1,240
Views
16
CrossRef citations to date
0
Altmetric
Original Article

Cost-effectiveness of everolimus vs sunitinib in treating patients with advanced, progressive pancreatic neuroendocrine tumors in the United States

, , , , &
Pages 55-64 | Accepted 08 Aug 2012, Published online: 03 Sep 2012

Abstract

Background:

Everolimus (Afinitor) and sunitinib (Sutent) were recently approved to treat patients with advanced, progressive pancreatic neuroendocrine tumors (pNETs). (Afinitor is a registered trademark of Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA; Sutent is a registered trademark of Pfizer Inc., New York, NY, USA.) This analysis examined the projected cost-effectiveness of everolimus vs sunitinib in this setting from a US payer perspective.

Methods:

A semi-Markov model was developed to simulate a cohort of patients with advanced, progressive pNET and to estimate the cost per life-year gained (LYG) and per quality-adjusted life-year (QALY) gained when treating with everolimus vs sunitinib. Efficacy data were based on a weight-adjusted indirect comparison of the agents using phase 3 trial data. Model health states included: stable disease with no adverse events, stable disease with adverse events, disease progression, and death. Therapy costs were based on wholesale acquisition cost. Other costs such as physician visits, tests, hospitalizations, and adverse event costs were obtained from literature and/or primary research. Utility inputs were based on primary research. Sensitivity analyses were conducted to test the model’s robustness.

Results:

In the base-case analysis, everolimus was associated with an incremental 0.448 LYG (0.304 QALYs) at an incremental cost of $12,673, resulting in an incremental cost-effectiveness ratio (ICER) of $28,281/LYG ($41,702/QALY gained). The ICER fell within the cost per QALY range for many widely used oncology drugs. Sensitivity analyses demonstrated that, overall, there is a trend that everolimus is cost-effective compared to sunitinib in this setting.

Limitations:

Results of the indirect analysis were not statistically significant (p > 0.05). Assumptions that treatment patterns are the same across therapies may not represent real-world practice.

Conclusions:

While the analysis is limited by its reliance on an indirect comparison of two phase 3 studies, everolimus is expected to be cost-effective relative to sunitinib in advanced, progressive pNET.

Introduction

Pancreatic neuroendocrine tumors (pNETs) are tumors that arise from well-differentiated mature endocrine cells or multipotent stem cells within the pancreasCitation1. The overall annual incidence of pNETs in the US is estimated to be 3.2 per million, of which 64% are diagnosed in an advanced stageCitation2. Advanced NET, defined as unresectable and/or metastatic disease, is a rare, progressive, and fatal malignancyCitation2. The median survival for patients with well- to moderately-differentiated pNET with distant metastases is 27 monthsCitation2.

Until recently, only streptozocin had been approved by the US Food and Drug Administration (FDA) for the treatment of pNET. The lack of approved therapies has contributed to inconsistent treatment approaches and the use of toxic agents such as chemotherapy. Everolimus (Afinitor; a registered trademark of Novartis Pharmaceuticals Corporation, East Hanover, NJ), recently approved by the FDA for treatment of progressive pNETs, represents a significant clinical advance in the treatment of advanced NET by controlling disease progression through inhibition of the mammalian target of rapamycin (mTOR) pathway. The pivotal RADIANT-3 trial demonstrated that everolimus 10 mg/day administered orally plus best supportive care was associated with a 65% reduction in disease progression risk (hazard ratio [HR] = 0.35; 95% confidence interval [CI]: 0.27, 0.45; p < 0.001) in patients with advanced pNET when compared to placebo. The median progression-free survival (PFS) was 11.0 months (95% CI: 8.4, 13.9) for everolimus-treated patients and 4.6 months (95% CI: 3.1, 5.4) for placebo-treated patients. Median overall survival (OS) was not reached at the time of analysisCitation3. Sunitinib (Sutent; a registered trademark of Pfizer Inc., New York, NY), an anti-vascular endothelial growth factor (VEGF) therapy, was also recently approved by the FDA to treat progressive, well-differentiated pNET in patients with unresectable locally advanced or metastatic disease. Its pivotal trial (A6181111) demonstrated that sunitinib 37.5 mg/day administered orally was associated with a 58% reduction in the risk of disease progression or death (HR: 0.42; 95% CI: 0.26, 0.66; p < 0.001). The median PFS was 11.4 months (95% CI: 7.4, 19.8) for sunitinib-treated patients and 5.5 months (95% CI: 3.6, 7.4) for placebo-treated patients. Early OS data reported in a recent New England Journal of Medicine publication of the A6181111 trial suggested that treatment was associated with a 59% reduction in the risk of death (HR: 0.41; 95% CI: 0.19, 0.89; p = 0.02)Citation4. However, subsequent analyses submitted to the European Medicines Agency (EMA) and FDA of more mature data were no longer statistically significant (HR: 0.74; 95% CI: 0.47, 1.17)Citation5.

