Second Malignancies after Chemotherapy



Second Malignancies after Chemotherapy


Daniel J. Becker

Thomas S. Uldrick

Alfred I. Neugut



The era of modern chemotherapy traces back to the 1940s when it was discovered that the nitrogen mustards suppressed both the lymphoid and the myeloid cell lines. By the 1960s, MOPP (mechlorethamine, vincristine, procarbazine, and prednisone) was first used to treat patients with Hodgkin lymphoma effectively. Today, multidrug regimens are routinely available to treat practically all forms of cancer. These therapies, however, can increase long-term complications, including the risk of developing a new cancer. Typical early reports were published in 1969 and 1970 with descriptions of bladder cancer after the use of chlornaphazine for polycythemia vera and the occurrence of acute myeloid leukemia after alkylating agents for multiple myeloma.1,2 Epidemiologic research with controlled observational studies over the ensuing decades has confirmed this consistent association of secondary leukemias with alkylating agents and drugs that target DNA-topoisomerase II, such as the epipodophyllotoxins.3,4 Other forms of chemotherapy, such as many of the antimetabolites, do not appear to be carcinogenic (Table 24-1). Advances in cytogenetic research have provided a model for subcategorization of secondary leukemias based on genetic pathways of leukemogenesis.5

In the following sections, we will focus on studies of patients treated with chemotherapy that have provided information on the risks and characteristics of secondary cancer. Neither the effects of long-term immunosuppression nor those of hormonal therapies, such as tamoxifen, are covered in this review.


LEUKEMIA AFTER CHEMOTHERAPY

Treatment-related acute myeloid leukemia (t-AML) is by far the most frequently reported cancer after chemotherapy. t-AML has been documented after the alkylating agent treatment of Hodgkin lymphoma,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32 multiple myeloma,33,34,35,36,37,38,39 non-Hodgkin lymphoma (NHL),40,41,42,43,44,45,46,47,48 breast cancer,49,50,51,52,53,54,55,56 ovarian cancer,57,58,59,60,61,62 lung cancer,63,64,65,66,67,68 testicular cancer,69,70,71,72,73,74,75,76,77,78,79 various childhood cancers,80,81,82,83,84,85,86,87,88,89,90,91 gastrointestinal cancer,92,93,94 brain cancer,95 and polycythemia vera.96

The magnitude of risk has been estimated by various methods. Relative risk (RR) has been calculated from adverse outcomes in randomized controlled trials as well as case-control studies within large cohorts. Standardized incidence ratios (SIR) compare the incidence within a treated cohort to the expected incidence based on general population data. The range of reported RRs and SIRs for leukemia after chemotherapy is wide, between 1- and 100-fold, depending on the disease being treated and the intensity of treatment. Because leukemia is rare, however, the absolute risk and the cumulative probability (actuarial risk) are often used to indicate the true population impact and often may have more relevance to the clinician and patient, whereas the RR and SIR are more helpful in establishing causality. For example, an RR of 2 for leukemia that is associated with cyclophosphamide therapy for breast cancer would correspond to an absolute risk or excess of approximately 5 leukemias in 10,000 patients over a 10-year period.49

The risk of t-AML after chemotherapy depends on many factors, including the drug(s) administered, duration of treatment, cumulative dose, dose intensity, age of the patient, and the concomitant use of radiotherapy. The risk estimates also differ by whether treatment-related myelodysplastic syndromes (t-MDS) are included. Similar to t-AML, these preleukemic conditions are frequently fatal.97,98

The association of chemotherapeutic agents with leukemia is related to their mechanism of action. The alkylating agents bind covalently to DNA and have been convincingly linked to leukemia in many studies, although details of the relation between alkylating agents and specific karyotypic subtypes are not understood. Some alkylating agents are less leukemogenic than others; for example, cyclophosphamide appears to be less leukemogenic than melphalan.34,59 MDS and t-AML developing after the use of cyclophosphamide for nonneoplastic conditions, such as rheumatoid arthritis99,100 and Wegener granulomatosis,101 has been described, supporting the theory that the drug causes the leukemia in patients with and without malignancies. Dose-intense cyclophosphamide has been shown to be more leukemogenic than lower doses in adjuvant breast cancer.1102

