Oncol Rev (2010) 4:223–232 DOI 10.1007/s12156-010-0058-8
REVIEW
Drug-induced QT interval prolongation in cancer patients Torben K. Becker • Sai-Ching J. Yeung
Received: 9 April 2010 / Accepted: 28 June 2010 / Published online: 9 July 2010 Springer-Verlag 2010
Abstract Cancer patients are at an increased risk for QT interval prolongation and subsequent potentially fatal Torsade de pointes tachycardia due to the multiple drugs used for treatment of malignancies and the associated symptoms and complications. Based on a systematic review of the literature, this article analyzes the risk for prolongation of the QT interval with antineoplastic agents and commonly used concomitant drugs. This includes anthracyclines, fluorouracil, alkylating agents, and new molecularly targeted therapeutics, such as vascular disruption agents. Medications used in the supportive care can also prolong QT intervals, such as methadone, 5-HT3-antagonists and antihistamines, some antibiotics, antifungals, and antivirals. We describe the presumed mechanism of QT interval prolongation, drug-specific considerations, as well as important clinical interactions. Multiple risk factors and drug–drug interactions increase this risk for dangerous arrhythmias. We propose a systematic approach to evaluate cancer patients for the risk of QT interval prolongation and how to prevent adverse effects. Keywords QT prolongation Long QT syndrome Chemotherapy Supportive care Risk assessment
T. K. Becker S.-C. J. Yeung (&) Department of General Internal Medicine, Ambulatory Treatment and Emergency Care, The University of Texas MD Anderson Cancer Center, Unit 1465, P.O. Box 301402, Houston, TX 77230-1402, USA e-mail:
[email protected]
Introduction Case scenario A 25-year-old man presents to the emergency room at a comprehensive cancer center with a chief complaint of loss of consciousness earlier that day. His medical history includes T-cell acute lymphoblastic leukemia for which he recently received chemotherapy. The patient is accompanied by his mother who reports that her son experienced a sudden loss of consciousness that morning as well as three prior episodes in the past week. An EKG shows prolongation of the QT interval to 490 ms (ms). The patient is admitted to telemetry for further medical evaluation. Syncope is a common presentation seen in the emergency room. It often challenges the clinician due to its extensive differential diagnosis and the difficulty to obtain a detailed and conclusive history from the patient [1]. The above case scenario showcased a Long QT Syndrome (LQTS) as the initial working diagnosis for the patient’s recurrent syncopal episodes. This diagnosis of LQTS demands a comprehensive diagnostic process to identify the root cause, which may be inherited or acquired. Additionally, certain treatment-related conditions need to be taken into consideration in cancer patients. LQTS is among multiple cardiac problems faced by cancer patients, but it puts these patients at an additional risk for sudden cardiac death and morbidities due to injuries sustained from syncopal falls. Cancer patients are often at an even higher risk for an adverse outcome because of general deconditioning, frequently coexisting electrolyte abnormalities, long-term sequelae of radiation to the chest, chemotherapy-induced cardiotoxicities and tissue changes, such as cardiomyopathy or pulmonary hypertension. This review describes the pharmaceutical agents commonly used in cancer patients which can cause LQTS and outlines the necessary clinical considerations.
123
224
Search and selection strategy The authors conjointly searched the PubMed database for the keywords, ‘‘QT interval’’, ‘‘QT interval prolongation’’, ‘‘prolonged QT interval’’ and ‘‘long QT syndrome’’ in combination with any of the keywords ‘‘cancer’’, ‘‘antineoplastic’’, ‘‘chemotherapy’’, ‘‘targeted therapy’’, ‘‘antibiotics’’, ‘‘antimicrobials’’, ‘‘analgesics’’, ‘‘antiemetic’’, ‘‘supportive care’’, ‘‘alternative medicine’’, ‘‘therapy’’, ‘‘genetics’’. Articles in the English, German, Spanish, and French languages were reviewed. Evaluation and subsequent selection for inclusion in this article were based on the following criteria: describes general genetics and pathophysiology of QT interval prolongation, describes therapeutic considerations for QT interval prolongation, describes relevance of QT interval prolongation in cancer patients, describes anti-cancer drugs which influence the QT interval, describes antimicrobials or analgesics or antiemetics which influence the QT interval, describes remedies used as alternative medicine in cancer patients which influence the QT interval.
Oncol Rev (2010) 4:223–232
leading to LQTS. Four congenital forms of the LQTS (Romano-Ward syndrome, Lange-Nielsen syndrome, Andersen-Tawil syndrome and Timothy syndrome) have been recognized to date. The Romano-Ward syndrome can remain unrecognized until adulthood; it is caused by mutated genes which code for a specific Na? channel or for the Ankyrin-2 protein. The Lange-Nielsen syndrome is accompanied by deafness and less common than the Romano-Ward syndrome which has an incidence of about 1:7,000. The Lange-Nielsen syndrome is usually detected while investigating deafness in a child. It is caused by inherited mutations in genes coding for K? channels. The severity of symptoms varies between clinically normal and sudden cardiac death. The Andersen-Tawil and Timothy syndromes represent extremely rare forms of congenital LQTS [7–9]. More than 700 mutations in 12 genes leading to LQTS are currently known [9]. Some data suggest that drug-induced LQTS variants can be regarded as a subclinical form of the inherited variants [10].