As there were no head-to-head trials comparing everolimus and sunitinib in advanced, progressive pNET, an indirect comparison was performed based upon the sunitinib A6181111 trial and a matched sub-set of the RADIANT-3 populationCitation6. A comparable study was not available for streptozocin, and thus an indirect comparison could not be performed against everolimus. The weight-adjusted indirect comparison against sunitinib was performed using a method of moments estimator, as previously used in a Signorovich et al.Citation7 study. The analysis demonstrated a trend towards improved PFS and OS with everolimus compared to sunitinib. To date, there have been no published economic evaluations of these treatments in pNET. Consequently, based on the results of the indirect survival comparison, this analysis examines the potential cost-effectiveness of everolimus vs sunitinib from a US payer (i.e., insurer) perspective.

Methods

Model structure

A semi-Markov model was developed in Microsoft Excel® (Microsoft Corp, Redmond, WA) to simulate two hypothetical patient cohorts with advanced, progressive pancreatic NET: one treated with everolimus and the other with sunitinib. The cohorts were modeled over a 20-year time horizon in monthly (30.4-day) cycles; the 30.4-day cycle length was calculated by dividing the number of days per year (365) by the number of months per year (12). A 20-year time horizon was chosen in order to allow 99.99% of patients in both arms to reach the absorbing state. Extrapolation of the efficacy data had produced a long tail for progression-free and overall survival, thus necessitating a long time horizon. Four health states were included in the model: stable disease with no adverse events, stable disease with adverse events, disease progression, and death ().

Figure 1.  Semi-Markov model health states.

Figure 1.  Semi-Markov model health states.

All patients started in the health state ‘stable disease with no adverse events’ and transitioned to the remaining health states according to PFS and OS estimates obtained from the indirect analysisCitation6. Movement among health states was unidirectional: patients who transitioned from stable disease to disease progression were unable to return to the former state. The probability of health state membership was estimated using a partitioned survival analysis, which calculates the mean time spent in a health state from the area under the appropriate survival curve (). This type of analysis has been described by Glasziou et al.Citation8,Citation9. In the model presented herein, Weibull cumulative distribution curves for the everolimus arm were constructed from the PFS (shape: 1.195, 95% CI: 0.990–1.429; scale: 447.188, 95% CI: 223.632–897.847) and OS (shape: 1.379, 95% CI: 1.111–1.724; scale: 1427.245, 95% CI: 765.095–2670.444) data for the matched sub-set of the RADIANT-3 population (derived in statistical software [SAS 9.2, SAS Institute, Cary, NC USA]); the area under the curve was calculated to derive the health-state membership at each cycle. As health states were mutually exclusive, the disease progression health-state membership was calculated as the complement of the summation of the stable disease and death states. The Weibull distribution was used because it is uniquely relevant in the clinical settingCitation10.

Figure 2.  Partitioned survival analysis of (a) everolimus patients and (b) sunitinib patients.

Figure 2.  Partitioned survival analysis of (a) everolimus patients and (b) sunitinib patients.

