23

24

25

26

27

28 29

30 31

32

33

34 35

abnormalities in acute leukemia and dysmyelopoietic syndromes in patients with previously treated malignant disease. Blood 1981; 58: 759–767. Le Beau MM, Albain KS, Larson RA, Vardiman JW, Davis EM, Blough RR, Golomb HM, Rowley JD. Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosome No. 5 and 7. J Clin Oncol 1986; 4: 325–345. Johansson B, Mertens F, Heim S, Kristoffersson U, Mitelman F. Cytogenetics of secondary myelodysplasia (sMDS) and acute nonlymphocytic leukemia (sANLL). Eur J Haematol 1991; 47: 17–27. Pedersen-Bjergaard J, Pedersen M, Roulston D, Philip P. Different genetic pathways in leukemogenesis for patients presenting with therapy-related myelodysplasia and therapy-related acute myeloid leukemia. Blood 1995; 86: 3542–3552. Hemminki K, Kallama S. Reactions of nitrogen mustards with DNA. IARC Scientific Publications 1986; 78: 55–70. Povirk LF, Shuker DE. DNA damage and mutagenesis induced by nitrogen mustards. Mutat Res 1994; 318: 205–226. Pedersen-Bjergard J, Timshel S, Andersen MK, Andersen AS, Philip P. Cytogenetically unrelated clones in therapy-related myelodysplasia and acute myeloid leukemia: experience from the Copenhagen series updated to 180 consecutive cases. Genes Chromosomes Cancer 1998; 23: 337–349. Zhao N, Stoffel A, Wang PW, Eisenbart JD, Espinosa R 3rd, Larson RA, Le Beau MM. Molecular delineation of the smallest commonly deleted region of chromosome 5 in malignant myeloid diseases to 1–1.5 Mb and preparation of a PAC-based physical map. Proc Natl Acad Sci USA 1997; 94: 6948–6953. Horrigan SK, Arbieva ZH, Xie HY, Kravarusic J, Fulton NC, Naik H, Le TT, Westbrook CA. Delineation of a minimal interval and identification of 9 candidates for a tumor suppressor gene in malignant myeloid disorders on 5q31. Blood 2000; 95: 2372– 2377. Do¨hner K, Brown J, Hehmann U, Hetzel C, Stewart J, Lowther G, Scholl C, Frohling S, Cuneo A, Tsui LC, Lichter P, Scherer SW, Dohner H. Molecular cytogenetic characterization of a critical region in bands 7q35-q36 commonly deleted in malignant myeloid disorders. Blood 1998; 92: 4031–4035. Liang H, Fairman J, Claxton DF, Nowell PC, Green ED, Nagarajan L. Molecular anatomy of chromosome 7q deletions in myeloid neoplasms: Evidence for multiple critical loci. Proc Natl Acad Sci USA 1998; 95: 3781–3785. Tosi S, Scherer SW, Giudici G, Czepulkowski B, Biondi A, Kearney L. Delineation of multiple deleted regions in 7q in myeloid disorders. Genes Chromosomes Cancer 1999; 25: 384–392. Christiansen DH, Andersen MK, Pedersen-Bjergaard J. Mutations with loss of heterozygosity of p53 are common in therapy-related MDS and AML after exposure to alkylating agents, and significantly associated with deletion or loss of 5q, a complex karyotype and a poor prognosis. J Clin Oncol 2001; 19: 1405–1413. Pedersen-Bjergaard J, Andersen MK, Christiansen DH, Nerlov C. Genetic pathways in therapy-related myelodysplasia and acute myeloid leukemia. Blood 2002; 99: 1909–1912. Horiike S, Misawa S, Kaneko H, Sasai Y, Kobayashi M, Fujii H, Tanaka S, Yagita M, Abe T, Kashima K, Taniwaki M. Distinct genetic involvement of the TP53 gene in therapy-related leukemia and myelodysplasia with chromosomal losses of nos 5 and/or 7 and its possible relationship to replication error phenotype. Leukemia 1999; 13: 1235–1242. Merlat A, Lai JL, Sterkers Y, Demory JL, Bauters F, Preudhomme C, Fenaux P. Therapy-related myelodysplastic syndrome and acute myeloid leukemia with 17p deletion. A report on 25 cases. Leukemia 1999; 13: 250–257. Andersen MK, Pedersen-Bjergaard J. Increased frequency of dicentric chromosomes in therapy-related MDS and AML compared to de novo disease is significantly related to previous treatment with alkylating agents and suggests a specific susceptibility to chromosome breakage at the centromere. Leukemia 2000; 14: 105–111. Andersen MK, Pedersen-Bjergaard J. Multi-color FISH in 54 patients with therapy-related MDS/AML. Blood 2001; 98: 579a (Abstr.). Andersen MK, Christiansen DH, Kirchhoff M, Pedersen-Bjergaard J. Duplication or amplification of chromosome band 11q23,

