Cardiotoxicity of Targeted Agents in Oncology: A Medical Oncology Perspective

Publication
Article
OncologyOncology Vol 28 No 6
Volume 28
Issue 6

Without question, targeted therapies have revolutionized the treatment of cancer across histologies.

Without question, targeted therapies have revolutionized the treatment of cancer across histologies. Given the prevalence of breast cancer and the fact that the human epidermal growth factor receptor 2 (HER2) alteration was discovered more than 25 years ago, HER2-directed therapies are arguably the best-studied of the biologics in terms of efficacy and cardiac-specific safety. For the ~25% of breast cancers that overexpress HER2, the addition of trastuzumab to chemotherapy has been shown to substantially improve survival in both the adjuvant and metastatic settings.[1-4] More recently, lapatinib, pertuzumab, and the antibody-drug conjugate ado-trastuzumab emtansine (T-DM1) have shown substantial activity against HER2-positive breast cancer. Fortunately, these therapies have been generally safe. However, as Bhave and colleagues point out, there is a recognized risk of cardiac toxicity that must be considered carefully. While several risk factors have been linked to the development of cardiotoxicity from HER2-targeted therapies, coadministration with anthracyclines augments this risk significantly.

Bhave and colleagues[5] present a thorough review of the cardiac complications associated with HER2-targeted therapies based on data primarily from the metastatic setting. However, a discussion relating to the use of trastuzumab in the curative setting is lacking. While the risk of cardiac toxicity may be outweighed by the known benefits achieved in patients with metastatic disease, the highest level of caution should be used when treating patients in the adjuvant setting, since a rather substantial proportion of women with early-stage disease are cured by local-regional therapy alone. Distinguishing patients who are cured and do not require systemic therapy from those who have distant micrometastases is still impossible. Consequently, a large number of patients continue to receive systemic therapy, and all the associated toxicities, unnecessarily. Therefore, the choice of an adjuvant treatment regimen should be based not only on efficacy but also on a critical evaluation of the risk of long-term toxicity, including heart damage. One way to mitigate this risk in the HER2-positive setting is to choose a chemotherapy backbone that does not cause additional cardiac toxicity. A nonanthracycline regimen-docetaxel, carboplatin, trastuzumab (TCH)-was tested in the Breast Cancer International Research Group (BCIRG)-006 adjuvant study and shown to confer a 0.4% risk of heart failure.[4] In contrast, 2% of patients in the doxorubicin-cyclophosphamide→docetaxel-trastuzumab (AC-TH) arm of that study developed heart failure, and a higher proportion of patients had a sustained decrease in left ventricular ejection fraction (LVEF) at 48 months. Furthermore, among the 1,073 patients randomly assigned to the AC-TH arm, 23 patients were unable to receive trastuzumab because they developed heart failure while receiving AC, presumably due to type I cardiac toxicity from doxorubicin.[4] Given the significant improvement in progression-free survival and overall survival associated with the addition of trastuzumab to adjuvant chemotherapy, choosing a chemotherapy regimen that maximizes the chances that patients can receive the full course of trastuzumab is critical. Three other studies have evaluated TCH in over 3,400 patients as a therapeutic backbone in the curative setting, providing more support for its safety and efficacy.[6-8]

One question that has arisen is whether the various HER2-targeted agents have differing levels of cardiac toxicity. The authors report a reduced incidence of cardiac events with lapatinib compared with trastuzumab and hypothesize that this is because lapatinib protects cardiomyocytes against apoptosis via induction of AMP-activated protein kinase (AMPK) activity. While this is one possibility, it should be recognized that almost all patients enrolled in the early studies evaluating lapatinib had already been exposed to trastuzumab, an anthracycline, or both.[9-11] Furthermore, to be eligible for study entry, patients had to have normal cardiac function. Therefore, the lower incidence may, in part, be a result of selection bias. Having said that, in the MA-31 study, in which a frontline taxane-lapatinib regimen was compared to a taxane-trastuzumab regimen, up to 3% of the trastuzumab-treated patients experienced a > 20% decrease in LVEF over the course of the study, compared to 0% in the lapatinib arm.[12] Interestingly, use of dual HER2-targeted therapy with pertuzumab and trastuzumab[13] does not appear to increase the risk of cardiac events. Although Bhave and colleagues do not specifically discuss the cardiac data available for the recently approved T-DM1, data from the EMILIA study are reassuring, as there was only a 1.7% incidence of LVEF reduction in T-DM1-treated patients (vs 1.6% in the lapatinib arm) and only 1 case of grade 3 cardiomyopathy (vs none in the lapatinib arm).[14]