Weibull curves for sunitinib were constructed by applying the hazard ratios for PFS and OS (0.84 [95% CI: 0.460–1.530] and 0.81 [95% CI: 0.490–1.310], respectively) reported in the indirect analysisCitation6 to the resultant transition ratesCitation11 from the everolimus-based parametric curves. As the survival curves incorporated in the model were time-dependent, the transition probabilities (and the transition rates) varied according to the cycle. A similar methodology utilizing Weibull distributions fitted to Kaplan-Meier curves for the baseline therapy, and hazard ratios to estimate the expected disease progression in the comparator was used in the Peninsula Technology Assessment Group’s (PenTAG) assessment of bevacizumab, sorafenib tosylate, sunitinib, and temsirolimus for renal cell carcinomaCitation12.

The proportion of everolimus-treated patients who experienced an adverse event (AE) in each cycle was calculated from the RADIANT-3 data. For the weight-adjusted matched population used in the indirect comparison with sunitinib, the proportion of everolimus patients who experienced an adverse event was derived by calculating the AE rate ratio between the pre- and post-matched everolimus population and applying the inverse to the former. The proportion of sunitinib patients who experienced an adverse event at each cycle was then derived by calculating the ratio of AE rates between the post-match everolimus population and the sunitinib population, and applying the inverse to the cycle-specific adverse event rate of the post-matched everolimus arm. It was assumed that presence in ‘stable disease with adverse events’ occurred for a single cycle as many of the AEs could be resolved within a hospitalization visit and did not lead to treatment discontinuation; presence in this state was cross-sectionally assessed for each model cycle. The adverse events included in the analysis were based on the grade 3/4 AEs that occurred in at least 2% of everolimus patients (irrespective of treatment)Citation13 and at least 2% of sunitinib patientsCitation5, as well as any AEs that had significantly different rates (p < 0.05) in the patient populations (of which there were eight), as identified in the indirect analysisCitation6. Adverse event status did not affect likelihood of progression or death, but was used to calculate the associated quality-of-life (QoL) reductions and increased costs.

Resource utilization

To estimate resource utilization, a survey of physicians with experience in NET treatment in the US was conductedCitation14. Physicians reported treatment practice patterns on patients (40 across all physicians) during three disease stages: baseline, initial progression, and second progression.

The cost-effectiveness analysis incorporated the resource utilization estimates from the initial progression stage for the model’s stable disease state as the RADIANT-3 patient population consists of persons with advanced, progressive NET. The analysis incorporated the survey’s second progression results in the model’s disease progression state (i.e., when the patients represented in the model progress a second time). The model did not incorporate the resource utilization data captured for the survey baseline disease stage, as patients in this category have not yet progressed. Results were analyzed using statistical software (Cary, NC).

Cost inputs

In the RADIANT-3 trial and in the A6181111 study, patients received, respectively, everolimus 10 mg/day or sunitinib 37.5 mg/day (or placebo in both studies) until disease progression or dose reduction/discontinuation due to tolerance issues. The model used the patient-level data to estimate the treatment dose intensity, calculated to be 85.9% for everolimusCitation13 and 91.3% for sunitinibCitation4. The drug cost of everolimus and sunitinib was based on 2011 wholesale acquisition cost, and was estimated to be $221 per day and $230 per day, respectively ()Citation15.

Table 1.  Drug acquisition costs.

Patients also received supportive care, which included symptomatic care (somatostatin analogs), physician visits, imaging and lab tests, hospitalizations, treatment for AEs (i.e., AE-specific hospitalizations), post-progression therapy, and end-of-life care (). Drug costs were derived from an electronic drug reference (Red Book for Windows 2011, Thomson Reuters, Greenwood Village, CO USACitation15); medical resource utilization costs were taken from procedure codes (Physicians' Fee and Coding Guide, MAG Mutual, Atlanta, GA USACitation16); and the Thomson Reuters© MS-DRG 2010 HandbookCitation17. Costs of AEs were based on the Healthcare Cost & Utilization ProjectCitation18 and secondary literatureCitation19–21. End-of-life care costs were calculated from literatureCitation22. As this analysis examined the cost-effectiveness of the two oncologics from a third-party payer’s perspective, other societal costs, such as patient time or productivity losses, were not considered.

Table 2.  Health state medical costs.