2181

Leukemia

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

2182

36 37 38 39 40

41

42

43

44

45

46

47 48 49 50 51

52

53

Leukemia

including the unrearranged MLL gene, is a recurrent abnormality in therapy-related MDS and AML, is closely related to mutation of the tp53 gene and to previous therapy with alkylating agents. Genes Chromosomes Cancer 2001; 31: 33–41. Bouffler SD, Kemp CJ, Balmain A, Cox R. Spontaneous and ionizing radiation-induced chromosomal abnormalities in p53 – different mice. Cancer Res 1995; 55: 3883–3889. Livingstone LR, White A, Sprouse J, Livanos E, Jacks T, Tisty TD. Altered cell cycle arrest and gene amplification potential accompany loss of wild-type p53. Cell 1992; 70: 923–935. Yin Y, Tainsky MA, Bischoff FZ, Strong LC, Wahl GM. Wild-type p53 restores cell cycle control and inhibits gene amplification in cells with mutant p53 alleles. Cell 1992; 70: 937–948. Castro PD, Liang JC, Nagarajan L. Deletions of chromosome 5q13.3 and 17p loci cooperate in myeloid neoplasms. Blood 2000; 95: 2138–2143. Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, Ohyashiki K, Toyama K, Aul C, Mufti G, Bennett J. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997; 89: 2079–2088. Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G, Rees J, Hann I, Stevens R, Burnett A, Goldstone A. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 patients entered into the MRC AML 10 trial. Blood 1998; 92: 2322–2333. Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A, Paietta E, Willman CL, Head DR, Rowe JM, Forman SJ, Appelbaum FR. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study. Blood 2000; 96: 4075–4083. Fenaux P, Jonveaux P, Quiquandon I, Lai JL, Pignon JM, Loucheux-Lefebvre MH, Bauters F, Berger R, Kerckaert JP. p53 gene mutations in acute myeloid leukemia with 17p monosomy. Blood 1991; 78: 1652–1657. Lai JL, Preudhomme C, Zandecki M, Flactif M, Vanrumbeke M, Lepelley P, Wattel E, Fenaux P. Myelodysplastic syndromes and acute myeloid leukemia with 17p deletion. An entity characterized by specific dysgranulopoiesis and a high incidence of p53 mutations. Leukemia 1995; 9: 370–381. Stirewalt DL, Kopecky KJ, Meshinchi S, Appelbaum FR, Slovak ML, Willman CL, Radich JP. FLT3, RAS and TP53 mutations in elderly patients with acute myeloid leukemia. Blood 2001; 97: 3589–3595. Mitelman F, Brandt L, Nilsson PG. Relation among occupational exposure to potential mutagenic/carcinogenic agents, clinical findings, and bone marrow chromosomes in acute nonlymphocytic leukemia. Blood 1978; 52: 1229–1237. Golomb HM, Alimena G, Rowley JD, Vardiman JW, Testa JR, Sovik C. Correlation of occupation and karyotype in adults with acute nonlymphocytic leukemia. Blood 1982; 60: 404–411. Narod SA, Dube´ ID. Occupational history and involvement of chromosomes 5 and 7 in acute nonlymphocytic leukemia. Cancer Genet Cytogenet 1989; 38: 261–269. Van den Berghe H, Cassiman JJ, David G, Fryns JP, Michaux JL, Sokal G. Distinct haematological disorder with deletion of long arm of no. 5 chromosome. Nature 1974; 251: 437–438. Boultwood J, Lewis S, Wainscoat JS. The 5q- syndrome (review). Blood 1994; 84: 3253–3260. Passmore SJ, Hann IM, Stiller CA, Ramani P, Swansbury GJ, Gibbons B, Reeves BR, Chessells JM. Pediatric myelodysplasia: A study of 68 children and a new prognostic scoring system. Blood 1995; 85: 1742–1750. Hasle H, Arico M, Basso G, Biondi A, Cantu Rajnoldi A, Creutzig U, Fenu S, Fonatsch C, Haas OA, Harbott J, Kardos G, Kerndrup G, Mann G, Niemeyer CM, Ptoszkova H, Ritter J, Slater R, Stary J, Stollmann-Gibbels B, Testi AM, van Wering ER, Zimmermann M. Myelodysplastic syndrome, juvenile myelomonocytic leukemia, and acute myeloid leukemia associated with complete or partial monosomy 7. Leukemia 1999; 13: 376–385. Ratain MJ, Kaminer LS, Bitran JD, Larson RA, Le Beau MM, Skosey C, Purl S, Hoffman PC, Wade J, Vardiman JW, Daly K, Rowley JD, Golomb HM. Acute nonlymphocytic leukemia following etoposide and cisplatin combination chemotherapy for advanced