Nitrosoreas have also been associated with secondary leukemias. In a randomized trial of 3,633 patients with gastrointestinal cancer, 14 leukemias occurred in 2,067 patients who were given semustine, whereas only 1 occurred in 1,566 patients who were given other therapies.92 Higher doses of semustine were more strongly associated with leukemia development.93 BCNU was linked to t-AML in a small clinical trial series of patients with brain cancer.95

The epipodophyllotoxins, etoposide and teniposide, used alone or in combination chemotherapy, increase the risk of leukemia.4,103,104,105 The epipodophyllotoxins bind directly to DNA-topoisomerase II, leading to chromosome breakage and cell death. Epipodophyllotoxin-related leukemias differ from alkylating agent-related leukemias. They develop after a shorter latency, involve translocations rather than deletions at the molecular level, and have a better prognosis. Other

chemotherapeutic agents that inhibit DNA-topoisomerase II, such as doxorubicin, epirubicin and mitoxantrone, may cause leukemia, especially in combination with alkylating agents.91,106,107,108,109 Distinguishing between the effects of chemotherapeutics frequently used in multidrug regimens is challenging. A recent review of mitoxantrone use in multiple sclerosis, a nonmalignant setting in which mitoxantrone has been used as a single agent, suggested increased risk of posttherapy leukemia, and a preponderance of acute promyelocytic leukemia (APL) (63%) among those cases.110








Table 24-1 Carcinogenicity of Selected Anticancer Drugs














































































































































































































Drug


Human Carcinogen


Animal Carcinogen


References


Alkylating Agents





Busulfan (Myleran)


+++


+


66,174


Carmustine (BCNU)


++


+++


95,174


Chlorambucil (Leukeran)


+++


+++


6,8,41,57,96,174,282


Chlornaphazine


+++


+


174, 239 [more]


Cisplatin


+++


+++


62,79,174,283


Cyclophosphamide (Cytoxan)


+++


+++


41,48,49,59,101,174,209,236,240,244


Dacarbazine (DTIC)


+


+++


174


Ifosfamide


ND


+


174


Lomustine (CCNU)


++


+++


9,12,174


Mechlorethamine (nitrogen mustard)


++


+++


7,12,13,174


Melphalan (phenylalanine mustard/Alkeran)


+++


+++


34,49,52,59,174


Mitomycin-C


+


+++


174


Prednimustine


++


±


41,284


Procarbazine


++


+++


8,174


Semustine (methyl-CCNU)


+++


+


92,93,174


Thiotepa


+++


+++


57,174


Treosulfan


+++


ND


57,60,174


Uracil mustard


+


+++


174


Antimetabolites





5-Fluorouracil (5-FU)




174


6-Mercaptopurine (6-MP)




174


Methotrexate




174,285


Mitotic inhibitors





Vinblastine


±



8,174


Vincristine




174


DNA intercalation





Dactinomycin (actinomycin D)



+


174


DNA strand breakage





Bleomycin




174


DNA intercalation and DNA topoisomerase II inhibitiona








Doxorubicin (Adriamycin)


+


+++


17,91,106,128,174


Epirubicin (4′-epi doxorubicin)


±



107,128


Mitoxantrone


±



128


DNA topoisomerase II inhibitiona





Etoposide (VP 16)


++



12,76,83,86


Teniposide (VM 26)


++



128


Razoxane


±



128


+++, carcinogenic; ++, probably carcinogenic; +, possibly carcinogenic; not known to be carcinogemoc; ±, uncertain due to limited, inadequate, or inconsistent data; ND.


a For practical purposes, these anticancer drugs could be classified as human leukemogens; however, the leukemogenic effect is most notably apparent in the presence of alkylating agent or cisplatin therapy.


