Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cardiovascular Toxicity of Multi-Tyrosine Kinase Inhibitors in Advanced Solid Tumors: A Population-Based Observational Study

  • Amirrtha Srikanthan,

    Affiliations Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada, Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada

  • Josee-Lyne Ethier,

    Affiliation Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada

  • Alberto Ocana,

    Affiliation Medical Oncology Department and Translational Research Unit, Albacete University Hospital, Edificio de Investigación, Calle Francisco Javier de Moya, Albacete, Spain

  • Bostjan Seruga,

    Affiliation Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia

  • Monika K. Krzyzanowska,

    Affiliation Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada

  • Eitan Amir

    eitan.amir@uhn.ca

    Affiliations Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada, Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada

Abstract

Background

Treatment with small molecule tyrosine kinase inhibitors (TKIs) has improved survival in many cancers, yet has been associated with an increased risk of adverse events. Warnings of cardiovascular events are common in drug labels of many TKIs. Despite these warnings, cardiovascular toxicity of patients treated with TKIs remains unclear. Here, we evaluate the cardiovascular outcomes of advanced cancer patients treated with small molecule tyrosine kinase inhibitors.

Methods

A population based cohort study was undertaken involving adults aged >18 years in Ontario, Canada, diagnosed with any advanced malignancy between 2006 and 2012. Data were extracted from linked administrative governmental databases. Adults with advanced cancer receiving TKIs were identified and followed throughout the time period. The main outcomes of interest were rates of hospitalization for ischemic heart disease (acute myocardial infarction and angina) or cerebrovascular accidents and death.

Results

1642 patients with a mean age of 62.5 years were studied; 1046 were treated with erlotinib, 166 with sorafenib and 430 with sunitinib. Over the 380 day median follow-up period (range 6-1970 days), 1.1% of all patients had ischemic heart events, 0.7% had cerebrovascular accidents and 72.1% died. Rates of cardiovascular events were similar to age and gender-matched individuals without cancer. In a subgroup analysis of treatment patients with a prior history of ischemic heart disease, 3.3% had ischemic heart events while 1.2% had cerebrovascular accidents.

Conclusions

TKIs do not appear to increase the cause-specific hazard of ischemic heart disease and cerebrovascular accidents compared to age and gender-matched individuals without advanced cancer.

Background

More than 90 tyrosine kinases have been shown to be critical to malignant transformation and tumor angiogenesis [1, 2]. Tyrosine kinase inhibitors (TKIs), which can target both receptor and cytoplasmic kinases [3] can improve cancer outcomes by exploiting activation of kinases in cancer cells [4]. A number of different TKIs have been studied and approved for use in both solid tumors and haematological malignancies [512]. Commonly used TKIs include erlotinib targeting epidermal growth factor receptor (EGFR), and sorafenib and sunitinib targeting mainly vascular endothelial growth factor receptor (VEGRF) and platelet-derived growth factor receptor (PDGFR) [13]. In addition to improved outcomes, TKIs are also relatively easy to administer [14].

Despite this targeted intent, TKIs usually affect multiple kinases [15, 16] and impact the function of non-malignant cells with resultant on- and off-target toxicities [13]. On-target toxicities, such as hypertension from VEGFR inhibitors, are due to class effects and are difficult to prevent [13]. Off-target toxicities occur when unintended targets are inhibited by the drug due to similarities with the intended target [13]. Both on- and off-target adverse events are described in clinical trials [17]. Non-cardiac toxicities from TKIs include skin toxicity, diarrhea, mucositis, pneumonitis and electrolyte abnormalities [13].

Hypertension, congestive heart failure, left ventricular systolic dysfunction and QT prolongation are common adverse cardiac toxicities associated with TKIs [3, 1823]. Cardiovascular events, such as cardiac ischemia, myocardial infarction and cerebrovascular accidents, have also emerged as concerning toxicities such that drug label warnings have been issued for TKIs [2426]. Given the broad patient populations eligible for TKI treatment, the improved survival seen with targeted agents [512], and the prevalence of cardiovascular disease in the population [27], recognition, management and prevention of TKI related cardiovascular events have emerged as important [19]. The development of strategies and guidelines to assess this emerging issue [21, 28, 29] and recommendations for cardiac safety monitoring of patients undergoing TKI treatment in clinical practice [3032] have therefore been recommended.