54

55

56

57

58

59

60

61 62 63 64

65 66

67 68

69

70 71 72 73

non-small cell carcinoma of the lung. Blood 1987; 70: 1412– 1417. Pui C-H, Behm FG, Raimondi SC, Dodge RK, George SL, Rivera GK, Mirro J Jr, Kalwinsky DK, Dahl GV, Murphy SB. Secondary acute myeloid leukemia in children treated for acute lymphoid leukemia. N Engl J Med 1989; 321: 136–142. Pedersen-Bjergaard J, Daugaard G, Hansen SW, Philip P, Larsen SO, Rorth M. Increased risk of myelodysplasia and leukaemia after etoposide, cisplatin, and bleomycin for germ-cell tumours. Lancet 1991; 338: 359–363. Tucker MA, Meadows AT, Boice JD Jr, Stovall M, Oberlin O, Stone BJ, Birch J, Voute PA, Hoover RN, Fraumeni JF Jr. Leukemia after therapy with alkylating agents for childhood cancer. J Natl Cancer Int 1987; 78: 459–464. Kaldor JM, Day NE, Pettersson F, Clarke EA, Pedersen D, Mehnert W, Bell J, Host H, Prior P, Karjalainen S, Neal F, Koch M, Band P, Choi W, Kirn VP, Arslan A, Zare´n B, Belch AR, Storm H, Kittelmann B, Fraser P, Stovall M. Leukemia following chemotherapy for ovarian cancer. N Engl J Med 1990; 322: 1–6. Pedersen-Bjergaard J, Sigsgaard TC, Nielsen D, Gjedde SB, Philip P, Hansen M, Larsen SO, Rorth M, Mouridsen H, Dombernowsky P. Acute monocytic or myelomonocytic leukemia with balanced chromosome translocations to band 11q23 after therapy with 4epi-doxorubicin and cisplatin or cyclophosphamide for breast cancer. J Clin Oncol 1992; 10: 1444–1451. Cremin P, Flattery M, McCann SR, Daly PA. Myelodysplasia and acute myeloid leukemia following adjuvant chemotherapy for breast cancer using mitoxantrone and methotrexate with or without mitomycin. Ann Oncol 1996; 7: 745–746. Melillo LMA, Sajeva MR, Musto P, Perla G, Cascavilla N, Minervini MM, D’Arena G, Carotenuto M. Acute myeloid leukemia following 3M (mitoxantrone, mitomycin and methotrexate) chemotherapy for advanced breast cancer. Leukemia 1997; 11: 2211– 2213. Kudo K, Yoshida H, Kiyoi H, Numata S, Horibe K, Naoe T. Etoposide-related acute promyelocytic leukemia. Leukemia 1998; 12: 1171–1175. Carli PM, Sgro C, Parchin-Geneste N, Isambert N, Mugneret F, Girodon F, Maynadie M. Increase therapy-related leukemia secondary to breast cancer. Leukemia 2000; 14: 1014–1017. Xue Y, Lu D, Guo Y, Lin B. Specific chromosomal translocations and therapy-related leukemia induced by bimolane therapy for psoriasis. Leukemia Res 1992; 16: 1113–1123. Pedersen-Bjergaard J, Philip P. Balanced translocations involving chromosome bands 11q23 and 21q22 are highly characteristic of myelodysplasia and leukemia following therapy with cytostatic agents targeting at DNA-topoisomerase II. Blood 1991; 78: 1147–1148. Larson RA, Le Beau MM, Ratain MJ, Rowley JD. Balanced translocations involving chromosome bands 11q23 and 21q22 in therapy-related leukemia. Blood 1992; 79: 1892–1893. Pedersen-Bjergaard J, Andersen MK, Johansson B. Balanced chromosome aberrations in leukemias following chemotherapy with DNA-topoisomerase II inhibitors. J Clin Oncol 1998; 16: 1897– 1898. Secker-Walker LM, Moorman AV, Bain BJ, Mehta AB. Secondary acute leukemia and myelodysplastic syndrome with 11q23 abnormalities. Leukemia 1998; 12: 840–844. Dissing M, Le Beau MM, Pedersen-Bjergaard J. Inversion of chromosome 16 and uncommon rearrangements of the CBFB and MYH11 genes in therapy-related acute myeloid leukemia: rare events related to DNA-topoisomerase II inhibitors? J Clin Oncol 1998; 16: 1890–1896. Detourmignies L, Castaigne S, Stoppa AM, Harousseau JL, Sadoun A, Janvier M, Demory JL, Sanz M, Berger R, Bauters F, Chomienne C, Fenaux P. Therapy-related acute promyelocytic leukemia: a report on 16 cases. J Clin Oncol 1992; 10: 1430–1435. Look TA. Oncogenic transcription factors in human leukemia. Science 1997; 278: 1059–1064. Strout MP, Caligiuri MA. Developments in cytogenetics and oncogenes in acute leukemia. Curr Opin Oncol 1997; 9: 8–17. Rowley JD. The role of chromosome translocations in leukemogenesis. Semin Hematol 1999; 36 (Suppl. 7): 59–72. Felix CA. Leukemias related to treatment with DNA topoisomerase II inhibitors. Med Pediatr Oncol 2001; 36: 525–535.

