[Skip to Navigation]
Sign In
Figure 1.  Hierarchy of Evidence for Surrogate End Point and Overall Survival Association
Hierarchy of Evidence for Surrogate End Point and Overall Survival Association
Figure 2.  Distribution of Data Sources Used in the Evaluated Surrogacy End Point Trials
Distribution of Data Sources Used in the Evaluated Surrogacy End Point Trials
Figure 3.  Correlations by Treatment Setting
Correlations by Treatment Setting

We scored strength of trial-level correlation according to a modification to surrogate criteria proposed by the Institute of Quality and Efficiency in Health Care34: low correlation (r ≤ 0.7), medium strength correlation (r > 0.7 to r < 0.85), and high correlation (r ≥ 0.85).

Table 1.  Summary of Trial-Level Surrogate Meta-analyses Testing the Validity of Surrogate End Points in Oncology
Summary of Trial-Level Surrogate Meta-analyses Testing the Validity of Surrogate End Points in Oncology
Table 2.  Included Meta-analyses of Both Published and Unpublished Reports
Included Meta-analyses of Both Published and Unpublished Reports
1.
Svensson  S, Menkes  DB, Lexchin  J.  Surrogate outcomes in clinical trials: a cautionary tale.  JAMA Intern Med. 2013;173(8):611-612.PubMedGoogle ScholarCrossref
2.
Prasad  V, Vandross  A.  Cardiovascular primary prevention: how high should we set the bar?  Arch Intern Med. 2012;172(8):656-659.PubMedGoogle ScholarCrossref
3.
Fleming  TR, DeMets  DL.  Surrogate end points in clinical trials: are we being misled?  Ann Intern Med. 1996;125(7):605-613.PubMedGoogle ScholarCrossref
4.
Messerli  FH, Bangalore  S.  ALTITUDE trial and dual RAS blockade: the alluring but soft science of the surrogate end point.  Am J Med. 2013;126(3):e1-e3.PubMedGoogle ScholarCrossref
5.
Yudkin  JS, Lipska  KJ, Montori  VM.  The idolatry of the surrogate.  BMJ. 2011;343:d7995.PubMedGoogle ScholarCrossref
6.
Ciani  O, Buyse  M, Garside  R,  et al.  Comparison of treatment effect sizes associated with surrogate and final patient relevant outcomes in randomised controlled trials: meta-epidemiological study.http://www.bmj.com/content/346/bmj.f457. Accessed May 12, 2015.
7.
Shi  Q, Sargent  DJ.  Meta-analysis for the evaluation of surrogate endpoints in cancer clinical trials.  Int J Clin Oncol. 2009;14(2):102-111.PubMedGoogle ScholarCrossref
8.
Fauber  JCE.  FDA approves cancer drugs without proof they're extending lives.http://www.jsonline.com/watchdog/watchdogreports/fda-approves-cancer-drugs-without-proof-theyre-extending-lives-b99348000z1-280437692.html. Accessed December 14, 2014.
9.
Korde  N, Zingone  A, Kwok  M,  et al Phase II clinical and correlative study of carfilzomib, lenalidomide, and dexamethasone (CRd) in newly diagnosed multiple myeloma (MM) patients.  J Clin Oncol. 2012(suppl);abstr e18568.Google Scholar
10.
Jakubowiak  AJ, Dytfeld  D, Griffith  KA,  et al.  A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma.  Blood. 2012;120(9):1801-1809.PubMedGoogle ScholarCrossref
11.
National Comprehensive Cancer Network.  Myeloma NgM [registration required].http://www.nccn.org/professionals/physician_gls/pdf/myeloma.pdf. Accessed May 12, 2015.
12.
Tillman  K, Burton  B, Jacques  LB, Phurrough  SE.  Compendia and anticancer therapy under Medicare.  Ann Intern Med. 2009;150(5):348-350.PubMedGoogle ScholarCrossref
13.
McGivney  WT.  Medicare recognizes NCCN compendium.  J Natl Compr Canc Netw. 2008;6(7):635.Google Scholar
14.
McGivney  WT.  NCCN guidelines and their impact on coverage policy.  J Natl Compr Canc Netw. 2010;8(6):625.Google Scholar
15.
Miller  K, Wang  M, Gralow  J,  et al.  Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer.  N Engl J Med. 2007;357(26):2666-2676.PubMedGoogle ScholarCrossref
16.
Carpenter  D, Kesselheim  AS, Joffe  S.  Reputation and precedent in the bevacizumab decision.  N Engl J Med. 2011;365(2):e3.PubMedGoogle ScholarCrossref
17.
Lassere  MN, Johnson  KR, Schiff  M, Rees  D.  Is blood pressure reduction a valid surrogate endpoint for stroke prevention? an analysis incorporating a systematic review of randomised controlled trials, a by-trial weighted errors-in-variables regression, the surrogate threshold effect (STE) and the Biomarker-Surrogacy (BioSurrogate) Evaluation Schema (BSES).  BMC Med Res Methodol. 2012;12(1):27.PubMedGoogle ScholarCrossref
18.
Torri  V, Simon  R, Russek-Cohen  E, Midthune  D, Friedman  M.  Statistical model to determine the relationship of response and survival in patients with advanced ovarian cancer treated with chemotherapy.  J Natl Cancer Inst. 1992;84(6):407-414.PubMedGoogle ScholarCrossref
19.
Freedman  LS, Graubard  BI, Schatzkin  A.  Statistical validation of intermediate endpoints for chronic diseases.  Stat Med. 1992;11(2):167-178.PubMedGoogle ScholarCrossref
20.
Buyse  M, Molenberghs  G, Burzykowski  T, Renard  D, Geys  H.  The validation of surrogate endpoints in meta-analyses of randomized experiments.  Biostatistics. 2000;1(1):49-67.PubMedGoogle ScholarCrossref
21.
Weir  CJ, Walley  RJ.  Statistical evaluation of biomarkers as surrogate endpoints: a literature review.  Stat Med. 2006;25(2):183-203.PubMedGoogle ScholarCrossref
22.
Elston  J, Taylor  RS.  Use of surrogate outcomes in cost-effectiveness models: a review of United Kingdom health technology assessment reports.  Int J Technol Assess Health Care. 2009;25(1):6-13.PubMedGoogle ScholarCrossref
23.
Buyse  M, Burzykowski  T, Michiels  S, Carroll  K.  Individual- and trial-level surrogacy in colorectal cancer.  Stat Methods Med Res. 2008;17(5):467-475.PubMedGoogle ScholarCrossref
24.
Prentice  RL.  Surrogate endpoints in clinical trials: definition and operational criteria.  Stat Med. 1989;8(4):431-440.PubMedGoogle ScholarCrossref
25.
Sargent  DJ, Wieand  HS, Haller  DG,  et al.  Disease-free survival versus overall survival as a primary end point for adjuvant colon cancer studies: individual patient data from 20,898 patients on 18 randomized trials.  J Clin Oncol. 2005;23(34):8664-8670.PubMedGoogle ScholarCrossref
26.
Pratt  CM, Moyé  LA.  The cardiac arrhythmia suppression trial: casting suppression in a different light.  Circulation. 1995;91(1):245-247.PubMedGoogle ScholarCrossref
27.
Echt  DS, Liebson  PR, Mitchell  LB,  et al.  Mortality and morbidity in patients receiving encainide, flecainide, or placebo: the Cardiac Arrhythmia Suppression Trial.  N Engl J Med. 1991;324(12):781-788.PubMedGoogle ScholarCrossref
28.
Laporte  S, Squifflet  P, Baroux  N,  et al.  Prediction of survival benefits from progression-free survival benefits in advanced non-small-cell lung cancer: evidence from a meta-analysis of 2334 patients from 5 randomised trials.  BMJ Open. 2013;3(3):e001802.PubMedGoogle ScholarCrossref
29.
Burzykowski  T, Buyse  M.  Surrogate threshold effect: an alternative measure for meta-analytic surrogate endpoint validation.  Pharm Stat. 2006;5(3):173-186.PubMedGoogle ScholarCrossref
30.
Buyse  M.  Use of meta-analysis for the validation of surrogate endpoints and biomarkers in cancer trials.  Cancer J. 2009;15(5):421-425.PubMedGoogle ScholarCrossref
31.
Sherrill  B, Kaye  JA, Sandin  R, Cappelleri  JC, Chen  C.  Review of meta-analyses evaluating surrogate endpoints for overall survival in oncology.  Onco Targets Ther. 2012;5:287-296.PubMedGoogle ScholarCrossref
32.
Davis  S, Tappenden  P, Cantrell  A.  A review of studies examining the relationship between progression-free survival and overall survival in advanced or metastatic cancer.http://www.nicedsu.org.uk/PFSOS%20Report.FINAL.06.08.12.pdf. Accessed May 12, 2015.
33.
Ciani  O, Davis  S, Tappenden  P,  et al.  Validation of surrogate endpoints in advanced solid tumors: systematic review of statistical methods, results, and implications for policy makers.  Int J Technol Assess Health Care. 2014;30(3):312-324.PubMedGoogle ScholarCrossref
34.
Institute for Quality and Efficiency in Health Care [Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen] (IQWiG).  Validity of surrogate endpoints in oncology: executive summary.http://www.iqwig.de/download/A10-05_Executive_Summary_v1-1_Surrogate_endpoints_in_oncology.pdf. Accessed December 20, 2014.
