Open Access

The mRNA expression of Il6 and Pdcd1 are predictive and protective factors for doxorubicin‑induced cardiotoxicity

  • Authors:
    • Syu-Ichi Kanno
    • Akiyoshi Hara
  • View Affiliations

  • Published online on: December 3, 2020     https://doi.org/10.3892/mmr.2020.11752
  • Article Number: 113
  • Copyright: © Kanno et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Anthracyclines, such as doxorubicin (DOX), have been widely used in the treatment of a number of different solid and hematological malignancies. However, these drugs can inflict cumulative dose‑dependent and irreversible damage to the heart, and can occasionally lead to heart failure. The cardiotoxic susceptibility varies among patients treated with anthracycline, and delays in the recognition of cardiotoxicity can result in poor prognoses. Accordingly, if the risk of cardiotoxicity could be predicted prior to drug administration, it would aid in safer and more effective chemotherapy treatment. The present study was carried out to identify genes that can predict DOX‑induced cardiotoxicity (DICT). In an in vivo study, mice cumulatively treated with DOX demonstrated increases in serum levels of cardiac enzymes (aspartate aminotransferase, lactate dehydrogenase, creatine kinase MB isoenzyme and troponin T), in addition to decreases in body and heart weights. These changes were indicative of DICT, but the severity of these effects varied among individual mice. In the current study, the correlation in these mice between the extent of DICT and circulating blood concentrations of relevant transcripts before DOX administration was analyzed. Among various candidate genes, the plasma mRNA levels of the genes encoding interleukin 6 (Il6) and programmed cell death 1 (Pdcd1) in blood exhibited significant and positive correlations with the severity of DICT. In an in vitro study using cardiomyocyte H9c2 cells, knockdown of Il6 or Pdcd1 by small interfering RNA was revealed to enhance DOX‑induced apoptosis, as determined by luminescent assays. These results suggested that the levels of transcription of Il6 and Pdcd1 in cardiomyocytes serve a protective role against DICT, and that the accumulation of these gene transcripts in blood is a predictive marker for DICT. To the best of our knowledge, this is the first report to demonstrate a role for Il6 and Pdcd1 mRNA expression in DICT.

Introduction

Anthracyclines are chemotherapy drugs that inhibit the activity of topoisomerase IIα, thereby negatively impacting DNA synthesis in cancer cells; anthracyclines have been widely used in the treatment of various solid and hematological malignancies (1,2). It is well known, however, that anthracyclines can inflict severe damage to the heart, and cardiotoxicity represents one of the most important adverse effects of this class of compounds (14). Anthracycline-induced cardiotoxicity is cumulative, dose dependent, and irreversible; therefore, these drugs increase the risk of heart failure, in turn leading to increased mortality and decreased quality of life in patients (14). Although the precise mechanism of cardiotoxicity is not fully understood, cardiac apoptosis (programmed cell death) mediated by either of the following two mechanisms has been proposed: One is the excessive production of reactive oxygen species, the other is DNA damage by inhibition of topoisomerase IIβ in cardiomyocytes (1,2,5).

Retrospective pooled analysis by Swain et al (3) documented that rates of heart failure caused by doxorubicin (DOX), a typical anthracycline drug, were 5, 26 and 48% at cumulative doses of 400, 500 and 700 mg/m2, respectively, providing a rationale for limiting the cumulative lifetime dose of DOX. Nevertheless, there are individual differences in the maximal cumulative dose tolerated, and anthracyclines can lead to the occurrence and development of heart failure even at lower doses (57). In addition, the risk of heart failure may be augmented by pre-existing loading conditions such as hypertension and valvular disease (8), and by combination with other chemotherapy drugs such as molecularly targeted drugs and immune checkpoint inhibitors (9). These facts indicate that anthracyclines have no safe treatment dose.

Several strategies for reducing the risk of heart failure in anthracycline-treated patients have been proposed, including pharmacological therapy to prevent cardiotoxicity (10). Although some antioxidants, such as vitamin E and probucol, have been observed to provide cardioprotective action in DOX-treated animal models (2,11), there is (to date) no direct evidence supporting clinically efficacy (12). Alternatively, it has been demonstrated (in both animals and humans) that standard drugs for heart failure, such as β-blockers and angiotensin-converting enzyme (ACE) inhibitors/angiotensin II receptor blockers (ARBs), are effective in attenuating DOX-induced cardiac damage (2,10,13). In fact, pharmacological therapy with these drugs is generally accepted as a preventive strategy against cardiac dysfunction in anthracycline-treated patients (10). It should be noted, however, that the response rate to the pharmacological therapy progressively decreases with increasing time delay between the end of chemotherapy to the onset of cardiac dysfunction (i.e., with delayed start of the pharmacological therapy) (13,14). The delayed recognition of cardiotoxicity results in a poor prognosis, presumably because of the accumulation of irreversible cardiac dysfunction and a lack of clinical response to the pharmacological therapy. Therefore, early detection of cardiac abnormalities and early adequate therapy would be extremely important in preventing the occurrence and development of heart failure after anthracycline therapy (15).

If we could predict the risk of cardiotoxicity prior to the administration of anthracycline, it would help to render chemotherapy in cancer patients safer and more effective. Genetic factors have been shown to determine individual susceptibility to DOX-induced cardiotoxicity (DICT) (16,17). Therefore, in the present study, we identified genes closely associated with DICT (i.e., predictive biomarkers) in mice in vivo, and then examined the roles of these genes in DOX-induced apoptosis of H9c2 cells (rat cardiac myoblasts) in vitro.

Materials and methods

Animals and chemicals

Four-week-old adult male C57BL/6J mice (22–24 g each), were obtained from Japan SLC (Hamamatsu, Japan). The animals were maintained on a 12-h/12-h light/dark cycle in a temperature- and humidity-controlled room. Experiments were conducted in accordance with the standards established by the Japanese Pharmacological Society and were approved by the Tohoku Medical and Pharmaceutical University of Institutional Animal Care and Use Committee (Experimental Protocol no. 18013). Animals were allowed free access to laboratory pellet chow (CE-2; CLEA Japan, Inc.) and water throughout the experiments. DOX was purchased from Sandoz K. K. All other reagents, unless stated, were of the highest grade available and were supplied by either Sigma Chemical Co. or Wako Pure Chemical Industries, Ltd.