Health state utility values

Health state utility scores (between 0 = death, and 1 = perfect health) were used to calculate quality-adjusted life-years (QALYs). A time trade-off (TTO) study was conducted in the UK to estimate the utility value of the stable disease and disease progression health statesCitation23. Disutility values associated with specific adverse events were obtained from the study, and were multiplied by the relative frequency of the corresponding event to yield a weighted average disutility value for each comparator’s adverse event profile (). It should be noted that the time trade-off study did not provide disutility values for all adverse events present in the model.

Table 3.  Health state utility values.

Base-case analysis

The effectiveness and total costs for each cohort were assessed and compared based upon the point estimates of the hazard ratios for PFS and OS from the indirect comparison. The analyses estimated the total and incremental life-years (LYs) and QALYs. Both costs and survival estimates were discounted annually. The model incorporates a discount rate of 3%, as supported by pharmacoeconomic guidelines published by the Academy of Managed Care PharmacyCitation24. Discounting began during the first year of the model. The ICERs were calculated as the cost per life-year gained and cost per QALY gained using the discounted total costs and survival time in the final model cycle (cycle 240).

Sensitivity analyses

One-way sensitivity analyses were performed to identify parameters driving the ICERs. Parameters tested included: discount rate of the cost and efficacy values, model time horizon, adverse event care costs, post-progression treatment costs, end-of-life care costs associated with death, health state utility values, and PFS and OS hazard ratios. Additionally, the consistency of the results across scenarios was evaluated by a probabilistic sensitivity analysis (PSA). The cost parameters were assumed to follow a gamma distribution, utility parameters to follow a beta distribution, the Weibull survival curve parameters to follow a normal distribution, and the hazard ratio parameters to follow a log-normal distribution (). The PSA was run for 1000 iterations. Results of the PSA were used to generate a cost-effectiveness scatterplot.

Table 4.  Model parameters varied in the PSA and their distributions.

Results

The base-case analysis projected everolimus and sunitinib to yield mean survival times of 3.298 life-years and 2.850 life-years, respectively. Everolimus treatment would incur an incremental cost of $12,673, 23% of which comprised active treatment costs associated with the predicted increase in PFS implied by the indirect comparison (). Estimated post-progression treatment costs were higher for the everolimus cohort than for the sunitinib cohort, which is also a result of the implied additional survival of the everolimus patients per the indirect analysis. Incremental cost-effectiveness ratios for the base-case analysis were $28,281/LYG and $41,702/QALY.

Table 5.  Base case results.

The one-way sensitivity analyses are summarized in . The PFS hazard ratio point estimate greatly influenced model results. Active treatment dose intensity, post-progression treatments costs, and adverse event costs were also influential on the results. The PSA yielded results that were consistent with the deterministic analysis. The probabilities that the ICER fell below $50,000/QALY and $100,000/QALY were 53% and 69%, respectively. The PSA yielded some instances when everolimus was dominated by sunitinib, as well as instances when everolimus was dominant (); however, the probability that everolimus had a gain in QALYs over sunitinib was 81%.

Table 6.  One-way sensitivity analysis results.

Figure 3.  Probabilistic sensitivity scatter plot. Note: The ellipse demonstrates the 95% confidence interval for the PSA results and the center of the triangle represents the base-case ICER.

Figure 3.  Probabilistic sensitivity scatter plot. Note: The ellipse demonstrates the 95% confidence interval for the PSA results and the center of the triangle represents the base-case ICER.

Discussion

While everolimus treatment of patients with advanced, progressive pNET has proven to be clinically efficacious, model results indicate that it is also likely to be cost-effective compared to sunitinib: specifically, in 81% of simulated projections, everolimus had an added benefit in terms of QALYs gained. Overall, the results of this analysis remained consistent across one-way sensitivity analyses (). The PFS hazard ratio has the greatest impact on model results. While it may seem counterintuitive that the lower bound of this parameter (the maximum expected efficacy for PFS) produced an ICER of $144,652, while the upper bound (the lowest expected efficacy for PFS) produced a dominant ICER, these results can be explained by the fact that the PFS and OS are not assumed to be correlated and are modeled independently in the analysis. Therefore, as the HR for PFS decreases, the PFS duration (and corresponding active treatment cost) for everolimus increases relative to sunitinib, while OS remains unaffected. This results in a higher ICER despite the improved efficacy. This result also reflects the fact that the change in QALYs in the denominator of the ICER is only slightly affected by improved efficacy in terms of PFS and is largely driven by OS. Overall, based on the available data in comparison to sunitinib (along with the associated variability of these data), everolimus has a high probability (69%) of being considered cost-effective at a willingness-to-pay threshold of $100,000/QALY.