35.
Cortazar  P, Zhang  L, Untch  M,  et al.  Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis.  Lancet. 2014;384(9938):164-172.PubMedGoogle ScholarCrossref
36.
Berruti  A, Amoroso  V, Gallo  F,  et al.  Pathologic complete response as a potential surrogate for the clinical outcome in patients with breast cancer after neoadjuvant therapy: a meta-regression of 29 randomized prospective studies.  J Clin Oncol. 2014;32(34):3883-3891.PubMedGoogle ScholarCrossref
37.
Ng  R, Pond  GR, Tang  PA, MacIntosh  PW, Siu  LL, Chen  EX.  Correlation of changes between 2-year disease-free survival and 5-year overall survival in adjuvant breast cancer trials from 1966 to 2006.  Ann Oncol. 2008;19(3):481-486.PubMedGoogle ScholarCrossref
38.
Burzykowski  T, Buyse  M, Yothers  G, Sakamoto  J, Sargent  D.  Exploring and validating surrogate endpoints in colorectal cancer.  Lifetime Data Anal. 2008;14(1):54-64.PubMedGoogle ScholarCrossref
39.
Oba  K, Paoletti  X, Alberts  S,  et al; GASTRIC group.  Disease-free survival as a surrogate for overall survival in adjuvant trials of gastric cancer: a meta-analysis.  J Natl Cancer Inst. 2013;105(21):1600-1607.PubMedGoogle ScholarCrossref
40.
Michiels  S, Le Maître  A, Buyse  M,  et al; MARCH and MACH-NC Collaborative Groups.  Surrogate endpoints for overall survival in locally advanced head and neck cancer: meta-analyses of individual patient data.  Lancet Oncol. 2009;10(4):341-350.PubMedGoogle ScholarCrossref
41.
Mauguen  A, Pignon  JP, Burdett  S,  et al; Surrogate Lung Project Collaborative Group.  Surrogate endpoints for overall survival in chemotherapy and radiotherapy trials in operable and locally advanced lung cancer: a re-analysis of meta-analyses of individual patients’ data.  Lancet Oncol. 2013;14(7):619-626.PubMedGoogle ScholarCrossref
42.
Bruzzi  P, Del Mastro  L, Sormani  MP,  et al.  Objective response to chemotherapy as a potential surrogate end point of survival in metastatic breast cancer patients.  J Clin Oncol. 2005;23(22):5117-5125.PubMedGoogle ScholarCrossref
43.
Hackshaw  A, Knight  A, Barrett-Lee  P, Leonard  R.  Surrogate markers and survival in women receiving first-line combination anthracycline chemotherapy for advanced breast cancer.  Br J Cancer. 2005;93(11):1215-1221.PubMedGoogle ScholarCrossref
44.
Burzykowski  T, Buyse  M, Piccart-Gebhart  MJ,  et al.  Evaluation of tumor response, disease control, progression-free survival, and time to progression as potential surrogate end points in metastatic breast cancer.  J Clin Oncol. 2008;26(12):1987-1992.PubMedGoogle ScholarCrossref
45.
Miksad  RA, Zietemann  V, Gothe  R,  et al.  Progression-free survival as a surrogate endpoint in advanced breast cancer.  Int J Technol Assess Health Care. 2008;24(4):371-383.PubMedGoogle ScholarCrossref
46.
Sherrill  B, Amonkar  M, Wu  Y,  et al.  Relationship between effects on time-to-disease progression and overall survival in studies of metastatic breast cancer.  Br J Cancer. 2008;99(10):1572-1578.PubMedGoogle ScholarCrossref
47.
Wilkerson  J, Fojo  T.  Progression-free survival is simply a measure of a drug’s effect while administered and is not a surrogate for overall survival.  Cancer J. 2009;15(5):379-385.PubMedGoogle ScholarCrossref
48.
Beauchemin  C, Cooper  D, Lapierre  ME, Yelle  L, Lachaine  J.  Progression-free survival as a potential surrogate for overall survival in metastatic breast cancer.  Onco Targets Ther. 2014;7:1101-1110.PubMedGoogle ScholarCrossref
49.
Petrelli  F, Barni  S.  Surrogate endpoints in metastatic breast cancer treated with targeted therapies: an analysis of the first-line phase III trials.  Med Oncol. 2014;31(1):776.PubMedGoogle ScholarCrossref
50.
Johnson  KR, Ringland  C, Stokes  BJ,  et al.  Response rate or time to progression as predictors of survival in trials of metastatic colorectal cancer or non-small-cell lung cancer: a meta-analysis.  Lancet Oncol. 2006;7(9):741-746.PubMedGoogle ScholarCrossref
51.
Chirila  C, Odom  D, Devercelli  G,  et al.  Meta-analysis of the association between progression-free survival and overall survival in metastatic colorectal cancer.  Int J Colorectal Dis. 2012;27(5):623-634.PubMedGoogle ScholarCrossref
52.
Sidhu  R, Rong  A, Dahlberg  S.  Evaluation of progression-free survival as a surrogate endpoint for survival in chemotherapy and targeted agent metastatic colorectal cancer trials.  Clin Cancer Res. 2013;19(5):969-976.PubMedGoogle ScholarCrossref
53.
Buyse  M, Thirion  P, Carlson  RW, Burzykowski  T, Molenberghs  G, Piedbois  P; Meta-Analysis Group in Cancer.  Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis.  Lancet. 2000;356(9227):373-378.PubMedGoogle ScholarCrossref
54.
Tang  PA, Bentzen  SM, Chen  EX, Siu  LL.  Surrogate end points for median overall survival in metastatic colorectal cancer: literature-based analysis from 39 randomized controlled trials of first-line chemotherapy.  J Clin Oncol. 2007;25(29):4562-4568.PubMedGoogle ScholarCrossref
55.
Giessen  C, Laubender  RP, Ankerst  DP,  et al.  Progression-free survival as a surrogate endpoint for median overall survival in metastatic colorectal cancer: literature-based analysis from 50 randomized first-line trials.  Clin Cancer Res. 2013;19(1):225-235.PubMedGoogle ScholarCrossref
56.
Shi  Q, de Gramont  A, Grothey  A,  et al.  Individual patient data analysis of progression-free survival versus overall survival as a first-line end point for metastatic colorectal cancer in modern randomized trials: findings from the analysis and research in cancers of the digestive system database.  J Clin Oncol. 2015;33(1):22-28.PubMedGoogle ScholarCrossref
57.
Giessen  C, Laubender  RP, Ankerst  DP,  et al.  Surrogate end points in second-line treatment for mCRC: a systematic literature-based analysis from 23 randomised trials.  Acta Oncol. 2015;54(2):187-193.PubMedGoogle ScholarCrossref
58.
Buyse  M, Burzykowski  T, Carroll  K,  et al.  Progression-free survival is a surrogate for survival in advanced colorectal cancer.  J Clin Oncol. 2007;25(33):5218-5224.PubMedGoogle ScholarCrossref
59.
Amir  E, Seruga  B, Kwong  R, Tannock  IF, Ocaña  A.  Poor correlation between progression-free and overall survival in modern clinical trials: are composite endpoints the answer?  Eur J Cancer. 2012;48(3):385-388.PubMedGoogle ScholarCrossref
60.
Paoletti  X, Oba  K, Bang  YJ,  et al; GASTRIC group.  Progression-free survival as a surrogate for overall survival in advanced/recurrent gastric cancer trials: a meta-analysis.  J Natl Cancer Inst. 2013;105(21):1667-1670.PubMedGoogle ScholarCrossref
61.
Shitara  K, Matsuo  K, Muro  K, Doi  T, Ohtsu  A.  Correlation between overall survival and other endpoints in clinical trials of second-line chemotherapy for patients with advanced gastric cancer.  Gastric Cancer. 2014;17(2):362-370.PubMedGoogle ScholarCrossref
62.
Flaherty  KT, Hennig  M, Lee  SJ,  et al.  Surrogate endpoints for overall survival in metastatic melanoma: a meta-analysis of randomised controlled trials.  Lancet Oncol. 2014;15(3):297-304.PubMedGoogle ScholarCrossref
63.
Lee  L, Wang  L, Crump  M.  Identification of potential surrogate end points in randomized clinical trials of aggressive and indolent non-Hodgkin’s lymphoma: correlation of complete response, time-to-event and overall survival end points.  Ann Oncol. 2011;22(6):1392-1403.PubMedGoogle ScholarCrossref
64.
Hayashi  H, Okamoto  I, Taguri  M, Morita  S, Nakagawa  K.  Postprogression survival in patients with advanced non-small-cell lung cancer who receive second-line or third-line chemotherapy.  Clin Lung Cancer. 2013;14(3):261-266.PubMedGoogle ScholarCrossref
65.
Delea  TE, Khuu  A, Heng  DY, Haas  T, Soulières  D.  Association between treatment effects on disease progression end points and overall survival in clinical studies of patients with metastatic renal cell carcinoma.  Br J Cancer. 2012;107(7):1059-1068.PubMedGoogle ScholarCrossref
