Late Effects of Cancer Chemotherapy



Late Effects of Cancer Chemotherapy


Brian A. Costello

Charles Erlichman



Although the potential of antineoplastic agents to induce new malignancies was suggested by Haddow1 in 1947 on the basis of the ability of chemical carcinogens to cause growth inhibition, convincing evidence for carcinogenic effects of these agents in humans has been reported only in the past 40 years. The major reasons for this belated recognition of the problem are the long latency periods seen for expression of drug-induced carcinogenicity in humans (3 to 4 years) and the brief survival of most patients treated with chemotherapy in the past. Only in the past four decades have a sizable number of patients with advanced malignancy been cured by chemotherapy or been treated with adjuvant chemotherapy; thus, sufficient time has elapsed and sufficient numbers of individuals are now at risk for second tumors to be seen in clinically significant numbers. Although survival benefits will undoubtedly continue to accrue from the use of these agents and will probably outweigh the risks of second neoplasms, concern for this complication is likely to grow as these antineoplastic drugs gain wider use in adjuvant programs and in nonneoplastic conditions such as solid organ transplantation or autoimmune disease.

Definition of the risk of carcinogenesis as the result of chemotherapy is a difficult task. First, prediction of carcinogenicity at the experimental level depends on test systems that examine the ability of chemicals to cause mutation of bacteria or mammalian cells, malignant transformation of mammalian cells, chromosomal aberrations, or tumors in mice or rats. Such tests are subject to interspecies variability in drug metabolism and target-tissue kinetics, and to other host-specific factors that influence susceptibilities to tumor development. These factors make extrapolation of the quantitative risk to humans a difficult, if not impossible, task. Second, the immune status of the patient is believed to play an important role in determining carcinogenicity, as indicated by the increased risk of lymphoid and cutaneous neoplasms in patients receiving immunosuppressive therapy; in cancer patients, the immune system is suppressed both as a result of the neoplastic process and as a consequence of therapy. This immunosuppression undoubtedly influences the risk of carcinogenesis but is not duplicated in test systems. Finally, the assessment of risk in humans at present is based partly on analyses of retrospective series, which often give incomplete information regarding key parameters of treatment (dose, duration) and which lack a control or untreated population. Such a control population is particularly important in risk assessment because an increased incidence of second tumors, such as acute myelocytic leukemia in patients with Hodgkin’s disease, may exist in the absence of treatment. More recently, analyses of randomized trials that compare adjuvant chemotherapy regimens with no additional treatment have been undertaken with respect to incidence of second malignancies.2, 3, 4, 5, 6 The information derived from these studies clarifies some of the confounding variables mentioned.

With these limitations in mind, this section considers available information concerning the carcinogenic potential of antitumor agents. This discussion examines the common pharmacologic properties shared by antineoplastic agents and classic carcinogens, specific predictions of carcinogenicity based on nonhuman test systems, and clinical evidence for an increased risk of second neoplasms in patients receiving these agents.