Clinical trial reports of cardiovascular toxicity are limited by inadequate power and are known to underrepresent at risk patients such as older individuals or those with significant comorbidities [33]. Patients enrolled in RCTs are highly selected and likely not representative of patients treated in general practice [34]. Efficacy and toxicity outcomes have been shown to be different between patients treated on and off clinical trials even when treated at the same institution at the same time [35]. Therefore, an assessment of the potential cardiovascular toxicities of TKIs in a population of unselected cancer patients is desirable.

Here, we report on a population based observational cohort study to assess the rates of cardiovascular and cerebrovascular outcomes and death among cancer patients receiving TKIs. We hypothesized that, compared to the general population, cardiovascular events and cerebrovascular accidents would be more prevalent in a population of patients with advanced cancer receiving TKIs, particularly among individuals with a prior history of ischemic heart disease (IHD).

Materials and Methods

Data Sources

The Ontario Health Insurance Plan (OHIP) is a publically funded health insurance program providing universal coverage for medically necessary care in Ontario, Canada—Canada’s most populous province with approximately 13.5 million residents [36]. Each resident is assigned a unique Ontario Health Insurance Number (OHIN), which was used to link multiple administrative health databases. Databases and data sets were held securely in a linked, de-identified form and analyzed through the Institute for Clinical Evaluative Sciences (ICES).

The Ontario Cancer Registry (OCR) is a passive registry of invasive cancer diagnoses of Ontario residents from 1964 onwards [37, 38]. This registry was used to identify patients with oncological diagnoses. The Ontario Drug Benefit (ODB) program reimburses prescription medication for all Ontario residents ≥ 65 years old. Individuals between 18–64 years of age are eligible for ODB support only if requiring long-term care, home care, governmental financial assistance, disability support or financial assistance (defined as high prescription drug costs relative to income) [39]. The ODB was used to determine exposure to TKI medications and exposure to cardiac medications prescribed before the first TKI prescription. TKI medications were prescribed in line with ODB Exceptional Access Program (EAP) eligibility for these drugs [40].

Oncologic drug availability is a multi-step process in Canada. After federal Health Canada approval, the national pan-Canadian Oncology Drug Review (pCODR) evaluates a new cancer agent’s efficacy and cost-effectiveness. A recommendation regarding funding the new agent is then issued. Individual provinces then review pCODR recommendations and manufacturer requests. Individual provincial funding decisions are made after consideration of expert reviews, governmental budgets and the public interest [41]. Once provincially approved by the Ontario Ministry of Health and Long-Term Care, an intravenous drug is available throughout Ontario through the New Drug Funding Program (NDFP). Oral agents are funded through the ODB, private insurance or self-pay mechanisms [42]. Prices of anti-cancer agents are negotiated privately between provinces and manufacturers and are details of agreements are not generally available publically.

The NDFP was used to identify individuals who received systemic treatment prior to or after TKI initiation. The Canadian Institute for Health Information Discharge Abstract Database (CIHI-DAD) holds a record of all discharges from acute care hospitals in Ontario. The National Ambulatory Care Reporting System (NACRS) provides a record of hospital and community based emergency and ambulatory care. The OHIP database also provides additional data of physician services from billing claims.

Baseline comorbidities were extracted from CIHI-DAD and NACRS using corresponding International Classification of Diseases codes (ICD10) in the 10 years preceding the index case. Baseline hypertension, IHD, congestive heart failure and diabetes were identified using validated algorithms. These algorithms use both in- and out-of-hospital diagnostic and billing codes [4346].

The Ontario Registered Persons Database (ORPD) provided demographic data such as vital status, postal code and date of death. Cause of death was not available in the database. Canadian Census data was used to establish median household neighbourhood income, which was used as a surrogate for socioeconomic status [47]. Limitations of available databases prevented determination of when death occurred relative to TKI treatment completion.

Study Design

The OCR was used to identify adults ≥ 18 years old from January 1, 2006 to September 30, 2012 with a first documented diagnosis of cancer. Prescription drug information was available for all adults ≥ 65 years old and limited for individuals between 18–64 years of age due to the aforementioned provincial criteria.

The ODB was used to identify all adults who were dispensed a TKI prescription at any point after their date of diagnosis. All individuals with any exposure to a TKI were initially included. The control group comprised all age and gender-matched individuals without cancer in the Ontario population during the time of interest. The control group was derived from the general population and was compared to the treatment group with respect to additional comorbidities. Drugs with fewer than 50 exposed patients were excluded from the analysis to reduce heterogeneity.