66.
Johnson  KR, Liauw  W, Lassere  MN.  Evaluating surrogacy metrics and investigating approval decisions of progression-free survival (PFS) in metastatic renal cell cancer: a systematic review.  Ann Oncol. 2015;26(3):485-496.PubMedGoogle ScholarCrossref
67.
Foster  NR, Qi  Y, Shi  Q,  et al.  Tumor response and progression-free survival as potential surrogate endpoints for overall survival in extensive stage small-cell lung cancer: findings on the basis of North Central Cancer Treatment Group trials.  Cancer. 2011;117(6):1262-1271.PubMedGoogle ScholarCrossref
68.
Hotta  K, Fujiwara  Y, Matsuo  K,  et al.  Time to progression as a surrogate marker for overall survival in patients with advanced non-small cell lung cancer.  J Thorac Oncol. 2009;4(3):311-317.PubMedGoogle ScholarCrossref
69.
Non-small Cell Lung Cancer Collaborative Group.  Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials.  BMJ. 1995;311(7010):899-909.PubMedGoogle ScholarCrossref
70.
Aupérin  A, Le Péchoux  C, Rolland  E,  et al.  Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer.  J Clin Oncol. 2010;28(13):2181-2190.PubMedGoogle ScholarCrossref
71.
Arriagada  R, Auperin  A, Burdett  S,  et al; NSCLC Meta-analyses Collaborative Group.  Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non-small-cell lung cancer: two meta-analyses of individual patient data.  Lancet. 2010;375(9722):1267-1277.PubMedGoogle ScholarCrossref
72.
NSCLC Meta-Analyses Collaborative Group.  Chemotherapy in addition to supportive care improves survival in advanced non-small-cell lung cancer: a systematic review and meta-analysis of individual patient data from 16 randomized controlled trials.  J Clin Oncol. 2008;26(28):4617-4625.PubMedGoogle ScholarCrossref
73.
Mauguen  A, Le Péchoux  C, Saunders  MI,  et al.  Hyperfractionated or accelerated radiotherapy in lung cancer: an individual patient data meta-analysis.  J Clin Oncol. 2012;30(22):2788-2797.PubMedGoogle ScholarCrossref
74.
Paoletti  X, Oba  K, Burzykowski  T,  et al; GASTRIC (Global Advanced/Adjuvant Stomach Tumor Research International Collaboration) Group.  Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis.  JAMA. 2010;303(17):1729-1737.PubMedGoogle ScholarCrossref
75.
Buyse  M, Sargent  DJ, Grothey  A, Matheson  A, de Gramont  A.  Biomarkers and surrogate end points: the challenge of statistical validation.  Nat Rev Clin Oncol. 2010;7(6):309-317.PubMedGoogle ScholarCrossref
76.
Booth  CM, Eisenhauer  EA.  Progression-free survival: meaningful or simply measurable?  J Clin Oncol. 2012;30(10):1030-1033.PubMedGoogle ScholarCrossref
77.
Booth  CM, Cescon  DW, Wang  L, Tannock  IF, Krzyzanowska  MK.  Evolution of the randomized controlled trial in oncology over three decades.  J Clin Oncol. 2008;26(33):5458-5464.PubMedGoogle ScholarCrossref
78.
Kay  A, Higgins  J, Day  AG, Meyer  RM, Booth  CM.  Randomized controlled trials in the era of molecular oncology: methodology, biomarkers, and end points.  Ann Oncol. 2012;23(6):1646-1651.PubMedGoogle ScholarCrossref
79.
Prasad  V, Cifu  A, Ioannidis  JP.  Reversals of established medical practices: evidence to abandon ship.  JAMA. 2012;307(1):37-38.PubMedGoogle ScholarCrossref
80.
Prasad  V, Vandross  A, Toomey  C,  et al.  A decade of reversal: an analysis of 146 contradicted medical practices.  Mayo Clin Proc. 2013;88(8):790-798.PubMedGoogle ScholarCrossref
81.
US Food and Drug Administration.  FDA approves Afinitor for advanced breast cancer. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm312965.htm. Accessed January 4, 2014.
82.
Piccart  M, Hortobagyi  GN, Campone  M,  et al.  Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2.  Ann Oncol. 2014;25(12):2357-2362.PubMedGoogle ScholarCrossref
83.
Finn  RS, Crown  JP, Lang  I,  et al.  The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study.  Lancet Oncol. 2015;16(1):25-35.PubMedGoogle ScholarCrossref
84.
Ning  YM, He  K, Dagher  R,  et al.  Liposomal doxorubicin in combination with bortezomib for relapsed or refractory multiple myeloma.  Oncology (Williston Park). 2007;21(12):1503-1508.PubMedGoogle Scholar
85.
Orlowski  RZ, Nagler  A, Sonneveld  P,  et al.  Final overall survival results of a randomized trial comparing bortezomib plus pegylated liposomal doxorubicin with bortezomib alone in subjects with relapsed or refractory multiple myeloma.  Blood. 2014;124(21):3448.Google Scholar
86.
Ioannidis  JP.  Effect of the statistical significance of results on the time to completion and publication of randomized efficacy trials.  JAMA. 1998;279(4):281-286.PubMedGoogle ScholarCrossref
87.
Krzyzanowska  MK, Pintilie  M, Tannock  IF.  Factors associated with failure to publish large randomized trials presented at an oncology meeting.  JAMA. 2003;290(4):495-501.PubMedGoogle ScholarCrossref
88.
Misakian  AL, Bero  LA.  Publication bias and research on passive smoking: comparison of published and unpublished studies.  JAMA. 1998;280(3):250-253.PubMedGoogle ScholarCrossref
89.
Dickersin  K, Min  YI, Meinert  CL.  Factors influencing publication of research results. Follow-up of applications submitted to two institutional review boards.  JAMA. 1992;267(3):374-378.PubMedGoogle ScholarCrossref
90.
Dwan  K, Altman  DG, Arnaiz  JA,  et al.  Systematic review of the empirical evidence of study publication bias and outcome reporting bias.  PLoS One. 2008;3(8):e3081.PubMedGoogle ScholarCrossref
91.
Rising  K, Bacchetti  P, Bero  L.  Reporting bias in drug trials submitted to the Food and Drug Administration: review of publication and presentation.  PLoS Med. 2008;5(11):e217.PubMedGoogle ScholarCrossref
92.
Jefferson  T, Jones  M, Doshi  P, Spencer  EA, Onakpoya  I, Heneghan  CJ.  Oseltamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments.  BMJ. 2014;348:g2545.PubMedGoogle ScholarCrossref
93.
Kirsch  I, Deacon  BJ, Huedo-Medina  TB, Scoboria  A, Moore  TJ, Johnson  BT.  Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration.  PLoS Med. 2008;5(2):e45.PubMedGoogle ScholarCrossref
94.
Chapman  PB, Hauschild  A, Robert  C,  et al; BRIM-3 Study Group.  Improved survival with vemurafenib in melanoma with BRAF V600E mutation.  N Engl J Med. 2011;364(26):2507-2516.PubMedGoogle ScholarCrossref
95.
Raymond  E, Dahan  L, Raoul  J-L,  et al.  Sunitinib malate for the treatment of pancreatic neuroendocrine tumors.  N Engl J Med. 2011;364(6):501-513.PubMedGoogle ScholarCrossref
96.
Kantoff  PW, Higano  CS, Shore  ND,  et al; IMPACT Study Investigators.  Sipuleucel-T immunotherapy for castration-resistant prostate cancer.  N Engl J Med. 2010;363(5):411-422.PubMedGoogle ScholarCrossref
97.
Baselga  J, Campone  M, Piccart  M,  et al.  Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer.  N Engl J Med. 2012;366(6):520-529.PubMedGoogle ScholarCrossref
98.
Burstein  HJ.  Bevacizumab for advanced breast cancer: all tied up with a RIBBON?  J Clin Oncol. 2011;29(10):1232-1235.PubMedGoogle ScholarCrossref
99.
Prasad  V.  Double-crossed: why crossover in clinical trials may be distorting medical science.  J Natl Compr Canc Netw. 2013;11(5):625-627.PubMedGoogle Scholar
100.
Prasad  V, Grady  C.  The misguided ethics of crossover trials.  Contemp Clin Trials. 2014;37(2):167-169.PubMedGoogle ScholarCrossref
101.
Lakdawalla  DN, Chou  JW, Linthicum  MT, MacEwan  JP, Zhang  J, Goldman  DP.  Evaluating expected costs and benefits of granting access to new treatments on the basis of progression-free survival in non–small-cell lung cancer [published online March 19, 2015].  JAMA Oncol. doi:10.1001/jamaoncol.2015.0203.Google Scholar
102.
Fojo  T, Mailankody  S, Lo  A.  Unintended consequences of expensive cancer therapeutics—the pursuit of marginal indications and a me-too mentality that stifles innovation and creativity: the John Conley Lecture.  JAMA Otolaryngol Head Neck Surg. 2014;140(12):1225-1236.PubMedGoogle ScholarCrossref
Original Investigation
Health Care Reform
August 2015