Drugs causing QT interval prolongation Definition and pathophysiology
Antineoplastic agents
The estimated overall incidence of the LQTS is ranging between 1:2,500 and 1:5,000 [2, 3]. As cancer is one of the leading causes of morbidity worldwide, these data need to be taken in consideration by oncologists as well. Even though the exact incidence among cancer patients remains unknown, the rate of EKG changes in cancer patients has been reported as high as 36% [4]. LQTS is characterized by a Bazett-corrected QT interval time (QTc) greater or equal 450 ms and T wave alterations and can lead to a potentially fatal polymorphic ventricular tachycardia known as Torsade de pointes (Tdp). Persons with a QTc greater or equal 500 ms are regarded at high risk for Tdp, but no threshold has been established as definitively ‘‘safe’’ [5, 6]. The QT interval in the EKG represents the sum of the action potential duration of the ventricular cardiomyocytes. The cardiac action potential during depolarization is characterized by inward sodium (Na?) and calcium (Ca??) currents. During repolarization, the inward Ca?? current decreases and the outward-rectifier potassium current via human ether-a-go-go-related potassium (hERG K?) channels increases. Therefore, drugs can induce LQTS either by hindering the repolarizing K? influx or by prolonging the depolarizing Ca?? or Na? currents. Indirect effects leading to QT interval prolongation include electrolyte disorders, especially hypokalemia, caused by pharmaceuticals and drug–drug interactions at the cytochrome P450 isoenzyme CYP3A4. The inhibition of this enzyme can increase the plasma levels of other direct ion channel modifiers, thereby
Many commonly used antineoplastic agents can prolong the QT interval (Table 1). Direct hERG K? channel inhibition is a common mechanism of the undesired QT interval prolongation, but direct damage of the cardiomyocytes and CYP450 enzyme interactions play important roles as well. From a drug development standpoint, hERG K? channels are regarded as ‘‘antitargets’’ (i.e., as unwanted targets because of the risk of LQTS associated with their blockade), but hERG K? channels are present in many other tissues than solely the heart, specifically also in cancer cells, where they may serve as a possible target for future anti-cancer drugs [11, 12]. Cumulative or daily dose, prior treatment, drug formulation, as well as concurrent electrolyte abnormalities also influence the likelihood and severity of QT interval prolongation.
123
5-Fluorouracil (5-FU) 5-FU is a pyrimidine antimetabolite used as standard chemotherapy for many solid tumors, such as breast, colon, pancreatic or ovarian cancer. Multiple EKG changes, which are usually rather nonspecific, but may include prolonged QT intervals, have been described in patients receiving 5-FU. These are dose-dependent (single application and/or total treatment course) and also correlate with the infusion rate. In addition, the potential for cardiotoxicity and especially arrhythmias is increased by its ability to cause coronary vasospasm [13–15].
Oncol Rev (2010) 4:223–232
225
Table 1 Antineoplastic agents which can cause a QT interval prolongation Pharmaceutical agent
Clinical aspects
5-Fluorouracil
Dose-dependency
Amsacrine (topoisomerase II inhibitor)
Causes concurrent hypomagnesemia
Anti-estrogens
Tamoxifen, but not clomiphene
Arsenic trioxide
High rate of Tdp if QT interval prolongation occurs
Anthracyclines
Magnitude of QT interval prolongation may predict development of acute heart failure, dose-dependency
Cyclophosphamide
Magnitude of QT interval prolongation may predict development of acute heart failure, dose-dependency, irradiation is a common risk factor
Histone deacetylase inhibitors
Dose-dependency not likely
Platinum compounds
Clinical relevance questionable
Trastuzumab
Increases toxicity of anthracyclines
Tyrosine kinase inhibitors and angiogenesis inhibitors (bevacizumab, dasatinib, vandetanib etc.)
QT interval prolongation occurs frequently
Amsacrine Amsacrine, a topoisomerase II inhibitor, is used to treat acute leukemia. It can cause QT interval prolongation directly and indirectly by hERG K? channel inhibition and concurrent hypomagnesemia, although only a few cases of LQTS have been described in the literature to date [16, 17]. Anti-estrogens Anti-estrogens compete with estrogen for binding sites in target tissues and are used for the treatment of estrogenreceptor positive breast cancer. A QT interval prolongation has been described for tamoxifen, but not for clomiphene. This has been attributed to tamoxifen’s ability to inhibit hERG K? channels [18, 19]. Arsenic trioxide Arsenic trioxide is used in refractory or relapsed acute promyelocytic leukemia and induces apoptosis by DNA fragmentation and protein degradation. QT interval prolongation is relatively in patients receiving arsenic trioxide. Case reports suggest a high rate of arsenic trioxide-induced LQTS which will precipitate episodes of Tdp [20, 21]. Anthracyclines Anthracyclines (doxorubicin, daunorubicin, and epirubicin) are antibiotics. Their cytostatic effect is due to DNA intercalation. They are frequently used in the treatment of hematologic (e.g., acute leukemia) and solid (e.g., breast cancer) malignancies. A prolonged QT interval is a well known side effect of treatment with anthracyclines and is thought to reflect cardiomyocyte damage. This is consistent with findings that the QT interval dispersion may predict the development of
acute heart failure. Cardiotoxicity and subsequent LQTS exhibit a cumulative dose-dependency. Application of liposomal anthracycline preparations and dexrazoxane is associated with less cardiotoxic effects [22–28]. Cyclophosphamide Cyclophosphamide, a nitrogen mustard alkylating agent, cross-links tumor cell DNA, and thus damages DNA and inhibits DNA replication. It is widely used for treatment of hematologic malignancies and solid tumors. QT interval prolongation is also commonly observed in patients treated with cyclophosphamide. Cardiomyocyte damage is usually considered to be the origin of this, even though it may not be the initial site of injury and direct ion channel interactions may exist. Previous irradiation to the chest is a common risk factor, likely due to cellular damage. The magnitude of the QT interval prolongation is also predictive for the future development of acute heart failure [14, 29–31]. Histone deacetylase inhibitors Histone deacetylase inhibitors (e.g., vorinostat), aim to reverse aberrant epigenetic changes associated with cancer, and are licensed in the United States for treatment of cutaneous T-cell lymphoma. They lead to hyperacetylation of histones and transcription factors, and thereby affect gene expression. LQTS cases have been observed with histone deacetylase inhibitors and data suggest that the occurrence of the QT interval prolongation is dose-independent [13, 32]. Platinum compounds Platinum compounds (cisplatin and carboplatin), commonly used for treatment of lung, ovarian and many other
123
226
Oncol Rev (2010) 4:223–232
types of cancer, form platinum–DNA coordination complexes or adducts in tumor cells, leading not only to inhibition of DNA replication and transcription, but also DNA damage response-associated apoptosis. Except in the context of severe hypomagnesemia associated with platinum compounds, QT interval prolongation has been reported rarely for platinum compounds. In addition, the clinical relevance is questionable, as the prolongation does not seem to lead to Tdp [13, 33, 34].