Common Features of Antineoplastic Agents and Chemical Carcinogens

The chemical induction of cancer in animals is thought to involve a multistage process with a long latency period. This process can be initiated by a variety of chemical structures that have at least one common thread in their mode of action: an interaction with DNA.7, 8, 9 Initiation results from irreversible genetic alterations, such as mutations or deletions in DNA.10 One of the most carefully studied systems of tumorogenesis is the induction of skin cancer in mice and rabbits by alkylating agents, polycyclic hydrocarbons, and ethyl carbamate. Repeated applications of these agents over long periods result in the development of benign or malignant tumors. Exposure to these compounds in limited doses, however, causes morphologic changes in the epithelium but does not result in tumors unless this stage of initiation is followed by the introduction of a promoter, such as a phorbol ester, an ingredient of croton oil. Promoters are not carcinogenic by themselves but lead to tumor production if applied after the initiating agent. Treatment with a specific promoter before exposure to the initiating agent does not result in tumor formation. This stage of promotion occurs over weeks and months and is reversible in its early stages. Promotion involves changes not in DNA structure but in the expression of the genome mediated through promoter-receptor interaction. The binding of promoter to receptor alters the expression of genes downstream. For example, estrogens and androgens may act as promoters by binding to the estrogen and androgen receptors, respectively, in liver or mammary tissue. The final stage of progression is irreversible and is characterized by karyotypic instability and malignant growth. Thus, carcinogenesis is a multistep process that may be arrested at intermediate stages, that requires a long latency period for induction, and that can be influenced by a promoting agent. For cancer patients, induction of second tumors may require not only an initiator (a DNA damaging agent) but also a promoter, a function
that may be fulfilled by a second chemotherapeutic agent, by radiotherapy, by a disease-related abnormality in metabolism or immune function, or by a somatic mutation or deletion that increases the risk of malignancy. Chemical carcinogens show a diversity of structures but share important metabolic features. Most are inert and require microsomal metabolic activation to positively charged (or electrophilic) intermediates that react with DNA bases. This characteristic of carcinogens is shared by certain antineoplastic agents such as cyclophosphamide, procarbazine, and mitomycin C and is essential in the antineoplastic action of these drugs. Other agents, such as L-phenylalanine mustard and nitrogen mustard, do not require metabolic activation to form alkylating species. Carcinogenicity has also been ascribed to ionizing irradiation, which produces free radicals, such as superoxide or hydroxyl radicals. A number of antitumor drugs have the same ability to promote formation of reactive oxygen intermediates, such agents include those that possess quinone functional groups (doxorubicin hydrochloride and plicamycin [mithramycin]) and those that bind electron-donating heavy metals (such as bleomycin sulfate and hydroxyurea).

Host factors, including enzymes such as glutathione S-transferases (GST), detoxify potentially mutagenic and toxic DNA-reactive electrophiles. Functional polymorphisms of GSTP1 (codon 105 Val allele) are associated with a higher risk of treatment (chemotherapy)-related acute myelogenous leukemia (AML). This risk is particularly relevant in patients receiving agents that are substrates for GSTP1.11 Other polymorphisms of drug-metabolizing enzymes, including cytochrome P450 3A4, NAD(P)H:quinine oxidoreductase and myeloperoxidase, may be markers of susceptibility to genotoxicity.12 Direct genotoxicity may not be the sole explanation for drug-induced carcinogenesis. Short and long interspersed elements (SINEs and LINEs) comprise one quarter of the human genome and are spread throughout the genome through retrotransposition. In retrotransposition, an element is transcribed into RNA and then converted back to DNA by reverse transcription. The copied DNA is then reinserted into a new location. Genotoxic agents and γ irradiation have been shown to induce SINE transcription and reverse transcriptase activity. This observation suggests that genotoxic exposure may lead to genomic mutation through both DNA damage and through potentially mutagenic mobile elements in the genome.13

The identification of oncogenes and suppressor genes has added another variable to the equation. Several possible mechanisms of actions have been proposed. The loss of one allele in a tumor suppressor gene such as p53 can potentially increase the risk of a drug-induced mutation in the other allele and development of the malignant phenotype. Oncogenes can be activated by a variety of mechanisms summarized in Table 42-1. Point mutations, chromosomal translocations, and gene amplification can alter expression of these genes. Just as altered oncogene expression and mutation occur with exposure to potential carcinogens, exposure to carcinogenic antitumor drugs likely alters oncogene expression and increases the risk of second malignancies.