The Strength of Association Between Surrogate End Points and Survival in Oncology: A Systematic Review of Trial-Level Meta-analyses

Author Affiliations
  • 1Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
  • 2Division of Medical Oncology, University of California, Los Angeles
JAMA Intern Med. 2015;175(8):1389-1398. doi:10.1001/jamainternmed.2015.2829
Abstract

Importance  The strength of association between surrogate end points and survival in oncology is important to understand because surrogate end points are frequently used in oncology clinical trials, supporting US Food and Drug Administration approvals and National Comprehensive Cancer Network guideline recommendations.

Objective  To identify and evaluate trial-level meta-analyses of randomized clinical trials quantifying the association between a surrogate end point and overall survival in medical oncology. Trial-level correlations test whether treatments that improve the surrogate end point also improve the final end point and are widely considered the strongest evidence to validate a surrogate end point.

Evidence Review  Our literature search was built on earlier reported data sets and updated with Google Scholar and MEDLINE searches conducted on December 26, 2014. For MEDLINE, search terms included (“regression” or “correlation”) and “surrogate” and “end point [or endpoint]” and (“oncology” or “cancer”). For Google scholar, search terms included (“regression” or “correlation”) and “surrogate end point [or endpoint]” and “overall survival” and “trial level.” A total of 108 abstracts were retrieved, and 62 articles were read in full in addition to articles identified through prior reviews.

Findings  We found 36 articles in which 65 specific correlations between a surrogate end point and survival were identified. Surrogate end points were studied in the neoadjuvant, adjuvant, locally advanced, and metastatic settings. The most common sources for trials included in the 36 articles were systematic reviews of the published literature (10 of 36; 28%), and published literature and meeting abstracts (14 of 36; 39%). Four meta-analyses (11%) used a convenience sample, and only 5 studies (14%) attempted to include unpublished trials by surveying clinical trial registries. Among these 5 studies, only 352 of 684 eligible trials (51.1%) were included in the analyses. More than half of reported correlations (34 of 65; 52%) were of low strength (r ≤ 0.7). Approximately a quarter (16 of 65; 25%) were of medium strength (r > 0.7 to r < 0.85), and 15 of 65 (23%) were highly correlated (r ≥ 0.85) with survival.

Conclusions and Relevance  Most trial-level validation studies of surrogate end points in oncology find low correlations with survival. All validation studies use only a subset of available trials. The evidence supporting the use of surrogate end points in oncology is limited.

Introduction

Although there is growing recognition that adopting new medical practices based on improvements in surrogate outcomes can lead to misleading conclusions,1-6 surrogate end points continue to play a prominent role in oncology.7 In the United States, most new cancer drugs are approved based on surrogate measures,8 such as response rate (RR) and progression-free survival (PFS), through the US Food and Drug Administration’s (FDA) Accelerated Approval pathway. In addition, clinical practice guidelines often expand treatment recommendations for approved drugs based on studies assessing surrogates. For example, the drug carfilzomib (Kyprolis; Onxy Pharmaceuticals) received FDA approval in 2012 based on RR for relapsed and/or refractory multiple myeloma. In 2013, the National Comprehensive Cancer Network (NCCN) expanded the drug’s indication to include untreated myeloma—based on RR in 2 small, uncontrolled phase 1/2 studies9,10 (category 2A).11 Notably, NCCN endorsement obliges many commercial insurers and Medicare to reimburse the therapy.12-14 Thus, surrogate end points are used both to introduce new cancer drugs and to promote new uses for the drugs we already have.

The use of surrogate end points in oncology has led to use of toxic drugs that do not improve survival. For example, bevacizumab (Avastin; Roche/Genentech) gained FDA accelerated approval for metastatic breast cancer in 2008 based on evidence that it could improve PFS.15 In 2011, that approval was withdrawn when multiple randomized trials confirmed that bevacizumab did not improve overall survival and that gains in PFS were more modest than those seen in the earlier trial.16 Nevertheless, Medicare and other insurers are still obliged to pay for bevacizumab because it remains endorsed by the NCCN for this indication.14

For surrogates such as PFS or tumor RR to be reliable, it is important that they are validated. Several authors have described statistical methods to validate surrogates,17-21 but the framework that uses a hierarchy (Figure 1) is the most useful.22 In this model, level 3 is surrogacy based on biological plausibility alone. Level 2 occurs when a strong correlation exists between the surrogate and the final end point across cohorts or at the level of the individual patient. The highest level is trial-level surrogacy, meaning that across many trials treatments that improve the surrogate end point also improve the final outcome. Trial-level surrogacy directly asks the question that faces regulators and guideline writers: “If a drug improves a surrogate, will it also improve survival?” Others have noted that individual patient correlations do not directly validate surrogate measures.23-25

Establishing trial-level (level 1) surrogacy requires a meta-analysis of all randomized trials on a question of interest, with each trial serving as a unique data point. The x coordinate is typically the improvement in the surrogate when a drug is used, and the y coordinate is the improvement in the final outcome. Regression analysis is typically performed to assess what correlation exists between the change in the surrogate end point and the change in overall survival, across the randomized studies. The correlation coefficient (r) for the analysis, ranging from 0 to 1, provides a measure of the strength of the surrogate-survival correlation.

Two general medical examples can illustrate this approach. Consider the question of hypertension (the surrogate), blood pressure control (the intervention), and adverse cardiovascular end points (the final outcome). A level 2 analysis would show that groups of patients with higher average blood pressure have worse cardiovascular outcomes or that this correlation is observed at an individual patient level. A level 1 or trial-level analysis would show that across many trials, drugs that lowered blood pressure also decreased cardiovascular events. If a drug lowered blood pressure by 10%, and another lowered it by 20%, we would “validate” hypertension as a surrogate if there were fewer cardiovascular events for the drug with greater blood pressure reduction. Looking at many blood pressure trials, one could perform regression analysis and draw a general conclusion about the relationship between blood pressure reduction and adverse outcomes. In fact, this analysis has been performed.17

The case of premature ventricular contractions (PVCs) postmyocardial infarction illustrates the importance of level 1 analysis. Although studies showed a correlation between frequent PVCs and death26 (level 2), and certain anti-arrhythmic drugs reduce PVCs, it did not follow that anti-arrhythmic drugs (level 1) that lowered PVCs reduced mortality. In fact, a randomized clinical trial showed that these drugs actually increased mortality.27 Thus, only a level 1 analysis asks the right question: “Can we can trust a reduction in PVCs as a reliable surrogate for improved survival in pharmacologic trials?” Because oncology typically has dozens of trials examining both surrogate end points and survival in many clinical situations, performing level 1 analyses is more straightforward than in many other medical fields. For these reasons, we only examined level 1 studies.

We sought to examine all level 1 studies aiming to validate a surrogate in oncology. Specifically, we asked what percentage of these meta-analyses used data from published and unpublished studies. We also ascertained the median number of studies used to establish surrogacy in these studies and the reported correlation coefficients (r).

Methods
Eligible Studies

We identified meta-analyses of randomized clinical trials quantifying the association between a surrogate end point—RR, pathologic complete response, locoregional control, disease-free survival, event-free survival, time to progression, and/or PFS—and overall survival (OS) in medical oncology. As such, we excluded studies of radiation therapies, surgery, procedures, or supportive measures.

We sought only studies that examined trial-level correlations in meta-analyses of randomized clinical trials—ie, level 1 surrogate studies. We placed no restriction on the setting of medical therapy; thus, we include analyses in the neoadjuvant, adjuvant, locally advanced, and metastatic settings.

We excluded all analyses that did not examine trial-level (level 1) surrogacy. Specifically, we excluded analyses that only studied individual patient level correlations or those that did not examine the difference between investigational and control arms, considering each trial arm as a unique data point (level 2). We excluded conference abstracts, letters to the editor, and descriptive reviews. Finally, we excluded meta-analyses that used each treating center or nation (rather than each trial) as a unique data point. We were not convinced that such analyses accurately capture the question of our study because the individual data points in such analyses are no longer independent (comparisons are both within and across trial) and may spuriously inflate trial-level correlation. Nevertheless, including these studies20,28-30 would not have changed our results.

Literature Search

We constructed our data set by updating prior systematic reviews. An analysis by Sherril et al,31 conducted in October 2010, systematically identified all studies prior to that date that studied surrogate end points in oncology. Over 1000 articles were screened in that investigation, and all articles assessing the relationship between a surrogate end point and overall survival were listed. We then evaluated this list to ensure that the articles performed trial-level correlation—all other analyses were discarded. We also drew on a prior report commissioned for the National Institutes of Clinical Excellence, which identified meta-analyses of surrogate end points in malignant solid tumors.32,33 Again, only trial-level analyses were selected from this set.

We built on this data set, updating it for the last 4 years. Specifically, Google Scholar and MEDLINE were searched with the following search terms on December 26, 2014: For MEDLINE, search terms included (“regression” or “correlation”) and “surrogate” and “end point [or endpoint]” and (“oncology” or “cancer”). For Google scholar, search terms included (“regression” or “correlation”) and “surrogate end point [or endpoint]” and “overall survival” and “trial level.” A total of 108 abstracts were retrieved, and 62 articles were read in full.