One class of antimetabolites, the thiopurines, including 6-MP and azathioprine, is also associated with t-AML. A review of > 180,000 solid organ transplant recipients reported an increased incidence of AML, and a dose-response relationship between azathioprine and AML development.111 Another review of 439 children treated for acute lymphocytic leukemia (ALL) suggested that the risk of leukemia correlated with levels of 6-MP metabolites in susceptible patients.112 These patients were treated with multiple cytotoxic agents and had a history of ALL, which confounds interpretation of any direct effect of 6-MP on leukemogenesis. Both alkylator and thiopurineassociated t-AML cases are associated with defective DNA mismatch repair.113

It is difficult to disentangle the separate effects of cumulative dose, dose intensity,114 and duration of administration, because these measures are highly intercorrelated; for example, patients who receive large cumulative exposures usually have long durations of treatment. Dose-response relationships, however, have been reported among patients who are given alkylating agents for Hodgkin lymphoma,6,22 breast cancer,49,102 gastrointestinal cancer,93 NHL,41,44 ovarian cancer,57,59,62,115 testicular cancer,79 and childhood cancer.17 Duration of treatment has been proposed as an additional factor, with continuous exposure of stem cells to a stream of cytotoxic agents, possibly enhancing the progression of a transformed cell.34 The period of highest risk of t-AML is 2 to 10 years after initial treatment. A shorter latency of 1 to 3 years, is seen with topoisomerase II inhibitors than with alkylating agents, frequently 5 to 7 years. In three large series, t-AML risk decreased over time, but remained significantly elevated for >10 years after treatment.25,47

The importance of gender and age on the risk of chemotherapy-related AML is unclear. A few series have indicated that women may be at higher risk than men for second cancer development,25,116,117 but other series have challenged this conclusion.6,7,13,118 Age at treatment has been associated with increased risk in several Hodgkin lymphoma cohorts, with treatment over the age of 40 years carrying a higher risk, perhaps 4-fold higher, than treatment at a youngerage.7,12,14,15,22 This interaction of age and risk, however, was not seen in one large series.25 Chemotherapy for childhood cancer also appears to carry a high risk of leukemia,119,120,121,122,123,124 with adolescents at higher risk than younger patients in one cohort.117

Because radiotherapy is often given in combination with chemotherapy, it is important to determine whether the two modalities together synergistically promote leukemia. One large study of patients with breast cancer suggested a possible interaction between radiation and chemotherapy,49 but such an interaction is not generally seen in Hodgkin lymphoma patients.11 The risk of t-AML is likely to be related to the extent to which bone marrow is exposed in radiation fields. Relatively small increased risks of t-AML have been observed after pelvic irradiation for the treatment of cervical, uterine, rectum, and anus in several large cancer registry-based studies.125,126,127 The risk of radiation-induced t-AML appears to peak 5 to 10 years after pelvic irradiation, with risk persisting beyond 10 years.


Pathogenesis

Historically, treatment-related leukemias have been grouped into two distinct categories: leukemias related to alkylating agents and those secondary to topoisomerase II inhibitors.128 Leukemia related to alkylating agents frequently exhibits deletion of chromosome 5q or 7q, whereas reciprocal translocations are associated with DNA-topoisomerase II inhibitors.128,129 Longer latency periods and more frequent presentation with MDS prior to leukemia also characterize alkylator-associated leukemias.130 Topoisomerase II is an enzyme that cuts doublestranded supercoiled DNA and then ligates the break, to relieve tension and facilitate transcription. Topoisomerase II inhibitors, including the epipodophyllotoxins (e.g., etoposide), anthracenediones (e.g., mitoxantrone), and anthracyclines (e.g., doxorubicin and epirubicin), interfere with the proper ligation of the topoisomerase II-induced double-strand break. One revealing study established that cases of APL (t(15;17)) in patients who received mitoxantrone for breast or laryngeal cancer had a characteristic chromosome 15 breakpoint at a location known to be cleaved by mitoxantrone-poisoned topoisomerase II.131 Similar chromosomal breakpoint “hotspots” at sites of epirubicin/topoisomerase II cleavage were noted in six cases of epirubicin-associated APL. These hotspots were distinct from the sites of mitoxantrone cleavage.132 Taken together, these observations offer a compelling mechanism for topoisomerase II inhibitor-mediated formation of leukemogenic translocations in APL after aberrant DNA splicing at topoisomerase cleavage sites and recombination by the nonhomologous endjoining machinery. This observation was reinforced by a study of t-APL patients who had received mitoxantrone for multiple sclerosis and had DNA breakpoints similar to those seen in the breast/laryngeal cancer-treated cohort.133 Etoposide and anthracyclines are also frequently associated with translocations involving 11q23,68,84,134,135 which lead to chimeric rearrangement of the MLL gene121,136 at a recognized site of epirubicin cleavage.137 The mechanism by which alkylators promote leukemogenesis is less clear but possibly related to selection for cells with poor mismatch repair pathways.138