tract). Due to the relatively high risk of Tdp, close EKG monitoring during therapy is warranted [13, 36–38]. In spite of this, patients who develop a prolonged QT interval during therapy with these agents may as well remain asymptomatic. Therefore, attention should be paid towards the identification of additional risk factors, which can help to identify patients at higher risk for life-threatening arrhythmias. Synergistic effects
Trastuzumab Trastuzumab is a monoclonal antibody, which binds to the human epidermal growth factor receptor 2 (HER2) in breast cancer patients, thereby inhibiting HER2-driven tumor cell proliferation and survival. It increases the toxicity of anthracyclines and thereby indirectly the risk for LQTS and Tdp. These effects are at least partially reversible [12, 22, 23, 35]. Tyrosine kinase inhibitors and angiogenesis inhibitors QT interval prolongation is commonly observed during treatment with tyrosine kinase inhibitors (e.g., dasatinib, imatinib, erlotinib, used as treatment for hematologic malignancies) and angiogenesis inhibitors, such as bevacizumab and vandetanib (typically used to treat non-small cell lung cancer and malignancies of the gastrointestinal
Even though anthracyclines are rather old drugs, the physician needs to be well aware of their potential for adverse effects. New targeted therapy agents, such as monoclonal antibodies and receptor blockers can increase the effect of QT prolongation of these older drugs or lead to cardiac toxicity by themselves [39]. As mentioned above, this has been described in particular for anthracyclines or paclitaxel in combination with trastuzumab [22, 23, 35, 40]. In fact, almost one third of post-marketing withdrawals of the new targeted therapy agents have been due to QT interval prolongation or Tdp [37]. Concomitant medication Other drugs commonly used in cancer patients for comorbid conditions and treatment-related complications may also cause LQTS (Table 2).
Table 2 Pharmaceutical agents often used concomitantly in cancer patients which can cause QT interval prolongation
Analgesics
Antiemetics
Antimicrobials
Alternative medicine
123
Pharmaceutical agent
Clinical aspects
Methadone
Most common QT interval prolonging drug within group of analgesics (5–30%), QT interval shortens rapidly after discontinuation
Buprenorphine
Less pronounced than methadone
Oxycodone
Dose-dependency
5-HT3-antagonists
QT interval shortens rapidly after discontinuation
Haloperidol
Hypokalemia and hypomagnesemia are strongest risk factors for Tdp, LQTS can occur even with low doses
Promethazine
Very low arrhythmogenic potential
Fluoroquinolones
Delayed onset of QT prolongation, patients who experience LQTS usually have multiple risk factors
Macrolides
Patients who experience LQTS usually have multiple risk factors
Telavancin
Clinical significance yet to be determined
Trimethoprim-sulfamethoxazole
LQTS very rare
Azoles
LQTS can occur even with low doses
Foscarnet
Can lead to severe hypocalcemia causing QT interval prolongation
Amantadine
High doses only
Pentamidine
Chemical similarity to procainamide
Cesium
Treatment consists of Prussian blue and electrolyte replacement
Oncol Rev (2010) 4:223–232
Analgesics Pain is a symptom faced by almost every cancer patient during the course of the disease. In accordance with the World Health Organization (WHO) analgesic ladder, it often requires multiple and very potent analgesic drugs to control pain and its associated symptoms. QT interval prolongation generally occurs most often (5–30%) with methadone due to CYP3A4 interactions and direct hERG K? channel inhibition. Therefore, a close EKG monitoring throughout the therapy is warranted. The QT interval shortens rapidly after discontinuation of methadone [33, 41– 46]. Buprenorphine is regarded as a safer alternative even in patients who developed Tdp while receiving methadone, as its QT interval prolonging effect is less pronounced [33, 46– 48]. Oxycodone dose-dependently inhibits hERG K? channel and can thus cause LQTS as well [33, 49]. Antiemetics Chemotherapy, pain, antibiotics and the tumor itself or its complications, among other substances and causes, can provoke nausea and emesis, which substantially decreases the perceived sense of wellbeing. Unfortunately, 5-HT3antagonists and promethazine, as some of the most commonly used antiemetics, are known to cause QT interval prolongation and may subsequently lead to cardiac arrhythmias. If LQTS is observed during therapy with 5-HT3-antagonists, a close monitoring for 8 h after discontinuation of the drug is sufficient, as the QT interval prolongation subsides quickly [33, 50, 51]. Promethazine can cause LQTS via direct hERG K? channel inhibition and CYP450 interactions, but clinical data suggest that its arrhythmogenic potential is very low [52, 53]. Haloperidol, often used in an antiemetic intention, causes QT interval prolongation as most antipsychotics. Accompanying hypokalemia and hypomagnesemia are the strongest risk factors for LQTS and subsequent Tdp. There are several reports of Tdp even in patients who only received a low dose of haloperidol [54–57]. Antimicrobials
227
consequences such as Tdp and ventricular fibrilation are relatively rare. The onset of the QT interval prolongation may be delayed [58–62]. Macrolides interact with hERG K? channels as well [58–60, 63]. The new antibiotic televancin also prolongs the QT interval, but no significant clinical adverse effects have been reported to date [64, 65]. LQTS seems to be a very rare complication of treatment with trimethoprim-sulfamethoxazole [60]. Special attention needs to be paid while using Azole antifungals, as a potential QT interval prolongation may happen independently of the dose. Direct hERG K? channel inhibition versus CYP3A4 interaction are debated as mechanism of actions, but the current findings are contradictory [66–70]. Foscarnet binds calcium; this can lead to a severe hypocalcemia, thus indirectly prolonging the QT interval. Cases of indirect QT interval prolongation have been reported with very high doses or overdosing of amantadine, as this agent causes hypokalemia [71]. The mechanisms of the QT interval prolonging effects of pentamidine are unknown, but its chemical similarity to the antiarrhythymic procainamide suggests interactions with cardiac ion channels [58, 60, 61, 72]. Alternative medicine Some cancer patients also use nutritional supplements or seek help from alternative medicine practitioners who may advise them to use specific ‘‘natural’’ remedies [73, 74]. Several cases of a cesium-induced life-threatening Tdp have been reported, the treatment in such situations consists of Prussian blue and electrolyte replacement [75, 76].