Carcinogenicity of Antineoplastic Agents Based on Nonclinical Testing

Five test systems for carcinogen exposure are available. Mutagenesis assays such as the Ames test attempt to quantify the frequency with which a chemical induces mutational events based on the assumption that mutagenicity correlates with the likelihood of causing cancer in animals. In extensive testing of a wide variety of agents previously documented to be carcinogens and noncarcinogens, 90% of the known carcinogens gave positive results in the Ames assay and 87% of the noncarcinogens were inactive.14,15 Many of the antineoplastic agents in use today have been examined in the Ames system and some of the results are incorporated in Table 42-2.16, 17, 18, 19 At least seven classes of agents known to contain carcinogenic compounds are poorly detected in the Ames system, including azo compounds, carbonyl, hydrazine, chloroethylene, steroid, and antimetabolite structures. Cytogenetic studies attempt to correlate drug-induced chromosomal aberrations such as sister chromatid exchanges (SCEs) with carcinogenicity.20 Assay of SCE, a type of chromosomal study that detects the exchange of small DNA fragments between sister chromatid pairs, has particular appeal because the effects of chemotherapeutic agents can be assessed in vivo by performing this test on peripheral lymphocytes from patients receiving antineoplastic therapy. Studies of lymphocytes from patients before and at intervals after chemotherapy have shown a marked increase in SCEs after the administration of lomustine (CCNU), dacarbazine, and mitomycin.20, 21, 22








TABLE 42.1 Oncogene activation




















Alteration


Effect


Base mutation in coding sequence


New gene product with altered activity


Base deletion in noncoding sequence


Altered regulation of normal gene product


Chromosomal translocation


Altered message and level of expression


Gene amplification


Increased gene expression


Adapted from Pitot HC. The molecular biology of carcinogenesis. Cancer 1993;72:962-970, with permission.


Tests of oncogenesis in tissue culture are based on the hypothesis that agents that produce neoplastic transformation in culture are likely to be carcinogenic in the whole animal. This approach assumes that the drug concentration, duration of exposure, and metabolism of the suspected carcinogen are relevant to the in vivo situation, which may not be valid. Carcinogenicity studies in animals to predict the risk in humans are usually conducted in rodents over extended periods and at great expense. The primary drawbacks of this system are the known species, sex, and age dependencies of drug metabolism in rodents and the lack of pharmacologic information that would allow an extrapolation of results from rodents to humans.

The fifth approach, a measure of carcinogen exposure, uses detection of carcinogen-macromolecular adducts or somatic gene mutation in either target tissue or peripheral blood elements in animals or in man. The assumption here is that adduct formation or mutation will lead to cancer.









TABLE 42.2 Results of testing antineoplastic agents in three systems for carcinogenicity























































































































Agent


Ames test


SCEs


Animal studies


Mechlorethamine hydrochloride


+


+


+


Cyclophosphamide


+


+a


+


Melphalan


+


+


+


Thiotriethylene phosphoramide (thiotepa)


+


+


+


Chlorambucil


NR


+


+


Procarbazine hydrochloride



+a



Lomustine (CCNU)


NR


+b



Doxorubicin hydrochloride


+


+


+


Streptozotocin


+


NR


+


Bleomycin sulfate



+c



Actinomycin D (Dactinomycin)



±


+


Mitomycin C


+


+


+


Dacarbazine (DTIC)


NR


b


+


Cisplatin


+


+


NR


5-Fluorouracil



NR


NR


6-Mercaptopurine


+



+


Cytosine arabinoside (ara-C)



NR



Vincristine sulfate



±



Vinblastine sulfate



NR


+


Pemetrexed



NR



Oxaliplatin


+


NR


NR


Methotrexate sodium



+


a Drug must be activated.

b Test done on patient lymphocytes after treatment with agent.

c Concentration giving positive results also causes significant numbers of other chromosomal aberrations.

+, Positive result reported in at least one study; −, no positive result reported; ±, slight decrease over control (which is of unknown significance); NR, no result reported.



Clinical Studies Implicating Antineoplastic Agents in Carcinogenesis

Although experimental evidence demonstrating the carcinogenic potential of many antineoplastic agents is abundant, the clinical evidence of this problem has been slower to appear. The fact that the rate of development of “secondary” cancers in patients with malignant lymphoma, pediatric cancers, ovarian cancer, and breast cancer is higher than that seen in an age-matched normal population has become clear. Many good reviews of this topic are now available in the medical literature.23, 24, 25, 26 Data reported more recently have come from hospital-based, national, and international tumor registries and from longer follow-up of chemotherapy and hormonal therapy studies.