To account for more of the genetic and clinical complexity of t-AML, Pedersen-Bjergaard and colleagues5 proposed subdividing the t-AMLs into eight distinct pathogenic pathways characterized by distinct genetic alterations. Many of the central regulators of cell growth, division, and apoptosis are implicated in the complex cytogenetic abnormalities in t-AML. RAS, RUNX1, and TP53 have all been noted to be mutated with high frequency in t-AML/t-MDS.139,140,141 RAS mutations in
particular have been associated with progression from t-MDS to t-AML.142 Investigation of the etiology and effect of the genetic abnormalities of t-AML and t-MDS is an area of active research.

Recent research on the pathogenesis of cancer explores epigenetic changes to the cancer genome, in the form of DNA methylation and histone deacetylation. Adding methyl groups to DNA promotor sequences and removing acetyl groups from histones decrease the translation of the affected genes and can have a profound effect on cell survival. Cyclin-dependent kinase inhibitor p15 is an important negative regulator of the cell cycle. In a study of tumor tissue from 81 t-AML patients, p15 methylation density and frequency increased with increasing stage of disease.143 Death-associated protein kinase (DAPK), a potential tumor suppressor gene, was frequently hypermethylated in one study of AML and MDS.144 Cases of t-AML in this study were significantly more likely to be associated with DAPK hypermethylation than cases of de novo AML. Studies of epigenetic abnormalities in t-AML are in their infancy but likely to contribute significantly to the understanding of this disease.


Prognosis

Population survival data from the 1970s and 1980s demonstrated a worse prognosis for t-AML when compared with de novo AML, with an estimated 12-month survival of 10% versus 30%.145 The genetic alterations of t-AML are also closely linked to both prognosis and treatment response. As with de novo AML, favorable risk cytogenetics in t-AML, including t(8;21), inv(16), and t(15;17), are associated with improved survival relative to poor risk and intermediate cytogenetics in t-AML.146,147 Intermediate risk cytogenetics include t(9;11) and other abnormalities not covered in poor or favorable risk. Poor risk cytogenetics include monosomy 5 or 7, deletion 5q or 7q, 11q23 abnormalities other than t(9;11), or complex karyotype consisting of >3 abnormalities in the absence of any of the favorable risk markers. t-AML is more likely than de novo AML to have poor risk cytogenetics, and prognosis within any subgroup is worse for t-AML patients than for de novo AML.147 Anthracyclines are a mainstay of induction therapy in AML and frequently cannot be used in t-AML patients because of previous exposure and cumulative cardiotoxicity. Diminished treatment options likely contribute to the poorer outcome of t-AML. In a recent follow-up of 121 patients with t-AML, median survival among those with favorable, intermediate, and unfavorable karyotypes was 26.7, 15.5, and 5.6 months, respectively.148 t-AML patients with favorable disease are a heterogeneous group with respect to prognosis. A recent analysis of 188 cases of AML with core-binding factor gene abnormalities (either t(8;21) or inv(16)) which included 17 patients with t-AML, reported inferior event-free and overall survival for the t-AML patients compared with matched AML controls.149 Another review of 106 patients with treatment-associated APL found outcomes similar to the outcomes of de novo APL patients.150 APL is unusually responsive to nonanthracycline medications, including all trans-retinoic acid and arsenic trioxide. These additional treatment options likely contribute to the improved outcomes for t-APL patients.