The conclusions from this analysis should be viewed in light of several limitations. Although the indirect analysis demonstrated a trend towards improved PFS and OS for everolimus vs sunitinib, the calculated HRs were not statistically significant (p > 0.05). Despite this uncertainty, the estimates indicate that it is more likely that everolimus will provide added benefit and be cost-effective compared to sunitinib, than the converse. The fact that the HRs were not statistically significant may explain why everolimus was dominated by sunitinib in rare instances of the PSA and was dominant in other instances.

Furthermore, due to limitations in the availability of adverse event reporting between the trials, the model accounts for AE costs in a manner that is likely biased against everolimus. The RADIANT-3 trial captured AEs irrespective of the relationship to the study treatment while the sunitinib trial captured treatment-emergent AEs: therefore, the model may be over-estimating the AE costs for the everolimus arm. As expected, sensitivity analysis results demonstrate that everolimus remains cost-effective compared to sunitinib when AE costs for both arms are assumed to be $0.

Regarding resource utilization, the analysis assumes that all patients in stable disease (both those with and without adverse events) have the same resource utilization rates, aside from the inclusion of adverse event hospitalizations for the former. It is also assumed that treatment patterns are the same across therapies, which may not represent real-world practice. Furthermore, as many of the physicians were unable to report data for the model’s ‘disease progression’ state, the majority of responses for this group were based on hypothetical treatment scenarios. Although the survey results may differ from real-world treatment patterns of patients with pNET, they offer informed assumptions. The limitations of this study are expected to affect both model treatment arms similarly.

Study results also depend on the selected utility values. This analysis used values from a time trade-off study executed through fielding a survey of healthy individuals’ responses to NET-related vignettes. This method may be limited as the instrument may lack sensitivity between disease state utilities because healthy individuals may not fully comprehend the changes in QoL. Furthermore, the utility values for ‘stable disease with adverse events’ were based on a weighted average accounting for nine AEs. Therefore, the entire adverse event profiles were not represented, which may lead to over- or under-estimation of the state utility value. The utility value-related limitations, however, likely apply equally to both arms. As noted above, results from the sensitivity analyses indicate that this variability did not greatly affect the overall results.

In light of the uncertainty, there exists the question of whether it is worth conducting more research to narrow the parameters’ confidence intervals, thus providing a higher level of confidence in results. Value-of-information analyses can address the benefits and costs of gathering more data: however, as argued by Claxton, if a decision must be made immediately, a decision maker’s best choice is generally based on the estimated direction and magnitude of the current differential, even when the difference is not statistically significant based on conventional practiceCitation25. In principle, for physicians and payers to make more informed decisions regarding the use of everolimus and sunitinib, they would need to have improved efficacy data. As previously mentioned, it would be ideal to have a head-to-head trial comparing the therapies. Both parties would also benefit from access to more comparable safety data between the therapies; physicians would benefit from a more accurate view of patient safety, and payers would benefit from the more precise AE cost estimates. Similarly, both groups would require more precise utility scores in order to make better informed decisions. This information would help physicians better assess patients’ QoL while on treatment, and it would help payers determine more accurately the incremental cost per QALY. A value-of-information analysis would be a useful next step to compare the costs of conducting this trial, with the potential benefits.