DOX administration and collection of samples

One week before the start of dosing, 50 µl of blood was collected from a cut on the tail vein of each mouse, and total RNA was extracted for reverse transcription-quantitative PCR (RT-qPCR). Mice were injected intraperitoneally with 3.33 mg/kg DOX every other day for 18 days, resulting in a cumulative DOX dose of 30 mg/kg. This cumulative dose of DOX in mice is estimated to be comparable to approximately 100 mg/m2 in humans (18), which is considerably lower when compared to clinical cardiotoxic doses (3). However, the cumulative dose of DOX we used is sufficient to cause damage to the murine heart, as supported by many other studies (1921); thus, the susceptibility to DICT may be greater in mice than in humans. The control mice were injected intraperitoneally with an equivalent volume of saline. Animals were euthanized by exsanguination via jugular puncture on the seventh day after the last DOX administration under inhalation anesthesia with 2% isoflurane; whole blood and heart samples were collected. A small portion of whole blood was transferred into ethylenediaminetetraacetic acid (EDTA)-coated tubes and used for measurement of hematological parameters. The remaining blood was immediately centrifuged at 1,000 × g for 10 min at 4°C to separate serum for measurement of cardiac injury parameters. Heart samples were immediately frozen and stored at −80°C until used for RNA isolation. As an index of cardiac hypertrophy, the body weight-normalized heart weight was calculated.

Measurements of hematological parameters and serum cardiac injury parameters

As hematological parameters, white blood cells (WBCs), red blood cells (RBCs) and platelets were counted, and hemoglobin (Hb) levels and hematocrit (Hct) were assessed using a hematology analyzer (EYM-230; Erma, Inc.). As indices of cardiac injury, serum levels of lactate dehydrogenase (LDH), aspartate aminotransferase (AST), creatine kinase MB isoenzyme (CK-MB), and troponin T were determined; the first two were measured using a colorimetric kit (Wako) as described in our previous reports (22,23), and the last two were measured using a diagnostic kit (Cloud-Clone Corp.) according to the supplier's instructions.

Cell culture

Rat heart-derived embryonic myoblast H9c2 (21) cells were obtained from DS Pharma Biomedical. Cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum, 100 U/ml penicillin G and 100 µg/ml streptomycin at 37°C in a humidified 5% CO2−95% air incubator under standard conditions. Staining with 0.2% Trypan blue was performed to determine viable cell counts. To maintain exponential growth, cells were seeded at a density of 5×104 cells/ml and were passaged every 3–4 days. For the remaining assays cells were cultured in 2-ml aliquots in 35-mm dishes.

RNA extraction and RT-qPCR

Total RNA was isolated from blood using a NucleoSpin® RNA blood kit (MACHEREY-NAGEL GmbH & Co. KG) according to the manufacturer's instructions. Total RNA was purified from mouse heart and H9c2 cells by extraction using ISOGEN reagent (Nippon Gene) according to the manufacturer's protocol. The quantity and the purity of extracted RNA were determined on a NanoDrop ND-1000 spectrophotometer. RNA samples with 260/280 ratios higher than 1.8 were used for experiments. Total RNA (100 ng) was converted into cDNA using the ReverTra Ace® qPCR RT Kit (TOYOBO, Osaka, Japan). Aliquots of the resulting cDNA preparations then were subjected to qPCR analysis using the KOD SYBR® qPCR Mix (Toyobo). A CFX Connect™ Real-Time PCR system (Bio-Rad Laboratories, Inc.) was used to determine mRNA expression levels of the genes encoding connective tissue growth factor (Ctgf), interleukin 6 (Il6), programmed cell death 1 gene (Pdcd1, also called PD-1), superoxide dismutase-3 (Sod3), glutathione peroxidase-3 (Gpx3), and metallothionein-1 (Mt1). The primer pairs used were obtained from the Takara Perfect Real Time Primers collection (Takara Bio). cDNA/Taq-polymerase was denatured at 98°C for 2 min, and then 40 amplification cycles (each cycle: At 98°C for 10 sec, at 60°C for 10 sec, and at 68°C for 30 sec) were performed. The primer sequences and GenBank accession numbers are listed in Table I. Transcript levels were normalized to those of the housekeeping gene Gapdh (encoding glyceraldehyde-3-phosphate dehydrogenase) using the following formulae: ΔCq=Cq target-Cq reference; and ΔΔCq=mean value of ΔCq sample-ΔCq control. Finally, the 2−ΔΔCq method (24) was used to calculate the differences in mRNA transcription levels. The results of all assays were checked against melting curves to confirm the presence of single PCR products. At least two independent experiments were conducted and samples were assessed in (at least) triplicate in each experiment.

Table I.

Primers used for reverse transcription-quantitative PCR (RT-qPCR) analysis.

Table I.

Primers used for reverse transcription-quantitative PCR (RT-qPCR) analysis.

A, Mouse

GeneGenBank accession numberF primer sequence (5′-3′)R primer sequence (5′-3′)
Gpx3NM_008161.3 TCAACGTAGCCAGCTACTGAGGTC CTGTTTGCCAAATTGGTTGGAA-
Il6NM_031168.1 CCACTTCACAAGTCGGAGGCTTA GCAAGTGCATCATCGTTGTTCATAC
Mt1NM_013602.3 TCTAAGCGTCACCACGACTTCA GTGCACTTGCAGTTCTTGCAG
Pdcd1NM_008798.2 TGGCAATCAGGGTGGCTTC GACTCAGGCGGTTCCAGTTCA
Sod3NM_011435.3 GTGTCCCAAGACAATCCCACAA GGGAGTACTCTCAAAGGTGCTCA
CtgfNM_010217.2 ACCCGAGTTACCAATGACAATACC CCGCAGAACTTAGCCCTGTATG
GapdhNM_008084.3 TGTGTCCGTCGTGGATCTGA TTGCTGTTGAAGTCGCAGGAG

B, Rat

GeneGenBank accession numberF primer sequence (5′-3′)R primer sequence (5′-3′)

Il6NM_012589.2 ATTGTATGAACAGCGATGATGCAC CCAGGTAGAAACGGAACTCCAGA
Pdcd1NM_001106927.1 GCGCTTGCACTGTTGAGTGAG TGCCCAACAATAGGATTCAGGAG
GapdhNM_017008.4 GGCACAGTCAAGGCTGAGAATG ATGGTGGTGAAGACGCCAGTA

[i] F, forward; R, reverse; Gpx3, glutathione peroxidase-3; Mt1, metallothionein-1; Pdcd1, programmed cell death 1; Sod3, superoxide dismutase-3; Ctgf, connective tissue growth factor.

