Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence

Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence

    Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence Steven E. Lipshultz,...

459KB Sizes 0 Downloads 42 Views

    Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence Steven E. Lipshultz, Vivian I. Franco, Stephen E. Sallan, Peter C. Adamson, Rudolf Steiner, Sandra M. Swain, Joseph Gligorov, Giorgio Minotti PII: DOI: Reference:

S1058-9813(14)00008-3 doi: 10.1016/j.ppedcard.2014.09.007 PPC 816

To appear in:

Progress in Pediatric cardiology

Please cite this article as: Lipshultz Steven E., Franco Vivian I., Sallan Stephen E., Adamson Peter C., Steiner Rudolf, Swain Sandra M., Gligorov Joseph, Minotti Giorgio, Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence, Progress in Pediatric cardiology (2014), doi: 10.1016/j.ppedcard.2014.09.007

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: an update of the evidence

T

Steven E. Lipshultz1, Vivian I. Franco1, Stephen E. Sallan2, Peter C. Adamson3,

1

SC R

IP

Rudolf Steiner4, Sandra M. Swain5, Joseph Gligorov6, Giorgio Minotti7 Department of Pediatrics, Wayne State University School of Medicine and the Children’s

Hospital of Michigan, Detroit, MI, USA; 2Division of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children’s Hospital, Department of Pediatrics, Harvard Medical 4

NU

School, Boston, MA, USA; 3The Children's Hospital of Philadelphia, Philadelphia, PA, USA; University of Zurich, Zurich 8006, Switzerland; 5National Surgical Adjuvant Breast and

MA

Bowel Project and Washington Cancer Institute, MedStar Washington Hospital Center, Washington, DC, USA; 6Medical Oncology Department, APHP Hôpital Tenon, Paris and UPMC, Institut Universitaire de Cancérologie, Paris, France; 7CIR and Drug Sciences,

CE P

Corresponding author

TE

D

University Campus Bio-Medico, Rome, Italy.

Steven E. Lipshultz, MD Department of Pediatrics

AC

Wayne State University School of Medicine Children’s Hospital of Michigan 3901 Beaubien Boulevard, Suite 1K40 Detroit, MI 48201 Telephone: 313-745-5870 Fax: 313-993-0390 E-mail: [email protected]

1

ACCEPTED MANUSCRIPT Abstract Advances in treating childhood cancers over the past 40 years have more than doubled 5-year

T

survival rates. More effective use of chemotherapeutic agents has been key to this success.

IP

However, the increase has come at a price: chronic conditions are significantly more

SC R

prevalent in long-term survivors of childhood cancer than they are in the general population, and managing these survivors can be challenging. In patients receiving anthracyclines, cardiotoxicity is the leading cause of morbidity and mortality after relapse and second

NU

malignancies. More than 50% of patients exposed to anthracyclines exhibit some form of

MA

cardiac dysfunction within 20 years after completing chemotherapy, and about 5% develop heart failure. These conditions greatly reduce the quality of life of the individual and also

D

consume substantial amounts of healthcare resources. Dexrazoxane has been used to reduce

TE

anthracycline-related cardiotoxicity in children with cancer, but in 2011, the European Medicines Agency determined, on what it acknowledged were limited data, that dexrazoxane

CE P

was contraindicated in children. Here, we review the evidence for the clinical effects of dexrazoxane in children. Studies published since 2011 have confirmed the efficacy of

AC

dexrazoxane in preventing or reducing anthracycline-related cardiotoxicity in children with cancer, and no new evidence of increased risks for recurrence of primary or second malignancies, or reductions in antitumor efficacy has been reported. As a result, we believe that dexrazoxane should be available to children with high-risk cancers to reduce the risk of cardiotoxicity associated with high-dose anthracycline treatment.

Keywords: anthracyclines, cancer survivorship, cardioprotection, cardiotoxicity, dexrazoxane, doxorubicin, secondary malignancies.

2

ACCEPTED MANUSCRIPT Introduction Advances in cancer treatment have markedly improved oncological outcomes for

T

most childhood cancers, to a point where over three-quarters of those treated will still be alive

IP

in 5 years. However, one repercussion of this success is a marked increase in health problems

SC R

related to anthracycline-induced cardiotoxicity that can present even decades after treatment [1]. After cancer relapse and secondary malignancies, cardiovascular-related problems are the leading cause of morbidity and mortality in childhood cancer survivors [2,3]. Cardiotoxicity

NU

is a limiting adverse consequence of cancer chemotherapy, and its management is pivotal to

MA

the survival, overall quality of life, and well-being of these patients. Dexrazoxane has been used to reduce anthracycline-related cardiotoxicity in children

D

with cancer. In 2011, the Committee for Medicinal Products for Human Use (CHMP)

TE

[European Medicines Agency (EMA)] evaluated what it acknowledged were limited data relating the use of dexrazoxane to prevent anthracycline (such as doxorubicin)-induced

CE P

cardiotoxicity in children [4]. This risk-benefit assessment was triggered by early reports suggesting an increase in secondary malignancies, myelosuppression, and infection in

AC

children treated with dexrazoxane. In particular, the Committee noted that: 

The efficacy of dexrazoxane in children had not been established.



Acute myeloid leukemia-myelodysplastic syndrome (AML/MDS) was a potential risk of dexrazoxane treatment.



Second malignant neoplasms (SMNs) were a potential risk of dexrazoxane treatment. The Committee concluded, “the safety and efficacy of dexrazoxane in children have

not been established and that dexrazoxane should therefore not be used in children due to the risk of second malignancies and potentially negative pharmacodynamic interactions with anthracyclines.” On the basis of these findings, the EMA recommended that the use of dexrazoxane in children and adolescents up to 18 years of age be contraindicated.

3

ACCEPTED MANUSCRIPT In this literature review we re-evaluate the risk-benefit of dexrazoxane in children and adolescents receiving anthracycline therapy using evidence from studies published after the

T

original 2011 EMA appraisal. A MEDLINE search on the use of dexrazoxane in children

IP

identified 77 articles published between January 1, 2011 and September 12, 2014, 15a of

SC R

which are included in this review. Also included are 10b presentations from major oncology scientific meetings (e.g. American Society of Clinical Oncology [ASCO] and the European

NU

Society of Medical Oncology [ESMO]) and key articles from earlier literature.

MA

Childhood cancers and anthracyclines: balancing efficacy with longer-term safety Anthracyclines such as doxorubicin are among common chemotherapeutic drugs used

D

to treat solid and hematological malignancies in children [1,5-7]. In a population-based study

TE

in the UK between 1960 and 1999, 5-year survival increased from 23% to 70% (for leukemias it increased from 6% to 74%), and anthracyclines were administered to 50% of

CE P

children (78% of those with leukemia) [5]. Similar figures have been reported in the US. Since the 1960s, advances in treatment have greatly reduced mortality rates from childhood

AC

cancers; the 5-year survival rate is now approximately 80% [8] with rates in excess of 90% being reported for diseases such as acute lymphoblastic leukemia (ALL), Hodgkin lymphoma and Wilms tumor [9]. A large proportion of these patients become long-term survivors [10], with an estimated 1 in 530 young adults aged 20- to 39-years-old identified as childhood cancer survivors [8]. The improved longevity of childhood cancer survivors in the US is highlighted by the fact that in the year 2010, 379,112 patients were still alive and, of these, about 70% were 20 years or older [8].

a

15 dexrazoxane full references: 1,16-18,22,31,40,43,50,60,70,78,82,83,89

b

10 dexrazoxane abstract references: 19,35,45,51,52,59,71-73,79

4

ACCEPTED MANUSCRIPT Several long-term studies have focused on the late effects of cancer treatment including anthracycline therapy. Childhood cancer survivors have a significantly higher risk

T

of premature death than that of the general population, and a wide range of health problems

IP

that can reduce their overall well-being and quality of life [1,3,5,11-14]. For example, in the

SC R

Childhood Cancer Survivor Study (Table 1), a retrospective analysis of 10,397 adult survivors of childhood cancer, 62% of patients had at least one chronic condition, and in 28% of these patients the condition was life-threatening (Grade 3 or 4 toxicity) [12].

NU

Clinical and subclinical cardiovascular damage, heart failure, coronary artery disease,

MA

and cerebrovascular events are treatment-related health complications in survivors of childhood cancer [3,5,12,15-17]. Cumulative doses of anthracycline of 300 mg/m2 or higher

D

increase the risk of cardiomyopathy, valve disorders, and conduction abnormalities over that

TE

of unexposed survivors; radiotherapy is an added risk factor [18-20]. Heart damage resulting from anthracycline chemotherapy is a serious problem because it reduces quality of life and

CE P

can lead to premature death. This problem is considered in more depth in the section on the cardioprotective effects of dexrazoxane.

AC

Anthracycline cardiotoxicity

Long-term cardiovascular complications of cancer therapies are a major concern in survivors of childhood cancer. These complications pose a threat to overall quality of life, but more specifically, as it relates to this review, to the cardiotoxicity associated with anthracycline chemotherapy [1,21,22], with or without radiotherapy [2,23,24]. The cardiotoxicity of anthracycline drugs, such as doxorubicin, is a major drawback for physicians treating children with cancer because these drugs are associated with an irreversible and dose-dependent loss of cardiomyocytes. Reported risk factors of cardiotoxicity in childhood cancer survivors include cumulative anthracycline doses greater than 300 mg/m2, combination therapy involving 5

ACCEPTED MANUSCRIPT anthracyclines with other toxic drugs such as the topoisomerase II inhibitors (e.g. etoposide), female sex, cardiovascular history, previous cytotoxic therapy, concomitant mediastinal or

T

cranial radiotherapy, younger age at diagnosis, longer follow-up, trisomy 21, and African-

IP

American race [17]. The correlation between high cumulative doses of anthracyclines and

SC R

cardiotoxicity is well established, but lower doses can also be cardiotoxic, with no safe dose established [18]. This was clearly shown in a study specifically designed to evaluate the effects of low-dose anthracyclines (≤100 mg/m2) on LV function in 91 children after a mean

NU

of 9.8 years from diagnosis, as a significant proportion demonstrated subclinical

MA

abnormalities [25].