The use of benchmarks or willingness-to-pay thresholds in healthcare economic analyses remains under debateCitation26–31. Some economists have proposed a threshold of $50,000/QALYCitation28; this benchmark assumes that economic efficiency from a societal perspective is the primary concern of decision-makers. However, multiple studies demonstrate that such a threshold may be inconsistent with societal preferences in the US; estimates of the willingness-to-pay threshold may approach or exceed $100,000/QALYCitation30,Citation31. Practicing clinicians have a different perspective. Nadler et al.Citation28 suggest that oncologists are willing to prescribe regimens that cost much more for the QALYs gained. Regarding bevacizumab use, for example, physicians indicated an average implied willingness-to-pay threshold of $320,000/QALY. Many economists would also argue that the average threshold should be higher. For example, Murphy and TopelCitation32 estimate the willingness-to-pay threshold peaks at $350,000 per life-year for a 50-year-old. Willingness-to-pay thresholds must be considered with the understanding that the extension of survival leads to a longer period during which patients incur healthcare expenditures. Therefore, it is important to consider the lifetime survival gain estimation of economic efficiency, as is done in this analysis.

The probabilistic sensitivity analysis results project that everolimus will be cost-effective compared to sunitinib under a range of acceptable willingness-to-pay thresholds. While further studies are warranted in broader populations, providers and decision-makers should take these results into consideration when treating patients with advanced, progressive pNET.

Conclusion

Although this analysis is limited due to its reliance on an indirect comparison of two phase 3 studies, everolimus is expected to be cost-effective relative to sunitinib in advanced, progressive pNET. Sensitivity analyses demonstrate that, overall, there is a trend that everolimus is cost-effective compared to sunitinib in patients with advanced, progressive pNET.

Transparency

Declaration of funding

Novartis Pharmaceuticals Corporation provided funding for this study. The sponsored research did not put limits on freedom to publish or the content of the publication.

Declaration of financial/other relationships

Roman Casciano, Maruit Chulikavit, and Allison Perrin are employees of The LA-SER Group, a consultancy that received compensation for the overall economic study design, the analysis, and preparation of this manuscript. Zhimei Liu and Xufang Wang are employees of and own stock in Novartis Pharmaceuticals Corporation. They also contributed to the analysis and manuscript preparation. Louis P. Garrison is a consultant for Novartis Pharmaceuticals Corporation and has received compensation for his contributions to analysis and the preparation of the manuscript.

Acknowledgments

No assistance in the preparation of this article to be declared. This study was presented in part ( and , , and ) at the American Society of Clinical Oncology, 2012 Gastrointestinal Cancers Symposium; Science and Multidisciplinary Management of GI Malignancies; January 19–21, San Francisco, CA. The poster was titled, Cost-Effectiveness of Treating Patients with Advanced Progressive Pancreatic Neuroendocrine Tumors with Everolimus Versus Sunitinib in the United States.