Considerations for risk assessment To evaluate an individual patient’s risk of developing a clinically relevant LQTS, several additional factors need to be taken into consideration (Table 3). A higher incidence Table 3 Risk factors for life-threatening events in patients with LQTS Risk factors for life-threatening events in patients with LQTS Female sex
Many cancer patients acquire infections during the course of their treatment, often because of a chemotherapyinduced neutropenia. In patients with hematologic malignancies, a prophylactic use of antimicrobials is therefore commonly indicated. Fluoroquinolones and macrolides are infamous for the potential QT interval prolongation due to the vast experience with these drugs, but this adverse reaction almost exclusively happens in patients with multiple risk factors for LQTS [58]. Fluoroquinolones cause a direct hERG K? channel inhibition, but severe
Older age ([40 years) Adolescent male History of symptoms Positive family history QT interval prolonging drugs Subtype of genetic mutations in LQT1–LQT12 genes Bradycardia Coronary artery disease Structural heart disease
123
228
of prolonged QT interval and Tdp has been shown repeatedly in women [77–81]. This is true for congenital variants of LQTS as well as for a drug-induced prolongation of the QT interval [77]. Data suggests that estrogens predispose to LQTS, while androgens reduce drug effects on cardiac repolarization. Gender-related differences in cardiac ion channels have been discussed as well [80]. However, these findings are not sufficient enough to completely account for the significant sex-related differences in the risk of arrhythmia. Older patients ([40 years) and male adolescents are at an increased risk for LQTS [82, 83]. Close genetic kinship to a population with certain founder mutations is consistent with an increased chance to suffer from LQTS [84, 85]. Ethnicity is debated as an additional factor, but the results remain inconclusive [86– 89]. Cancer patients commonly face several symptoms which require pharmacologic interventions at once; therefore, it is important to evaluate the patient’s medication profile to identify all drugs that may prolong the QT interval. Also, as many of the QT interval prolonging drugs are metabolized by the cytochrome P450 isoenzyme CYP3A4, interaction of these or inhibition of CYP3A4 increases the risk for a life-threatening Tdp [90]. A preexisting structural or vascular heart disease is another risk factor for a clinically relevant prolongation of the QT interval [61, 86, 90–92]. Certain chemotherapeutic agents, such as anthracyclines, alkylating agents, fluorouracil, bevacizumab or trastuzumab, and mediastinal radiation treatment can lead to structural heart damage. Therefore, a continuous monitoring of cardiac function may be necessary even for patients without any past cardiovascular history [14, 23, 31, 40]. Electrolyte disorders, more specifically hypokalemia as well as hypomagnesemia and hypocalcemia, are very common in cancer patients due to malnutrition, polypharmacy, or infusion therapy and increase the risk of Tdp [67, 90, 93].
Clinical consequences LQTS may have serious or even fatal consequences for a cancer patient. The risk for LQTS is considerable in this patient population due to the high potential for multiple drug interactions, cancer-related pathologies and treatmentassociated long-term sequelae. Therefore, we propose a multi-step screening approach to select cancer patients for further testing and prevention of LQTS and its potential complications (Fig. 1). Additional diagnostic and preventive steps should be based on these findings, thus avoiding unnecessary and costly measures, while ensuring a high-level of patient safety. Initially, all cancer patients should be screened for a family history and a past medical history positive for a
123
Oncol Rev (2010) 4:223–232
Fig. 1 Risk assessment for LQTS in cancer patients: A systematic analysis of risk factors determines the appropriate diagnostic and preventive measures and influences the therapeutic regimen
diagnosis of LQTS and genetic syndromes associated with QT interval prolongation, unexplained cases of sudden death or syncopes. Past cardiovascular problems or cardiac risk factors including again the family history as well as previous cardiotoxic treatments complete this step. An EKG should be obtained for all cancer patients to have a baseline measurement of the QT interval length. Depending on the information obtained from the medical history, further workup may be necessary, such as Holter EKGs for unexplained syncopes, molecular testing for patients with a suspicion for an inherited LQTS or echocardiographic imaging, and stress testing in patients with a presumed or confirmed structural heart disease. In cases of chemotherapy-induced cardiomyopathy, brain natriuretic peptide and other biomarker testing may be helpful in assessing the risk, too [22, 94]. Careful evaluation of the patient for presyncopal events, syncopes, and palpitations is a cornerstone to early diagnosis and intervention. Caregivers, such as nurses and family members, need to be aware of these signs and should report them immediately to the physician. For drugs which dose-dependently increase the QT interval, patients need to be monitored with daily EKGs and, if additional risk factors are present (Table 3), may require telemetry monitoring during dose-increased phases. In patients with a confirmed LQTS, the following preventive measures are considered standard of care [95–98]: • • • • •
avoidance of strenuous physical activity avoidance of vagal stimulation avoidance of emotional stress use of beta-blockers in patients with a genetic form of LQTS avoidance/cautious use of beta-blockers in patients with acquired LQTS
Oncol Rev (2010) 4:223–232
The use of beta-blockers in patients with long QT syndrome aims at blunting the adrenergic surge associated with emotional or physical stress by reducing the QT hysteresis (the adaptation delay of the QT interval to heart rate changes) [98]. However, beta-blockers also reduce the heart rate and thus prolong the QT interval. Therefore, we recommend a selective use of this drug in high-risk patients, i.e., individuals with a confirmed genetic form of LQTS (LQT1 genotype, in particular [99]). For patients who remain symptomatic and refractory to standard initial therapy, the necessity to perform a left cardiac sympathetic denervation and to implant an implantable cardioverter-defibrillator needs to be considered [8, 95, 96, 100]. If a cancer patient develops QT interval prolongation during his treatment course, the triggering drugs should be identified and discontinued, if possible. Otherwise, close clinical monitoring is necessary. Potassium levels should be kept in a high-normal range and magnesium levels need to be kept above 2.0 mg/dl to reduce the risk of hypokalemia- or hypomagnesemia-associated Tdp. If necessary, electrolyte replacement needs to be adjusted to other existing conditions, such as renal insufficiency. Also, genetic testing for LQTS mutations should be considered, as drug-induced LQTS may reflect a subclinical form of the inherited variants [10].