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

74 Earnshaw WC, Halligan B, Cooke CA, Heck MM, Liu LF. Topoisomerase II is a structural component of mitotic chromosome scaffolds. J Cell Biol 1985; 100: 1706–1715. 75 Richardson C, Jasin M. Frequent chromosomal translocations induced by DNA double-strand breaks. Nature 2000; 405: 697– 700. 76 Zechiedrich EL, Osheroff N. Eukaryotic topoisomerase recognize nucleic acid topology by preferentially interacting with DNA crossovers. EMBO J 1990; 13: 4555–4562. 77 Sperry AO, Blasquez VC, Garrard WT. Dysfunction of chromosomal loop attachment sites: Illegitimate recombination linked to matrix association regions and topoisomerase II. Proc Natl Acad Sci USA 1989; 86: 5497–5501. 78 Lui LF. DNA topoisomerase poisons as antitumor drugs. Ann Rev Biochem 1989; 58: 351–375. 79 Pommier Y, Kohn K. Topoisomerase II inhibition by antitumor intercalators and demethylepipodophyllotoxins. In: Glazer RI (ed.). Development in Cancer Chemotherapy. CRC: Boca Raton, FL, 1989, p 175. 80 Chen C-L, Fuscoe JC, Liu Q, Relling MV. Etoposide causes illegitimate V(D)J recombination in human lymphoid leukemic cells. Blood 1996; 88: 2210–2218. 81 Aplan PD, Chervinsky DS, Stanulla M, Burhans WC. Site-specific DNA cleavage within the MLL breakpoint cluster region induced by topoisomerase II inhibitors. Blood 1996; 87: 2649–2658. 82 Stanulla M, Wang J, Chervinsky DS, Aplan PD. Topoisomerase II inhibitors induce DNA double-strand breaks at a specific site within the AML1 locus. Leukemia 1997; 11: 490–496. 83 Felix CA, Lange BJ, Hosler MR, Fertala J, Bjornsti MA. Chromosome band 11q23 translocation breakpoints are DNA topoisomerase II cleavage sites. Cancer Res 1995; 55: 4287–4292. 84 Strissel PL, Strick R, Rowley JD, Zeleznik-Le NJ. An in vivo topopisomerase II cleavage site and a DNase I hypersensitive site colocalize near exon 9 in the MLL breakpoint cluster region. Blood 1998; 92: 3793–3803. 85 Ahuja HG, Felix CA, Aplan P. Potential role for DNA topoisomerase II poisons in the generation of t(11,20)(p15;q11) translocations. Genes Chromosomes Cancer 2000; 29: 96–105. 86 Pui C-H, Relling, MV, Rivera GK, Hancock ML, Raimondi SC, Heslop HE, Santana VM, Ribeiro RC, Sandlund JT, Mahmoud HH, Evans WE, Crist WM, Krance RA. Epipodophyllotoxin-related acute myeloid leukemia: a study of 35 cases. Leukemia, 1995; 9: 1990–1996. 87 Ross JA, Potter JD, Reaman GH, Pendergrass TW, Robison LL. Maternal exposure to potential inhibitors of DNA topoisomerase II and infant leukemia (United States): a report from the Children’s Cancer Group. Cancer Causes Control 1996; 7: 581–590. 88 Strick R, Strissel PL, Borgers S, Smith SL, Rowley JD. Dietary bioflavonoids induce cleavage in the MLL gene and may contribute to infant leukemia. Proc Natl Acad Sci USA 2000; 97: 4790–4795. 89 Higuchi M, O’Brien D, Kumaravelu P, Lenny N, Yeoh EJ, Downing JR. Expression of a conditional AML1-ETO oncogene bypasses embryonic lethality and establishes a murine model of human t(8;21) acute myeloid leukemia. Cancer Cell 2002; 1: 63–74. 90 Miyamoto T, Nagafuji K, Akashi K, Harada M, Kyo T, Akashi T, Takenaka K, Mizuno S, Gondo H, Okamura T, Dohy H, Niho Y. Persistence of multipotent progenitors expressing AML1/ETO transcripts in long-term remission patients with t(8;21) acute myelogenous leukemia. Blood 1996; 87: 4789–4796. 91 Nakao M, Yokota S, Iwai T, Kaneko H, Horiike S, Kashima K, Sonoda Y, Fujimoto T, Misawa S. Internal tandem duplication of the FLT3 gene found in acute myeloid leukemia. Leukemia 1996; 10: 1911–1918. 92 Horiike S, Yokota S, Nakao M, Iwai T, Sasai Y, Kaneko H, Taniwaki M, Kashima K, Fujii H, Abe T, Misawa S. Tandem duplications of the FLT3 receptor gene are associated with leukemic transformation of myelodysplasia. Leukemia 1997; 11: 1442– 1446. 93 Yokota S, Kiyoi H, Nakao M, Iwai T, Misawa S, Okuda T, Sonoda Y, Abe T, Kahsima K, Matsuo Y, Naoe T. Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines. Leukemia 1997; 11: 1605–1609.