The impact of treatment on the cause specific hazard of IHD and cerebrovascular disease and on the hazard of death was evaluated. Outcomes were determined by identifying hospitalizations with a most responsible diagnosis of acute myocardial infarction (AMI), angina (ICD10 I20-I22 or I24) or cerebrovascular accident (ICD10 I60-I69 or G45). The outcome of IHD was defined as a composite of AMI and angina.

Statistical Analysis

Descriptive statistical analyses were utilized and frequency of occurrence and percentage was calculated for each of the independent variables. Continuous baseline variables were compared between the treatment and control group using Wilcoxon Rank Sum test. Categorical baseline variables were compared using the χ2 statistic. Time-to-event analyses were performed using the control population as the reference for each of the outcomes of interest. Time-to-event was defined as the time from first prescription of TKI to the event of interest. Hazard ratios (HR) and 95% confidence intervals (CI) were calculated. Pre-stratified subgroup analyses were performed based on the presence of previously diagnosed IHD. Patients were censored if an event of interest did not occur prior to September 30, 2012 (the end of follow-up). In the analysis of cause specific hazard for IHD or cerebrovascular disease, patients were censored at death or if they received systemic therapy or radiation during the year after TKI initiation. Kaplan-Meier survival curves were plotted to illustrate overall survival free of death, free of IHD and free of cerebrovascular accidents. All analyses were performed using SAS software, version 9.2 (SAS Institute Inc., Cary, NC). Statistical significance was defined using a two-tailed p-value of <0.05. Data cells involving ≤ 5 patients were not included in keeping with ICES’ privacy regulations.

Ethics Statement

The ICES review board and privacy office approved this study prior to commencement. Consent was not obtained from individual patients; however, all patient information was anonymized and de-identified by ICES prior to receipt by the investigators for analysis.

Results

Seven TKIs were publically funded and in use between 2006 and 2012 in Ontario. Data for these drugs comprised 1046 patients treated with erlotinib, 430 with sunitinib, 166 with sorafenib, 46 with gefitinib, 11 with everolimus, 9 with temsirolimus and 5 with lapatinib. Data for gefitinib, everolimus, temsirolimus and lapatinib were excluded and consequently, a cohort of 1642 patients exposed to erlotinib, sunitinib and sorafenib were included in the analysis. These treatment patients were compared to 128,415 age and gender matched individuals without cancer who served as controls. The mean age of the TKI-treated patients was 65.2 years and 338 (20.6%) were identified as having baseline IHD, including 99 individuals (6.0%) who had a previous AMI (Table 1).

thumbnail
Table 1. Baseline Characteristics of the TKI-Treated Patients and Control Group.

https://doi.org/10.1371/journal.pone.0122735.t001

Baseline Comparison of TKI-treated Individuals to Individuals Without Cancer

Baseline characteristics of the treatment and control group are shown in Table 1. The control group of age and gender-matched individuals without cancer was of similar age (65.4 versus 65.2, p = 0.61), but less likely to have prior IHD (18.0% versus 20.6%, p = 0.007), venous thromboembolism (1.5% versus 8.7%, p<0.001), renal disease (4.6% versus 5.7%, p = 0.028), and cardiac medication usage (angiotensin converting enzyme inhibitors, 20.4% versus 22.5%, p = 0.031; angiotensin receptor blockers, 8.5% versus 11.7%, p<0.001; beta-blockers, 15.8% versus 18.9%, p<0.001; calcium channel blockers, 17.0% versus 22.4%, p<0.001 and anti-dyslipidemia medications, 26.8% versus 32.4%, p<0.001). Baseline diagnosis of diabetes was not available; however, baseline usage of both oral hypoglycaemic agents (9.3% versus 15.1%, p<0.001) and insulin (2.6% versus 3.7%, p = 0.005) was less common among the control group. Usage of aspirin was greater among control patients (2.9% vs. 4.2%, p = 0.013). All TKI-treated patients had a stage IV cancer diagnosis, in keeping with ODB EAP eligibility criteria [40].