Genetic Predisposition

Efforts to understand host factors that increase susceptibility to iatrogenic malignancies have focused on genetic polymorphisms in genes known to be involved in drug metabolism and DNA repair.151 Candidate genes that are involved in drug metabolism include glutathione S-transferases (GST), nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase (NQO1), and cytochrome P450 (CYP)152 as well as genes involved in nucleotide excision DNA repair, base excision DNA repair, DNA mismatch repair, and cell-death signaling.153 Epipodophyllotoxins and cyclophosphamide are substrates for CYP3A. Several case-control studies have reported an association between wild-type CYP3A and leukemia.103,154 GST participates in inactivating many chemotherapeutics, including doxorubicin, etoposide, cyclophosphamide, and mitoxantrone. GST also detoxifies potentially mutagenic chemotherapy metabolites, and specific polymorphisms of GST P1 are found more frequently in t-AML compared with AML.155 NQO1 participates in epipodophyllotoxin breakdown and is more frequently mutated in patients with t-AML relative to patients with AML.156,157

Genes involved in DNA repair are also implicated in susceptibility to t-AML. The xeroderma pigmentosum group D gene codes for a helicase involved in DNA nucleotide excision repair. A functional lysine to glutamine polymorphism at codon 751 was evaluated in 341 elderly patients with AML. Heterozygotes and glutamine homozygotes had worse prognoses and increased risk of developing t-AML.158 Both RAD51 and XRCC3 participate in the homologous repair of doublestranded DNA breaks. Abnormalities in both genes are associated with AML and more strongly associated with t-AML.159 Abnormalities of DNA mismatch repair, also essential to maintaining genetic integrity, have been implicated in development of t-AML. A case-control study of 133 patients with therapyrelated cancers reported an odds ratio of 5.31 (95% CI, 1.40 to 20.15) for the development of t-AML among those exposed to methylating agents who had a particular variant of MLH1 mismatch repair gene.160 These studies support the hypothesis that genetically determined variation in the pharmacokinetics of chemotherapeutic agents or the ability to repair DNA damage caused by chemotherapy may alter the risk of chemotherapyrelated secondary leukemias. Future translational research is needed to evaluate the possible risk stratification or prevention strategies based on pharmacogenetic profiles.



ACUTE LEUKEMIA AFTER HODGKIN LYMPHOMA

The treatment of Hodgkin lymphoma disease with radiation chemotherapy and combination chemotherapy is one of the great triumphs of modern cancer therapy. In 1960, the diagnosis of Hodgkin lymphoma meant nearly certain death within a few years; today, patients are mostly cured. This success, however, has resulted in long-term complications of clinical importance, including increased second cancer development.172,173 More studies have been published describing the risk of AML after Hodgkin lymphoma than after any other cancer (Table 24-2). Several large series of many thousands of patients report >1,000 secondary leukemias.6,7,8


Type of Chemotherapeutic Agent

Patients with Hodgkin lymphoma can be treated with combinations of drugs and may receive additional cytostatic chemicals for relapse; it is, therefore, difficult to attribute independent effects to individual drugs. Combination chemotherapy, particularly MOPP, has been convincingly linked to secondary leukemias, with mechlorethamine (nitrogen mustard) the most potent leukemogen of the four agents,114 although procarbazine is also a leukemogen in animals.174 Mechlorethamine was the strongest predictor of leukemia risk in most,6,7,12 but not all, studies.8 Doxorubicin, bleomycin, vinblastine, and decarbazine (ABVD) appears to be less leukemogenic than MOPP,15,175 with the cumulative risks estimated at 1.5% and 5.9%, respectively.8 Among 12,411 patients in the International Database on Hodgkin lymphoma, an RR of 17 was linked to MOPP combination chemotherapy.7 The PAVe regimen (procarbazine, melphalan, and vinblastine) appears to be less leukemogenic, with oral melphalan replacing mechlorethamine.14,176 It is encouraging that the chemotherapy regimens introduced in the 1980s appear to carry a lower risk of leukemia than earlier drug combinations did.11 A recent evaluation of 35,511 survivors of Hodgkin lymphoma from international registries reported an excess absolute risk (EAR) of 6.2 cases of AML per 10,000 patient-years (95% CI, 5.4 to 7.1).177 EAR was significantly elevated throughout the 15 years of follow-up, but decreased over time. Throughout the follow-up, the EAR for patients treated between 1984 and 2001 was significantly less than the EAR for patients treated between 1970 and 1984.177 Highdose chemotherapy and autoSCT have become an important therapeutic option in selected patients with relapsed Hodgkin lymphoma.178 Current evidence does not suggest an increased incidence of t-AML among Hodgkin lymphoma patients treated with autologous transplant.179