In 110 children up to 16 years old who were treated with doxorubicin, daunorubicin,

D

or both, 15 experienced cardiac dysfunction within a month after treatment and 28 within a

TE

year [26]. Factors associated with higher incidences of cardiotoxicity included high cumulative dose of doxorubicin, co-treatment with daunorubicin, and AML (which had

CE P

double the risk vs. children with ALL, Hodgkin lymphoma, or non-Hodgkin lymphoma). Seven patients (16%) died from cardiotoxicity in this short-term study, highlighting the need

AC

for better diagnosis, monitoring, and management, especially because the cardiotoxic effects of anthracyclines tend to be progressive and more cases are likely to occur with time. The interplay between the various factors that can adversely impact the cardiovascular system of children with cancer, and the type of cardiotoxicity they cause is both complex and poorly understood [27]. More thorough evaluation of anticancer therapies, including radiotherapy, and the specific cardiovascular changes they are responsible for may help determine the most appropriate diagnostic and screening procedures for childhood cancer survivors [27].

Epidemiology

6

ACCEPTED MANUSCRIPT Formal estimates of the prevalence of cardiotoxicity in children treated with anthracyclines are lacking. Some of the most comprehensive epidemiological data come from

T

population-based studies in Europe and the US [3,5,12,15,28,29]. In the US, for example, the

IP

number of childhood cancer survivors was almost 380,000 in 2010, an increase of 15% from

SC R

the estimated number in 2005 [1,28]. More than 20% had survived for more than 30 years after diagnosis. Survivors of childhood cancer have about a 75% chance of experiencing an eventual treatment-related chronic health problem [12]. Compared with control subjects,

NU

survivors are:

15 times more likely to have heart failure [12]



10 times as likely to experience coronary artery disease [12]



9 times as likely to have a cerebrovascular event [12]



8 times as likely to die from cardiovascular-related disease [29]

TE

D

MA



In addition, 30 years after diagnosis, cardiac-related deaths exceed those caused by

[30].

CE P

cancer recurrence [3], and 45 years after diagnosis 13% of excess deaths are cardiac related

AC

The Childhood Cancer Survivor Study determined the prevalence of a first occurrence of cardiac disorders [29]. Although the frequency of disorders was low overall, the rates were markedly higher for childhood cancer survivors (Figure 1). Compared to their siblings, survivors had the following hazard ratios: 6.3 for pericardial disease, 5.9 for heart failure, 5.0 for myocardial infarction, and 4.8 for valvular abnormalities. The risks for heart failure, valve disease, and pericardial disease were 2 to 5 times as high in patients treated with anthracyclines, and the cumulative risk appears to continue to increase up to 30 years after diagnosis [31]. In total, more than 50% of patients exposed to anthracyclines show some evidence of cardiac dysfunction within 20 years after chemotherapy, with about 5% experiencing heart failure [29,32].

7

ACCEPTED MANUSCRIPT

Pathogenesis

T

Anthracycline-induced cardiotoxicity ranges from acute changes in QT interval

IP

prolongation and cardiac arrhythmias to longer-term changes in coronary vasomotion,

SC R

myocardial ischemia, myocarditis, pericarditis, contractile dysfunction, cardiomyopathy, and heart failure [33,34]. The pathophysiological mechanisms of chemotherapy-related cardiotoxicity are unclear but appear to be multifactorial [17]. Anthracyclines enter

NU

cardiomyocytes by passive diffusion, causing myocardial cell damage or loss through

MA

apoptosis or necrosis [35]. In children with ALL, at a median follow-up of 21 years after doxorubicin treatment, left ventricular (LV) fractional shortening remained decreased, and

D

LV dimension adjusted for body-surface area, which was initially elevated, was significantly

TE

decreased at the last follow-up visit [36]. In later years, the LV thickness-dimension ratio, which was initially reduced, started to increase as a result of wall thickness increasing

CE P

relative to a decreased chamber size (rather than myocardial hypertrophy). The net result of these effects was a progressive reduction in adjusted LV mass and cavity size, which appears

AC

to be a long-term risk in children treated with high cumulative doses of anthracyclines [36]. The molecular and genetic mechanisms of anthracycline-related cardiotoxicity are not completely understood, but the generation of iron-dependent, reactive free radicals that induce lipid peroxidation with consequent membrane damage is plausible [37]. Other possible contributory mechanisms include effects on topoisomerase directly (mediated through topoisomerase 2β) [38,39], decreased activity of Na+, K+-ATPase, mitochondrial damage, DNA damage, derangement of the calcium current with the inhibition of the sarcoplasmic reticulum, accumulation of tumor suppressor protein, and mitochondrial apoptosis in the heart and cardiomyocytes [17,34,40].

8

ACCEPTED MANUSCRIPT Management A major challenge to pediatric oncologists when initially treating children with

T

anthracyclines is incurring the risk of dose-related, progressive cardiotoxicity, which can lead

IP

to reduced LV performance and ultimately to heart failure and possible death [41-44]. For

SC R

example, it was reported that among 3575 patients with Hodgkin lymphoma, after a median follow-up of 22 years since treatment, 1328 had died; 52% from cancer and 15.5% from cardiac disease [45]. The insidious nature of the cardiovascular changes and the fact that

NU

overt cardiac disease may present many decades after cancer treatment pose an important

MA

diagnostic dilemma. Waiting for signs or symptoms of cardiotoxicity to appear is clearly unwise because anthracycline-induced cardiomyopathy is associated with a 2-year mortality

D

rate as high as 60% [40]. Thus, several organizations, including the Children’s Oncology

TE

Group (COG) in North America, have created comprehensive surveillance guidelines for these patients which involves periodic lifetime screening based upon age at treatment,

CE P

cumulative anthracycline dosage and whether or not the heart was irradiated [31,42,44,4649]. In addition to taking a medical history, a physical examination for cardiovascular

AC

symptoms and signs of heart failure, long-term, periodic electrocardiographic and echocardiographic monitoring, especially of LV systolic function, is also recommended. Recently, two research groups independently evaluated the health economic benefits of adhering to the consensus-based COG long-term follow-up guidelines, which advocate lifetime echocardiographic screening for asymptomatic LV dysfunction [47-49]. Using a simulation of life histories and a Markov health states model, applied to 12 risk profiles Wong and colleagues found that compared with no screening, the COG guidelines resulted in an incremental cost-effectiveness ratio of US$ 61,500 and extended life expectancy by 6 months and quality-adjusted life years (QALYs) by 1.6 months, and reduced the cumulative incidence of heart failure by 18% at 30 years after cancer diagnosis [47]. Less frequent

9

ACCEPTED MANUSCRIPT screening achieved most of the clinical benefits associated with following the COG guidelines, and proved to be more cost-effective [47]. In contrast, Yeh and colleagues

T

categorized patients as low- or high-risk based upon the cumulative anthracycline dosage and

IP

a cut point of 250 mg/m2 [48]. In this simulation the lifetime risk for systolic heart failure was

SC R

18.8% in 5-year childhood cancer survivors (aged 15 years) without routine cardiac assessment. Routine cardiac follow-up reduced lifetime risk by 2.3% and 8.7% with assessments every 10 years and annually, respectively, and this resulted in incremental cost-

NU

effectiveness ratios of US$111,600 and US$278,600, respectively. Again, this group found

MA

that less frequent assessments might be a more cost-effective approach [48]. Apart from imaging techniques, sensitive diagnostic tools as predictors of subclinical

D

cardiotoxicity are clearly of interest in treating these patients [50]. Early detection may help

TE

improve long-term outcomes by prompting early treatment. In this regard, serum biomarkers of myocardial damage, such as elevated concentrations of cardiac troponin T and troponin I,

CE P

which detect acute damage, and of N-terminal pro-brain natriuretic peptide (a marker of increased LV wall stress) to monitor chronic changes, may prove useful [22,31,42].

AC

Currently, interest is growing in potential genomic predictors of anthracycline cardiotoxicity. Although this research is in its early stages, results suggest that the allele G of the CBR3 gene involved in the carbonyl reductase pathway may be increased in patients with cardiotoxicity [51], and certain plasma microRNAs [52] may be up-regulated after anthracycline administration. Among survivors of childhood high-risk ALL, the risk of doxorubicin-related myocardial injury was nine times as high in carriers, as that of noncarriers, of mutations in the hemochromatosis gene C282Y allele. This mutation is associated with hereditary hemochromatosis, a genetic disorder that leads to cellular iron overload [53]. Since iron binds to doxorubicin to form a free radical complex, having increased iron may result in more free radical formation and associated injury. There is also evidence that low

10

ACCEPTED MANUSCRIPT cardiac topoisomerase 2β expression may be predictive of reduced cardiotoxicity and that peripheral blood leukocyte levels might therefore be a useful surrogate marker [39].

T

However, prospective studies are required to validate the suitability of these genetic

IP

biomarkers for clinical application.

SC R

Children determined to be at moderate to high risk of cardiotoxicity are most likely to benefit from three evidence-based specific preventive strategies to reduce the likelihood of cardiotoxicity. These include: (1) limiting the cumulative anthracycline dose: (2) minimizing

NU

cardiac exposure to radiotherapy: and (3) using a cardioprotectant such as dexrazoxane

MA

[22,31,44,54]. Other less-cardiotoxic drugs or formulations are also being investigated.