Ischemic Cardiac and Cerebrovascular Outcomes and Death

Within the 380-day median follow-up period after TKI therapy initiation, 18 (1.1%) of the 1642 treatment patients developed an ischemic heart disease event requiring hospitalization, 11 (0.7%) developed a cerebrovascular accident requiring hospitalization and 1184 (72.1%) individuals died. Of the 18 cases of ischemic heart disease, 11 occurred in erlotinib treated patients, 5 in sunitinib treated patients and 2 in sorafenib treated patients. Of the 11 cases of cerebrovascular accidents, 8 occurred in erlotinib treated patients, 2 in sunitinib treated patients and 1 in sorafenib treated patients. These proportions closely mirrored the relative frequency of drug use in the population.

Cardiovascular events predominantly occurred late in follow-up (see Fig. 1). Of the 338 patients with baseline IHD, 11 (3.3%) had ischemic heart events during follow-up while 4 (1.2%) had cerebrovascular accidents and 245 (72.5%) died. Of the 1304 patients without baseline IHD, 7 (0.5%) had ischemic heart events, 7 (0.5%) had cerebrovascular accidents and 939 (72.0%) died. A comparison of time to cardiovascular event between patients with and without prior IHD is shown in Fig. 2. Compared to those without prior IHD, there was a numerical, but non-significantly higher hazard of cardiovascular events in those with prior IHD (HR 1.59, 95% CI 0.76–3.33, p = 0.22). Power to detect differences between these groups was low (20.3% assuming alpha = 0.05).

thumbnail
Fig 1. Kaplan Meier curve for time to cardiovascular event in tyrosine kinase inhibitor (TKI)-treated group.

https://doi.org/10.1371/journal.pone.0122735.g001

thumbnail
Fig 2. Kaplan Meier curves for time to cardiovascular event in tyrosine kinase inhibitor (TKI)-treated group based on history of ischemic heart disease (IHD).

https://doi.org/10.1371/journal.pone.0122735.g002

Comparison of Outcomes to Individuals Without Cancer

Compared to age and gender-matched non-cancer patients, patients exposed to TKI had similar rates of IHD and cerebrovascular accidents (Table 2), but a significantly higher hazard of death (Fig. 3). Results were similar for both the whole study population and the subgroup with IHD (Table 2). Due to small event numbers, additional subgroup analyses based on duration of treatment or type of TKI were not undertaken.

thumbnail
Table 2. Ischemic Cardiac and Cerebrovascular Outcomes and Death—Compared to the Control Group.

https://doi.org/10.1371/journal.pone.0122735.t002

thumbnail
Fig 3. Kaplan Meier curves for overall survival in tyrosine kinase inhibitor (TKI)-treated and control groups.

https://doi.org/10.1371/journal.pone.0122735.g003

Discussion

While data are available to inform about TKI-induced cardiac toxicity among patients treated in clinical trials, less is known about such toxicities in patients treated in routine clinical practice. This population-based study of all patients treated in Ontario, Canada demonstrates that TKI use among advanced cancer patients did not show significantly higher rates of cardiovascular adverse events relative to the general population. Furthermore, those events which did occur appeared to be more frequent later in follow-up. As expected, in the stratified analyses, a trend for higher rates of ischemic heart disease and cerebrovascular accidents was seen among individuals with a history of prior IHD. Despite TKI-treated patients having higher rates of baseline cardiac comorbidities and usage of cardiac medications than the general population, rates of cardiac events were low overall, while death events (presumably from the underlying malignancy) were frequent.

Although aggressive management of TKI-induced cardiac toxicity and cardiovascular risk factors is advocated [3032, 48], our study suggests in advanced cancer patients, given the overwhelming mortality from malignancy, aggressive cardiovascular risk factor management is unlikely to significantly impact survival. These results are consistent with recent randomized data showing that discontinuation of preventative cardiac medications does not lead to worse survival and may improve quality of life in metastatic patients [49].

The practice of aggressive cardiac risk factor management may have contributed to the low rates of cardiovascular events and cerebrovascular accidents in this study. Toxicity has been suggested as a pharmacodynamics marker for targeted anti-neoplastic drugs [50]. On-target side effects, such as arterial hypertension, can potentially serve as a biomarker for efficacy [51, 52]. Although a risk factor for cardio- and cerebrovascular disease, aggressive management with anti-hypertensive medications is advocated for as opposed to dose reduction or discontinuation [31, 32]. When cardiovascular side effects represent off-target side effects (which are not pharmacodynamics markers), dose maintenance is still advocated to maximize oncologic control through on-target mechanisms [31, 32].