Data relating to the cardiac impact of antiangiogenic agents such as bevacizumab, sunitinib, and sorafenib are also reviewed. The authors point out the hypertensive effects, risks of thromboembolic disease, and incidence of congestive heart failure with these biologics. However, they did not include in their review the recent meta-analysis of the safety of bevacizumab reported by Cortes and colleagues that included 3,784 patients enrolled in phase III trials evaluating bevacizumab plus chemotherapy in metastatic breast cancer.[15] The incidence of grade 3/4 left ventricular dysfunction was over 2.2 times more common in patients receiving bevacizumab. This and other serious toxicities, coupled with the lack of demonstration of an overall survival benefit in patients with metastatic breast cancer, led to the withdrawal of FDA approval for bevacizumab in this disease setting, underscoring the need to carefully evaluate the therapeutic index of new agents. Antiangiogenic agents continue to play an important role in the treatment of other cancers; however, appropriate patient selection and clinician awareness of the potential toxicities are key to optimizing the safe use of these therapies.

Bhave and colleagues summarize well many other effective targeted cancer therapeutics currently in use. Many of these carry the risk of infusion reactions and venous thromboembolic events, but their risk of cardiotoxicity per se is less well defined. Additionally, several of the drugs reviewed were tested in patients with preexisting cardiac disease, making it more difficult to gauge the risk of heart toxicity in a low-risk patient. With so many approved biologic options, patients may receive treatment with such agents for years. Careful comparative assessment of toxicity and long-term follow-up are needed to understand the impacts of these therapies, alone or in combination with chemotherapy, on cardiac function.

What is clearly lacking is a consensus statement relating to the appropriate screening, monitoring, and management of the cardiac effects of each of these drugs specifically. That said, the European Society for Medical Oncology has published a rather thorough clinical practice guideline for the management of cardiac toxicity associated with chemotherapy, radiation, and targeted agents.[16] Although Bhave and colleagues do not specifically reference this guideline, they do mention others published by the American College of Cardiology and the American Heart Association. A thorough evaluation and summary statement of the existing guidelines would be most useful to a practicing clinician. In addition, it would be helpful to have a monitoring algorithm for patients who are being treated in the metastatic setting (in some cases for many years) with a targeted agent that is known to have cardiac toxicity. For example, how frequently and for how long should echocardiograms be performed in an asymptomatic patient?

The cardiac impact of biologic therapies is a critically important topic of discussion, not only for the practicing oncologist but also for investigators and scientists, especially as the most effective agents are moved to the adjuvant curative setting. Awareness of the potential side effects, recognition of signs and symptoms, ability to differentiate cardiac from noncardiac causes of side effects, and an understanding of how to intervene are all crucial for providing quality patient care. As the field continues to improve survival outcomes with new and exciting therapies, long-term follow-up not only to determine efficacy but also for safety is needed. Standardized guidelines to ensure consistent, quality care are imperative.

Financial Disclosure:Dr. Hurvitz receives research funding from Genentech/Roche and GlaxoSmithKline (paid to the University of California, Los Angeles), and her travel to conferences at which she was an invited speaker has been reimbursed by Genentech/Roche. Dr. Wong has no significant financial interest in or other relationship with the manufacturer of any product or provider of any service mentioned in this article.

References:

1. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783-92.

2. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659-72.

3. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673-84.

4. Slamon D, Eiermann W, Robert N, et al. Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med. 2011;365:1273-83.

5. Bhave M, Akhter N, Rosen ST. Cardiovascular toxicity of biologic agents for cancer therapy. Oncology (Williston Park). 2014;28:XXX-XXX.

6. Hurvitz C, Miller J, Dichmann R, et al. Final analysis of a phase II 3-arm randomized trial of neoadjuvant trastuzumab or lapatinib or the combination of trastuzumab and lapatinib, followed by six cycles of docetaxel and carboplatin with trastuzumab and/or lapatinib in patients with HER2+ breast cancer (TRIO-US B07). San Antonio Breast Cancer Symposium. Dec 10–14, 2013; San Antonio, TX:abstr S1-02.

7. Schneeweiss A, Chia S, Hickish T, et al. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol. 2013;24:2278-84.

8. Slamon DJ, Swain SM, Buyse M, et al. Primary results from BETH, a phase 3 controlled study of adjuvant chemotherapy and trastuzumab ± bevacizumab in patients with HER2-positive, node-positive or high risk node-negative breast cancer. San Antonio Breast Cancer Symposium. Dec 10–14, 2013; San Antonio, TX:abstr S1-03.

9. Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355:2733-43.

10. Blackwell KL, Burstein HJ, Storniolo AM, et al. Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol. 2010;28:1124-30.

11. Blackwell KL, Burstein HJ, Storniolo AM, et al. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol. 2012;30:2585-92.

12. Gelmon KA, Boyle F, Kaufman B, et al. Open-label phase III randomized controlled trial comparing taxane-based chemotherapy (Tax) with lapatinib (L) or trastuzumab (T) as first-line therapy for women with HER2+ metastatic breast cancer: Interim analysis (IA) of NCIC CTG MA.31/GSK EGF 108919. J Clin Oncol. 2012;30(suppl):abstr LBA671.

13. Baselga J, Cortes J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109-19.

14. Verma S, Miles D, Gianni L, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367:1783-91.

15. Cortes J, Calvo V, Ramirez-Merino N, et al. Adverse events risk associated with bevacizumab addition to breast cancer chemotherapy: a meta-analysis. Ann Oncol. 2012;23:1130-7.

16. Curigliano G, Cardinale D, Suter T, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155-66.

Articles in this issue

Fractionation in Breast Cancer Radiotherapy for Conservative Treatment: Are We Really Done Learning?
Consider a Single Intraoperative Fraction for Patients Eligible for Hypofractionated Regimens?
Cardiovascular Toxicity of Newer Chemotherapeutic Agents: The Heart of the Matter
Cardiotoxicity of Targeted Agents in Oncology: A Medical Oncology Perspective
It's Time to Have ‘The Talk’: Cost Communication and Patient-Centered Care
It's Time to Have ‘The Talk’: Cost Communication and Patient-Centered Care
Double Trouble: A Case of Concurrent De Novo T790M and L858R EGFR Mutations in Treatment-Naive Advanced Non–Small-Cell Lung Cancer
Double Trouble: A Case of Concurrent De Novo T790M and L858R EGFR Mutations in Treatment-Naive Advanced Non–Small-Cell Lung Cancer
EGJ and Esophageal Cancers: Choosing Induction Therapy so as to Err on the Side of Overtreatment Rather Than Undertreatment When Staging Is Imperfect
Esophagogastric Junction and Gastric Adenocarcinoma: Current Challenges and Future Directions
Cardiovascular Toxicity of Biologic Agents for Cancer Therapy
Cardiovascular Toxicity of Biologic Agents for Cancer Therapy
Esophagogastric Junction and Gastric Adenocarcinoma: Neoadjuvant and Adjuvant Therapy, and Future Directions
Esophagogastric Junction and Gastric Adenocarcinoma: Neoadjuvant and Adjuvant Therapy, and Future Directions
The Role of Radical Prostatectomy and Lymph Node Dissection in the Treatment of Young Men With High-Grade Node-Positive Prostate Cancer:  There May Be No RCTs-but There Are Good Reasons to Include Surgery
The Role of Radical Prostatectomy and Lymph Node Dissection in the Treatment of Young Men With High-Grade Node-Positive Prostate Cancer: There May Be No RCTs-but There Are Good Reasons to Include Surgery
The Role of Radical Prostatectomy and Lymph Node Dissection in the Treatment of Young Men With High-Grade Node-Positive Prostate Cancer: Less Is More-the Benefits of Surgery Do Not Yet Outweigh Potential Harms
The Role of Radical Prostatectomy and Lymph Node Dissection in the Treatment of Young Men With High-Grade Node-Positive Prostate Cancer: Less Is More-the Benefits of Surgery Do Not Yet Outweigh Potential Harms
Hypofractionation for Breast Cancer: Lessons Learned From Our Neighbors to the North and Across the Pond
Hypofractionation for Breast Cancer: Lessons Learned From Our Neighbors to the North and Across the Pond
The Challenge of Managing Increasingly Complex Cancer Toxicity
Related Videos
Pegulicianine-guided breast cancer surgery may allow practices to de-escalate subsequent radiotherapy, says Barbara Smith, MD, PhD.
Barbara Smith, MD, PhD, spoke about the potential use of pegulicianine-guided breast cancer surgery based on reports from the phase 3 INSITE trial.
Carey Anders, MD, an expert on breast cancer
Carey Anders, MD, an expert on breast cancer
Carey Anders, MD, an expert on breast cancer
Carey K. Anders, MD, an expert on breast cancer
A panel of 4 experts on breast cancer seated at a long table
A panel of 4 experts on breast cancer seated at a long table