End Points Assessed

Descriptive statistics were ascertained for the following end points: the percentage of meta-analyses that used data from published and unpublished studies, the number of individual randomized trials included in each surrogate meta-analyses, and the reported correlation coefficients. We considered convenience sample to mean a set of trials that the authors were able to obtain readily. We only credited authors with performing a search of the published literature, meeting abstracts, and/or unpublished trials if they performed a systematic search. We scored strength of trial-level correlation according to a modification to surrogate criteria proposed by the Institute of Quality and Efficiency in Health Care34: low correlation (r ≤ 0.7), medium strength correlation (r > 0.7 to r < 0.85), and high correlation (r ≥ 0.85). The specific cut points were adapted to function even when confidence intervals were not presented. Finally, for meta-analyses using published trials, abstracts, and registered studies, we investigated what percentage of eligible studies were ultimately included in analyses.

Results

We identified 36 articles20,23,25,30,35-68 that met our inclusion criteria and were considered trial-level or level 1 analyses. Claims of surrogacy were examined for 19 distinct clinical questions in the neoadjuvant, adjuvant, metastatic, and locally advanced settings (eTable in the Supplement).

The sources of randomized clinical trials included in the 36 articles most commonly were systematic reviews of the published literature (10 of 36; 28%) and published literature and meeting abstracts (14 of 36; 39%). Four meta-analyses (11%) used a convenience sample, and only 5 studies (14%) attempted to include unpublished trials by surveying clinical trial registries. Figure 2 summarizes search strategies.

Across the 36 studies, we noted 65 unique trial-level analyses. The median number of individual randomized clinical trials used in these analyses was 21, with as few as 9 and as many as 191 trials. For each analysis, we documented the study characteristics and the correlation coefficient (r) for all trial-level analyses (Table 1). There were 65 trial-level correlations scored according to criteria proposed by the Institute of Quality and Efficiency in Health Care (Figure 3).34 More than half of trial-level correlations (34 of 65; 52%) were of low strength (r ≤ 0.7). Approximately a quarter (16 of 65; 25%) were of medium strength (r > 0.7 to r < 0.85), and 15 of 65 (23%) were highly correlated (r ≥ 0.85) with survival.

We further examined the 5 studies39,41,48,66,68 that analyzed both published and unpublished trials. Although these articles searched the broadest possible collection of trials, they were unable to obtain data from all relevant studies. Table 2 lists the numbers of eligible and included trials for each of these meta-analyses. Notably, only 352 of 684 eligible studies (51.1%) were ultimately included. Reasons for omission included missing or lost data,41,74 missing information on progression,68 or missing median PFS or median OS reports.48,66

We also examined the 15 correlations of high strength. Six occurred in the adjuvant setting,23,25,38-41 6 in the metastatic setting,41,47,52,55,62,67 and 3 in the locally advanced setting.40,41 The median number of individual randomized trials included in these analyses was 14. Notably, 3 high correlations occurred in the same setting—adjuvant colorectal cancer—and 4 others occurred in settings where multiple other studies had found lesser correlations, specifically: metastatic breast47 and colon cancer52,55,58 (Table 1). In both these settings, the largest analyses conducted to date found low correlation.48,50

Discussion

Most trial-level meta-analyses in oncology found low correlation between a surrogate end point and overall survival, and all were based on only a subset of clinical trial evidence. Our findings call into question the widespread use of surrogate end points in oncology as a basis for treatment decisions.

Trial-level or level 1 meta-analyses are the highest form of evidence that surrogate end points can predict efficacy regarding final outcomes, such as overall survival, for new therapies or new combinations (Figure 1)22,23,75 The majority of these claims resulted in correlations of low (52%) or medium strength (25%) (Figure 3), which are generally considered insufficient evidence on which to base clinical or regulatory decisions.34

We were unable to identify any analyses that looked at all randomized trials on a topic. Most analyses relied on published articles (28%) or articles and meeting abstracts (39%), and only 5 authors (14%) sought unpublished trials. Even when authors were able to identify published and unpublished studies, they were unable to obtain data from all eligible trials. Only 51.5% of eligible studies were ultimately included in even the most rigorous meta-analyses (Table 2). The major barriers to including more studies that we noted were that primary investigators did not provide data, or primary study publications did not include information that the meta-analysts needed, such as the rates of surrogate improvement or improvement in overall survival.

Our findings suggest that most correlations of surrogacy in oncology are based on only a subset of potentially informative trials. Unpublished trials (and those that do not report data that meta-analysts require) may have poorer correlations than those that are published, and such discrepancies may contribute to reluctance among sponsors and authors to submit those findings to journals and conferences and to a poorer likelihood of acceptance.

The use of surrogate end points in oncology, particularly PFS, has grown in recent years and it is frequently the primary end point of cancer clinical trials and the basis for regulatory approval of novel agents.8,76-78 Surrogate end points are also used as the basis of many NCCN recommendations, which obliges many insurers and Medicare to cover those drugs for those indications.12-14 Our analysis confirms that the use of this end point occurs beyond the specific settings in which it has been validated.76 Simply because a surrogate is measureable does not make it predictive or meaningful,76 and our analysis builds on a prior study that concludes that the association between surrogates and survival in oncology is generally poor.33

High-profile medical reversals, such as the case of bevacizumab in breast cancer, emphasize the dangers of excessive reliance on unvalidated surrogates.79,80 Although bevacizumab was approved for breast cancer on the basis of an improvement in PFS,15,16 this surrogate-survival correlation is not well supported. We identified 8 meta-analyses examining whether gains in PFS predict overall survival in metastatic breast cancer. Six reported low correlation; 1 reported medium correlation; and only 1 reported a strong correlation (Table 1). Despite this evidence, in 2012 another drug, everolimus, was approved for metastatic breast cancer based on improvement in PFS.81 In 2014, updated follow-up from the everolimus study did not find an overall survival benefit.82 In 2015, palbociclib received accelerated approval for metastatic breast cancer on the basis of improvement in PFS. Again, there is no demonstration of an overall survival advantage.83 Both palbociclib and everolimus remain on the market.

In 2007, liposomal doxorubicin received FDA approval for treatment of multiple myeloma based on a delay in time to progression when given in combination with bortezomib compared with bortezomib alone.84 In our investigation, we were unable to identify any analysis validating this surrogate and survival in multiple myeloma, and an updated analysis of liposomal doxorubicin confirms that the drug confers no survival benefit.85 Liposomal doxorubicin remains FDA approved.

The FDA’s use of pathologic complete response as grounds for accelerated approval in the neoadjuvant setting (the basis for the approval of pertuzumab [Perjeta; Roche/Genentech]) also appears to run counter to both meta-analyses we identified. Both meta-analyses on this topic found that improvements in pathologic complete response were poorly correlated with subsequent event-free survival and OS35,36 (Table 1).

Limitations

There are several limitations to our study. First, the specific thresholds we used to grade the strength of correlation were adapted from a guidance document but have not been externally validated and are subject to disagreement. Although we believe that few would dispute that a correlation coefficient (r) below 0.7 is weak support for a surrogate end point, some may be critical of the thresholds for medium and high strength. For this reason, we provide all coefficients in Table 1 to permit alternative analyses.

Second, we cannot say whether correlations will improve or weaken based on a more comprehensive assessment of clinical trials. However, prior research has found that unpublished trials are different than published studies.86-91 Just as meta-analyses on specific clinical topics may change with consideration of published and unpublished evidence,92,93 the strength of surrogacy may also change with a more comprehensive collection of data. However, this is an assumption and should be treated as such. Unfortunately, we were unable to identify even 1 study that considered the totality of the evidence.

Third, the use of crossover in cancer clinical trials is thought to complicate the role of surrogacy in oncology. Unidirectional crossover from the control or placebo arm of cancer trials to the experimental drug (but not vice versa) is thought to explain why some cancer drugs that improve PFS fail to improve OS. However, crossover is unlikely to affect our analysis because most of the individual randomized trials included in the surrogate validation studies we examined occurred prior to the widespread use of crossover.

Moreover, there are several reasons to question the common narrative regarding crossover. First, effective cancer drugs can show overall survival benefits despite crossover.94-96 Second, favorable PFS but negative OS results seen with everolimus,97 palbociclib,83 and bevacizumab98 in metastatic breast cancer occurred in the absence of crossover. And finally, there are several alternative interpretations for the effect of crossover, including that crossover can hide the harms of a drug or a mask a lack of clinical benefit.99,100 For these reasons, we do not believe that crossover alters our findings.

Finally, a recent study suggests that approving new drugs based on surrogates (even inaccurate ones) may provide a greater benefit for society than waiting for OS data.101 The authors found favorable societal benefits for approving non–small-cell lung cancer (NSCLC) drugs with PFS benefits of 3 months or more. This occurred despite the fact that the authors noted a poor correlation (as we also did in Table 1) between net mean PFS and OS in NSCLC cancer (r = 0.56). The major limitation of this analysis, like other modeling studies, is that it rests on a number of assumptions. For instance, the authors likely overestimate the delay between PFS and OS results by using date of publication rather than the data cutoff date. Also the authors draw broad conclusions from a very small set of trials, ie, the few NSCLC trials with greater than 3-month PFS gains from an overall set of 27 trials. Finally, the model does not account for the wider unintended consequences of approval based on weak surrogates, which may encourage the pharmaceutical industry to pursue even more cancer drugs with marginal to no benefits.102

Conclusions

Most trial-level validation studies of surrogate end points in oncology find low or medium strength correlations with overall survival. All validation studies use only a subset of available trials. The evidence that surrogate end points predict overall survival in oncology is limited.