Dexrazoxane and anthracycline-related cardiotoxicity in children and adolescents

D

Dexrazoxane is a bisketopiperazine that undergoes stepwise hydrolysis of its two

TE

piperazine rings to form a one-ring open intermediate; the latter eventually hydrolyses to a

CE P

diacid-diamide that is very similar to ethylenediaminetetraacetic acid (EDTA). The open-ring derivative of dexrazoxane acts as a chelating agent that reduces the number of metal ions that can complex with anthracyclines. As a consequence, it is believed to interfere with iron-

AC

mediated free radical generation, thereby decreasing tissue damage [33,55]. In animal studies, dexrazoxane reduced or prevented anthracycline-induced cardiotoxicity [56,57]. It also reduced acetaminophen-induced hepatotoxicity, alloxaninduced diabetes, bleomycin pulmonary toxicity, and hyperoxia-induced pulmonary damage. It seems likely that these toxicities involve some form of iron-dependent, oxygen-radical production in the pathogenesis of the cellular damage, lending support to the theory that dexrazoxane acts as a free-radical scavenger [34,57]. There are also data showing that dexrazoxane can change the configuration of topoisomerase 2β, preventing the binding of anthracyclines to the enzyme, and this may also help explain its cardioprotective properties [39,58]. Preclinical studies highlighted irreversible doxorubicin-associated cardiac 11

ACCEPTED MANUSCRIPT mitochondrial dysfunction, which was reflected by an increase in mitochondrial DNA in doxorubicin-treated survivors [57]. Patients treated with dexrazoxane had significantly lower

T

mitochondrial DNA copy numbers per cell compared with patients not receiving the

IP

cardioprotectant [59]. To investigate some of these effects further a pilot placebo-controlled

SC R

study assessing the cardioprotective and antiapoptotic effects of dexrazoxane in children <1year-old undergoing heart surgery has been initiated [60]. This study will evaluate postoperative time to resolution of organ failure, development of low cardiac output

NU

syndrome, length of cardiac hospitalization, and echocardiographic indices of cardiac

MA

dysfunction [60].

D

Cardioprotective effects

TE

In the early 1980s, dexrazoxane had shown potential as a cardioprotective agent against anthracycline-related cardiotoxicity in animal models [61] and subsequently in

CE P

women with breast cancer [62,63] and in children with solid tumors or relapsed AML given high cumulative doses of anthracycline (550 to 1150 mg/m2) [64]. After this pediatric study,

AC

the cardioprotective properties of dexrazoxane have been more formally evaluated in children and adolescents treated with anthracyclines (Table 2) [21,22,65-73]. Studies prior to the EMA evaluation in 2011 provided evidence for less subclinical cardiotoxicity [65], improvement in biomarkers of cardiotoxicity [66], better myocardial responses and LV performance [66-69] and fewer cardiac events [69] in children, adolescents, and young adults treated with dexrazoxane plus doxorubicin compared with those on doxorubicin alone. Data on the cardioprotective effects of dexrazoxane are accumulating steadily. Since the EMA review, clinical studies have reported improvements in predictors of cardiotoxicity such as biomarkers [22], as well as significantly less impact on LV structure and performance, in patients treated with anthracyclines and dexrazoxane compared with those

12

ACCEPTED MANUSCRIPT not receiving dexrazoxane (Table 2) [22,70-73]. In early studies of children treated with anthracyclines, the impact of dexrazoxane on clinical outcomes, such as heart failure, could

T

not be ascertained due to the limited duration of follow-up and the progressive nature of

IP

cardiac dysfunction that can take years to develop. However, in a meta-analysis of random

SC R

trials by the Cochrane Group that combined findings from adults and children, heart failure occurred in 11 of 769 patients (1.4%) receiving dexrazoxane plus anthracycline and in 69 of 792 (8.7%) receiving anthracycline alone [16], indicating a clear cardioprotective benefit of

NU

dexrazoxane [risk ratio (RR), 0.18; 95% CI, 0.10 to 0.32; P <0.001].

MA

In another meta-analysis that also combined studies of adults and children, similar findings for dexrazoxane were reported [40]. Finally, a review of only children treated with

D

anthracyclines indicated that dexrazoxane reduced clinical cardiotoxicity (RR, 0.29; P =

CE P

Clinical response

TE

0.001) and clinical plus subclinical cardiotoxicity [RR, 0.43; P <0.001] [71].

One early study, from a number of randomized, prospective clinical trials in adults

AC

with breast cancer, suggested that women treated with dexrazoxane might have lower clinical response rates than those of controls not on dexrazoxane. Interestingly, the time to progression and survival were not significantly different between treatment arms [74]. No other studies in adults or children have confirmed this finding of a reduction in anthracycline antitumor activity, as highlighted in the meta-analysis by the Cochrane group that evaluated response rates (complete and partial remission) and survival times [16]. Of 10 studies involving children and adolescents with a variety of cancers treated with anthracyclines, all 10 found similar response rates and survival times in groups treated with or without dexrazoxane (Table 3) [21, 65,68, 69,75-79]. Mean event-free survival rates (between 5 and 10 years after chemotherapy) ranged from 71% to 87% in dexrazoxane-treated patients and

13

ACCEPTED MANUSCRIPT from 73% to 86% in patients treated without dexrazoxane (Table 3). The 5-year, cardiacevent-free survival rates were 69% and 46%, respectively [69]. In a pooled analysis of three

T

trials involving children treated with doxorubicin for ALL or Hodgkin lymphoma (low- to

IP

intermediate-stage or advanced disease), dexrazoxane was not associated with a risk of

SC R

mortality (dexrazoxane+, 12.8% vs. dexrazoxane-, 12.2% at 10 years; HR 1.02, 95% CI 0.721.43) or relapse (10-year cumulative incidence: dexrazoxane +, 15.6% vs. dexrazoxane-,

MA

Tolerability and second malignancies

NU

18.8%; HR 0.82, 95% CI 0.60-1.10), nor with differential causes of death [79].

The overall tolerability and safety of dexrazoxane has been extensively reviewed

D

[16,80]. Van Dalen et al. reported that dexrazoxane was well tolerated [16]. An earlier review

TE

had reported that dexrazoxane was associated with a higher incidence of leukopenia (78% vs. 68%; P <0.01), which was dose-related and rapidly reversible [80]. Leukopenia may increase

CE P

the risk of serious infections and should be monitored [4]. Furthermore, the Cochrane review concluded that it could not rule out other undesirable adverse effects for dexrazoxane, given

AC

the nature of the patient population and the multiple potent therapies that were being administered [16].

The safety of dexrazoxane was formally reviewed by the EMA, which raised concerns about an increased risk of SMNs in children, including those with AML or MDS and solid tumors [4]. These conclusions were largely based on the findings of Tebbi and colleagues from two randomized trials involving 478 patients (<21 years old) treated for Hodgkin lymphoma as part of the Pediatric Oncology Group (POG) series of clinical trials (Table 4) [81]. At a median follow-up of 58 months, SMNs had developed in 10 patients, 8 of whom had received dexrazoxane [81]. The mean (SD) cumulative incidence rate for SMNs was 3.43% (1.2%) in the dexrazoxane group and 0.85% (0.6%) in the doxorubicin control

14

ACCEPTED MANUSCRIPT group (P = 0.06). In another POG clinical trial of more than 350 patients (<22 years old) with T-cell ALL who were treated with doxorubicin, with or without dexrazoxane (randomized),

T

SMNs developed in only 2.8% of all patients, and no association with dexrazoxane

IP

administration was apparent [77].

SC R

Tebbi and colleagues published the details of a long-term follow-up of 255 patients (of the original 355 <22 years old at the time of treatment) with Hodgkin lymphoma [78]. In the original study, six SMNs were reported, and the updated analysis documented eight new

NU

SMNs. Five of the SMNs occurred as first events (three cases of AML, one thyroid

MA

carcinoma, and one osteosarcoma) and the remaining three occurred after relapse. In the case of the primary SMNs, four were in patients receiving dexrazoxane. The authors attributed

D

these four cases to the concomitant administration of three topoisomerase II inhibitors

TE

(doxorubicin, etoposide, and dexrazoxane) rather than to the effect of any single agent. Since the EMA evaluation was published, several studies have reported no

CE P

statistically or clinically relevant increase in the incidence of SMNs in children or adolescents treated with dexrazoxane (Table 4). Seif and co-workers reported the largest evaluation of

AC

anthracycline therapy to date, a retrospective analysis of 15,532 cancer patients (30% <5 years old; 70% >3 to <15 years old) from 43 Pediatric Health Information System (PHIS) children’s hospitals [82]. Of these, 1406 received dexrazoxane for cardioprotection. The secondary AML rate was 0.21% in the dexrazoxane group and 0.55% in the non-dexrazoxane group. They found no association between dexrazoxane and the risk of SMNs, but they did find an association between etoposide exposure and secondary AML (OR, 2.36; 95% CI, 1.48 to 3.79; P <0.001). Moreover, in an adjusted model, which included etoposide, dexrazoxane was not significantly associated with secondary AML [82]. These findings are consistent with a pooled analysis by Vrooman and co-workers involving more than 500 children with highor very high-risk ALL who received dexrazoxane. The cumulative incidence for SMNs was

15

ACCEPTED MANUSCRIPT 0.24% after 3.8 years [83]. The authors concluded that SMNs were rare in these children and

T

recommended the continued use of dexrazoxane in their treatment.

IP

Discussion

SC R

Over the past 40+ years, oncologists have successfully used anthracyclines to treat several hematological and solid-tumor cancers. This use is possibly best exemplified in pediatric oncology, where 5-year survival rates have more than doubled over this time period.

NU

Today, more children treated for many different cancers live a relatively normal lifespan, a

MA

success that brings a different set of problems. The field of cancer survivorship is a rapidly growing medical specialty. Unlike treatments for many other childhood diseases that do not

D

affect long-term health and well-being, the delayed effects of chemotherapy and radiotherapy

TE

often accompany improved survival [84]. The possible longer-term complications of therapy in survivors of childhood cancer may affect multiple organ systems, including the

CE P

cardiovascular, pulmonary, endocrine, renal/urologic, and central nervous systems. In addition, these individuals remain at risk for a relapse of their primary malignancy or a

AC

second malignancy [85].