One method that is used to identify treatment factors involved in the development of new cancers from a registry is the “nested” case-control study. Patients in the registry who develop a second cancer are compared with others who did not. These comparisons have provided a better estimate of the risks and identified factors that influence the development of second cancers. Clinical information about the total dose of drugs, concomitant therapy, and the duration of treatment is important in estimating risk. For some drugs such as the alkylating agents or etoposide, a threshold exists above which the risk of neoplasia rises sharply. Another issue in assessing the true risk of second cancers from cytotoxic agents is the existence of other factors that may also influence their development. An underlying increased incidence of second malignancy is found independent of therapy in patients with retinoblastoma, Wilms’ tumor, multiple myeloma, Hodgkin’s disease, and other tumors such as those associated with the hereditary nonpolyposis colorectal cancer syndrome. Other therapies used to treat the cancer, particularly radiation therapy, also impact the development of secondary cancers. An increase in solid tumors after therapy for Hodgkin’s disease and testicular cancer is most likely related to radiation rather than chemotherapy. In many reports, combination treatment regimens or regimens using irradiation and chemotherapy were used. Thus, the carcinogenic effects cannot necessarily be ascribed to one compound of the regimen with certainty, although the use of the nested case-control method may allow conclusions to be drawn regarding the carcinogenicity of different components of the regimen.

Interpretation of studies in this area must also take into consideration the statistical methods used to assess relative risk (RR).27 The use of a person-years-of-risk analysis assumes that the yearly incidence of second malignancies is constant for the entire follow-up period and does not allow for the fact that a patient must live a certain
time through the latency period before the occurrence of a second malignancy. Such an analysis allows a reasonable estimate of the carcinogenic effects of a single therapy, but its use when comparing two treatments results in bias against the treatment that leads to a longer survival. Many studies compare the risk of cancer in the treated group with that of an age-matched cohort in the normal population to determine an RR. For a tumor that is uncommon in this age-matched population, a 5-fold to 10-fold increase in the RR sounds impressive but may only translate into a problem for fewer than 1% of patients who received therapy. On the other hand, small increases in RR for the more common solid tumors such as lung or breast cancer translate into a much greater problem in terms of absolute risk. One method that is useful in determining the overall impact of a secondary cancer in a population is to describe it in terms of the number of new cancers that occur per 10,000 patients treated.

Based on information currently available, one can categorize antineoplastic agents into high, moderate, low, and unknown risk groups on the basis of their oncogenic potential in humans (Table 42-3). This classification is based on reports of second malignancy in patients treated for both hematologic and solid tumors, with additional information coming from trials of cytotoxic agents in patients with immune diseases or after organ transplantation. Given that the latency period for the development of secondary cancers can range from 1 year (e.g., for etoposide-induced leukemias) to 20 years for solid tumors, the risk for many newer agents such as paclitaxel, docetaxel, irinotecan hydrochloride, gemcitabine hydrochloride, pemetrexed, and oxaliplatin cannot yet be properly determined. Furthermore, the impact of the new targeted agents such as imatinib, gefitinib, and erlotinib on formation of secondary malignancies is unclear. Neither gefitinib nor erlotinib has demonstrated genotoxic potential with in vitro or in vivo assays. Imatinib has been shown to induce benign and malignant tumors of preputial/clitoral gland, kidney, and urinary bladder in rats. This has not been demonstrated in humans.28 A true assessment of agents primarily used in palliative therapy is also difficult because most patients may not survive long enough for problems such as second cancers to manifest.