Back to top
Article Information

Accepted for Publication: April 11, 2015.

Corresponding Author: Vinay Prasad, MD, MPH, National Cancer Institute, National Institutes of Health, 10 Center Dr, 10/12N226, Bethesda, MD 20892 (vinayak.prasad@nih.gov).

Published Online: June 22, 2015. doi:10.1001/jamainternmed.2015.2829.

Author Contributions: Dr Prasad had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Study concept and design: Prasad.

Acquisition, analysis, or interpretation of data: Prasad, Kim, Burotto, Vandross.

Drafting of the manuscript: Prasad, Kim.

Critical revision of the manuscript for important intellectual content: Prasad, Kim, Burotto, Vandross.

Statistical analysis: Prasad, Kim.

Administrative, technical, or material support: Prasad, Burotto, Vandross.

Study supervision: Prasad.

Conflict of Interest Disclosures: None reported.

Disclaimer: The views and opinions of Drs Prasad, Kim, and Burotto do not reflect those of the National Cancer Institute.

References
1.
Svensson  S, Menkes  DB, Lexchin  J.  Surrogate outcomes in clinical trials: a cautionary tale.  JAMA Intern Med. 2013;173(8):611-612.PubMedGoogle ScholarCrossref
2.
Prasad  V, Vandross  A.  Cardiovascular primary prevention: how high should we set the bar?  Arch Intern Med. 2012;172(8):656-659.PubMedGoogle ScholarCrossref
3.
Fleming  TR, DeMets  DL.  Surrogate end points in clinical trials: are we being misled?  Ann Intern Med. 1996;125(7):605-613.PubMedGoogle ScholarCrossref
4.
Messerli  FH, Bangalore  S.  ALTITUDE trial and dual RAS blockade: the alluring but soft science of the surrogate end point.  Am J Med. 2013;126(3):e1-e3.PubMedGoogle ScholarCrossref
5.
Yudkin  JS, Lipska  KJ, Montori  VM.  The idolatry of the surrogate.  BMJ. 2011;343:d7995.PubMedGoogle ScholarCrossref
6.
Ciani  O, Buyse  M, Garside  R,  et al.  Comparison of treatment effect sizes associated with surrogate and final patient relevant outcomes in randomised controlled trials: meta-epidemiological study.http://www.bmj.com/content/346/bmj.f457. Accessed May 12, 2015.
7.
Shi  Q, Sargent  DJ.  Meta-analysis for the evaluation of surrogate endpoints in cancer clinical trials.  Int J Clin Oncol. 2009;14(2):102-111.PubMedGoogle ScholarCrossref
8.
Fauber  JCE.  FDA approves cancer drugs without proof they're extending lives.http://www.jsonline.com/watchdog/watchdogreports/fda-approves-cancer-drugs-without-proof-theyre-extending-lives-b99348000z1-280437692.html. Accessed December 14, 2014.
9.
Korde  N, Zingone  A, Kwok  M,  et al Phase II clinical and correlative study of carfilzomib, lenalidomide, and dexamethasone (CRd) in newly diagnosed multiple myeloma (MM) patients.  J Clin Oncol. 2012(suppl);abstr e18568.Google Scholar
10.
Jakubowiak  AJ, Dytfeld  D, Griffith  KA,  et al.  A phase 1/2 study of carfilzomib in combination with lenalidomide and low-dose dexamethasone as a frontline treatment for multiple myeloma.  Blood. 2012;120(9):1801-1809.PubMedGoogle ScholarCrossref
11.
National Comprehensive Cancer Network.  Myeloma NgM [registration required].http://www.nccn.org/professionals/physician_gls/pdf/myeloma.pdf. Accessed May 12, 2015.
12.
Tillman  K, Burton  B, Jacques  LB, Phurrough  SE.  Compendia and anticancer therapy under Medicare.  Ann Intern Med. 2009;150(5):348-350.PubMedGoogle ScholarCrossref
13.
McGivney  WT.  Medicare recognizes NCCN compendium.  J Natl Compr Canc Netw. 2008;6(7):635.Google Scholar
14.
McGivney  WT.  NCCN guidelines and their impact on coverage policy.  J Natl Compr Canc Netw. 2010;8(6):625.Google Scholar
15.
Miller  K, Wang  M, Gralow  J,  et al.  Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer.  N Engl J Med. 2007;357(26):2666-2676.PubMedGoogle ScholarCrossref
16.
Carpenter  D, Kesselheim  AS, Joffe  S.  Reputation and precedent in the bevacizumab decision.  N Engl J Med. 2011;365(2):e3.PubMedGoogle ScholarCrossref
17.
Lassere  MN, Johnson  KR, Schiff  M, Rees  D.  Is blood pressure reduction a valid surrogate endpoint for stroke prevention? an analysis incorporating a systematic review of randomised controlled trials, a by-trial weighted errors-in-variables regression, the surrogate threshold effect (STE) and the Biomarker-Surrogacy (BioSurrogate) Evaluation Schema (BSES).  BMC Med Res Methodol. 2012;12(1):27.PubMedGoogle ScholarCrossref
18.
Torri  V, Simon  R, Russek-Cohen  E, Midthune  D, Friedman  M.  Statistical model to determine the relationship of response and survival in patients with advanced ovarian cancer treated with chemotherapy.  J Natl Cancer Inst. 1992;84(6):407-414.PubMedGoogle ScholarCrossref
19.
Freedman  LS, Graubard  BI, Schatzkin  A.  Statistical validation of intermediate endpoints for chronic diseases.  Stat Med. 1992;11(2):167-178.PubMedGoogle ScholarCrossref
20.
Buyse  M, Molenberghs  G, Burzykowski  T, Renard  D, Geys  H.  The validation of surrogate endpoints in meta-analyses of randomized experiments.  Biostatistics. 2000;1(1):49-67.PubMedGoogle ScholarCrossref
21.
Weir  CJ, Walley  RJ.  Statistical evaluation of biomarkers as surrogate endpoints: a literature review.  Stat Med. 2006;25(2):183-203.PubMedGoogle ScholarCrossref
22.
Elston  J, Taylor  RS.  Use of surrogate outcomes in cost-effectiveness models: a review of United Kingdom health technology assessment reports.  Int J Technol Assess Health Care. 2009;25(1):6-13.PubMedGoogle ScholarCrossref
23.
Buyse  M, Burzykowski  T, Michiels  S, Carroll  K.  Individual- and trial-level surrogacy in colorectal cancer.  Stat Methods Med Res. 2008;17(5):467-475.PubMedGoogle ScholarCrossref
24.
Prentice  RL.  Surrogate endpoints in clinical trials: definition and operational criteria.  Stat Med. 1989;8(4):431-440.PubMedGoogle ScholarCrossref
25.
Sargent  DJ, Wieand  HS, Haller  DG,  et al.  Disease-free survival versus overall survival as a primary end point for adjuvant colon cancer studies: individual patient data from 20,898 patients on 18 randomized trials.  J Clin Oncol. 2005;23(34):8664-8670.PubMedGoogle ScholarCrossref
26.
Pratt  CM, Moyé  LA.  The cardiac arrhythmia suppression trial: casting suppression in a different light.  Circulation. 1995;91(1):245-247.PubMedGoogle ScholarCrossref
27.
Echt  DS, Liebson  PR, Mitchell  LB,  et al.  Mortality and morbidity in patients receiving encainide, flecainide, or placebo: the Cardiac Arrhythmia Suppression Trial.  N Engl J Med. 1991;324(12):781-788.PubMedGoogle ScholarCrossref
28.
Laporte  S, Squifflet  P, Baroux  N,  et al.  Prediction of survival benefits from progression-free survival benefits in advanced non-small-cell lung cancer: evidence from a meta-analysis of 2334 patients from 5 randomised trials.  BMJ Open. 2013;3(3):e001802.PubMedGoogle ScholarCrossref
29.
Burzykowski  T, Buyse  M.  Surrogate threshold effect: an alternative measure for meta-analytic surrogate endpoint validation.  Pharm Stat. 2006;5(3):173-186.PubMedGoogle ScholarCrossref
30.
Buyse  M.  Use of meta-analysis for the validation of surrogate endpoints and biomarkers in cancer trials.  Cancer J. 2009;15(5):421-425.PubMedGoogle ScholarCrossref
31.
Sherrill  B, Kaye  JA, Sandin  R, Cappelleri  JC, Chen  C.  Review of meta-analyses evaluating surrogate endpoints for overall survival in oncology.  Onco Targets Ther. 2012;5:287-296.PubMedGoogle ScholarCrossref
32.
Davis  S, Tappenden  P, Cantrell  A.  A review of studies examining the relationship between progression-free survival and overall survival in advanced or metastatic cancer.http://www.nicedsu.org.uk/PFSOS%20Report.FINAL.06.08.12.pdf. Accessed May 12, 2015.
33.
Ciani  O, Davis  S, Tappenden  P,  et al.  Validation of surrogate endpoints in advanced solid tumors: systematic review of statistical methods, results, and implications for policy makers.  Int J Technol Assess Health Care. 2014;30(3):312-324.PubMedGoogle ScholarCrossref
34.
Institute for Quality and Efficiency in Health Care [Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen] (IQWiG).  Validity of surrogate endpoints in oncology: executive summary.http://www.iqwig.de/download/A10-05_Executive_Summary_v1-1_Surrogate_endpoints_in_oncology.pdf. Accessed December 20, 2014.