Il6 and Pdcd1 knockdown

Small interfering RNA (siRNA)-Il6 (siIl6) and siRNA-Pdcd1 (siPdcd1) were transfected into H9c2 cells using the HyperFect transfection reagent (Qiagen) according to the manufacturer's protocol. A non-targeting siRNA (Mock) was used as a vehicle control to assess the non-sequence-specific effects of transfected siRNAs. The siRNAs used were siIl6, a FlexiTube siRNA (ID no. SI01525356, Qiagen) and siPdcd1, a Silencer® Select Pre-designed siRNA Product (ID no. s154115, Thermo Fisher Scientific, Inc.); the negative control siRNA was obtained as AllStars Neg. Control siRNA (ID no. AM4611, Qiagen). Transfections consisted of 4×104 cells combined with a given siRNA (final concentration, 10 nM) in the presence of HyperFect reagent; the mixtures then were incubated for 24 h before assessment of Il6 or Pdcd1 expression.

IL-6 and PDCD1 immunofluorescence

Cells transfected with siRNAs were seeded into the Lab-Tek® 8-well chambered cover glass system plates (Thermo Fisher Scientific, Inc.) at 4×104 cells/ml and incubated overnight under standard culture conditions. The chambered slides were washed twice with phosphate-buffered saline (PBS) adjusted to pH 7.4 and fixed in ice-cold 1:1 methanol:acetone for 30 min. The slides were immersed for 10 min in 1% goat serum and 0.25% Triton X-100 in PBS and then transferred to Blocking One Histo (Nacalai Tesque) for 10 min. The slides then were washed with PBS containing 0.1% Tween-20, incubated with primary antibody anti-IL-6 rabbit polyclonal antibody (cat. no. NB600-1131, Novus Biologicals) or anti-PDCD1 mouse monoclonal antibody (cat. no. 66220-1-Ig, Proteintech®) at 1:1,000 in PBS for 1 h at room temperature, washed with PBS, and incubated with Alexa Fluor-conjugated secondary antibodies (Thermo Fisher Scientific, Inc.) for 1 h. After rinsing with PBS, a drop of UltraCruz™ Mounting Medium with DAPI (Santa Cruz Biotechnology, Inc.) was added to each well. Cells were observed under a confocal fluorescence microscope C-1 (Nikon) and cell components were visualized by fluorescent intensity in blue (405 nm) for nuclear DNA, green (488 nm) for PDCD1-positive cells, and red (594 nm) for IL-6-positive cells.

Apoptosis assay

Annexin V-mediated detection of phosphatidylserine (PS) was used to identify cells at early stages of apoptosis, because the redistribution of PS from the internal membranes to the external membrane surface occurs in early apoptosis (25). Several previous reports have employed an annexin V assay to determine apoptosis in DOX-treated cardiomyocytes (26,27). On the other hand, caspase-3/7 is known to be a key downstream effector in the apoptosis pathway (28). To test both aspects of cell death, we assessed the DOX-induced apoptosis using the RealTime-Glo™ Annexin V Apoptosis assay (Promega Corp.) and the Caspase-Glo® 3/7 Assay (Promega Corp.). Briefly, the H9c2 cells were transfected with siRNAs, distributed in a 96-well plate at a density of 4×103 cells per well, and allowed to adhere overnight. For the RealTime-Glo™ Annexin V Apoptosis assay, the adhered cells were incubated with 1 µM DOX in the presences of annexin V-luciferase reagents and time-dependent increases in luminescence were monitored, reflecting the apoptotic process. Similarly, apoptosis was evaluated after incubation for 24 h with the concentration of DOX at 0.01, 0.03, 0.1, 0.3 or 1 µM. These concentrations were set based on the results of a previous study (29), in which DOX at 0.1 µM or higher was shown to induce apoptosis of cardiomyocyte cells. For the Caspase-Glo® 3/7 Assay, the adhered cells were incubated with DOX at 0.1, 0.3 or 1 µM for 4 or 8 h; Caspase-Glo® 3/7 reagents then were added to each well and the contents were gently mixed. The resulting luminescent intensity was measured using a Varioskan™ LUX multimode microplate reader (Thermo Fisher Scientific, Inc.).

Statistical analysis

Data are expressed as mean ± standard error of the mean (SEM). Differences between two groups were compared using one-way analysis of variance (ANOVA). Multiple comparisons were performed by one-way ANOVA followed by Tukey post hoc test. P-value of less than 0.05 was considered significant. Correlation analyses were performed using the Spearman's rank correlation coefficient method. Statistical analyses were performed using Ekuseru-Toukei 2012 software (Social Survey Research Information Co., Ltd.).