TABLE 42.3 Categorization of antineoplastic agents according to carcinogenic risk in humans




























High risk


Moderate risk


Low risk


Unknown


Melphalan


Doxorubicin hydrochloride


Vinca alkaloids


Bleomycin sulfate


Mechlorethamine hydrochloride


Thiotriethylene phosphoramide (thiotepa)


Methotrexate sodium
Cytosine arabinoside


Taxanes
Busulfan


Nitrosoureas
Etoposide
Teniposide
Azathioprine


Cyclophosphamide


5-Fluorouracil


Gemcitabine hydrochloride
Irinotecan hydrochloride
Mitoxantrone hydrochloride
Pemetrexed



Procarbazine hydrochloride
Dacarbazine (DTIC)
Cisplatin


L-asparaginase
Carboplatin


Oxaliplatin



Second Malignancies in Specific Populations of Cancer Patients


Pediatric Patients

Long-term survival is now possible for many patients with pediatric malignancies. Overall, the risk of developing a second cancer 20 years after childhood cancer has been estimated at 8% to 20%.29,30 The Childhood Cancer Survivor Study reported the RR of second malignancies in survivors who received treatment between 1970 and 1986 as follows: non-Hodgkin’s lymphoma (NHL), 3.2; leukemia, 5.7; and Hodgkin’s disease, 9.7.31 A more recent report updated the results through January 1, 2006 and the 30-year cumulative incident for second malignant neoplasms was found to be 9.3%.32 Other large cohorts have recently been reported. In a British retrospective cohort study of 16,541 patients treated for childhood cancer and who survived 3 years or more, 278 second malignant neoplasms were identified.33 These patients were found to have an increased risk for developing a malignancy beyond that of the general population, as evidenced by a standardized incidence ratio (SIR) of 6.2. The cumulative risk of a second malignant neoplasm by 25 years was found to be 4.2%. Another study of childhood cancer survivors used Surveillance, Epidemiology, and End Results (SEER) cancer registries and included a total of 25,965 children diagnosed with cancer and who survived 2 months or more. These patients were observed for an average of 8.9 years and a total of 433 new primary cancers were identified. The most common subsequent primary cancers were noted to be that of the female breast, central nervous system, bone, thyroid, and soft tissue. Other common malignancies were melanoma and acute nonlymphocytic leukemia (ANLL). The authors found a nearly sixfold increase risk of developing a new cancer as compared to the general population.34 Most recently a very large series of 47,697 patients treated for childhood cancer in the Nordic countries was published. This cohort included children and adolescents treated between 1943 and 2005 for childhood cancer. In the period of surveillance, 1,180 asynchronous second cancers were observed. As in the other study, this indicated an increased
risk of developing second primary cancers for patients previously treated for childhood malignancy. The SIR in this study was 3.3.35 Further analysis from the childhood cancer survivor study reveals that survivors of childhood cancer are 15.2 times more likely to die of a subsequent cancer than match controls from the general population. The risk factors for death due to subsequent malignancy include previous therapy with alkylating agents, epipodophyllotoxins, or radiotherapy.36

One consistent finding has been an association between treatment with etoposide or teniposide and secondary AML, often with monocytic features. One series examined 205 children with acute lymphoblastic leukemia (ALL) who were treated with a four-drug induction consisting of prednisone, L-asparaginase, vincristine, and daunorubicin hydrochloride followed by maintenance therapy with oral 6-mercaptopurine, methotrexate, L-asparaginase, etoposide, and cytarabine. The etoposide was given twice weekly. The risk of secondary AML at 4 years was 5.9% ± 3.2%. Because none of these children received alkylating agent therapy or irradiation, etoposide was most likely responsible for these secondary leukemias.37 In another series Pui et al. reported secondary AML in 21 of the 734 patients, with an overall cumulative risk at 6 years of 3.8% (range, 2.3% to 6.1%).38 At the Dana-Farber Cancer Institute, when no epipodophyllotoxin was used, only 2 of 752 patients with ALL who entered complete remission after induction therapy developed AML after a median follow-up of 4 years.39

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

Stay updated, free articles. Join our Telegram channel

May 27, 2016 | Posted by in ONCOLOGY | Comments Off on Late Effects of Cancer Chemotherapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access