Fortunately, most cardiovascular toxicities specific to TKIs appear reversible with aggressive and early management [13, 32, 48]. This reversibility is increasingly important in the management of subgroups of patients with good prognoses. Renal cell carcinoma, for example, can behave heterogeneously, with patients with good prognosis disease remaining on TKIs for several years [53]. In such subgroups of patients, particularly if cardiac risk factors exist, aggressive cardiac optimization may prevent deaths from cardiovascular outcomes. As many of the events in this study occurred later in the follow-up period, this finding is applicable to individuals who remain on TKIs for long periods of time. TKI usage is also being increasingly used in the adjuvant setting for prolonged durations [54], thus further necessitating minimization of long-term adverse effects.

Other known baseline cardiovascular risk factors may have contributed to death events, yet may not have been adequately captured. Risk factors such as diabetes and renal disease [55, 56] were not included in the validated algorithm used to establish baseline IHD [46]. Those with baseline diabetes or renal disease may not have been as aggressively managed for these comorbidities. Often, concerns of hypoglycaemia result in relaxed glycemic control [57, 58]. Those without overt baseline IHD, yet with other cardiovascular risk factors may be at higher likelihood for adverse events. Due to small numbers, a subgroup analysis to assess this was not possible.

The validated IHD algorithm used in this study assessed two physician billing codes (with one of the billing codes being from a physician in a hospital or emergency room setting) or one hospital discharge abstract to identify patients with IHD [46]. This algorithm excluded family physician diagnosis of angina or silent MI, which can be exclusively managed as an outpatient, and may have led to under-reporting of the prevalence of IHD [46].

This study has limitations. First, there is the potential for selection bias. Medications included in our analysis were available only to patients meeting certain criteria consistent with the registration trial supporting marketing of this drug. The effect of these drugs on individuals not meeting these criteria is unclear. Additionally, the NDFP only captures new or more expensive systemic treatments administered throughout the province. The proportion of cancer patients receiving chemotherapy prior to TKI initiation may be higher than captured through this study. The impact on subsequent cardiac outcomes is unlikely given the currently high proportion of chemotherapy use identified. Also, as a passive cancer registry, the OCR may not identify all cancer cases, as non-registry personnel may not be familiar with all reporting criteria and terminology. Second, we were unable to capture individuals treated with TKIs funded by mechanisms other than ODB, such as private drug insurance and self-pay options. Individuals with private drug coverage may also represent a different distribution of socioeconomic status (SES) compared to those relying on public healthcare coverage. SES has been shown to impact the survival of oncology patients, with lower SES being linked to poorer outcomes and decreased survival [5961]. Thirdly, as a population-based study, expansion of the sample size to increase event rates is not possible. Finally, granularity of detail is not available in the ICES administrative databases. Therefore, information relating to previous radiation treatment, dose of TKI, duration of treatment and cause of death was not available in most cases. This limitation, may lead to some uncertainty regarding the results.

In summary, individuals treated with TKIs have a significantly higher hazard of death relative to the general population. Cause specific hazards of IHD and of cerebrovascular accidents are not increased. Our results are consistent with recent randomized data suggesting that discontinuation of cardio-protective medications is safe, presumably since the absolute rate of cardiac events is very low. The increased mortality identified in this study is likely reflective of the underlying malignant process. More careful surveillance and management of cardiac risks is likely only warranted in the subgroup of patients with an expected prolonged survival.

Author Contributions

Conceived and designed the experiments: AO BS MK EA. Performed the experiments: AO BS EA MK. Analyzed the data: AS JLE AO BS MK EA. Contributed reagents/materials/analysis tools: AO BS MK EA. Wrote the paper: AS JLE AO BS MK EA.