Results

DICT and DOX-induced hematotoxicity

Toxic effects of DOX on the heart and in blood cells were evaluated in mice treated with a cumulative dose of 30 mg/kg. The body weight and heart weight of DOX-treated mice were almost 30% lower than those of the control mice (Fig. 1A and B), whereas the body weight-normalized heart weight did not differ significantly between these two groups (P>0.05; Fig. 1C), suggesting that cardiac hypertrophy is absent in the DOX-treated mice. Gene expression analyses of pooled heart samples revealed that the mRNA level of Ctgf (which encodes a pro-fibrotic cytokine) was significantly increased in DOX-treated mice compared to control animals (P<0.01; Fig. 1D). These findings suggested that cardiac tissue is the primary target of DOX cardiotoxicity. To further clarify the effects of DOX on the heart, we measured the serum levels of specific biomarkers for myocardial injury (including CK-MB and troponin T) as well as the serum levels of more general markers for inflammation (AST and LDH) (Fig. 2A-D). Mice that received DOX showed higher levels of not only AST and LDH, but also CK-MB and troponin T, as compared to control mice, supporting the use of the specific biomarkers as indicators of DICT. We also measured the mRNA levels of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in blood and heart, other biomarkers, at the first screening. Unfortunately, there was poor reproducibility in the measurements, so we did not adopt the data regarding natriuretic peptides.

Because DOX also can lead to hematotoxicity (30), some hematological parameters were measured in the small portion of whole blood collected from mice of both groups. In the DOX-treated mice, WBC values decreased to 45.8% of those in control mice, while platelet counts increased to 115.7% of those in control mice. Nevertheless, post-dose values of other parameters, such as RBC, Hb, and Hct, in DOX-treated mice did not differ significantly from those values in control mice (P>0.05) (data not shown), suggesting that DOX did not cause anemia or hypoglobulinemia.

Correlation of DICT and mRNA expression in blood

We next examined the correlation between the severity of DICT and blood mRNA expression of genes prior to DOX administration. Among 54 DOX-treated mice, twelve animals (22.2%) had no significant change in serum levels of cardiac injury parameters (P>0.05), while apparent cardiotoxicity (as evidenced by increases in the levels of the parameters associated with cardiotoxicity) was observed in 42 mice (77.8%), including ten mice (18.5%) that were found dead. The death was considered due to severe heart failure, because survival rate has conventionally been used as an index of DICT (3133). Most of surviving mice that escaped from lethal DICT would develop heart failure or cardiotoxicity. To facilitate correlation analysis, susceptibility to DOX-induced heart damage was scored as 0 to 8 based on separate changes in parameters (AST, LDH, CK-MB, and troponin T) in surviving mice as follows: 0, no change in either parameter (no damage detected); 1, increase in either AST or LDH=1 (such that increases in both yielded a value of 2); 3, increase in either CK-MB or troponin T=3 (such that increases in both yielded a value of 6); and 8, increases in all of four parameters. For example, if three parameters except for troponin T increased in a blood sample, its DICT score was determined as 5 (AST=1, LDH=1 and CK-MB=3). In addition, death was given a score of 10 as a maximum cardiac injury.

As a first screen for mRNA expression, we used RT-qPCR to assess the blood levels of transcripts from 32 candidate genes, including those encoding apoptosis-, autophagy- and oxidative stress-related proteins, each of which might contribute to the pathological mechanism(s) of DICT. The results revealed that the susceptibility to DOX-induced heart damage (as described above) was significantly and positively correlated with blood mRNA levels of Il6 (P<0.01) and Pdcd1 (P<0.05) (Fig. 3), as well as with the blood mRNA levels of the genes encoding Sod3 (P<0.01), Gpx3 (P<0.05) and Mt1 (P<0.05) (Fig. 4) prior to drug administration, suggesting that the products of these five genes may have some role in DICT. We chose to focus on the roles of Il6 and Pdcd1, given that previous work has suggested that cell signaling mediated by IL-6 and PDCD1 contributes to cytoprotection in the heart and other tissues (3439). Therefore, in vitro experiments were performed to clarify the roles of Il6 and Pdcd1 in DICT.

Effects of Il6 or Pdcd1 knockdown on DOX-induced apoptosis in vitro

We next assessed the potential roles of Il6 and Pdcd1 on DOX-induced toxicity of H9c2 cells (embryonic rat cardiac myoblasts), a line that is commonly used as a model for studies of DICT. Because cell death by apoptosis is involved in the primary mechanism of DICT (1,2,5), we studied effects of knockdown of Il6 or Pdcd1 on apoptotic response to DOX. The knockdown of endogenous Il6 and Pdcd1 was achieved by transfection with gene-specific siRNAs (Fig. S1). Knockdown efficacy was estimated by RT-qPCR; mRNA expression of Il6 and Pdcd1 were 0.32±0.03 and 0.23±0.05, respectively, when expression following transfection with the control siRNA was defined as 1.00 (Fig. S1A and B). Mock cells (vehicle control cells) exhibited no significant change in the accumulation of either mRNA (P>0.05). The knockdown of Il6 and Pdcd1 was confirmed by assessing levels of the two proteins by immunofluorescence analysis (Fig. S1C and D).

In mock cells exposed to DOX (1 µM), apoptosis (as determined by a luminescence assay) increased with incubation time. The DOX-induced apoptosis, however, was significantly increased in Il6 knockdown cells (P<0.01) and in Pdcd1 knockdown cells (P<0.05) than in mock cells within the first 10 h of incubation (Fig. 5A and B). Thus, DOX-induced apoptosis during the first 10 h of exposure was significantly enhanced by either Il6 or Pdcd1 knockdown, although the effects were weakened or undetectable for later time points (Fig. 5A-C). The activity of caspase-3/7, a protease enzyme that plays an essential role in apoptotic processes, also was increased in a DOX concentration- and exposure time-dependent manner in mock cells exposed to DOX (0.1–1 µM), confirming the ability of the corresponding assay to detect DOX-induced apoptosis. The increase in activity, however, was potentiated in each of the knockdown cells compared to the mock cells (Fig. 6A and B); for example, after incubation for 4 h with 0.1 µM DOX, activities in Il6 and Pdcd1 knockdown cells were elevated by ~30 and ~60%, respectively, compared to mock cells (Fig. 6A). Thus, by two separate assays, DOX-induced apoptosis was enhanced by knockdown of Il6 or Pdcd1, indicating that these genes have a preventive role against DICT.