References

  • Ong SL, Garcea G, Pollard CA, et al. A fuller understanding of pancreatic neuroendocrine tumours combined with aggressive management improves outcome. Pancreatology 2009;9:583-600
  • Yao JC, Hassan M, Phan A, et al. One hundred years after "carcinoid": epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008;26:3063-72
  • Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011;364(6):514-23
  • Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011;364:501-13
  • US Food and Drug Administration. FDA Briefing Document: Oncology Drug Advisory Committee Meeting for Sutent (suntinib malate). United States Food and Drug Administration, Silver Spring, MD 2011
  • Signorovitch J, Swallow E, Kantor E, et al. Overall survival with everolimus, sunitinib, and placebo for advanced pancreatic neuroendocrine tumors: a matching-adjusted indirect comparison of randomized trials. J Clin Oncol 2012;30(4 [February 1 Supplement]):abstr 237
  • Signorovitch JE, Wu EQ, Yu AP, et al. Comparative effectiveness without head-to-head trials: a method for matching-adjusted indirect comparisons applied to psoriasis treatment with adalimumab or etanercept. Pharmacoeconomics 2010;28:935-45
  • Glasziou PP, Simes RJ, Gelber RD. Quality adjusted survival analysis. Stat Medicine 1990;9:1259-76
  • Glasziou PP, Cole BF, Gelber RD, et al. Quality adjusted survival analysis with repeated quality of life measures. Stat Medicine 1998;17:1215-29
  • Carroll KJ. On the use and utility of the Weibull model in the analysis of survival data. Control Clin Trials 2003;24:682-701
  • Briggs A, Claxton K, Sculpher M. Further developments in decision analytic models for economic evaluation. Decision modelling for health economic evaluation. New York: Oxford Univresity Press Inc., 2006. p 45-76
  • Thompson Coon J, Hoyle M, Green C, et al. Bevacizumab, sorafenib tosylate, sunitinib and temsirolimus for renal cell carcinoma: a systematic review and economic evaluation. Health Technol Assess 2010;14:1-184, iii–iv
  • Wojcik M, McComas J, Sachs C, et al. A randomized double-blind phase III study of RAD001 10 mg/d plus best supportive care versus placebo plus best supportive care in the treatment of patients with advanced pancreatic neuroendocrine tumor (NET): Clinical Study Report [Data on file]. Novartis, 2010
  • Casciano R, Chulikavit M, Parikh R, et al. Resource utilization among neuroendocrine tumor patients in the United States. J Clin Oncol 2012;30(4 [February 1 Supplement]):abstr 251
  • RED BOOK™ for Windows® [database on CD-ROM]. Annual 2011 Edition. Greenwood Village, Colo: Thomson Reuters (Healthcare) Inc.
  • Physicians’ Fee and Coding Guide 2011. MAG Mutual Healthcare Solutions, Inc, Atlanta, GA 2011
  • Thomson Reuters. MS-DRG Handbook 2010. Comparative clinical and financial benchmarks. Thomson Reuters, Ann Arbor, MI.
  • Healthcare Cost & Utilization Project (HCUP). Agency for Healthcare Research and Quality 2009. http://www.ahrq.gov/data/hcup/. Accessed May/June, 2011
  • Mickisch G, Gore M, Escudier B, et al. Costs of managing adverse events in the treatment of first-line metastatic renal cell carcinoma: bevacizumab in combination with interferon-alpha2a compared with sunitinib. Br J Cancer 2010;102:80-6
  • Weiner M, Ross S, Mathew J, et al. Estimating the costs of chemotherapy-associated adverse event clusters. Health Serv Outcomes Res Method 2007;7:1-21
  • Yu AP, Wu EQ, Birnbaum HG, et al. Short-term economic impact of body weight change among patients with type 2 diabetes treated with antidiabetic agents: analysis using claims, laboratory, and medical record data. Curr Med Res Opin 2007;23:2157-2169
  • Zhang B, Wright AA, Nilsson ME, et al. Associations between advanced cancer patients' end-of-life conversations and cost experiences in the final week of life. J Clin Oncol 2008;26(15 [May 20 Supplement]):abstr 9530
  • Swinburn P, Lloyd A. Neuroendocrine Tumours (NET) Utility Values. 2011. [Data on file at Novartis].
  • The AMCP format for formulary submissions: version 3.0. Academy of Managed Care Pharmacy, Alexandria, VA 2009
  • Claxton K. The irrelevance of inference: a decision-making approach to the stochastic evaluation of health care technologies. J Health Econ 1999;18:341-64
  • Rawlins M, Culyer A. National Institute for Clinical Excellence and its value judgments. BMJ 2004;329:224-7
  • Eichler HG, Kong SX, Gerth WC, et al. Use of cost-effectiveness analysis in health-care resource allocation decision-making: how are cost-effectiveness thresholds expected to emerge? Value Health 2004;7:518-28
  • Nadler E, Eckert B, Neumann PJ. Do oncologists believe new cancer drugs offer good value? Oncologist 2006;11:90-5
  • Remák E, Charbonneau C, Négrier S, et al. Economic evaluation of sunitinib malate for the first-line treatment of metastatic renal cell carcinoma. J Clin Oncol 2008;26:3995-4000
  • Braithwaite RS, Meltzer DO, King JT Jr, et al. What does the value of modern medicine say about the $50,000 per quality-adjusted life-year decision rule? Med Care 2008;46:349-56
  • Hirth RA, Chernew ME, Miller E, et al. Willingness to pay for a quality-adjusted life year: in search of a standard. Med Decis Making 2000;20:332-42
  • Murphy K, Topel R. The value of health and longevity. J Pol Econ 2006;114:871-903
  • Briggs A, Claxton K, Sculpher M. Making decision models probabilistic. Decision modelling for health economic evaluation. New York: Oxford University Press Inc., 2006. p 77-120

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.