Conclusion LQTS, although not an ‘‘everyday practice problem’’, is a potentially fatal, but preventable side effect of medications typically used in cancer patients. As with every medication, physicians need to be aware of the potential side effects, common and rare ones, to weigh the risks and benefits for an individual patient. QT interval prolongation and subsequent Tdp are relevant adverse effects that may occur with typical treatment regimens used in a curative intention. Even though many patients remain asymptomatic, some suffer from potentially fatal Tdp. In patients with a non-curable condition, the focus of care shifts towards quality of life. The prevention of LQTS and its deadly sequelae should be within the focus of this care, as the simple selection of a different antibiotic or analgesic is often the only necessary measure. Therefore physicians working with cancer patients in a palliative situation should be aware when supportive care measures may pose additional risks, how to prevent them as well as how to recognize and treat them. Risk assessment and stratification are complex tasks, as multiple factors need to be taken into consideration, such as genetics, drug–drug interactions and preexisting cardiac conditions. In addition, fluctuating parameters, e.g., drug
229
dosages and electrolyte abnormalities, may influence the severity of symptoms and the response to therapeutic interventions. Multiple other drugs used in conditions not directly caused by or related to the malignancy and its treatment may lead to a prolonged QT interval, either directly or via enzyme inhibition. Therefore, the evaluation for a possible or worsening QT interval prolongation still varies from case to case. We propose an approach to facilitate and structure such a complex task. Early recognition, treatment and monitoring of patients at risk is a complex but manageable task in the medical care of cancer patients.
References 1. Casini-Raggi V, Bandinelli G, Lagi A (2002) Vasovagal syncope in emergency room patients: analysis of a metropolitan area registry. Neuroepidemiology 21:287–291 2. Johnson JN, Ackerman MJ (2009) QTc: how long is too long? Br J Sports Med 43:657–662 3. Shah M, Akar FG, Tomaselli GF (2005) Molecular basis of arrhythmias. Circulation 112:2517–2529 4. Yeh ET, Bickford CL (2009) Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management. J Am Coll Cardiol 53:2231–2247 5. Priori SG, Schwartz PJ, Napolitano C, Bloise R, Ronchetti E, Grillo M, Vicentini A, Spazzolini C, Nastoli J, Bottelli G, Folli R, Cappelletti D (2003) Risk stratification in the long-QT syndrome. N Engl J Med 348:1866–1874 6. Bednar MM, Harrigan EP, Anziano RJ, Camm AJ, Ruskin JN (2001) The QT interval. Prog Cardiovasc Dis 43:1–45 7. Donaldson MR, Yoon G, Fu YH, Ptacek LJ (2004) AndersenTawil syndrome: a model of clinical variability, pleiotropy, and genetic heterogeneity. Ann Med 36(Suppl 1):92–97 8. Crotti L, Celano G, Dagradi F, Schwartz PJ (2008) Congenital long QT syndrome. Orphanet J Rare Dis 3:18 9. Hedley PL, Jorgensen P, Schlamowitz S, Wangari R, MoolmanSmook J, Brink PA, Kanters JK, Corfield VA, Christiansen M (2009) The genetic basis of long QT and short QT syndromes: a mutation update. Hum Mutat 30:1486–1511 10. Itoh H, Sakaguchi T, Ding WG, Watanabe E, Watanabe I, Nishio Y, Makiyama T, Ohno S, Akao M, Higashi Y, Zenda N, Kubota T, Mori C, Okajima K, Haruna T, Miyamoto A, Kawamura M, Ishida K, Nagaoka I, Oka Y, Nakazawa Y, Yao T, Jo H, Sugimoto Y, Ashihara T, Hayashi H, Ito M, Imoto K, Matsuura H, Horie M (2009) Latent genetic backgrounds and molecular pathogenesis in drug-induced long-QT syndrome. Circ Arrhythm Electrophysiol 2:511–523 11. Raschi E, Vasina V, Poluzzi E, De Ponti F (2008) The hERG K? channel: target and antitarget strategies in drug development. Pharmacol Res 57:181–195 12. Raschi E, Vasina V, Ursino MG, Boriani G, Martoni A, De Ponti F (2010) Anticancer drugs and cardiotoxicity: insights and perspectives in the era of targeted therapy. Pharmacol Ther 125:196–218 13. Bagnes C, Panchuk PN, Recondo G (2009) Antineoplastic chemotherapy induced QTc prolongation. Curr Drug Saf 5:93–96 14. Pai VB, Nahata MC (2000) Cardiotoxicity of chemotherapeutic agents: incidence, treatment and prevention. Drug Saf 22:263– 302
123
230 15. de Forni M, Bugat R, Sorbette F, Delay M, Bachaud JM, Chevreau C (1990) Cardiotoxicity of continuous intravenous infusion of 5-fluorouracil: clinical study, prevention and physiopathology. Apropos of 13 cases. Bull Cancer 77:429–438 16. Seymour JF (1993) Induction of hypomagnesemia during Amsacrine treatment. Am J Hematol 42:262–267 17. Thomas D, Hammerling BC, Wu K, Wimmer AB, Ficker EK, Kirsch GE, Kochan MC, Wible BA, Scholz EP, Zitron E, Kathofer S, Kreye VA, Katus HA, Schoels W, Karle CA, Kiehn J (2004) Inhibition of cardiac HERG currents by the DNA topoisomerase II inhibitor amsacrine: mode of action. Br J Pharmacol 142:485–494 18. Slovacek L, Ansorgova V, Macingova Z, Haman L, Petera J (2008) Tamoxifen-induced QT interval prolongation. J Clin Pharm Ther 33:453–455 19. Yuill KH, Borg JJ, Ridley JM, Milnes JT, Witchel HJ, Paul AA, Kozlowski RZ, Hancox JC (2004) Potent inhibition of human cardiac potassium (HERG) channels by the anti-estrogen agent clomiphene-without QT interval prolongation. Biochem Biophys Res Commun 318:556–561 20. Ohnishi K, Yoshida H, Shigeno K, Nakamura S, Fujisawa S, Naito K, Shinjo K, Fujita Y, Matsui H, Sahara N, Takeshita A, Satoh H, Terada H, Ohno R (2002) Arsenic trioxide therapy for relapsed or refractory Japanese patients with acute promyelocytic leukemia: need for careful electrocardiogram