Discussion

DICT can result in acute or chronic adverse effects. Both acute and chronic toxicity may lead to cardiac dysfunction or cardiomyopathy, eventually leading to severe heart failure and death (40,41). While acute cardiotoxicity following treatment with a high dose of DOX is now rare (40), late-onset chronic cardiotoxicity induced by the cumulative DOX dose remains common, occurring in up to 65% of patients (42). In the present study, we used a chronic cardiotoxicity model, in which DICT was established in mouse by repeated administration of DOX.

DOX-treated mice showed elevations (compared to control mice) in serum levels of CK-MB and troponin T, specific markers of myocardial cell injury, as well as in serum levels of non-specific cytosolic enzymes, such as AST and LDH. Although we did not assess the early cardiotoxicity markers after DOX administration, the data on early markers may help to find new information and knowledge about DICT. We need consider these points in our next study. The mice also had elevated (compared to control mice) cardiac tissue levels of the Ctgf transcript, which is associated with fibrosis after heart tissue damage. These biochemical changes indicate that DOX induces lethal injury in myocardial cells. It should be noted, however, that no myocardial injury was found in a subset (22.2%) of DOX-treated mice, although the majority of mice displayed apparent DICT, including death. Thus, individual differences are likely to exist in sensitivity of the heart to DOX. Clinical findings also have indicated that individuals vary greatly in their sensitivity to DOX (57); for instance, some patients are highly tolerant to DOX at cumulative doses exceeding 1,000 mg/m2, while others exhibit cardiotoxicity at cumulative doses below 400 mg/m2 (5).

It is possible that differences among individuals are attributable to genetic factors, which can be associated with the pathogenesis of DICT. To assess this possibility, we analyzed the correlation between mRNA expression of candidate genes in blood before DOX administration and the severity of DICT (using a score based on the changes in parameters known to be indicative of cardiac injury). Among 32 candidate genes, we have identified the five genes (Il6, Pdcd1, Sod3, Gpx3 and Mt1) that were correlated with susceptibility to DICT. According to previous reports (3439), cell signaling mediated by IL-6 and PDCD1 may be involved in protection from myocardial damage, such as ischemic damage, and therefore we investigated the roles of Il6 and Pdcd1 in DICT. As shown in Fig. 3, higher mRNA expression of Il6 or Pdcd1 was positively correlated with the DICT score, indicating that the expression levels of these genes might serve as predictive markers for DICT. In addition, these results led to the expectation that the expression of Il6 or Pdcd1 (at the mRNA or protein level) may be factors influencing the development of DICT. In the next experiment, therefore, DOX toxicity was assessed in cells of the rat cardiomyocyte line H9c2 in which Il6 or Pdcd1 expression had been subjected to knockdown via transfection with gene-specific siRNAs (Fig. S1). The cell toxicity of DOX was evaluated by monitoring apoptosis, a process known to be one of the primary mechanisms of DICT (1,2,5). Apoptosis was assessed by an annexin V assay, previously demonstrated to show high sensitivity and specificity for cell death (26), and by a caspase-3/7 assay known to be indicative of apoptotic processes (28). Contrary to our expectation, cells subjected to Il6 and Pdcd1 knockdown exhibited increased (rather than decreased) levels of DOX-induced apoptosis, suggesting that both of these genes play a protective role against the DOX-induced cardiomyocyte apoptosis, at least in this rat cardiomyocyte line.

IL-6 has been shown to be a pro-inflammatory cytokine with cardioprotective potential (35). Indeed, many reports have stated that the expression of this cytokine in cardiomyocytes attenuates myocardial ischemia-reperfusion damage (34,35,39). However, consistent with our results for blood concentrations of Il6 transcripts in mice, the circulating levels of IL-6 have been shown to be elevated in patients with congestive heart failure (43), and IL-6 is known to be released from the border zone of myocardial infarcts (44). The increased circulating levels of Il6 transcript and protein may reflect the release of these gene products into blood as the result of a compensatory mechanism(s) intended to reduce the vulnerability of heart tissue to various stresses such as ischemia and chemical stimuli, leading to cardioprotective action. In contrast, IL-6 has also been found to act as a deleterious cytokine associated with oxidative stress on the heart. The cytokine is indicated to induce apoptosis in cardiomyocytes (45), in agreement with our findings in terms of role of IL-6 on apoptosis. Furthermore, involvement of IL-6 (and oxidative stress) in the pathogenesis of cardiac heart failure (46) and atrial fibrillation (47) has been reported. The cardiac role of IL-6 is complex and remains poorly understood (48).

PDCD1, an immune inhibitory receptor, has been shown to inhibit lymphocyte activation and cytokine production (49). A previous study in mouse demonstrated that neutralization of PDCD1 with an anti-PDCD1 monoclonal antibody enhances DOX-induced nephropathy, suggesting that the PDCD1 pathway protects renal tissue from DOX-associated toxicity (38). That finding would be in agreement with our results suggesting the beneficial action of PDCD1 in preventing DOX-induced cardiotoxicity, although the underlying mechanism(s) of this effect remain unclear. Further studies will be needed to determine the details of the pathways and mechanisms whereby IL-6 and PDCD1 counteract DOX toxicity.

Extensive efforts have been made in various model systems to understand the role of gene expression in the mechanism(s) of DICT pathogenesis (21,5055). These studies assessed early and time-dependent molecular changes that occur following DOX administration, but did not include assessment of the levels of the tested molecules prior to DOX administration. For instance, troponins, which are sensitive tissue-specific markers of heart damage, are known to accumulate to elevated levels in blood only after cardiac tissue damage has occurred (56), but (to our knowledge) the pre-exposure levels of troponins have not previously been investigated in the context of DOX exposure. Invasive and noninvasive clinical approaches are currently being tested for use in predicting cardiotoxicity in DOX-treated cancer patients, but there have been technical complications (57,58). In that context, the results obtained in the present study may contribute to the early prediction of DICT and to new therapeutic strategies of cardioprotection. To our knowledge, the present report is the first to demonstrate a role for Il6 and Pdcd1 as predictive and protective factors in DICT. If animals with diverse genetic backgrounds are used, further genes associated with DICT may be found. Nevertheless, we used C57BL/6J inbred mice to minimize the individual differences including age, body weight, physical condition and genetic background, all of which could significantly influence the DICT. Although it is difficult to apply our findings to a human population in terms of genetic diversity, we believe that Il6 and Pdcd1 may also provide a beneficial role in cardioprotection in clinical chemotherapy with DOX.