35.
Cortazar  P, Zhang  L, Untch  M,  et al.  Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis.  Lancet. 2014;384(9938):164-172.PubMedGoogle ScholarCrossref
36.
Berruti  A, Amoroso  V, Gallo  F,  et al.  Pathologic complete response as a potential surrogate for the clinical outcome in patients with breast cancer after neoadjuvant therapy: a meta-regression of 29 randomized prospective studies.  J Clin Oncol. 2014;32(34):3883-3891.PubMedGoogle ScholarCrossref
37.
Ng  R, Pond  GR, Tang  PA, MacIntosh  PW, Siu  LL, Chen  EX.  Correlation of changes between 2-year disease-free survival and 5-year overall survival in adjuvant breast cancer trials from 1966 to 2006.  Ann Oncol. 2008;19(3):481-486.PubMedGoogle ScholarCrossref
38.
Burzykowski  T, Buyse  M, Yothers  G, Sakamoto  J, Sargent  D.  Exploring and validating surrogate endpoints in colorectal cancer.  Lifetime Data Anal. 2008;14(1):54-64.PubMedGoogle ScholarCrossref
39.
Oba  K, Paoletti  X, Alberts  S,  et al; GASTRIC group.  Disease-free survival as a surrogate for overall survival in adjuvant trials of gastric cancer: a meta-analysis.  J Natl Cancer Inst. 2013;105(21):1600-1607.PubMedGoogle ScholarCrossref
40.
Michiels  S, Le Maître  A, Buyse  M,  et al; MARCH and MACH-NC Collaborative Groups.  Surrogate endpoints for overall survival in locally advanced head and neck cancer: meta-analyses of individual patient data.  Lancet Oncol. 2009;10(4):341-350.PubMedGoogle ScholarCrossref
41.
Mauguen  A, Pignon  JP, Burdett  S,  et al; Surrogate Lung Project Collaborative Group.  Surrogate endpoints for overall survival in chemotherapy and radiotherapy trials in operable and locally advanced lung cancer: a re-analysis of meta-analyses of individual patients’ data.  Lancet Oncol. 2013;14(7):619-626.PubMedGoogle ScholarCrossref
42.
Bruzzi  P, Del Mastro  L, Sormani  MP,  et al.  Objective response to chemotherapy as a potential surrogate end point of survival in metastatic breast cancer patients.  J Clin Oncol. 2005;23(22):5117-5125.PubMedGoogle ScholarCrossref
43.
Hackshaw  A, Knight  A, Barrett-Lee  P, Leonard  R.  Surrogate markers and survival in women receiving first-line combination anthracycline chemotherapy for advanced breast cancer.  Br J Cancer. 2005;93(11):1215-1221.PubMedGoogle ScholarCrossref
44.
Burzykowski  T, Buyse  M, Piccart-Gebhart  MJ,  et al.  Evaluation of tumor response, disease control, progression-free survival, and time to progression as potential surrogate end points in metastatic breast cancer.  J Clin Oncol. 2008;26(12):1987-1992.PubMedGoogle ScholarCrossref
45.
Miksad  RA, Zietemann  V, Gothe  R,  et al.  Progression-free survival as a surrogate endpoint in advanced breast cancer.  Int J Technol Assess Health Care. 2008;24(4):371-383.PubMedGoogle ScholarCrossref
46.
Sherrill  B, Amonkar  M, Wu  Y,  et al.  Relationship between effects on time-to-disease progression and overall survival in studies of metastatic breast cancer.  Br J Cancer. 2008;99(10):1572-1578.PubMedGoogle ScholarCrossref
47.
Wilkerson  J, Fojo  T.  Progression-free survival is simply a measure of a drug’s effect while administered and is not a surrogate for overall survival.  Cancer J. 2009;15(5):379-385.PubMedGoogle ScholarCrossref
48.
Beauchemin  C, Cooper  D, Lapierre  ME, Yelle  L, Lachaine  J.  Progression-free survival as a potential surrogate for overall survival in metastatic breast cancer.  Onco Targets Ther. 2014;7:1101-1110.PubMedGoogle ScholarCrossref
49.
Petrelli  F, Barni  S.  Surrogate endpoints in metastatic breast cancer treated with targeted therapies: an analysis of the first-line phase III trials.  Med Oncol. 2014;31(1):776.PubMedGoogle ScholarCrossref
50.
Johnson  KR, Ringland  C, Stokes  BJ,  et al.  Response rate or time to progression as predictors of survival in trials of metastatic colorectal cancer or non-small-cell lung cancer: a meta-analysis.  Lancet Oncol. 2006;7(9):741-746.PubMedGoogle ScholarCrossref
51.
Chirila  C, Odom  D, Devercelli  G,  et al.  Meta-analysis of the association between progression-free survival and overall survival in metastatic colorectal cancer.  Int J Colorectal Dis. 2012;27(5):623-634.PubMedGoogle ScholarCrossref
52.
Sidhu  R, Rong  A, Dahlberg  S.  Evaluation of progression-free survival as a surrogate endpoint for survival in chemotherapy and targeted agent metastatic colorectal cancer trials.  Clin Cancer Res. 2013;19(5):969-976.PubMedGoogle ScholarCrossref
53.
Buyse  M, Thirion  P, Carlson  RW, Burzykowski  T, Molenberghs  G, Piedbois  P; Meta-Analysis Group in Cancer.  Relation between tumour response to first-line chemotherapy and survival in advanced colorectal cancer: a meta-analysis.  Lancet. 2000;356(9227):373-378.PubMedGoogle ScholarCrossref
54.
Tang  PA, Bentzen  SM, Chen  EX, Siu  LL.  Surrogate end points for median overall survival in metastatic colorectal cancer: literature-based analysis from 39 randomized controlled trials of first-line chemotherapy.  J Clin Oncol. 2007;25(29):4562-4568.PubMedGoogle ScholarCrossref
55.
Giessen  C, Laubender  RP, Ankerst  DP,  et al.  Progression-free survival as a surrogate endpoint for median overall survival in metastatic colorectal cancer: literature-based analysis from 50 randomized first-line trials.  Clin Cancer Res. 2013;19(1):225-235.PubMedGoogle ScholarCrossref
56.
Shi  Q, de Gramont  A, Grothey  A,  et al.  Individual patient data analysis of progression-free survival versus overall survival as a first-line end point for metastatic colorectal cancer in modern randomized trials: findings from the analysis and research in cancers of the digestive system database.  J Clin Oncol. 2015;33(1):22-28.PubMedGoogle ScholarCrossref
57.
Giessen  C, Laubender  RP, Ankerst  DP,  et al.  Surrogate end points in second-line treatment for mCRC: a systematic literature-based analysis from 23 randomised trials.  Acta Oncol. 2015;54(2):187-193.PubMedGoogle ScholarCrossref
58.
Buyse  M, Burzykowski  T, Carroll  K,  et al.  Progression-free survival is a surrogate for survival in advanced colorectal cancer.  J Clin Oncol. 2007;25(33):5218-5224.PubMedGoogle ScholarCrossref
59.
Amir  E, Seruga  B, Kwong  R, Tannock  IF, Ocaña  A.  Poor correlation between progression-free and overall survival in modern clinical trials: are composite endpoints the answer?  Eur J Cancer. 2012;48(3):385-388.PubMedGoogle ScholarCrossref
60.
Paoletti  X, Oba  K, Bang  YJ,  et al; GASTRIC group.  Progression-free survival as a surrogate for overall survival in advanced/recurrent gastric cancer trials: a meta-analysis.  J Natl Cancer Inst. 2013;105(21):1667-1670.PubMedGoogle ScholarCrossref
61.
Shitara  K, Matsuo  K, Muro  K, Doi  T, Ohtsu  A.  Correlation between overall survival and other endpoints in clinical trials of second-line chemotherapy for patients with advanced gastric cancer.  Gastric Cancer. 2014;17(2):362-370.PubMedGoogle ScholarCrossref
62.
Flaherty  KT, Hennig  M, Lee  SJ,  et al.  Surrogate endpoints for overall survival in metastatic melanoma: a meta-analysis of randomised controlled trials.  Lancet Oncol. 2014;15(3):297-304.PubMedGoogle ScholarCrossref
63.
Lee  L, Wang  L, Crump  M.  Identification of potential surrogate end points in randomized clinical trials of aggressive and indolent non-Hodgkin’s lymphoma: correlation of complete response, time-to-event and overall survival end points.  Ann Oncol. 2011;22(6):1392-1403.PubMedGoogle ScholarCrossref
64.
Hayashi  H, Okamoto  I, Taguri  M, Morita  S, Nakagawa  K.  Postprogression survival in patients with advanced non-small-cell lung cancer who receive second-line or third-line chemotherapy.  Clin Lung Cancer. 2013;14(3):261-266.PubMedGoogle ScholarCrossref
65.
Delea  TE, Khuu  A, Heng  DY, Haas  T, Soulières  D.  Association between treatment effects on disease progression end points and overall survival in clinical studies of patients with metastatic renal cell carcinoma.  Br J Cancer. 2012;107(7):1059-1068.PubMedGoogle ScholarCrossref
66.