After treatment with anthracyclines, the greatest non-cancer-related risk is cardiac toxicity, the severity of which is principally related to cumulative dose, and it can occur from the first administration of drugs such as doxorubicin. Anthracycline-induced cardiotoxicity can be acute (occurring during the first week of therapy), early (<1 year after beginning treatment) or late (>1 year after beginning treatment) [34]. Late-stage cardiotoxicity can be the most challenging because it is progressive and may lead to severe LV contractile dysfunction, cardiomyopathy, and heart failure, and it has a high mortality rate [33,34]. Despite the fact that severe forms of cardiomyopathy are more apparent with higher

16

ACCEPTED MANUSCRIPT cumulative-doses of anthracyclines, adverse cardiovascular changes have also been observed at lower dosages, suggesting that there is no safe dose [1,25,31,40].

T

Management strategies for children at moderate-to high-risk of cardiotoxicity include

IP

limiting the cumulative anthracycline dose, substituting a potentially less cardiotoxic drug or

SC R

formulation, minimizing cardiac exposure to radiotherapy, and using a cardioprotective agent such as dexrazoxane before each dose of anthracycline [1,31,40,42]. There is no evidence that administering doxorubicin by continuous intravenous infusion to children provides any

NU

benefit over bolus infusion in terms of cardioprotection or clinical outcomes [86], and it is

MA

therefore not recommended because continuous infusion increases the risk of infection, mucositis, and thromboembolic events, requires longer hospital stays, and is more costly

D

[40,86].

TE

In terms of pharmacotherapy, numerous studies have documented the effectiveness of dexrazoxane against anthracycline LV cardiotoxicity, and dexrazoxane is thought to be the

CE P

only cardioprotectant proven to be effective against anthracycline treatment in randomised trials [17]. In a recent review Herrmann and Lerman [87] suggested that certain statins, beta-

AC

blockers and angiotensin antagonists were as effective as dexrazoxane in preventing anthracycline-induced cardiotoxicity. These conclusions were based upon a meta-analysis [40], which looked mostly at short-term effects and only included one small study for dexrazoxane in childhood cancer from 1996 [65]. We believe that the substantive evidence for dexrazoxane that we have included in this review highlights longer-term cardioprotection that has not been shown for any other agent as yet. Early diagnosis may also be important in optimizing outcomes in long-term survivors of childhood cancer. Apart from research into some of the more traditional surveillance methods based on cardiac performance (echocardiography) or predictive markers of cardiac damage (cardiac troponins T and I and N-terminal pro-brain natriuretic peptide

17

ACCEPTED MANUSCRIPT concentrations), recent research explored genomic predictors of anthracycline cardiotoxicity, such as allele G of the CBR3 gene, which is involved in the carbonyl reductase pathway,

T

certain plasma microRNAs, and mutations of the C282Y allele of the hemochromatosis gene

IP

[51-53].

SC R

The decision to use of dexrazoxane in children and adolescents has been complicated since the EMA contraindicated its use in these age groups [4]. At present, there appears to be a trans-Atlantic polarization of regulatory views about the clinical benefit of dexrazoxane: a

NU

drug contraindicated in children and adolescents on one side, and in continued use (albeit

MA

very limited, outside of clinical trials) in some US and Canadian children’s hospitals where it is viewed as safe and effective. Indeed, dexrazoxane was recently (August 2014) designated an orphan drug by the FDA for the rare disease “prevention of cardiomyopathy for children

TE

D

and adolescents 0 through 16 years of age treated with anthracycline” [88]. Walker and colleagues performed a retrospective cohort study to investigate patterns

CE P

of dexrazoxane usage in pediatric patients with ALL or AML included in the Pediatric Health Information Systems (PHIS) database in the US [89]. Overall, only 2.4% of ALL patients

AC

(207 of 8733) and 2.0% of AML patients (52 of 2556) received dexrazoxane outside of clinical trials. They found that dexrazoxane administration was limited, and prescribing practices varied widely within and between institutions. The authors suggested that the low off-label usage of dexrazoxane was potentially also related to issues raised by the EMA. During the >3 years since the EMA deemed dexrazoxane to be contraindicated in children, a number of relevant studies have been published that address the EMA’s main concerns: 

Is dexrazoxane an effective cardioprotectant?



Is dexrazoxane associated with an increased risk of SMNs?

18

ACCEPTED MANUSCRIPT 

Does dexrazoxane negatively impact clinical outcomes (response rates or survival times)?

T

In this review, we have integrated published new evidence into the existing

IP

knowledge base of dexrazoxane in pediatric oncology. For all three questions above, the new

SC R

studies summarized in Tables 2 through 4 reinforce earlier findings supporting the cardioprotective properties of dexrazoxane in children and adolescents treated with

NU

anthracyclines. Furthermore, these new studies, including a systematic review undertaken by the Cochrane group confirm equivalent clinical responses and event-free survival over

MA

extended follow-up periods, in dexrazoxane-treated patients [16]. A follow-up of the COG studies the EMA cited as concerning for reduced oncologic efficacy has shown reassuring

D

results [76]. Finally, no new evidence of an increased risk of SMNs with dexrazoxane has

TE

been published, and a large retrospective cohort study of 15,532 cancer patients found no

CE P

association between dexrazoxane and risk of SMNs [82]. This analysis did show an association between etoposide exposure and secondary AML, and others have noted that coadministration of etoposide may be an added risk factor in children treated with dexrazoxane

AC

[83]. These authors reported no SMNs in 487 children with high-risk ALL who were treated with doxorubicin plus dexrazoxane without etoposide, and a single SMN occurred in a veryhigh risk patient who received etoposide. A similar view has been expressed by Tebbi and colleagues [78] in a follow-up to their 2007 study, which was cited in the EMA review of dexrazoxane [4,81]. Tebbi and colleagues more recently concluded that the difference in the number of SMNs in the two treatment groups may have been attributable to the concomitant administration of 3 topoisomerase II inhibitors (doxorubicin, etoposide, and dexrazoxane) rather than the effect of any single agent [78]. They surmised that combining the three drugs could exceed a threshold above which topoisomerase inhibition caused DNA damage in normal tissues. Clearly, further study of the risks associated with concomitant administration 19

ACCEPTED MANUSCRIPT of topoisomerase II inhibitors is needed, though in clinical practice for many pediatric

T

cancers such regimens are generally not employed.

IP

Conclusions

SC R

The 40+ years that anthracyclines have been used in pediatric oncology includes two populations of treated children; those recently diagnosed with cancer, on the one hand, and older patients who received anthracycline therapy as children on the other. For patients who

NU

have already received high-dose anthracycline chemotherapy, routine monitoring of LV

MA

function and cardiac biomarkers, combined with appropriate symptomatic treatment as necessary, may represent best clinical practice.

D

For patients about to be treated with anthracyclines for the first time, several actions

TE

can help limit future cardiovascular problems, but must be done in a manner that minimizes the risk of compromizing established anti-cancer efficacy. Such actions include employing

CE P

lower cumulative anthracycline doses and using anthracycline analogs or liposomal formulations. However, for children requiring higher doses of anthracyclines to achieve

AC

improved cancer-free survival, cardioprotective therapy is needed and there is now convincing evidence of the cardioprotective effects of dexrazoxane [16,68,72,79]. The DanaFarber Cancer Institute Childhood Acute Lymphoblastic Leukemia Consortium, applying the results from clinical trials, including their own studies, now incorporates dexrazoxane into current and future clinical trial protocols involving anthracycline therapy. Similarly, the Children’s Oncology Group includes administration of dexrazoxane in all its research protocols that require treatment with ≥150 mg/m2 doxorubicin or require anthracycline administration at any dose with planned radiation treatment portals that may impact the heart. As the cardiotoxic effects of anthracyclines can be immediate, and the damage to the

20

ACCEPTED MANUSCRIPT cardiomyocytes irreversible and cumulative, dexrazoxane should be administered before the first anthracycline dose and before each therapy cycle [40,86].

T

In sum, the evidence from clinical studies published between 2011 and August 2014

IP

is now compelling that dexrazoxane is an effective cardioprotectant in children and

SC R

adolescents with cancer receiving anthracycline-based treatment. Collectively, these studies confirm and expand upon the safety and efficacy profile of dexrazoxane in children with cancer. Dexrazoxane-induced leukopenia does not result in treatment delays but should be

NU

monitored. No new evidence supporting the claim that dexrazoxane may interfere with the

MA

antitumor efficacy of chemotherapy or that it increases the incidence of SMNs has been published. Cardiotoxicity was the theme for an International Colloquium held in Rome in

D

March 2014. In several sessions that covered cardioprotection and reviewed all the new

TE

evidence for the use of dexrazoxane in children, the consensus favored extending its clinical use [90]. We also encourage its use in protocols involving higher-dose (≥150 mg/m2)

CE P

anthracycline-based chemotherapy in treating childhood cancers, such as high-risk ALL, while the search for less-cardiotoxic drugs and more effective prevention and treatment

AC

strategies continues.

Acknowledgements

We thank Dr. Steve Clissold who provided medical writing services on behalf of Scientific Writing & Publishing Services, Ltd. Vivian Franco is supported by funding from The Michael Garil Fund. Dr. Steven Lipshultz was a paid consultant to The Clinigen Group to help organize the expert panel on cardio-oncology in Newark, NJ in July 2014. Dr. Steven Lipshultz was also supported in part by grants from the National Institutes of Health (HL072705, HL078522, HL053392, CA127642, CA068484, HD052104, AI50274, HD052102, HL087708, HL079233, HL004537, HL087000, HL007188, HL094100, 21

ACCEPTED MANUSCRIPT HL095127, and HD80002), the Children’s Cardiomyopathy Foundation, the Women’s Cancer Association of the University of Miami, the Lance Armstrong Foundation, the STOP

AC

CE P

TE

D

MA

NU

SC R

IP

T

Children’s Cancer Foundation, the Scott Howard Fund, and the Michael Garil Fund.

22

ACCEPTED MANUSCRIPT References 1. Lipshultz SE, Cochran TR, Franco VI, Miller TL. Treatment-related cardiotoxicity in

T

survivors of childhood cancer. Nat Rev Clin Oncol 2013;10:697-710.