Pharmacological strategies for preventing DICT have also been proposed in many studies; β-blockers, ACE inhibitors, ARBs and statins can reduce DICT in animals and in humans (1,10,13). It is noteworthy that dexrazoxane, which reduces oxidative stress (by iron chelation) and inhibits topoisomerase IIβ, protects the heart from anthracycline-induced toxicity (21,59,60). In fact, dexrazoxane is licensed in many parts of the world for two different indications; prevention of cardiotoxicity from anthracycline-based chemotherapy, and prevention of tissue injuries after extravasation of anthracycline (60). Based on information on predictive gene expression in individual patients, a pharmacological approach may be useful for further effective protection against DICT.

In conclusion, the pre-existing level of expression of both Il6 and Pdcd1 in cardiomyocytes may play an important role in protection against DOX-induced damage, such that the expression of these genes in blood serves as a predictive marker for DICT. These findings may provide useful information for prevention of cardiotoxicity in cancer patients receiving DOX therapy.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The current study was supported in part by a Grant-in-Aid for Scientific Research (C) (grant no. KAKENHI 17K08458) from the Japan Society for the Promotion of Science, and by a Matching Fund Subsidy for Private Universities from the Ministry of Education, Culture, Sports, Science and Technology of Japan.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

SK performed the experimental work, data collection and writing of the paper. AH participated in the study design, interpreted the data and revised the paper. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Experiments were conducted in accordance with the standards established by the Japanese Pharmacological Society and were approved by the Tohoku Medical and Pharmaceutical University of Institutional Animal Care and Use Committee (Experimental Protocol no. 18013).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

DOX

doxorubicin

ACE

angiotensin-converting enzyme

ARBs

angiotensin II receptor blockers

PDCD1

programmed cell death 1

Sod3

superoxide dismutase-3

Gpx3

glutathione peroxidase-3

Mt1

metallothionein-1

DICT

DOX-induced cardiotoxicity

LDH

lactate dehydrogenase

AST

aspartate aminotransferase

CK-MB

creatine kinase MB isoenzyme

References

1 

McGowan JV, Chung R, Maulik A, Piotrowska I, Walker JM and Yellon DM: Anthracycline chemotherapy and cardiotoxicity. Cardiovasc Drugs Ther. 31:63–75. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Nebigil CG and Désaubry L: Updates in anthracycline-mediated cardiotoxicity. Front Pharmacol. 9:12622018. View Article : Google Scholar : PubMed/NCBI

3 

Swain SM, Whaley FS and Ewer MS: Congestive heart failure in patients treated with doxorubicin: A retrospective analysis of three trials. Cancer. 97:2869–2879. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Ewer MS and Ewer SM: Cardiotoxicity of anticancer treatments: What the cardiologist needs to know. Nat Rev Cardiol. 7:564–575. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Jungsuwadee P: Doxorubicin-induced cardiomyopathy: An update beyond oxidative stress and myocardial cell death. Cardiovasc Reg Med. 3:e11272016.

6 

Ohtani K, Fujino T, Ide T, Funakoshi K, Sakamoto I, Hiasa KI, Higo T, Kamezaki K, Akashi K and Tsutsui H: Recovery from left ventricular dysfunction was associated with the early introduction of heart failure medical treatment in cancer patients with anthracycline-induced cardiotoxicity. Clin Res Cardiol. 108:600–611. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Raj S, Franco VI and Lipshultz SE: Anthracycline-induced cardiotoxicity: A review of pathophysiology, diagnosis, and treatment. Curr Treat Options Cardiovasc Med. 16:3152014. View Article : Google Scholar : PubMed/NCBI

8 

Zamorano JL, Lancellotti P, Muñoz DR, Aboyans V, Asteggiano R, Galderisi M, Habib G, Lenihan DJ, Lip GY, Lyon AR, et al: 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC committee for practice guidelines. Kardiol Pol. 74:1193–1233. 2016.(In Polish). View Article : Google Scholar : PubMed/NCBI

9 

Glass CK and Mitchell RN: Winning the battle, but losing the war: Mechanisms and morphology of cancer-therapy-associated cardiovascular toxicity. Cardiovasc Pathol. 30:55–63. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Totzeck M, Mincu RI, Heusch G and Rassaf T: Heart failure from cancer therapy: Can we prevent it? ESC Heart Fail. 6:856–862. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Hadi N, Yousif NG, Al-amran FG, Huntei NK, Mohammad BI and Ali SJ: Vitamin E and telmisartan attenuates doxorubicin induced cardiac injury in rat through down regulation of inflammatory response. BMC Cardiovasc Disord. 12:632012. View Article : Google Scholar : PubMed/NCBI