monitoring. Leukemia 16:617–622 21. Ohnishi K, Yoshida H, Shigeno K, Nakamura S, Fujisawa S, Naito K, Shinjo K, Fujita Y, Matsui H, Takeshita A, Sugiyama S, Satoh H, Terada H, Ohno R (2000) Prolongation of the QT interval and ventricular tachycardia in patients treated with arsenic trioxide for acute promyelocytic leukemia. Ann Intern Med 133:881–885 22. Youssef G, Links M (2005) The prevention and management of cardiovascular complications of chemotherapy in patients with cancer. Am J Cardiovasc Drugs 5:233–243 23. Jensen BV (2006) Cardiotoxic consequences of anthracyclinecontaining therapy in patients with breast cancer. Semin Oncol 33:S15–S21 24. Schwartz CL, Hobbie WL, Truesdell S, Constine LC, Clark EB (1993) Corrected QT interval prolongation in anthracyclinetreated survivors of childhood cancer. J Clin Oncol 11:1906– 1910 25. Nakamae H, Hino M, Akahori M, Terada Y, Yamane T, Ohta K, Hayashi T, Tsumura K (2004) Predictive value of QT dispersion for acute heart failure after autologous and allogeneic hematopoietic stem cell transplantation. Am J Hematol 76:1–7 26. Nakamae H, Tsumura K, Akahori M, Terada Y, Yamane T, Hayashi T, Saito I, Kaneko M, Okamoto N, Ichihara Y, Ohta K, Hino M (2004) QT dispersion correlates with systolic rather than diastolic parameters in patients receiving anthracycline treatment. Intern Med 43:379–387 27. Poprach A, Petrakova K, Vyskocil J, Lakomy R, Nemecek R, Kocak I, Kocakova I, Vyzula R (2008) Cardiotoxicity of drugs used in oncology. Klin Onkol 21:288–293 28. Barrett-Lee PJ, Dixon JM, Farrell C, Jones A, Leonard R, Murray N, Palmieri C, Plummer CJ, Stanley A, Verrill MW (2009) Expert opinion on the use of anthracyclines in patients with advanced breast cancer at cardiac risk. Ann Oncol 20:816– 827 29. Kupari M, Volin L, Suokas A, Timonen T, Hekali P, Ruutu T (1990) Cardiac involvement in bone marrow transplantation: electrocardiographic changes, arrhythmias, heart failure and autopsy findings. Bone Marrow Transplant 5:91–98 30. Nakamae H, Tsumura K, Hino M, Hayashi T, Tatsumi N (2000) QT dispersion as a predictor of acute heart failure after highdose cyclophosphamide. Lancet 355:805–806
123
Oncol Rev (2010) 4:223–232 31. Morandi P, Ruffini PA, Benvenuto GM, La Vecchia L, Mezzena G, Raimondi R (2001) Serum cardiac troponin I levels and ECG/ Echo monitoring in breast cancer patients undergoing high-dose (7 g/m(2)) cyclophosphamide. Bone Marrow Transplant 28:277–282 32. Shah MH, Binkley P, Chan K, Xiao J, Arbogast D, Collamore M, Farra Y, Young D, Grever M (2006) Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin Cancer Res 12:3997–4003 33. Keller GA, Ponte ML, Di Girolamo G (2009) Other drugs acting on nervous system associated with QT-interval prolongation. Curr Drug Saf 5:105–111 34. Woei Chung L, Liao YM, Hsieh CY, Lin CY (2009) Oxaliplatin-induced long QT syndrome in a patient with appendiceal adenocarcinoma. Acta Oncol 48:156–157 35. Ewer SM, Ewer MS (2008) Cardiotoxicity profile of trastuzumab. Drug Saf 31:459–467 36. Heymach JV, Johnson BE, Prager D, Csada E, Roubec J, Pesek M, Spasova I, Belani CP, Bodrogi I, Gadgeel S, Kennedy SJ, Hou J, Herbst RS (2007) Randomized, placebo-controlled phase II study of vandetanib plus docetaxel in previously treated non small-cell lung cancer. J Clin Oncol 25:4270–4277 37. Ederhy S, Cohen A, Dufaitre G, Izzedine H, Massard C, Meuleman C, Besse B, Berthelot E, Boccara F, Soria JC (2009) QT interval prolongation among patients treated with angiogenesis inhibitors. Target Oncol 4:89–97 38. Orphanos GS, Ioannidis GN, Ardavanis AG (2009) Cardiotoxicity induced by tyrosine kinase inhibitors. Acta Oncol 48:964– 970 39. Strevel EL, Ing DJ, Siu LL (2007) Molecularly targeted oncology therapeutics and prolongation of the QT interval. J Clin Oncol 25:3362–3371 40. Tan-Chiu E, Yothers G, Romond E, Geyer CE Jr, Ewer M, Keefe D, Shannon RP, Swain SM, Brown A, Fehrenbacher L, Vogel VG, Seay TE, Rastogi P, Mamounas EP, Wolmark N, Bryant J (2005) Assessment of cardiac dysfunction in a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel, with or without trastuzumab as adjuvant therapy in node-positive, human epidermal growth factor receptor 2-overexpressing breast cancer: NSABP B-31. J Clin Oncol 23:7811–7819 41. Krantz MJ, Kutinsky IB, Robertson AD, Mehler PS (2003) Doserelated effects of methadone on QT prolongation in a series of patients with torsade de pointes. Pharmacotherapy 23:802–805 42. Almehmi A, Malas AM, Yousufuddin M, Rosencrance JG (2004) Methadone-induced torsade de pointes in a patient with normal baseline QT interval. W V Med J 100:147–148 43. Ower K, Morley-Forster P, Moulin D (2005) Fluctuating QTc interval in an asymptomatic patient treated with methadone for chronic pain. J Opioid Manag 1:73–76 44. Sticherling C, Schaer BA, Ammann P, Maeder M, Osswald S (2005) Methadone-induced Torsade de pointes tachycardias. Swiss Med Wkly 135:282–285 45. George S, Moreira K, Fapohunda M (2008) Methadone and the heart: what the clinician needs to know. Curr Drug Abuse Rev 1:297–302 46. Anchersen K, Clausen T, Gossop M, Hansteen V, Waal H (2009) Prevalence and clinical relevance of corrected QT interval prolongation during methadone and buprenorphine treatment: a mortality assessment study. Addiction 104:993–999 47. Wedam EF, Bigelow GE, Johnson RE, Nuzzo PA, Haigney MC (2007) QT-interval effects of methadone, levomethadyl, and buprenorphine in a randomized trial. Arch Intern Med 167:2469–2475 48. Esses JL, Rosman J, Do LT, Schweitzer P, Hanon S (2008) Successful transition to buprenorphine in a patient with
Oncol Rev (2010) 4:223–232
49.
50.
51.
52.
53.
54. 55.
56. 57.
58. 59.