IP

2. Tukenova M, Guibout C, Oberlin O, et al. Role of cancer treatment in long-term overall

SC R

and cardiovascular mortality after childhood cancer. J Clin Oncol 2010;28:1308-15. 3. Mertens AC, Liu Q, Neglia JP, et al. Cause-specific late mortality among 5-year

NU

survivors of childhood cancer: the Childhood Cancer Survivor Study. J Natl Cancer Inst 2008;100:1368-79.

MA

4. European Medicines Agency. Assessment report dexrazoxane-containing medicinal products, EMA, 2011. Available at www.ema.europs.eu [last accessed August 30,

D

2014].

TE

5. Curry HL, Parkes SE, Powell JE, Mann JR. Caring for survivors of childhood cancers:

CE P

the size of the problem. Eur J Cancer 2006;42:501-8. 6. Barry E, Alvarez JA, Scully RE, Miller TL, Lipshultz SE. Anthracycline-induced cardiotoxicity: course, prevention and management. Expert Opin Pharmacother

AC

2007;8:1039-58.

7. van Dalen EC, Raphaël MF, Caron HN, Kremer LCM. Treatment including anthracyclines versus treatment not including anthracyclines for childhood cancer. Cochrane Database of Systematic Reviews 2014, Issue 9. Art. No.: CD006647. DOI:10.1002/14651858.CD006647.pub4. 8. Ward E, DeSantis C, Robbins A, Kohler B, Jemal A. Childhood and adolescent cancer statistics, 2014. CA Cancer J Clin 2014;64:83-103. 9. Howlader N, Noone AM, Krapcho M, et al. SEER cancer statistics review, 1975–2011. National Cancer Institute. April, 2014 Based on November 2013 data submission. Available at: http://seer.cancer.gov/csr/1975_2011/ [last accessed August 30, 2014]. 23

ACCEPTED MANUSCRIPT 10. Hinkle AS, Proukou C, French CA, et al. A clinic-based comprehensive care model for studying late effects in long-term survivors of pediatric illness. Pediatrics

T

2004;113:1141-5.

IP

11. Möller TR, Garwicz S, Barlow L, on behalf of the Association of the Nordic Cancer

SC R

Registries; Nordic Society for Pediatric Hematology and Oncology. Decreasing late mortality among five-year survivors of cancer in childhood and adolescence: a population-based study in the Nordic countries. J Clin Oncol 2001;19:3173-81.

NU

12. Oeffinger KC, Mertens AC, Sklar CA, et al. Chronic health conditions in adult survivors

MA

of childhood cancer. N Engl J Med 2006;355:1572-82. 13. Yeh JM, Nekhlyudov L, Goldie SJ, Mertens AC, Diller L. A model-based estimate of

D

cumulative excess mortality in survivors of childhood cancer. Ann Intern Med

TE

2010;152:409-17.

14. Hunger SP, Lu X, Devidas M, et al. Improved survival for children and adolescents with

CE P

acute lymphoblastic leukemia between 1990 and 2005: a report from the children's oncology group. J Clin Oncol 2012;30:1663-9.

AC

15. Gatta G, Rossi S, Foschi R, on behalf of the EUROCARE Working Group. Survival and cure trends for European children, adolescents and young adults diagnosed with acute lymphoblastic leukemia from 1982 to 2002. Haematologica 2013;98:744-52. 16. van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev 2011;15;(6):CD003917. 17. Harake D, Franco VI, Henkel JM, Miller TL, Lipshultz SE. Cardiotoxicity in childhood cancer survivors: strategies for prevention and management. Future Cardiol 2012;8:64770.

24

ACCEPTED MANUSCRIPT 18. Lipshultz SE, Diamond MB, Franco VI, et al. Managing chemotherapy-related cardiotoxicity in survivors of childhood cancers. Paediatr Drugs 2014 Aug 19. [Epub

T

ahead of print].

IP

19. Mulrooney DA, Armstrong GT, Huang S, et al. Cardiac outcomes in aging survivors of

SC R

childhood cancer exposed to cardiotoxic therapy: a report from the St. Jude Lifetime (SJLIFE) Cohort Study. J Clin Oncol 2014;32 (Suppl. 15):10025 (abstr). 20. Nysom K, Holm K, Lipsitz SR, et al. Relationship between cumulative anthracycline

NU

dose and late cardiotoxicity in childhood acute lymphoblastic leukemia. J Clin Oncol

MA

1998;16:545-50.

21. Lipshultz SE, Rifai N, Dalton VM, et al. The effect of dexrazoxane on myocardial

D

injury in doxorubicin-treated children with acute lymphoblastic leukemia. N Engl J Med

TE

2004; 351:145–53.

22. Lipshultz SE, Miller TL, Scully RE, et al. Changes in cardiac biomarkers during

CE P

doxorubicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: associations with long-term echocardiographic outcomes. J Clin Oncol 2012;30:1042-9.

AC

23. Adams MJ, Lipshultz SE, Schwartz C, Fajardo LF, Coen V, Constine LS. Radiationassociated cardiovascular disease: manifestations and management. Semin Radiat Oncol 2003;13:346-56.

24. Landy DC, Miller TL, Lipsitz SR, et al. Cranial irradiation as an additional risk factor for anthracycline cardiotoxicity in childhood cancer survivors: an analysis from the Cardiac Risk Factors in Childhood Cancer Survivors Study. Pediatr Cardiol 2013;34:826-34. 25. Leger K, Slone T, Lemler M, et al. Subclinical cardiotoxicity in childhood cancer survivors exposed to very low dose anthracycline therapy. Pediatr Blood Cancer 2014 Aug 30. doi:10.1002/pbc.25206. [Epub ahead of print].

25

ACCEPTED MANUSCRIPT 26. Shaikh AS, Saleem AF, Mohsin SS, Matloob M, Ahmed MA. Anthracycline-induced cardiotoxicity: prospective cohort study from Pakistan. BMJ Open 2013;3:e003663.

T

27. Lipshultz SE, Adams MJ. Cardiotoxicity after childhood cancer: beginning with the end

IP

in mind. J Clin Oncol 2010;28:1276-80.

SC R

28. Mariotto AB, Rowland JH, Yabroff KR, et al. Long-term survivors of childhood cancers in the United States. Cancer Epidemiol Biomarkers Prev 2009;18:1033-40. 29. Mulrooney DA, Yeazel MW, Kawashima T, et al. Cardiac outcomes in a cohort of

NU

survivors of childhood and adolescent cancer retrospective analysis of the Childhood

MA

Cancer Survivor Study cohort. BJM 2009;339:b4606. 30. Reulen RC, Winter DL, Frobisher C, et al. Long-term cause-specific mortality among

D

survivors of childhood cancer. JAMA 2010;304:172-9.

TE

31. Dillenburg RF, Nathan P, Mertens L. Educational paper: decreasing the burden of cardiovascular disease in childhood cancer survivors: an update for the pediatrician. Eur

CE P

J Pediatr. 2013;172:1149-60.

32. Steinherz LJ, Steinherz PG, Tan CT, Heller G, Murphy ML. Cardiac toxicity 4 to 20

AC

years after completing anthracycline therapy. JAMA 1991;266:1672-7. 33. Zuppinger C, Timolati F, Suter TM. Pathophysiology and diagnosis of cancer drug induced cardiomyopathy. Cardiovasc Toxicol 2007;7:61-6. 34. Raj S, Franco VI, Lipshultz SE. Anthracycline-induced cardiotoxicity: a review of pathophysiology, diagnosis, and treatment. Curr Treat Options Cardiovasc Med 2014;16:315. 35. Lipshultz SE, Rusconi P, Scully RE. Chapter 18: Assessment of cardiotoxicity during anticancer therapy. In Januzzi JL, Bayes-Genis A (eds). NT-proBNP as a biomarker in cardiovascular diseases. Prous Science SA, Barcelona, Spain: 2007; pp 193-98.

26

ACCEPTED MANUSCRIPT 36. Lipshultz SE, Scully RE, Stevenson KE, et al. Hearts too small for body size after doxorubicin for childhood ALL: Grinch syndrome. J Clin Oncol 2014;32 (Suppl.

T

15):10021 (abstr).

IP

37. Wouters KA, Kremer LC, Miller TL, Herman EH, Lipshultz SE. Protecting against

SC R

anthracycline-induced myocardial damage: a review of the most promising strategies. Br J Haematol 2005;131:561-78.

38. Zhang S, Liu X, Bawa-Khalfe T, et al. Identification of the molecular basis of

NU

doxorubicin-induced cardiotoxicity. Nat Med 2012;18:1639-42.

MA

39. Vejpongsa P, Yeh ET. Prevention of anthracycline-induced cardiotoxicity: challenges and opportunities. J Am Coll Cardiol 2014;64:938-45.

D

40. Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of

2013;49:2900-9.

TE

cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer

CE P

41. Lipshultz SE, Lipsitz SR, Sallan SE, et al. Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia. J Clin

AC

Oncol 2005;23:2629-36.

42. Lipshultz SE, Adams MJ, Colan SD, et al. Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention, and research directions: a scientific statement from the American Heart Association. Circulation 2013;128:1927-95. 43. Schlitt A, Jordan K, Vordermark D, Schwamborn J, Langer T, Thomssen C. Cardiotoxicity and oncological treatments. Dtsch Arztebl Int 2014;111:161-8. 44. Truong J, Yan AT, Cramarossa G, Chan KK. Chemotherapy-induced cardiotoxicity: detection, prevention, and management. Can J Cardiol 2014;30:869-78.

27

ACCEPTED MANUSCRIPT 45. Schaapveld M, van Eggermond AM, Janus CPM, et al. Cause-specific mortality among patients with Hodgkin lymphoma (HL) up to 40 years after treatment. J Clin Oncol

T

2014;32 (Suppl. 15): 9517 (abstr).

IP

46. Landier W, Bhatia S, Eshelman DA, et al. Development of risk-based guidelines for

SC R

pediatric cancer survivors: the Children's Oncology Group Long-Term Follow-Up Guidelines from the Children's Oncology Group Late Effects Committee and Nursing Discipline. J Clin Oncol 2004;22:4979-90.