94 Schnittger S, Schoch C, Kern W, Staib P, Wuchter C, Sauerland MC, Serve H, Buechner T, Haferlach T, Hiddemann W. FLT3 length mutations in AML correlation to cytogenetics, FAB-subtype and prognosis in 652 patients. Blood 2000; 96 (Suppl. 1): 826a (Abstr.). 95 Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE, Belton AA, Walker H, Wheatley K, Bowen DT, Burnett AK, Goldstone AH, Linch DC. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood 2001; 98: 1752–1759. 96 Caligiuri MA, Strout MP, Lawrence D, Arthur DC, Baer MR, Yu F, Knuutila S, Mrozek K, Oberkircher AR, Marcucci G, de la Chapelle A, Elonen E, Block AW, Rao PN, Herzig GP, Powell BL, Ruutu T, Schiffer CA, Bloomfield CD. Rearrangement of ALL1 (MLL) in acute myeloid leukemia with normal cytogenetics. Cancer Res 1998; 58: 55–59. 97 Schnittger S, Kinkelin U, Schoch C, Heinecke A, Haase D, Haferlach T, Buchner T, Wormann B, Hiddemann W, Griesinger F. Screening for MLL tandem duplication in 387 unselected patients with AML identify a prognostically unfavourable subset of AML. Leukemia 2000; 14: 798–804. 98 Virtaneva K, Wright FA, Tanner SM, Yuan B, Lemon WJ, Caligiuri MA, Bloomfield CD, de la Chapelle A, Krahe R. Expression profiling reveals fundamental biological differences in acute myeloid leukemia with isolated trisomy 8 and normal cytogenetics. Proc Natl Acad Sci USA 2001; 98: 1124–1129. 99 Christiansen DH, Pedersen-Bjergaard J. Internal tandem duplications of the FLT3 and MLL genes are mainly observed in atypical cases of therapy-related acute myeloid leukemia with a normal karyotype and are unrelated to type of previous therapy. Leukemia 2001; 15: 1848–1851. 100 Albin M, Bjo¨rk J, Welinder H, Tinnerberg H, Mauritzson N, Johansson B, Billstrom R, Stromberg U, Mikoczy Z, Ahlgren T, Nilsson PG, Mitelman F, Hagmar L. Acute myeloid leukemia and clonal chromosome aberrations in relation to past exposure to organic solvents. Scand J Work Environ Health 2000; 26: 482– 491. 101 Pedersen-Bjergaard J, Philip P, Larsen SO, Jensen G, Byrsting K. Chromosome aberrations and prognostic factors in therapyrelated myelodysplasia and acute nonlymphocytic leukemia. Blood 1990; 76: 1083–1091. 102 Boice JD Jr, Day NE, Andersen A, Brinton LA, Brown R, Choi NW, Clarke EA, Coleman MP, Curtis RE, Flannery JT, Hakama M, Hakulinen T, Howe GR, Jensen OM, Kleinerman RA, Magnin D, Magnus K, Makela K, Malker B, Miller AB, Nelson N, Patterson CC, Pettersson F, Pompe-Kirn V, Primic-Zakelj M, Prior P, Ravnihar B, Skeet RG, Skjerven JE, Smith PG, Sok M, Spengler RF, Storm HH, Stovall M, Tomkins GWO, Wall C. Second cancers following radiation treatment for cervical cancer. An international collaboration among cancer registries. J Natl Cancer Inst 1985; 74: 955–975. 103 Curtis RE, Boice JD Jr, Stovall M, Bernstein L, Greenberg RS, Flannery JT, Schwartz AG, Weyer P, Moloney WC, Hoover RN. Risk of leukemia after chemotherapy and radiation treatment for breast cancer. N Engl J Med 1992; 326: 1745–1751. 104 Curtis RE, Boice JD Jr, Stovall M, Bernstein L, Holowaty E, Karjalainen S, Langmark F, Nasca PC, Schwartz AG, Schymura MJ, Storm HH, Toogood P, Weyer P, Moloney WC. Relationship of leukemia risk to radiation dose following cancer of the uterine corpus. J Natl Cancer Inst 1994; 86: 1315–1324. 105 Gundestrup M, Andersen MK, Sveinbjornsdottir E, Rafnsson V, Storm HH, Pedersen-Bjergaard J. Cytogenetics of myelodysplasia and acute myeloid leukaemia in aircrew and people treated with radiotherapy. Lancet 2000; 256: 2158. 106 Mitelman F, Johansson B, Mertens F. Mitelman database of chromosome aberrations in cancer (2000). http://cgap.nci.nih.gov/ Chromosomes/Mitelman (accessed on Jan 23, 2001). 107 Farber E. Chemical carcinogenesis. N Engl J Med 1981; 305: 1379–1389. 108 Vigliani EC, Saita G. Benzene and leukemia. N Engl J Med 1964; 271: 872–876.