References

  1. 1. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298(5600):1912–34. pmid:12471243
  2. 2. Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. The New England journal of medicine. 2005;353(2):172–87. pmid:16014887
  3. 3. Chen MH, Kerkela R, Force T. Mechanisms of cardiac dysfunction associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118(1):84–95. pmid:18591451
  4. 4. Steeghs N, Nortier JW, Gelderblom H. Small molecule tyrosine kinase inhibitors in the treatment of solid tumors: an update of recent developments. Annals of surgical oncology. 2007;14(2):942–53. pmid:17103252
  5. 5. Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368(9544):1329–38. pmid:17046465
  6. 6. Druker BJ, Guilhot F, O'Brien SG, Gathmann I, Kantarjian H, Gattermann N, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. The New England journal of medicine. 2006;355(23):2408–17. pmid:17151364
  7. 7. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. The New England journal of medicine. 2006;355(26):2733–43. pmid:17192538
  8. 8. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. The New England journal of medicine. 2004;350(23):2335–42. pmid:15175435
  9. 9. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. The New England journal of medicine. 2007;356(2):115–24. pmid:17215529
  10. 10. Sherbenou DW, Druker BJ. Applying the discovery of the Philadelphia chromosome. The Journal of clinical investigation. 2007;117(8):2067–74. pmid:17671641
  11. 11. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. The New England journal of medicine. 2001;344(11):783–92. pmid:11248153
  12. 12. Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, Thongprasert S, et al. Erlotinib in previously treated non-small-cell lung cancer. The New England journal of medicine. 2005;353(2):123–32. pmid:16014882
  13. 13. Dy GK, Adjei AA. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA: a cancer journal for clinicians. 2013;63(4):249–79.
  14. 14. Lenihan DJ. Tyrosine kinase inhibitors: can promising new therapy associated with cardiac toxicity strengthen the concept of teamwork? Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(32):5154–5.
  15. 15. Broekman F, Giovannetti E, Peters GJ. Tyrosine kinase inhibitors: Multi-targeted or single-targeted? World journal of clinical oncology. 2011;2(2):80–93. pmid:21603317
  16. 16. Levitzki A. Tyrosine kinase inhibitors: views of selectivity, sensitivity, and clinical performance. Annual review of pharmacology and toxicology. 2013;53:161–85. pmid:23043437
  17. 17. Niraula S, Seruga B, Ocana A, Shao T, Goldstein R, Tannock IF, et al. The price we pay for progress: a meta-analysis of harms of newly approved anticancer drugs. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012;30(24):3012–9.
  18. 18. Chen MH. Cardiac dysfunction induced by novel targeted anticancer therapy: an emerging issue. Current cardiology reports. 2009;11(3):167–74. pmid:19379636
  19. 19. Khakoo AY, Yeh ET. Therapy insight: Management of cardiovascular disease in patients with cancer and cardiac complications of cancer therapy. Nature clinical practice Oncology. 2008;5(11):655–67. pmid:18797437
  20. 20. Kim TD, le Coutre P, Schwarz M, Grille P, Levitin M, Fateh-Moghadam S, et al. Clinical cardiac safety profile of nilotinib. Haematologica. 2012;97(6):883–9. pmid:22271904
  21. 21. Steingart RM, Yadav N, Manrique C, Carver JR, Liu J. Cancer survivorship: cardiotoxic therapy in the adult cancer patient; cardiac outcomes with recommendations for patient management. Seminars in oncology. 2013;40(6):690–708. pmid:24331191
  22. 22. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2010;28(6):1061–8.
  23. 23. Strevel EL, Ing DJ, Siu LL. Molecularly targeted oncology therapeutics and prolongation of the QT interval. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2007;25(22):3362–71.
  24. 24. Bayer HealthCare Pharmaceuticals Inc. Nexavar: Highlights of Prescribing Information Whippany, New Jersey: Bayer HealthCare Pharmaceuticals Inc 2013 [updated July 17, 2014; cited 2014 July 17]. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021923s016lbl.pdf.
  25. 25. Genentech USA Inc. Tarceva: Highlights of Prescribing Information San Francisco, CA: Genentech USA, Inc., A Member of the Roche Group; 2014 [updated July 17, 2014; cited 2014 July 17]. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/021743s019lbl.pdf.