NU

47. Wong FL, Bhatia S, Landier W, et al. Cost-effectiveness of the children’s oncology

MA

group long-term follow-up screening guidelines for childhood cancer survivors at risk for treatment-related heart failure. Ann Intern Med 2014;160:672-83.

D

48. Yeh JM, Nohria A, Diller L. Routine echocardiography screening for asymptomatic left

TE

ventricular dysfunction in childhood cancer survivors: a model-based estimation of the clinical and economic effects. Ann Intern Med 2014;160:661-71.

CE P

49. Steingart RM, Liu JE, Oeffinger KC. Cost-effectiveness of screening for asymptomatic left ventricular dysfunction in childhood cancer survivors. Ann Intern Med

AC

2014;160:731-2.

50. Franco VI, Henkel JM, Miller TL, Lipshultz SE. Cardiovascular effects in childhood cancer survivors treated with anthracyclines. Cardiol Res Pract 2011:134679. 51. Reinbolt RE, Patel R, Pan XJ, et al. Risk factors for anthracycline associated cardiotoxicity. J Clin Oncol 2014;32 (Suppl 15):e20583 (abstr). 52. Leger KJ, Leonard D, de Lemos JA, Nielson D, Mammen PP, Winick NJ. Plasma microRNAs: novel markers of cardiotoxicity in children undergoing anthracycline chemotherapy. J Clin Oncol 2014;32 (Suppl 15):10083 (abstr).

28

ACCEPTED MANUSCRIPT 53. Lipshultz SE, Lipsitz SR, Kutok JL, et al. Impact of hemochromatosis gene mutations on cardiac status in doxorubicin-treated survivors of childhood high-risk leukemia.

T

Cancer 2013;119:3555-62.

IP

54. Zerra P, Cochran TR, Franco VI, Lipshultz SE. An expert opinion on pharmacologic

SC R

approaches to reducing the cardiotoxicity of childhood acute lymphoblastic leukemia therapies. Expert Opin Pharmacother 2013;14:1497-513.

55. Hochster HS. Clinical pharmacology of dexrazoxane. Semin Oncol 1998;25 (Suppl

NU

10):37-42.

MA

56. Herman EH, Ferrans VJ. Examination of the potential long-lasting protective effect of ICRF-187 against anthracycline-induced chronic cardiomyopathy. Cancer Treat Rev

D

1990;17:155-60.

TE

57. Hasinoff BB, Herman EH. Dexrazoxane: how it works in cardiac and tumor cells. Is it a prodrug or is it a drug? Cardiovasc Toxicol 2007;7:140-4.

CE P

58. Lyu YL, Kerrigan JE, Lin CP, et al. Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane.

AC

Cancer Res 2007;67:8839-46. 59. Lipshultz SE, Miller TL, Gerschenson M, et al. Effect of dexrazoxane on impaired mitochondrial structure and function in doxorubicin-treated childhood ALL survivors. J Clin Oncol 2012;30 (Suppl): 9530 (abstr). 60. Stromberg D, Raymond T, Samuel D, et al. Use of the cardioprotectants thymosin β4 and dexrazoxane during congenital heart surgery: proposal for a randomized, doubleblind, clinical trial. Ann N Y Acad Sci 2012;1270:59-65. 61. Herman EH, Ferrans VJ, Myers CE, Van Vleet JF. Comparison of the effectiveness of (+/-)-1,2-bis(3,5-dioxopiperazinyl-1-yl)propane (ICRF-187) and N-acetylcysteine in preventing chronic doxorubicin cardiotoxicity in beagles. Cancer Res 1985;45:276-81.

29

ACCEPTED MANUSCRIPT 62. Speyer JL, Green MD, Kramer E, et al. Protective effect of the bispiperazinedione ICRF-187 against doxorubicin-induced cardiac toxicity in women with advanced breast

T

cancer. N Engl J Med 1988;319:745-52.

IP

63. Speyer JL, Green MD, Zeleniuch-Jacquotte A, et al. ICRF-187 permits longer treatment

SC R

with doxorubicin in women with breast cancer. J Clin Oncol 1992;10:117-27. 64. Bu'Lock FA, Gabriel HM, Oakhill A, Mott MG, Martin RP. Cardioprotection by ICRF187 against high dose anthracycline toxicity in children with malignant disease. Br

NU

Heart J 1993;70:185-8.

MA

65. Wexler LH, Andrich MP, Venzon D, et al. Randomized trial of the cardioprotective agent ICRF-187 in pediatric sarcoma patients treated with doxorubicin. J Clin Oncol

D

1996;14:362-72.

TE

66. Paiva MG, Petrilli AS, Moisés VA, Macedo CR, Tanaka C, Campos O. Cardioprotective effect of dexrazoxane during treatment with doxorubicin: a study using

CE P

low-dose dobutamine stress echocardiography. Pediatr Blood Cancer 2005;45:902-8. 67. de Matos Neto RP, Petrilli AS, Silva CM, et al. Left ventricular systolic function

AC

assessed by echocardiography in children and adolescents with osteosarcoma treated with doxorubicin alone or in combination with dexrazoxane. Arq Bras Cardiol 2006;87:763-71.

68. Lipshultz SE, Scully RE, Lipsitz SR, et al. Assessment of dexrazoxane as a cardioprotectant in doxorubicin-treated children with high-risk acute lymphoblastic leukaemia: long-term follow-up of a prospective, randomised, multicentre trial. Lancet Oncol 2010;11:950-61. 69. Choi HS, Park ES, Kang HJ, et al. Dexrazoxane for preventing anthracycline cardiotoxicity in children with solid tumors. J Korean Med Sci 2010;25:1336-42.

30

ACCEPTED MANUSCRIPT 70. Kang M, Kim KI, Song YC, Shin WG, Oh JM. Cardioprotective effect of early dexrazoxane use in anthracycline treated pediatric patients. J Chemother 2012;24:292-6.

T

71. Shaikh F, Alexander S, Dupuis L, Gupta AA, Nathan PC. Cardiotoxicity and second

IP

malignant neoplasms associated with dexrazoxane in children and adolescents: a

SC R

systematic review of randomized trials and nonrandomized studies. J Clin Oncol 2014;32 (Suppl 15):10093 (abstr).

72. Asselin BL, Devidas M, Zhou T, Camitta BM, Lipshultz SE, and Children's Oncology

NU

Group ALL Committee. Cardioprotection and safety of dexrazoxane (DRZ) in children

MA

treated for newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL) or advanced stage lymphoblastic leukemia (T-LL). J Clin Oncol 2012;30 (Suppl);9504 (abstr).

D

73. Kopp LM, Bernstein ML, Schwartz, et al. The effects of dexrazoxane on cardiac status

TE

and second malignant neoplasms (SMN) in doxorubicin-treated patients with osteosarcoma (OS). J Clin Oncol 2012;30 (Suppl);94877 (abstr).

CE P

74. Swain SM, Whaley FS, Gerber MC, et al. Cardioprotection with dexrazoxane for doxorubicin-containing therapy in advanced breast cancer. J Clin Oncol 1997;15:1318-

AC

32.

75. Barry EV, Vrooman LM, Dahlberg SE, et al. Absence of secondary malignant neoplasms in children with high-risk acute lymphoblastic leukemia treated with dexrazoxane. J Clin Oncol 2008;26:1106-11. 76. Schwartz CL, Constine LS, Villaluna D, et al. A risk-adapted, response-based approach using ABVE-PC for children and adolescents with intermediate- and high-risk Hodgkin lymphoma: the results of P9425. Blood 2009;114:2051-9. 77. Salzer WL, Devidas M, Carroll WL, et al. Long-term results of the pediatric oncology group studies for childhood acute lymphoblastic leukemia 1984-2001: a report from the children's oncology group. Leukemia 2010;24:355-70.

31

ACCEPTED MANUSCRIPT 78. Tebbi CK, Mendenhall NP, London WB, et al. Response-dependent and reduced treatment in lower risk Hodgkin lymphoma in children and adolescents, results of

T

P9426: a report from the Children's Oncology Group. Pediatr Blood Cancer

IP

2012;59:1259-65.

SC R

79. Chow EJ, Asselin B, Schwartz CL, et al. Late mortality and relapse after dexrazoxane (DRZ) treatment: an update from the Children’s Oncology Group (COG). J Clin Oncol 2014;32 (Suppl 15):10024 (abstr).

NU

80. Cvetkovic RS, Scott LJ. Dexrazoxane, a review of its use for cardioprotection during

MA

anthracycline chemotherapy. Drugs 2005;65:1005-24. 81. Tebbi CK, London WB, Friedman D, et al. Dexrazoxane-associated risk for acute

D

myeloid leukemia/myelodysplastic syndrome and other secondary malignancies in

TE

pediatric Hodgkin's disease. J Clin Oncol 2007;25:493-500. 82. Seif AE, Walker DM, Li Y, et al. Dexrazoxane exposure and risk of secondary acute

CE P

myeloid leukemia in pediatric oncology patients. Pediatr Blood Cancer 2014 Mar 26. doi:10.1002/pbc.25043. [Epub ahead of print].

AC

83. Vrooman LM, Neuberg DS, Stevenson KE, et al. The low incidence of secondary acute myelogenous leukaemia in children and adolescents treated with dexrazoxane for acute lymphoblastic leukaemia: a report from the Dana-Farber Cancer Institute ALL Consortium. Eur J Cancer 2011;47:1373-9. 84. Friedman DL, Meadows AT. Late effects of childhood cancer therapy. Pediatr Clin North Am 2002;49:1083-106. 85. Henderson TO, Friedman DL, Meadows AT. Childhood cancer survivors: transition to adult-focused risk-based care. Pediatrics 2010;126:129-36.

32

ACCEPTED MANUSCRIPT 86. Lipshultz SE, Miller TL, Lipsitz SR, et al. Continuous versus bolus infusion of doxorubicin in children with ALL: long-term cardiac outcomes. Pediatrics

T

2012;130:1003-11.

IP

87. Herrmann J, Lerman A. An update on cardio-oncology. Trends Cardiovasc Med 2014

SC R

Jul 22, pii: S1050-1738(14)00062-0. doi: 10.1016/j.tcm.2014.07.003. [Epub ahead of print].