60.
61.
62.
63. 64.
65. 66.
67.
68.
methadone-induced torsades de pointes. J Interv Card Electrophysiol 23:117–119 Fanoe S, Jensen GB, Sjogren P, Korsgaard MP, Grunnet M (2009) Oxycodone is associated with dose-dependent QTc prolongation in patients and low-affinity inhibiting of hERG activity in vitro. Br J Clin Pharmacol 67:172–179 Benedict CR, Arbogast R, Martin L, Patton L, Morrill B, Hahne W (1996) Single-blind study of the effects of intravenous dolasetron mesylate versus ondansetron on electrocardiographic parameters in normal volunteers. J Cardiovasc Pharmacol 28:53–59 Keefe DL (2002) The cardiotoxic potential of the 5-HT(3) receptor antagonist antiemetics: is there cause for concern? Oncologist 7:65–72 Jo SH, Hong HK, Chong SH, Lee HS, Choe H (2009) H(1) antihistamine drug promethazine directly blocks hERG K(?) channel. Pharmacol Res 60:429–437 Owczuk R, Twardowski P, Dylczyk-Sommer A, Wujtewicz MA, Sawicka W, Drogoszewska B, Wujtewicz M (2009) Influence of promethazine on cardiac repolarisation: a doubleblind, midazolam-controlled study. Anaesthesia 64:609–614 Alvarez PA, Pahissa J (2009) QT alterations in psychopharmacology: proven candidates and suspects. Curr Drug Saf 5:97–104 Freeman BD, Dixon DJ, Coopersmith CM, Zehnbauer BA, Buchman TG (2008) Pharmacoepidemiology of QT-interval prolonging drug administration in critically ill patients. Pharmacoepidemiol Drug Saf 17:971–981 Jackson T, Ditmanson L, Phibbs B (1997) Torsade de pointes and low-dose oral haloperidol. Arch Intern Med 157:2013–2015 Raghu KG, Singh R, Prathapan A, Yadav GK (2009) Modulation of haloperidol induced electrophysiological alterations on cardiac action potential by various risk factors and gender difference. Chem Biol Interact 180:454–459 Simko J, Csilek A, Karaszi J, Lorincz I (2008) Proarrhythmic potential of antimicrobial agents. Infection 36:194–206 Iannini PB (2002) Cardiotoxicity of macrolides, ketolides and fluoroquinolones that prolong the QTc interval. Expert Opin Drug Saf 1:121–128 Bril F, Gonzalez CD, Di Girolamo G (2009) Antimicrobial agents-associated with QT interval prolongation. Curr Drug Saf 5:85–92 Owens RC Jr, Nolin TD (2006) Antimicrobial-associated QT interval prolongation: pointes of interest. Clin Infect Dis 43:1603–1611 Alexandrou AJ, Duncan RS, Sullivan A, Hancox JC, Leishman DJ, Witchel HJ, Leaney JL (2006) Mechanism of hERG K? channel blockade by the fluoroquinolone antibiotic moxifloxacin. Br J Pharmacol 147:905–916 Hensey C, Keane D (2008) Clarithromycin induced torsade de pointes. Ir J Med Sci 177:67–68 Barriere S, Genter F, Spencer E, Kitt M, Hoelscher D, Morganroth J (2004) Effects of a new antibacterial, telavancin, on cardiac repolarization (QTc interval duration) in healthy subjects. J Clin Pharmacol 44:689–695 Smith WJ, Drew RH (2009) Telavancin: a new lipoglycopeptide for gram-positive infections. Drugs Today (Barc) 45:159–173 Haffner S, Lapp H, Thurmann PA (2002) Adverse drug reactions. QT-prolongation and torsade-de-pointes arrhythmias. Dtsch Med Wochenschr 127:1022–1024 Chakravarty C, Singh PM, Trikha A, Arora MK (2009) Fluconazole-induced recurrent ventricular fibrillation leading to multiple cardiac arrests. Anaesth Intensive Care 37:477–480 Pham CP, de Feiter PW, van der Kuy PH, van Mook WN (2006) Long QTc interval and torsade de pointes caused by fluconazole. Ann Pharmacother 40:1456–1461
231 69. Takemasa H, Nagatomo T, Abe H, Kawakami K, Igarashi T, Tsurugi T, Kabashima N, Tamura M, Okazaki M, Delisle BP, January CT, Otsuji Y (2008) Coexistence of hERG current block and disruption of protein trafficking in ketoconazole-induced long QT syndrome. Br J Pharmacol 153:439–447 70. Tholakanahalli VN, Potti A, Hanley JF, Merliss AD (2001) Fluconazole-induced torsade de pointes. Ann Pharmacother 35:432–434 71. Schwartz M, Patel M, Kazzi Z, Morgan B (2008) Cardiotoxicity after massive amantadine overdose. J Med Toxicol 4:173–179 72. Cortese LM, Gasser RA Jr, Bjornson DC, Dacey MJ, Oster CN (1992) Prolonged recurrence of pentamidine-induced torsades de pointes. Ann Pharmacother 26:1365–1369 73. Ernst E (2001) A primer of complementary and alternative medicine commonly used by cancer patients. Med J Aust 174:88–92 74. Mao JJ, Farrar JT, Xie SX, Bowman MA, Armstrong K (2007) Use of complementary and alternative medicine and prayer among a national sample of cancer survivors compared to other populations without cancer. Complement Ther Med 15:21–29 75. Chan CK, Chan MH, Tse ML, Chan IH, Cheung RC, Lam CW, Lau FL (2009) Life-threatening Torsades de Pointes resulting from ‘‘natural’’ cancer treatment. Clin Toxicol (Phila) 47:592– 594 76. O’Brien CE, Harik N, James LP, Seib PM, Stowe CD (2008) Cesium-induced QT-interval prolongation in an adolescent. Pharmacotherapy 28:1059–1065 77. Engelstein ED (2003) Long QT syndrome: a preventable cause of sudden death in women. Curr Womens Health Rep 3:126–134 78. Letsas KP, Efremidis M, Kounas SP, Pappas LK, Gavrielatos G, Alexanian IP, Dimopoulos NP, Filippatos GS, Sideris A, Kardaras F (2009) Clinical characteristics of patients with druginduced QT interval prolongation and torsade de pointes: identification of risk factors. Clin Res