  26. 26. Pfizer Inc. Sutent: Highlights of Prescribing Information New York: Pfizer Inc.; 2013 [updated July 17, 2014; cited 2014 July 17]. Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021938s024s025lbl.pdf.
  27. 27. Heart and Stroke Foundation. Statistics: Heart Disease 2014 [updated July 18, 2014; cited 2014 July 18]. Available from: http://www.heartandstroke.com/site/c.ikIQLcMWJtE/b.3483991/k.34A8/Statistics.htm - heartdisease.
  28. 28. Carver JR, Shapiro CL, Ng A, Jacobs L, Schwartz C, Virgo KS, et al. American Society of Clinical Oncology clinical evidence review on the ongoing care of adult cancer survivors: cardiac and pulmonary late effects. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2007;25(25):3991–4008.
  29. 29. Eschenhagen T, Force T, Ewer MS, de Keulenaer GW, Suter TM, Anker SD, et al. Cardiovascular side effects of cancer therapies: a position statement from the Heart Failure Association of the European Society of Cardiology. European journal of heart failure. 2011;13(1):1–10. pmid:21169385
  30. 30. Schmidinger M, Zielinski CC, Vogl UM, Bojic A, Bojic M, Schukro C, et al. Cardiac toxicity of sunitinib and sorafenib in patients with metastatic renal cell carcinoma. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(32):5204–12.
  31. 31. Maitland ML, Bakris GL, Black HR, Chen HX, Durand JB, Elliott WJ, et al. Initial assessment, surveillance, and management of blood pressure in patients receiving vascular endothelial growth factor signaling pathway inhibitors. Journal of the National Cancer Institute. 2010;102(9):596–604. pmid:20351338
  32. 32. Lenihan DJ, Kowey PR. Overview and management of cardiac adverse events associated with tyrosine kinase inhibitors. The oncologist. 2013;18(8):900–8. pmid:23918069
  33. 33. Van Spall HG, Toren A, Kiss A, Fowler RA. Eligibility criteria of randomized controlled trials published in high-impact general medical journals: a systematic sampling review. JAMA: the journal of the American Medical Association. 2007;297(11):1233–40. pmid:17374817
  34. 34. Ocana A, Amir E, Seruga B. Clinical research: show us the data. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2011;29(9):1099–100.
  35. 35. Templeton AJ, Vera-Badillo FE, Wang L, Attalla M, De Gouveia P, Leibowitz-Amit R, et al. Translating clinical trials to clinical practice: outcomes of men with metastatic castration resistant prostate cancer treated with docetaxel and prednisone in and out of clinical trials. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2013;24(12):2972–7. pmid:24126362
  36. 36. Government of Canada. Population by year, by province and territory 2013 [updated June 15, 2014; cited 2014 2014-06-15]. Available from: http://www.statcan.gc.ca/tables-tableaux/sum-som/l01/cst01/demo02a-eng.htm.
  37. 37. Clarke EA, Marrett LD, Kreiger N. Cancer registration in Ontario: a computer approach. IARC scientific publications. 1991;(95):246–57. pmid:1894327
  38. 38. Robles SC, Marrett LD, Clarke EA, Risch HA. An application of capture-recapture methods to the estimation of completeness of cancer registration. Journal of clinical epidemiology. 1988;41(5):495–501. pmid:3367181
  39. 39. Levy AR, O'Brien BJ, Sellors C, Grootendorst P, Willison D. Coding accuracy of administrative drug claims in the Ontario Drug Benefit database. The Canadian journal of clinical pharmacology = Journal canadien de pharmacologie clinique. 2003;10(2):67–71. pmid:12879144
  40. 40. Ontario Ministry of Health and Long-Term Care. Ontario Public Drug Programs: Exceptional Access Program (EAP) Reimbursement Criteria 2013 [cited 2014 December 3]. Available from: http://www.health.gov.on.ca/en/pro/programs/drugs/eap_criteria_list.aspx.
  41. 41. Ontario Ministry of Health and Long-Term Care. Ontario Public Drug Programs: How Drugs Are Approved: Queen's Printer For Ontario; 2013 [updated May 23; cited 2014 October 7]. Available from: http://www.health.gov.on.ca/en/pro/programs/drugs/how_drugs_approv/how_drugs_approv.aspx.
  42. 42. Cancer Care Ontario. Cancer Drugs—Frequently Asked Questions Toronto, Ontario2014 [updated January 6; cited 2014 October 7]. Available from: https://www.cancercare.on.ca/cms/One.aspx?portalId=1377&pageId=11829-1.
  43. 43. Hux JE, Ivis F, Flintoft V, Bica A. Diabetes in Ontario: determination of prevalence and incidence using a validated administrative data algorithm. Diabetes care. 2002;25(3):512–6. pmid:11874939