88. FDA. Orphan drug designations and approvals. Available at:

NU

www.accessdata.fda.gov/scripts/opdlisting/oopd/OOPD_Results_2.cfm?_Index_Numbe

MA

r=441314 [last accessed September 2014].

89. Walker DM, Fisher BT, Seif AE, et al. Dexrazoxane use in pediatric patients with acute

D

lymphoblastic or myeloid leukemia from 1999 and 2009: analysis of a national cohort of

2013;60:616-20.

TE

patients in the Pediatric Health Information Systems database. Pediatr Blood Cancer

CE P

90. Ewer M, Gianni L, Pane F, et al. Report on the international colloquium on cardiooncology (Rome, 12-14 March 2014). Ecancermedicalscience 2014 May 29;8:433. doi:

AC

10.3332/ecancer.2014.433.

33

ACCEPTED MANUSCRIPT Table 1. Prevalence and relative risk of selected severe (Grade 3) or life-threatening (Grade 4) health conditions in 10,397 survivors of childhood cancer, compared with their siblings- diagnosed between January 1, 1970 and December 31, 1986 [adapted

Relative risk (95% CI)

SC R

Prevalence, % 1.61

Heart failure

1.24

Second malignant neoplasm

2.38

Severe cognitive dysfunction

0.65

10.5 (2.6 to 43.0)

Coronary artery disease

1.11

10.4 (4.1 to 25.9)

1.56

9.3 (4.1 to 21.2)

0.52

8.9 (2.2 to 36.6)

1.96

6.3 (3.3 to 11.8)

2.92

5.8 (3.5 to 9.5)

2.79

3.5 (2.7 to 5.2)

NU

Major joint replacement

MA

Health condition

IP

T

from Oeffinger et al. 2006 [12]].

Cerebrovascular events

D

Renal failure

TE

Hearing loss

15.1 (4.8 to 47.9) 14.8 (7.2 to 30.4)

AC

Ovarian failure

CE P

Eye problems

54.0 (7.6 to 386.3)

34

ACCEPTED MANUSCRIPT Table 2. Summary of studies investigating the cardioprotective effects of dexrazoxane (DRZ) in children and adolescents with hematological or solid tumors receiving a chemotherapeutic regimen containing doxorubicin. DOX and DRZ dosage

Results and comments

T

Patients

Wexler et al. 1996 [65]

Sarcoma ≤25 y

DOX 50-70 mg/m2 with (n=18) or without (n=15) DRZ (20:1 dose ratio)

Lipshultz et al. 2004a [21]

ALL <18 y

DOX 30 mg/m2 with (n=105) or without (n=101) DRZ 300 mg/m2

Paiva et al. 2005 [66]

Osteosarcoma <28 y

Untreated control group (n=21) and DOX alone (mean dose 348 mg/m2; n=19) or DOX (mean dose 397 mg/m2; n=18) + DRZ (10:1 dose ratio)

de Matos Neto et al. 2006 [67]

CE P

Reference

Osteosarcoma <21 y

DOX 60-70 mg/m2 with (n=18) or without (n=37) DRZ (20:1 dose ratio)

The cumulative dose of DOX was approximately 15% higher in the DRZ group vs. the non-DRZ group. Systolic dysfunction was similar in the 2 groups, but the DRZ group had significantly (P=0.03) better LV performance as assessed by the mean fractional shortening % at 3 follow-up visits [37.2% vs. 35.7%; 38.5% vs. 35.0%; and 38.2% vs. 35.3%, respectively].

Lipshultz et al. 2010a [68]

ALL <18 y

DOX 30 mg/m2 with (n=68) or without (n=66) DRZ 300 mg/m2

After 5 y, mean LVFS and LVESD Z scores were significantly worse than normal in the DOX group [-0.82 and 0.57, respectively], but not in the DOX + DRZ group [-0.41 and 0.15].

Choi et al. 2010 [69]

Solid tumors ≤14 y

DOX 30 mg/m2 with (n=47) or without (n=42) DRZ (10:1 dose ratio)

There were significantly fewer cardiac events with DOX + DRZ than with DOX (27.7% vs. 52.4%; P=0.02 at mean follow-up of 54 m and 86 m, respectively). DOX + DRZ was also associated with less severe CHF (6.4% vs. 14.3%; P=0.049). 5-y cardiac event-free survival was 69.2% for DOX + DRZ and 45.8% for DOX (P=0.04). DRZ

AC

TE

D

MA

NU

SC R

IP

Patients who received DRZ were less likely to experience subclinical cardiotoxicity (22% vs. 67%; P<0.01), had a smaller reduction in LVEF per 100 mg/m2 DOX (1.0 vs. 2.7 percentages points; P=0.02), and received a higher median dose of DOX. Elevated troponin T in 55 of 158 pts. Pts on DOX alone were more likely to have elevated troponin T levels (50% vs. 21%; P<0.001) and extremely elevated troponin T levels (32% vs. 10%; P<0.001). Median follow-up was 2.7 y and event-free survival was 83% in both groups Myocardial responses during low-dose dobutamine stress echocardiography in patients treated with DOX + DRZ were similar to those of patients not on chemotherapy and better than those treated with DOX alone. Both ejection fraction (8.3%) and shortening fraction (7.0%) were significantly (P ≤0.02) lower in the DOXonly group vs. the control group (13% and 11%) and DOX + DRZ group (13.2% and 11%). Patients treated with DRZ had better systolic performance than those on DOX alone, suggesting a cardioprotective effect.

35

ACCEPTED MANUSCRIPT reduced the incidence and severity of early and late anthracycline toxicity. ALL (high risk) <18 y

DOX 30 mg/m2 with (n=105) or without (n=100) DRZ 300 mg/m2

Kang et al. 2012 [70]

Children with cancer <18 y

Anthracycline alone (n=123) or combined with DRZ [lowdose, <100 mg/m2, n=85; high-dose, >100 mg/m2, n=50

Shaikh et al. 2014 [71]

Children treated for cancer and with DRZ as a cardiac protectant

Cardiac troponin (CT) levels were increased 47% and 13% (P=0.05) and N-terminal probrain natriuretic peptide (BNP) by 48% and 20% (P=0.07) in the DOX-only and DOX + DRZ groups, respectively. The increases in CT were associated with abnormally reduced LV mass and end-diastolic posterior wall thickness-to-dimension ratio after 4 y (P<0.01). Increases in BNP were related to abnormal LV thickness-to-dimension ratio, suggesting LV remodeling after 4 y (P=0.01). CT and BNP hold promise as biomarkers for anthracycline-induced cardiotoxicity.

AC

CE P

TE

D

MA

NU

SC R

IP

T

Lipshultz et al. 2012a [22]

During chemotherapy, dose-limiting cardiotoxicity was significantly higher (P=0.006) in the DOX-only group (7.3%) vs. the high- (2.0%) and low-dose (0%) anthracycline groups treated with DRZ. In addition, LVEF and shortening fractions were greater in the low-dose anthracycline + DRZ group vs. the anthracycline only group. Early use of DRZ protects against the development of cardiotoxicity during anthracycline therapy in children with cancer.

Anthracycline alone or combined with DRZ Systematic review limited information

In 2 of 5 RCTs, DRZ was associated with improved shortening fraction Z-score (mean difference 0.61, P=0.002) and thickness dimension Z-score (mean difference 0.66, P<0.001). DRZ was associated with a reduction in clinical cardiotoxicity (relative risk 0.29, P=0.001) and subclinical cardiotoxicity (relative risk 0.43, P<0.001).

Asselin et al. 2012b [72]

ALL (T-cell) ≤21 y

DOX 30 mg/m2 with (n=273) or without (n=264) DRZ 300 mg/m2

After 3 years, Z-scores for LV fractional shortening (-0.05 vs. -0.77), LV wall thickness (-0.13 vs. -0.69) an LV thicknessto-dimension ratio (-0.09 vs. -0.75) were better for the DRZ group vs. the DOX control group. LV fractional shortening remained significantly improved in the DRZ group in a small number of patients after 4-6 y follow-up.

Kopp et al. 2014c [73]

Localized or metastatic osteosarcoma in children and adolescents

In patients with metastatic disease, DOX 375 mg/m2; in patients with localized disease, DOX 450-600 mg/m2. DRZ (10:1 dose ratio) was given for cardioprotection.

None of the 47 patients with metastatic disease evaluated for cardiac effects showed clinical evidence of cardiotoxicity. In 242 patients with localized disease, 1 had a measurable serum troponin-T level and 1 had clinical evidence of cardiotoxicity (grade 3). In this large group of patients with osteosarcoma, DRZ was an effective cardioprotectant.

36

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

Abbreviations: ALL, acute lymphoblastic leukemia; CHF, congestive heart failure; DOX, doxorubicin; DRZ, dexrazoxane; ESD, end-systolic dimension; LV, left ventricular; LVEF, left ventricular ejection fraction; LVFS, left ventricular fractional shortening; m, months; RCT, randomized clinical trial; y, years a Protocol Dana-Farber 95-01; b Protocol POG 9404; c Protocol AOST0121 and POG9754

37

ACCEPTED MANUSCRIPT Table 3. Summary of studies investigating oncological outcomes in children and adolescents with hematological or solid tumors treated with a chemotherapy regimen containing doxorubicin, with or without dexrazoxane. Patients Sarcoma ≤25 y

DOX and DRZ dosage DOX 50-70 mg/m2 with (n=18) or without (n=15) DRZ (20:1 dose ratio)

Lipshultz et al. 2004a [21]

ALL <18 y

DOX 30 mg/m2 with (n=105) or without (n=101) DRZ 300 mg/m2

The median follow up was 2.7 y and the event-free survival at 2.5 y was 83% in both groups (P=0.87).