Dose-Response

The number of cycles or treatments received is directly related to AML risk.6,9,12 Attempts have been made to correlate the risk of AML with measures of cumulative dose, and it is clear that the higher the dose, the higher therisk.12,13,17,22,118 Although quantitative data are sparse, an RR of 10 was reported after 60 mg per m2 mechlorethamine13 in the Union International Contra Cancer series, compared with 0 mg per m2. In the Netherlands, an RR of 10 was linked to 100 mg, which reached an RR of 84 at approximately 220 mg.12 Assuming a body surface area of 1.8 m2 for patients in the Netherlands, these estimates are similar. Although most modern regimens for Hodgkin lymphoma use less leukemogenic agents, high doses of cyclophosphamide in combination with procarbazine and etoposide have been well studied in both the standard and the dose-escalated BEACOPP (bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone) regimens by the German Hodgkin Study Group in patients with advanced stage and high-risk Hodgkin lymphoma. With 10-year followup, standard and escalated BEACOPP have estimated 10-year cumulative incidence of t-AML of 2.2% and 3.0%, respectively; both greater than a 0.4% cumulative incidence seen in patients receiving a regimen with less intense alkylating agents and no etoposide, ABVD/COPP (doxorubicin, bleomycin, vinblastine, dacarbazine, alternating with cyclophosphamide, vincristine, procarbazine, and prednisone).180,181 The risk of t-AML,
therefore, remains an important consideration in treatment choices for patients with Hodgkin lymphoma.








Table 24-2 Acute Leukemia after Hodgkin Disease



















































































































































Study (Reference)


No. of Patients


No. of Observed Leukemias


SIR or RR


Actuarial Risk (%/y)


IARC6


9,552


163


9.0 (RR)



IDHD7


2,411


158


27.5 (SIR)


2.4/20


Canada/US8


9,280


122


23.9 (RR)



BNLI9,10,25


5,519


45


21.1 (SIR)


1.7/20


GHSG286


5,411


46



1/5


Netherlands11,12


1,939


31


34.7 (SIR)


4.0/15


UICC13


1,681


18


64.0 (RR)


2.3/10


Nordic26


1,641


7


17.0 (SIR)


0.8/30


Stanford14


1,507


28


66.0 (SIR)


4.2/10


LESG24,27,287


1,380


24



2.8/14


Milan15


1,329


19



3.6/10


Harvard32


1,319


23


82.5 (SIR)


1.4/10


German-Austrian30,288


1,245


10



0.6/22


Oslo16


1,152


9


24.3 (SIR)



Munich28


1,120


8



1.0/15


Houston18


1,013


14




Gustave Roussy19


892


8


27.6 (SIR)


1.7/15


CALGB20


798


10


133.0


4.0/10


Stanford pediatric29


694


8



1.5/20


SWOG21


659


21



6.2/10


Denmark22


391


17



9.9/10


NCI 1964-198123


473


9



6.0/10 (est)


-, not reported.


BNLI, British National Lymphoma Investigation; CALGB, Cancer and Leukemia Group B; GHSG, German Hodgkin Lymphoma Study Group; IARC, International Agency for Research on Cancer; IDHD, International Database on Hodgkin Disease; LESG, Late Effects Study Group; NCI, National Cancer Institute; SWOG, Southwest Oncology Group; UICC, Union International Contra Cancer.


These institutions are not all independent series. For example, the International Database on Hodgkin Disease also includes data from the British National Lymphoma Investigation.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 19, 2016 | Posted by in ONCOLOGY | Comments Off on Second Malignancies after Chemotherapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access