  44. 44. Schultz SE, Rothwell DM, Chen Z, Tu K. Identifying cases of congestive heart failure from administrative data: a validation study using primary care patient records. Chronic diseases and injuries in Canada. 2013;33(3):160–6. pmid:23735455
  45. 45. Tu K, Campbell NR, Chen ZL, Cauch-Dudek KJ, McAlister FA. Accuracy of administrative databases in identifying patients with hypertension. Open medicine: a peer-reviewed, independent, open-access journal. 2007;1(1):e18–26.
  46. 46. Tu K, Mitiku T, Lee DS, Guo H, Tu JV. Validation of physician billing and hospitalization data to identify patients with ischemic heart disease using data from the Electronic Medical Record Administrative data Linked Database (EMRALD). The Canadian journal of cardiology. 2010;26(7):e225–8. pmid:20847968
  47. 47. Alter DA, Naylor CD, Austin P, Tu JV. Effects of socioeconomic status on access to invasive cardiac procedures and on mortality after acute myocardial infarction. The New England journal of medicine. 1999;341(18):1359–67. pmid:10536129
  48. 48. Suter TM, Ewer MS. Cancer drugs and the heart: importance and management. European heart journal. 2013;34(15):1102–11. pmid:22789916
  49. 49. Abernethy APK, Jean; Blatchford, Patrick Jud Blatchford; Ritchie, Christine; Fairclough, Diane; Hanson, Laura; Bull, Janet; PCRC Investigators. Managing comorbidities in oncology: A multisite randomized controlled trial of continuing versus discontinuing statins in the setting of life-limiting illness. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(5s):(suppl; abstr LBA9514).
  50. 50. Dienstmann R, Brana I, Rodon J, Tabernero J. Toxicity as a biomarker of efficacy of molecular targeted therapies: focus on EGFR and VEGF inhibiting anticancer drugs. The oncologist. 2011;16(12):1729–40. pmid:22135123
  51. 51. George S, Reichardt P, Lechner T, Li S, Cohen DP, Demetri GD. Hypertension as a potential biomarker of efficacy in patients with gastrointestinal stromal tumor treated with sunitinib. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2012;23(12):3180–7. pmid:22858558
  52. 52. Zee YKM N.; Kumaran G.; Swindell R.; Saunders M. P.; Clamp A. R.; Valle J. W.; Wilson G.; Jayson G. C. and Hasan J. Hypertension (HTN) and proteinuria (PTN) as biomarkers of efficacy in antiangiogenic therapy for metastatic colorectal cancer (mCRC). J Clin Oncol, 2010 ASCO Annual Meeting Abstracts. 2010;Vol 28(No 15_suppl (May 20 Supplement)).
  53. 53. Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2009;27(34):5794–9.
  54. 54. Joensuu H, Eriksson M, Sundby Hall K, Hartmann JT, Pink D, Schutte J, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA: the journal of the American Medical Association. 2012;307(12):1265–72. pmid:22453568
  55. 55. Ray KK, Seshasai SR, Wijesuriya S, Sivakumaran R, Nethercott S, Preiss D, et al. Effect of intensive control of glucose on cardiovascular outcomes and death in patients with diabetes mellitus: a meta-analysis of randomised controlled trials. Lancet. 2009;373(9677):1765–72. pmid:19465231
  56. 56. London G, Covic A, Goldsmith D, Wiecek A, Suleymanlar G, Ortiz A, et al. Arterial aging and arterial disease: interplay between central hemodynamics, cardiac work, and organ flow-implications for CKD and cardiovascular disease. Kidney international supplements. 2011;1(1):10–2. pmid:25018896
  57. 57. Bertoluci MC, Pimazoni-Netto A, Pires AC, Pesaro AE, Schaan BD, Caramelli B, et al. Diabetes and cardiovascular disease: from evidence to clinical practice—position statement 2014 of Brazilian Diabetes Society. Diabetology & metabolic syndrome. 2014;6:58.
  58. 58. Canadian Diabetes Association Clinical Practice Guidelines Expert C, Cheng AY. Canadian Diabetes Association 2013 clinical practice guidelines for the prevention and management of diabetes in Canada. Introduction. Canadian journal of diabetes. 2013;37 (Suppl 1):S1–3. pmid:24070926
  59. 59. Reames BN, Birkmeyer NJ, Dimick JB, Ghaferi AA. Socioeconomic disparities in mortality after cancer surgery: failure to rescue. JAMA surgery. 2014;149(5):475–81. pmid:24623106
  60. 60. Shariff-Marco S, Yang J, John EM, Sangaramoorthy M, Hertz A, Koo J, et al. Impact of neighborhood and individual socioeconomic status on survival after breast cancer varies by race/ethnicity: the Neighborhood and Breast Cancer Study. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2014;23(5):793–811.
  61. 61. Tao L, Foran JM, Clarke CA, Gomez SL, Keegan TH. Socioeconomic disparities in mortality after diffuse large B-cell lymphoma in the modern treatment era. Blood. 2014;123(23):3553–62. pmid:24705494