Barry et al. 2008a [75]

ALL (high-risk) <18 y

DOX chemotherapy 30 mg/m2 alone (n=100) or with DRZ (n=105)

Median follow-up 6.2 y, there was no difference between the DOX + DRZ group and the DOX group in terms of clinical outcome: CR 96 vs. 95%; CCR 77 vs. 76%; or relapse 18 vs. 16%.

Schwartz et al. 2009c [76]

Intermediate- and high-risk HD <22 y

DOX 30 mg/m2 with (n=107) or without (n=109) DRZ 300 mg/m2

Choi et al. 2010 [69]

Solid tumors ≤14 y

DOX 30 mg/m2 with (n=47) or without (n=42) DRZ (10:1 dose ratio)

The 5-y cardiac event-free survival rate was 69.2% in the DRZ group and 45.8% in the DOX control group (P=0.04)

ALL <18 y

DOX 30 mg/m2 with (n=68) or without (n=66) DRZ 300 mg/m2

With a median follow-up for recurrence and death of 8.7 y, event-free survival was 76% in the DRZ group and 77% in the DOX control group (P=0.99).

ALL <22 y

DOX with (187) or without (176) DRZ

5- and 10-y event free survival was 74.4% and 73.0%, in the DOX control group and 73.6% and 71.4% in the DRZ group (P=0.85).

Tebbi et al. 2012d [78]

HD (low-risk) ≤21 y

DOX chemotherapy 25 mg/m2 with (n=127) or without (n=128) DRZ 250 mg/m2

Mean (SD) 8-y event-free survival was 86.8% (3.1%) in the DRZ group and 85.7% (3.3) in the DOX control group (P=0.7). 45% of patients achieved CR after 2 cycles of chemotherapy.

Chow et al. 2014e Pooled analysis [79]

ALL (n=537) advanced HD (n=216), low/intermediate HD (n=255)

DOX 100-360 mg/m2 with (n=507) or without DRZ (10:1 dose ratio; n=501).

With a median follow-up of about 8 y, DRZ + DOX and DOX alone produced similar results in terms of overall mortality (12.8% vs. 12.2% at 10 y; hazard ratio, 1.02; 95% CI 0.72-1.43) and relapse (15.6% vs. 18.8% at 10 y; hazard ratio, 0.82; 95% CI 0.60-1.10). The original cancer accounted for 75.8% of all deaths.

Salzer et al. 2010b [77]

IP

SC R

NU

MA

D

TE

CE P

AC

Lipshultz et al. 2010a [68]

Results and comments 81% of the DOX control group achieved objective responses [3 CR and 10 PR in 16 patients] vs. 80% [4 CR and 12 PR in 20 patients] in the DRZ group. There were also no differences in event-free survival, overall survival, and median survival times.

T

Wexler et al. 1996 [65]

Reference

5-y event-free survival was 83% in the DRZ group and 86% in the DOX control group. CR rates after chemotherapy (70 vs. 60%, respectively) and radiation therapy (94 vs. 86%) were not significantly affected by DRZ.

38

ACCEPTED MANUSCRIPT Asselin et al 2012b [72]

ALL (T-cell) ≤21 y

DOX 30 mg/m2 with (n=273) or without (n=264) DRZ 300 mg/m2

No difference in mean (SD) 5-y event-free survival between the DRZ (76.7% [2.7%]) and DOX control (76.5% [2.8%]) groups (P=0.9).

IP

T

Abbreviations: ALL, acute lymphoblastic leukemia; CCR, complete continuous response; CI, confidence interval; CR, complete response; DOX, doxorubicin; DRZ, dexrazoxane; HD, Hodgkin’s disease/lymphoma; m, months; PR, partial response; y, years Protocol Dana-Farber 95-01; b Protocol POG 9404; c Protocol POG 9425; d Protocol POG 9426; e Protocols POG 9404, POG9425, and 9429.

AC

CE P

TE

D

MA

NU

SC R

a

39

ACCEPTED MANUSCRIPT Table 4. Summary of studies investigating the development of second malignant neoplasms in children and adolescents with hematological or solid tumors treated with a chemotherapy regimen containing doxorubicin, with or without dexrazoxane. Patients

DOX and DRZ dosage

Results and comments

T

Reference Tebbi et al. 2007f [81]

HD ≤25 y

DOX 25-30 mg/m2 (ABVE) alone (n=239) or with DRZ 300 mg/m2 (n=239)

Barry et al. 2008a [75]

ALL (high-risk) <18 y

DOX chemotherapy 30 mg/m2 with (n=105) or without (n=100) DRZ

Median follow-up 6.2 y, the 5-y CIR for SMNs was zero for the DOX + DRZ group with no significant difference between the 2 groups. DRZ was not associated with an increased risk for SMNs

Schwartz et al. 2009c [76]

Intermediate- and high-risk HD <22 y

DOX 30 mg/m2 with (n=107) or without (n=109) DRZ 300 mg/m2

SMNs occurred in 3/107 patients in the DRZ group and 1/109 patients in the DOX control group

Salzer et al. 2010b [77]

ALL (T-cell) <22 y

DOX with (173) or without (159) DRZ

The CIR for SMNs after 5 and 10 y, respectively, was 1.3% and 1.3%, in the DOX control group and 2.3% and 4.2% in the DRZ group (p=0.15).

Vrooman et al. 2011g [83]

ALL (high or very-high risk <18 y). Pooled analysis (3 studies including Barry et al 2008)

TE

D

MA

NU

SC R

IP

Median follow-up 58 m, 4-y CIRs: all SMNs [DOX 0.85 ± 0.6% DOX + DRZ 3.43 ± 1.2% (P=0.06)]; AML/MDS [DOX 0.85 ± 0.6% DOX + DRZ 2.55 ± 1.0% (P=0.16). Adding DRZ to DOX to chemotherapy may increase risk for SMNs

After 3.8 y median follow up (range 0.2 to 13.6 y) mean (SD) overall estimated cumulative incidence of SMNs was 0.24% [0.24%] (95% CI 0.02-1.29%). Thus, in a large population of children with high-risk ALL who received DRZ as a cardioprotectant, the occurrence of SMNs was rare.

HD (low-risk) ≤21 y

DOX 25 mg/m2 with (n=127) or without (n=128) DRZ 250 mg/m2

There were 5 primary SMNs and 3 occurred after relapse. Of the 5 primary SMNs, 4 occurred in the DRZ group and 1 in the DOX control group. The authors attributed the difference to the concomitant use of 3 topoisomerase II inhibitors (including etoposide) rather than to one specific agent.

Seif et al. 2014 [82]

Children with newly diagnosed cancer other than AML in the PHIS database

All children receiving anthracyclines were followed and exposure to DRZ and secondary AML monitored

Of 15,532 children in the cohort exposed to anthracyclines, 1406 received DRZ. The rate for secondary AML was 0.21% in the DRZ group and 0.55% in the group not receiving DRZ. DRZ was not associated with an increased risk of secondary AML.

Chow et al. 2014e [79]

ALL (n=537), advanced HD (n=216), low or intermediate HD (n=255). Pooled analysis [Tebbi et al. 2007, 2012;

DOX 100-360 mg/m2 with (n=507) or without DRZ (10:1 dose ratio; n=501).

With a median follow-up of about 8 y, DRZ + DOX was not associated with an increased risk of second cancers vs. DOX alone (10 vs. 9; hazard ratio, 1.08; 95% CI 0.44-2.67).

Tebbi et al. 2012d [78]

AC

CE P

DOX 30 mg/m2 with DRZ 300 mg/m2 (n=553)

40

ACCEPTED MANUSCRIPT

Children with cancer treated with DRZ

Kopp et al. 2014h [73]

Localized or metastatic osteosarcoma in children and adolescents

11 SMNs were recorded; 8 in the DRZ group and 3 in the DOX control group (P=0.17). At 5 y the mean cumulative incidences were 0.7 ± 0.5% vs. 0.8 ± 0.5% and at 10 y 1.8 ± 0.9% vs. 1.2 ± 0.7%, respectively. DRZ was not associated with an increased risk of SMNs.

Anthracycline alone or combined with DRZ. Systematic review with limited information.

In 5 RCTs there were16/625 (2.5%) SMNs with DRX vs. 6/619 (1.0%) SMNs without DRZ [P=0.06]. In NRSs, there were 6/860 (0.7%) with DRX vs. 18/1,825 (1.0%) SMNs without DRZ [P=0.06].

T

Shaikh et al. 2014 [71]

DOX 30 mg/m2 with (n=273) or without (n=264) DRZ 300 mg/m2

IP

Salzer et al. 2010] ALL (T-cell) ≤21 y

SC R

Asselin et al 2014b [72]

AC

CE P

TE

D

MA

NU

In patients with metastatic SMNs occurred in 3 of 96 patients with disease, DOX 375 mg/m2, metastatic disease and 2 of 272 patients with and in those with localized localized disease. In all, 5/398 patients disease, DOX 450-600 (1.4%) had SMNs, which compares mg/m2. DRZ (10:1 dose favorably with the historical rate of 1%-2% ratio) was given for in the centers involved in the study cardioprotection. Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CI, confidence interval; CIR, cumulative incidence rate; DOX, doxorubicin; DRZ, dexrazoxane; m, months; MDS, myelodysplastic syndrome; NRSs, non-randomized studies; RCTs, randomized clinical trials; SMN, second malignant neoplasm; y, years a Protocol Dana-Farber 95-01; b Protocol POG 9404; c Protocol POG 9425; d Protocol POG 9426; e Protocols POG 9404, POG9425, and 9429; f Protocol POG 9425 and 9426; g Protocols 95-01, 00-01 and 05-01; h Protocols POG9754 and AOST 0121.

41

ACCEPTED MANUSCRIPT Figure 1. Prevalence of cardiac events among 14,358, 5-year survivors of cancer diagnosed before the age of 21 years and a control group of 3,899 siblings of the cancer survivors [adapted from Mulrooney et al. 2009 [29]].

IP

T

2

SC R

1

0

MA

Myocardial Infarction

Pericardial disease

Controls

Valvular disease

CE P

TE

D

Congestive Heart Failure

NU

0.5

Cancer survivors

AC

Prevalence (%)

1.5

42