12 

Yamanaka S, Tatsumi T, Shiraishi J, Mano A, Keira N, Matoba S, Asayama J, Fushiki S, Fliss H and Nakagawa M: Amlodipine inhibits doxorubicin-induced apoptosis in neonatal rat cardiac myocytes. J Am Coll Cardiol. 41:870–878. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Cardinale D, Colombo A, Lamantia G, Colombo N, Civelli M, De Giacomi G, Rubino M, Veglia F, Fiorentini C and Cipolla CM: Anthracycline-induced cardiomyopathy: Clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 55:213–220. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, Ganame J, Sebag IA, Agler DA, Badano LP, et al: Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: A report from the American society of echocardiography and the european association of cardiovascular imaging. J Am Soc Echocardiogr. 27:911–939. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Cappetta D, Esposito G, Coppini R, Piegari E, Russo R, Ciuffreda LP, Rivellino A, Santini L, Rafaniello C, Scavone C, et al: Effects of ranolazine in a model of doxorubicin-induced left ventricle diastolic dysfunction. Br J Pharmacol. 174:3696–3712. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Henriksen PA: Anthracycline cardiotoxicity: An update on mechanisms, monitoring and prevention. Heart. 104:971–977. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Aminkeng F, Ross CJ, Rassekh SR, Hwang S, Rieder MJ, Bhavsar AP, Smith A, Sanatani S, Gelmon KA, Bernstein D, et al: Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol. 82:683–695. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Nair AB and Jacob S: A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 7:27–31. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Neilan TG, Jassal DS, Scully MF, Chen G, Deflandre C, McAllister H, Kay E, Austin SC, Halpern EF, Harmey JH and Fitzgerald DJ: Iloprost attenuates doxorubicin-induced cardiac injury in a murine model without compromising tumour suppression. Eur Heart J. 27:1251–1256. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Jenkins GR, Lee T, Moland CL, Vijay V, Herman EH, Lewis SM, Davis KJ, Muskhelishvili L, Kerr S, Fucoe JC and Desai VG: Sex-related differential susceptibility to doxorubicin-induced cardiotoxicity in B6C3F1 mice. Toxicol Appl Pharmacol. 310:159–174. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Vijay V, Moland CL, Han T, Fuscoe JC, Lee T, Herman EH, Jenkins GR, Lewis SM, Cummings CA, Gao Y, et al: Early transcriptional changes in cardiac mitochondria during chronic doxorubicin exposure and mitigation by dexrazoxane in mice. Toxicol Appl Pharmacol. 295:68–84. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Kanno S, Ishikawa M, Takayanagi M, Takayanagi Y and Sasaki K: Potentiation of acetaminophen hepatotoxicity and mortality by doxapram in mice. Biol Pharm Bull. 21:934–937. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Kanno S, Tomizawa A, Hiura T, Osanai Y, Kakuta M, Kitajima Y, Koiwai K, Ohtake T, Ujibe M and Ishikawa M: Melatonin protects on toxicity by acetaminophen but not on pharmacological effects in mice. Biol Pharm Bull. 29:472–476. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Blankenberg FG, Tait JF and Strauss HW: Apoptotic cell death: Its implications for imaging in the next millennium. Eur J Nucl Med. 27:359–367. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Bennink RJ, van den Hoff MJ, van Hemert FJ, de Bruin KM, Spijkerboer AL, Vanderheyden JL, Steinmetz N and van Eck-Smit BL: Annexin V imaging of acute doxorubicin cardiotoxicity (apoptosis) in rats. J Nucl Med. 45:842–848. 2004.PubMed/NCBI

27 

Schwartz RG, Jain D and Storozynsky E: Traditional and novel methods to assess and prevent chemotherapy-related cardiac dysfunction noninvasively. J Nucl Cardiol. 20:443–464. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Salvesen GS and Dixit VM: Caspases: Intracellular signaling by proteolysis. Cell. 91:443–446. 1997. View Article : Google Scholar : PubMed/NCBI

29 

He H, Liu C, Wu Y, Zhang X, Fan J and Cao Y: A multiscale physiologically-based pharmacokinetic model for doxorubicin to explore its mechanisms of cytotoxicity and cardiotoxicity in human physiological contexts. Pharm Res. 35:1742018. View Article : Google Scholar : PubMed/NCBI

30 

Eppstein DA, Kurahara CG, Bruno NA and Terrell TG: Prevention of doxorubicin-induced hematotoxicity in mice by interleukin 1. Cancer Res. 49:3955–3960. 1989.PubMed/NCBI

31 

Li K, Sung RY, Huang WZ, Yang M, Pong NH, Lee SM, Chan WY, Zhao H, To MY, Fok TF, et al: Thrombopoietin protects against in vitro and in vivo cardiotoxicity induced by doxorubicin. Circulation. 113:2211–2220. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Liu X, Chen Z, Chua CC, Ma YS, Youngberg GA, Hamdy R and Chua BH: Melatonin as an effective protector against doxorubicin-induced cardiotoxicity. Am J Physiol Heart Circ Physiol. 283:H254–H263. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Montgomery MD, Chan T, Swigart PM, Myagmar BE, Dash R and Simpson PC: An alpha-1A adrenergic receptor agonist prevents acute doxorubicin cardiomyopathy in male mice. PLoS One. 12:e01684092017. View Article : Google Scholar : PubMed/NCBI

34 

Dawn B, Xuan YT, Guo Y, Rezazadeh A, Stein AB, Hunt G, Wu WJ, Tan W and Bolli R: IL-6 plays an obligatory role in late preconditioning via JAK-STAT signaling and upregulation of iNOS and COX-2. Cardiovasc Res. 64:61–71. 2004. View Article : Google Scholar : PubMed/NCBI

35 

McGinnis GR, Ballmann C, Peters B, Nanayakkara G, Roberts M, Amin R and Quindry JC: Interleukin-6 mediates exercise preconditioning against myocardial ischemia reperfusion injury. Am J Physiol Heart Circ Physiol. 308:H1423–H1433. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N and Honjo T: Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 291:319–322. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, Ishida M, Hiai H, Matsumori A, Minato N and Honjo T: Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 9:1477–1483. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Qin XH, Lee VW, Wang YP, Zheng GP, Wang Y, Alexander SI and Harris DC: A protective role for programmed death 1 in progression of murine adriamycin nephropathy. Kidney Int. 70:1244–1250. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Smart N, Mojet MH, Latchman DS, Marber MS, Duchen MR and Heads RJ: IL-6 induces PI 3-kinase and nitric oxide-dependent protection and preserves mitochondrial function in cardiomyocytes. Cardiovasc Res. 69:164–177. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Sheng CC, Amiri-Kordestani L, Palmby T, Force T, Hong CC, Wu JC, Croce K, Kim G and Moslehi J: 21st Century cardio-oncology: Identifying cardiac safety signals in the era of personalized medicine. JACC Basic Transl Sci. 1:386–398. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Zhang YW, Shi J, Li YJ and Wei L: Cardiomyocyte death in doxorubicin-induced cardiotoxicity. Arch Immunol Ther Exp (Warsz). 57:435–445. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Lipshultz SE, Colan SD, Gelber RD, Perez-Atayde AR, Sallan SE and Sanders SP: Late cardiac effects of doxorubicin therapy for acute lymphoblastic leukemia in childhood. N Engl J Med. 324:808–815. 1991. View Article : Google Scholar : PubMed/NCBI