Johnson  KR, Liauw  W, Lassere  MN.  Evaluating surrogacy metrics and investigating approval decisions of progression-free survival (PFS) in metastatic renal cell cancer: a systematic review.  Ann Oncol. 2015;26(3):485-496.PubMedGoogle ScholarCrossref
67.
Foster  NR, Qi  Y, Shi  Q,  et al.  Tumor response and progression-free survival as potential surrogate endpoints for overall survival in extensive stage small-cell lung cancer: findings on the basis of North Central Cancer Treatment Group trials.  Cancer. 2011;117(6):1262-1271.PubMedGoogle ScholarCrossref
68.
Hotta  K, Fujiwara  Y, Matsuo  K,  et al.  Time to progression as a surrogate marker for overall survival in patients with advanced non-small cell lung cancer.  J Thorac Oncol. 2009;4(3):311-317.PubMedGoogle ScholarCrossref
69.
Non-small Cell Lung Cancer Collaborative Group.  Chemotherapy in non-small cell lung cancer: a meta-analysis using updated data on individual patients from 52 randomised clinical trials.  BMJ. 1995;311(7010):899-909.PubMedGoogle ScholarCrossref
70.
Aupérin  A, Le Péchoux  C, Rolland  E,  et al.  Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer.  J Clin Oncol. 2010;28(13):2181-2190.PubMedGoogle ScholarCrossref
71.
Arriagada  R, Auperin  A, Burdett  S,  et al; NSCLC Meta-analyses Collaborative Group.  Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non-small-cell lung cancer: two meta-analyses of individual patient data.  Lancet. 2010;375(9722):1267-1277.PubMedGoogle ScholarCrossref
72.
NSCLC Meta-Analyses Collaborative Group.  Chemotherapy in addition to supportive care improves survival in advanced non-small-cell lung cancer: a systematic review and meta-analysis of individual patient data from 16 randomized controlled trials.  J Clin Oncol. 2008;26(28):4617-4625.PubMedGoogle ScholarCrossref
73.
Mauguen  A, Le Péchoux  C, Saunders  MI,  et al.  Hyperfractionated or accelerated radiotherapy in lung cancer: an individual patient data meta-analysis.  J Clin Oncol. 2012;30(22):2788-2797.PubMedGoogle ScholarCrossref
74.
Paoletti  X, Oba  K, Burzykowski  T,  et al; GASTRIC (Global Advanced/Adjuvant Stomach Tumor Research International Collaboration) Group.  Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis.  JAMA. 2010;303(17):1729-1737.PubMedGoogle ScholarCrossref
75.
Buyse  M, Sargent  DJ, Grothey  A, Matheson  A, de Gramont  A.  Biomarkers and surrogate end points: the challenge of statistical validation.  Nat Rev Clin Oncol. 2010;7(6):309-317.PubMedGoogle ScholarCrossref
76.
Booth  CM, Eisenhauer  EA.  Progression-free survival: meaningful or simply measurable?  J Clin Oncol. 2012;30(10):1030-1033.PubMedGoogle ScholarCrossref
77.
Booth  CM, Cescon  DW, Wang  L, Tannock  IF, Krzyzanowska  MK.  Evolution of the randomized controlled trial in oncology over three decades.  J Clin Oncol. 2008;26(33):5458-5464.PubMedGoogle ScholarCrossref
78.
Kay  A, Higgins  J, Day  AG, Meyer  RM, Booth  CM.  Randomized controlled trials in the era of molecular oncology: methodology, biomarkers, and end points.  Ann Oncol. 2012;23(6):1646-1651.PubMedGoogle ScholarCrossref
79.
Prasad  V, Cifu  A, Ioannidis  JP.  Reversals of established medical practices: evidence to abandon ship.  JAMA. 2012;307(1):37-38.PubMedGoogle ScholarCrossref
80.
Prasad  V, Vandross  A, Toomey  C,  et al.  A decade of reversal: an analysis of 146 contradicted medical practices.  Mayo Clin Proc. 2013;88(8):790-798.PubMedGoogle ScholarCrossref
81.
US Food and Drug Administration.  FDA approves Afinitor for advanced breast cancer. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm312965.htm. Accessed January 4, 2014.
82.
Piccart  M, Hortobagyi  GN, Campone  M,  et al.  Everolimus plus exemestane for hormone-receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: overall survival results from BOLERO-2.  Ann Oncol. 2014;25(12):2357-2362.PubMedGoogle ScholarCrossref
83.
Finn  RS, Crown  JP, Lang  I,  et al.  The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study.  Lancet Oncol. 2015;16(1):25-35.PubMedGoogle ScholarCrossref
84.
Ning  YM, He  K, Dagher  R,  et al.  Liposomal doxorubicin in combination with bortezomib for relapsed or refractory multiple myeloma.  Oncology (Williston Park). 2007;21(12):1503-1508.PubMedGoogle Scholar
85.
Orlowski  RZ, Nagler  A, Sonneveld  P,  et al.  Final overall survival results of a randomized trial comparing bortezomib plus pegylated liposomal doxorubicin with bortezomib alone in subjects with relapsed or refractory multiple myeloma.  Blood. 2014;124(21):3448.Google Scholar
86.
Ioannidis  JP.  Effect of the statistical significance of results on the time to completion and publication of randomized efficacy trials.  JAMA. 1998;279(4):281-286.PubMedGoogle ScholarCrossref
87.
Krzyzanowska  MK, Pintilie  M, Tannock  IF.  Factors associated with failure to publish large randomized trials presented at an oncology meeting.  JAMA. 2003;290(4):495-501.PubMedGoogle ScholarCrossref
88.
Misakian  AL, Bero  LA.  Publication bias and research on passive smoking: comparison of published and unpublished studies.  JAMA. 1998;280(3):250-253.PubMedGoogle ScholarCrossref
89.
Dickersin  K, Min  YI, Meinert  CL.  Factors influencing publication of research results. Follow-up of applications submitted to two institutional review boards.  JAMA. 1992;267(3):374-378.PubMedGoogle ScholarCrossref
90.
Dwan  K, Altman  DG, Arnaiz  JA,  et al.  Systematic review of the empirical evidence of study publication bias and outcome reporting bias.  PLoS One. 2008;3(8):e3081.PubMedGoogle ScholarCrossref
91.
Rising  K, Bacchetti  P, Bero  L.  Reporting bias in drug trials submitted to the Food and Drug Administration: review of publication and presentation.  PLoS Med. 2008;5(11):e217.PubMedGoogle ScholarCrossref
92.
Jefferson  T, Jones  M, Doshi  P, Spencer  EA, Onakpoya  I, Heneghan  CJ.  Oseltamivir for influenza in adults and children: systematic review of clinical study reports and summary of regulatory comments.  BMJ. 2014;348:g2545.PubMedGoogle ScholarCrossref
93.
Kirsch  I, Deacon  BJ, Huedo-Medina  TB, Scoboria  A, Moore  TJ, Johnson  BT.  Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration.  PLoS Med. 2008;5(2):e45.PubMedGoogle ScholarCrossref
94.
Chapman  PB, Hauschild  A, Robert  C,  et al; BRIM-3 Study Group.  Improved survival with vemurafenib in melanoma with BRAF V600E mutation.  N Engl J Med. 2011;364(26):2507-2516.PubMedGoogle ScholarCrossref
95.
Raymond  E, Dahan  L, Raoul  J-L,  et al.  Sunitinib malate for the treatment of pancreatic neuroendocrine tumors.  N Engl J Med. 2011;364(6):501-513.PubMedGoogle ScholarCrossref
96.
Kantoff  PW, Higano  CS, Shore  ND,  et al; IMPACT Study Investigators.  Sipuleucel-T immunotherapy for castration-resistant prostate cancer.  N Engl J Med. 2010;363(5):411-422.PubMedGoogle ScholarCrossref
97.
Baselga  J, Campone  M, Piccart  M,  et al.  Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer.  N Engl J Med. 2012;366(6):520-529.PubMedGoogle ScholarCrossref
98.
Burstein  HJ.  Bevacizumab for advanced breast cancer: all tied up with a RIBBON?  J Clin Oncol. 2011;29(10):1232-1235.PubMedGoogle ScholarCrossref
99.
Prasad  V.  Double-crossed: why crossover in clinical trials may be distorting medical science.  J Natl Compr Canc Netw. 2013;11(5):625-627.PubMedGoogle Scholar
100.
Prasad  V, Grady  C.  The misguided ethics of crossover trials.  Contemp Clin Trials. 2014;37(2):167-169.PubMedGoogle ScholarCrossref
101.
Lakdawalla  DN, Chou  JW, Linthicum  MT, MacEwan  JP, Zhang  J, Goldman  DP.  Evaluating expected costs and benefits of granting access to new treatments on the basis of progression-free survival in non–small-cell lung cancer [published online March 19, 2015].  JAMA Oncol. doi:10.1001/jamaoncol.2015.0203.Google Scholar
102.
Fojo  T, Mailankody  S, Lo  A.  Unintended consequences of expensive cancer therapeutics—the pursuit of marginal indications and a me-too mentality that stifles innovation and creativity: the John Conley Lecture.  JAMA Otolaryngol Head Neck Surg. 2014;140(12):1225-1236.PubMedGoogle ScholarCrossref
×