2183

Leukemia

Causality of genetic changes in MDS and AML J Pedersen-Bjergaard et al

2184

Leukemia

109 Infante PF, Rinsky RA, Wagoner JK, Young RJ. Leukaemia in benzene workers. Lancet 1977; 2: 76–78. 110 Aksoy M, Erdem S. Follow-up study on the mortality and the development of leukemia in 44 pancytopenic patients with chronic exposure to benzene. Blood 1978; 52: 285–292. 111 Rinsky RA, Smith AB, Hornung R, Filloon TG, Young RJ, Okun AH, Landrigan PJ. Benzene and leukemia an epidemiologic risk assessment. N Engl J Med 1987; 316: 1044–1050. 112 Kinlen LJ, Rogot E. Leukaemia and smoking habits amoung United States veterans. Br Med J 1988; 297: 657–659. 113 McLaughlin JK. Cigarette smoking and leukemia. J Natl Cancer Inst 1989; 81: 1262–1263. 114 Garfinkel L, Boffetta P. Association between smoking and leuke-

mia in two American Cancer Society prospective studies. Cancer 1990; 65: 2356–2360. 115 Alexander FE, Patheal SL, Biondi A, Brandalise S, Cabrera ME, Chan LC, Chen Z, Cimino G, Cordoba JC, Gu LJ, Hussein H, Ishii E, Kamel AM, Labra S, Magalhaes IQ, Mizutani S, Petridou E, de Oliveira MP, Yuen P, Wiemels JL, Greaves MF. Transplacental chemical exposure and risk of infant leukemia with MLL gene fusion. Cancer Res 2001; 61: 2542–2546. 116 McDonald TA, Holland NT, Skibola C, Duramad P, Smith MT. Hypothesis: phenol and hydroquinone derived mainly from diet and gastrointestinal flora activity are causal factors in leukemia. Leukemia 2001; 15: 10–20.