43 

Tsutamoto T, Hisanaga T, Wada A, Maeda K, Ohnishi M, Fukai D, Mabuchi N, Sawaki M and Kinoshita M: Interleukin-6 spillover in the peripheral circulation increases with the severity of heart failure, and the high plasma level of interleukin-6 is an important prognostic predictor in patients with congestive heart failure. J Am Coll Cardiol. 31:391–398. 1998. View Article : Google Scholar : PubMed/NCBI

44 

Gwechenberger M, Mendoza LH, Youker KA, Frangogiannis NG, Smith CW, Michael LH and Entman ML: Cardiac myocytes produce interleukin-6 in culture and in viable border zone of reperfused infarctions. Circulation. 99:546–551. 1999. View Article : Google Scholar : PubMed/NCBI

45 

Sun Y: Oxidative stress and cardiac repair/remodeling following infarction. Am J Med Sci. 334:197–205. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Neri M, Fineschi V, Di Paolo M, Pomara C, Riezzo I, Turillazzi E and Cerretani D: Cardiac oxidative stress and inflammatory cytokines response after myocardial infarction. Curr Vasc Pharmacol. 13:26–36. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Li JY, He Y, Ke HH, Jin Y, Jiang ZY and Zhong GQ: Plasma oxidative stress and inflammatory biomarkers are associated with the sizes of the left atrium and pulmonary vein in atrial fibrillation patients. Clin Cardiol. 40:89–94. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Hartman MHT, Groot HE, Leach IM, Karper JC and van der Harst P: Translational overview of cytokine inhibition in acute myocardial infarction and chronic heart failure. Trends Cardiovasc Med. 28:369–379. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Okazaki T, Maeda A, Nishimura H, Kurosaki T and Honjo T: PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci USA. 98:13866–13871. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Berthiaume JM and Wallace KB: Persistent alterations to the gene expression profile of the heart subsequent to chronic doxorubicin treatment. Cardiovasc Toxicol. 7:178–191. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Holmgren G, Synnergren J, Bogestål Y, Améen C, Åkesson K, Holmgren S, Lindahl A and Sartipy P: Identification of novel biomarkers for doxorubicin-induced toxicity in human cardiomyocytes derived from pluripotent stem cells. Toxicology. 328:102–111. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Thompson KL, Rosenzweig BA, Zhang J, Knapton AD, Honchel R, Lipshultz SE, Retief J, Sistare FD and Herman EH: Early alterations in heart gene expression profiles associated with doxorubicin cardiotoxicity in rats. Cancer Chemother Pharmacol. 66:303–314. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Todorova VK, Beggs ML, Delongchamp RR, Dhakal I, Makhoul I, Wei JY and Klimberg VS: Transcriptome profiling of peripheral blood cells identifies potential biomarkers for doxorubicin cardiotoxicity in a rat model. PLoS One. 7:e483982012. View Article : Google Scholar : PubMed/NCBI

54 

Yi X, Bekeredjian R, DeFilippis NJ, Siddiquee Z, Fernandez E and Shohet RV: Transcriptional analysis of doxorubicin-induced cardiotoxicity. Am J Physiol Heart Circ Physiol. 290:H1098–H1102. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Zhao WJ, Wei SN, Zeng XJ, Xia YL, Du J and Li HH: Gene expression profiling identifies the novel role of immunoproteasome in doxorubicin-induced cardiotoxicity. Toxicology. 333:76–88. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Bertinchant JP, Polge A, Juan JM, Oliva-Lauraire MC, Giuliani I, Marty-Double C, Burdy JY, Fabbro-Peray P, Laprade M, Bali JP, et al: Evaluation of cardiac troponin I and T levels as markers of myocardial damage in doxorubicin-induced cardiomyopathy rats, and their relationship with echocardiographic and histological findings. Clin Chim Acta. 329:39–51. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Shan K, Lincoff AM and Young JB: Anthracycline-induced cardiotoxicity. Ann Intern Med. 125:47–58. 1996. View Article : Google Scholar : PubMed/NCBI

58 

Gharib MI and Burnett AK: Chemotherapy-induced cardiotoxicity: Current practice and prospects of prophylaxis. Eur J Heart Fail. 4:235–242. 2002. View Article : Google Scholar : PubMed/NCBI

59 

Ganatra S, Nohria A, Shah S, Groarke JD, Sharma A, Venesy D, Patten R, Gunturu K, Zarwan C, Neilan TG, et al: Upfront dexrazoxane for the reduction of anthracycline-induced cardiotoxicity in adults with preexisting cardiomyopathy and cancer: A consecutive case series. Cardiooncology. 5:12019.PubMed/NCBI

60 

Langer SW: Dexrazoxane for the treatment of chemotherapy-related side effects. Cancer Manag Res. 6:357–363. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 23 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kanno S and Kanno S: The mRNA expression of <em>Il6</em> and <em>Pdcd1</em> are predictive and protective factors for doxorubicin‑induced cardiotoxicity. Mol Med Rep 23: 113, 2021
APA
Kanno, S., & Kanno, S. (2021). The mRNA expression of <em>Il6</em> and <em>Pdcd1</em> are predictive and protective factors for doxorubicin‑induced cardiotoxicity. Molecular Medicine Reports, 23, 113. https://doi.org/10.3892/mmr.2020.11752
MLA
Kanno, S., Hara, A."The mRNA expression of <em>Il6</em> and <em>Pdcd1</em> are predictive and protective factors for doxorubicin‑induced cardiotoxicity". Molecular Medicine Reports 23.2 (2021): 113.
Chicago
Kanno, S., Hara, A."The mRNA expression of <em>Il6</em> and <em>Pdcd1</em> are predictive and protective factors for doxorubicin‑induced cardiotoxicity". Molecular Medicine Reports 23, no. 2 (2021): 113. https://doi.org/10.3892/mmr.2020.11752