Cardiol 98:208–212 79. Abi-Gerges N, Philp K, Pollard C, Wakefield I, Hammond TG, Valentin JP (2004) Sex differences in ventricular repolarization: from cardiac electrophysiology to Torsades de Pointes. Fundam Clin Pharmacol 18:139–151 80. Drici MD, Clement N (2001) Is gender a risk factor for adverse drug reactions? The example of drug-induced long QT syndrome. Drug Saf 24:575–585 81. Hreiche R, Morissette P, Turgeon J (2008) Drug-induced long QT syndrome in women: review of current evidence and remaining gaps. Gend Med 5:124–135 82. Locati EH, Zareba W, Moss AJ, Schwartz PJ, Vincent GM, Lehmann MH, Towbin JA, Priori SG, Napolitano C, Robinson JL, Andrews M, Timothy K, Hall WJ (1998) Age- and sexrelated differences in clinical manifestations in patients with congenital long-QT syndrome: findings from the International LQTS Registry. Circulation 97:2237–2244 83. Sze E, Moss AJ, Goldenberg I, McNitt S, Jons C, Zareba W, Qi M, Robinson JL (2008) Long QT syndrome in patients over 40 years of age: increased risk for LQTS-related cardiac events in patients with coronary disease. Ann Noninvasive Electrocardiol 13:327–331 84. Arbour L, Rezazadeh S, Eldstrom J, Weget-Simms G, Rupps R, Dyer Z, Tibbits G, Accili E, Casey B, Kmetic A, Sanatani S, Fedida D (2008) A KCNQ1 V205M missense mutation causes a high rate of long QT syndrome in a First Nations community of northern British Columbia: a community-based approach to understanding the impact. Genet Med 10:545–550 85. Brink PA, Schwartz PJ (2009) Of founder populations, long QT syndrome, and destiny. Heart Rhythm 6:S25–S33 86. Heradien M, Goosen A, Moolman-Smook JC, Brink PA (2007) Race and gender representation of hypertrophic cardiomyopathy
123
232
87.
88.
89.
90.
91.
92.
93.
Oncol Rev (2010) 4:223–232 or long QT syndrome cases in a South African research setting. Cardiovasc J Afr 18:312–315 Dekker JM, Crow RS, Hannan PJ, Schouten EG, Folsom AR (2004) Heart rate-corrected QT interval prolongation predicts risk of coronary heart disease in black and white middle-aged men and women: the ARIC study. J Am Coll Cardiol 43:565– 571 Liu JF, Goldenberg I, Moss AJ, Shimizu W, Wilde AA, Hofman N, McNitt S, Zareba W, Miyamato Y, Robinson JL, Andrews ML (2008) Phenotypic variability in Caucasian and Japanese patients with matched LQT1 mutations. Ann Noninvasive Electrocardiol 13:234–241 Crotti L, Spazzolini C, Schwartz PJ, Shimizu W, Denjoy I, Schulze-Bahr E, Zaklyazminskaya EV, Swan H, Ackerman MJ, Moss AJ, Wilde AA, Horie M, Brink PA, Insolia R, De Ferrari GM, Crimi G (2007) The common long-QT syndrome mutation KCNQ1/A341V causes unusually severe clinical manifestations in patients with different ethnic backgrounds: toward a mutation-specific risk stratification. Circulation 116:2366–2375 Wolbrette DL (2004) Drugs that cause Torsades de pointes and increase the risk of sudden cardiac death. Curr Cardiol Rep 6:379–384 Akylbekova EL, Crow RS, Johnson WD, Buxbaum SG, Njemanze S, Fox E, Sarpong DF, Taylor HA, Newton-Cheh C (2009) Clinical correlates and heritability of QT interval duration in blacks: the Jackson Heart Study. Circ Arrhythm Electrophysiol 2:427–432 Schimpf R, Veltmann C, Wolpert C, Borggrefe M (2009) Channelopathies: Brugada syndrome, long QT syndrome, short QT syndrome, and CPVT. Herz 34:281–288 Stringer J, Welsh C, Tommasello A (2009) Methadone-associated Q-T interval prolongation and torsades de pointes. Am J Health Syst Pharm 66:825–833
123
94. Broeyer, FJ, Osanto S, Ritsema van Eck HJ, van Steijn AQ, Ballieux BE, Schoemaker RC, Cohen AF, Burggraaf J (2008) Evaluation of biomarkers for cardiotoxicity of anthracyclinbased chemotherapy. J Cancer Res Clin Oncol 134:961–968 95. Kaufman ES (2009) Mechanisms and clinical management of inherited channelopathies: long QT syndrome, Brugada syndrome, catecholaminergic polymorphic ventricular tachycardia, and short QT syndrome. Heart Rhythm 6:S51–S55 96. Monnig G, Kobe J, Loher A, Eckardt L, Wedekind H, Scheld HH, Haverkamp W, Milberg P, Breithardt G, Schulze-Bahr E, Bocker D (2005) Implantable cardioverter-defibrillator therapy in patients with congenital long-QT syndrome: a long-term follow-up. Heart Rhythm 2:497–504 97. Hobbs JB, Peterson DR, Moss AJ, McNitt S, Zareba W, Goldenberg I, Qi M, Robinson JL, Sauer AJ, Ackerman MJ, Benhorin J, Kaufman ES, Locati EH, Napolitano C, Priori SG, Towbin JA, Vincent GM, Zhang L (2006) Risk of aborted cardiac arrest or sudden cardiac death during adolescence in the long-QT syndrome. JAMA 296:1249–1254 98. Krahn AD, Yee R, Chauhan V, Skanes AC, Wang J, Hegele RA, Klein GJ (2002) Beta blockers normalize QT hysteresis in long QT syndrome. Am Heart J 143:528–534 99. Priori SG, Napolitano C, Schwartz PJ, Grillo M, Ronchetti E, Moncalvo C, Tulipani C, Veia A, Bottelli G, Nastoli J (2004) Association of long QT syndrome loci and cardiac events among patients treated with beta-blockers. JAMA 292:1341–1344 100. Proclemer A, Ghidina M, Facchin D, Rebellato L, Corrado D, Gasparini M, Gregori D (2009) Use of implantable cardioverterdefibrillator in inherited arrhythmogenic diseases: data from Italian ICD Registry for the years 2001–2006. Pacing Clin Electrophysiol 32:434–445