analyzingthepivotaltrialthatcomparedsunitinibandifn...

9
Predictive Biomarkers and Personalized Medicine Analyzing the Pivotal Trial That Compared Sunitinib and IFN-a in Renal Cell Carcinoma, Using a Method That Assesses Tumor Regression and Growth Wilfred D. Stein 1,2 , Julia Wilkerson 1 , Sindy T. Kim 3 , Xin Huang 3 , Robert J. Motzer 4 , Antonio Tito Fojo 1 , and Susan E. Bates 1 Abstract Purpose: We applied a method that analyzes tumor response, quantifying the rates of tumor growth (g) and regression (d), using tumor measurements obtained while patients receive therapy. We used data from the phase III trial comparing sunitinib and IFN-a in metastatic renal cell carcinoma (mRCC) patients. Methods: The analysis used an equation that extracts d and g. Results: For sunitinib, overall survival (OS) was strongly correlated with log g (Rsq ¼ 0.44, P < 0.0001); much less with log d (Rsq ¼ 0.04; P ¼ 0.0002). The median g of tumors in these patients (0.00082 per days; log g ¼3.09) was about half that (P < 0.001) of tumors in patients receiving IFN-a (0.0015 per day; log g ¼ 2.81). With IFN-a, the OS/log g correlation (Rsq ¼ 0.14) was weaker. Values of g from measurements obtained by study investigators or central review were highly correlated (Rsq ¼ 0.80). No advantage resulted in including data from central review in regressions. Furthermore, g can be estimated accurately four months before treatment discontinuation. Extrapolating g in a model that incorporates survival generates the hypothesis that g increased after discontinuation of sunitinib but did not accelerate. Conclusions: In patients with mRCC, sunitinib reduced tumor growth rate, g, more than did IFN-a. Correlating g with OS confirms earlier analyses suggesting g may be an important clinical trial endpoint, to be explored prospectively and in individual patients. Clin Cancer Res; 18(8); 2374–81. Ó2012 AACR. Introduction In 2010, approximately 570,000 people died of cancer in the United States, most from chemotherapy-refractory solid tumors, including more than 13,000 from metastatic renal cell carcinoma (mRCC; ref. 1). With the advent of sorafenib, sunitinib, temsirolimus, bevacizumab with IFN, everoli- mus, pazopanib, and now axitinib therapy of mRCC has improved (2–7). However, these therapies are not curative, underscoring the need for alternative treatment strategies and novel decision paradigms (8). We developed a method to analyze tumor responses to therapy, quantifying the rate of tumor regression (decay, d) and growth (g), using measurements obtained while patients receive therapy (9–12). The rate constants are derived using data collected in clinical trials. In responding tumors, regression dominates from start of therapy until nadir, growth dominating after nadir. If tumors do not respond, growth dominates throughout. Previous analyses of phase II studies in mRCC used computed tomography (CT) measurements of tumors (9, 11); in prostate cancer, serum prostate-specific antigen (10, 12) was used. For those, growth rate constants, g, correlated with overall survival (OS), while, surprisingly, regression rate constants, d, did not. Here, we used randomized phase III trial data comparing sunitinib with IFN-a in untreated mRCC (3, 13). We illustrate the value of the growth rate constant as a clinical trial endpoint, reflecting the impact of sunitinib on tumor growth. Materials and Methods Clinical trial and study design The study, an international, multicenter, randomized, phase III trial, compared sunitinib (SUTENT, Pfizer), with IFN-a (3, 13). OS was calculated from randomization date until date of death. Tumor measurements from CT scans were recorded as the sum of longest diameters (LD) of Authors' Afliations: 1 Medical Oncology Branch, National Cancer Insti- tute, NIH, Bethesda, Maryland; 2 Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel; 3 Pzer, Inc., La Jolla, California; and 4 Memorial Sloan-Kettering Cancer Center, New York, New York Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). This article was presented, in part, as a poster at the 2010 ASCO annual meeting. Corresponding Author: Medical Oncology Branch, NCI, NIH, Building 10, RM 12N226, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-402- 1357; Fax: 301-402-1608; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-2275 Ó2012 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 18(8) April 15, 2012 2374 on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

Upload: lythuan

Post on 30-Apr-2019

214 views

Category:

Documents


0 download

TRANSCRIPT

Predictive Biomarkers and Personalized Medicine

Analyzing thePivotal Trial ThatComparedSunitiniband IFN-ain Renal Cell Carcinoma, Using a Method That AssessesTumor Regression and Growth

Wilfred D. Stein1,2, Julia Wilkerson1, Sindy T. Kim3, Xin Huang3, Robert J. Motzer4,Antonio Tito Fojo1, and Susan E. Bates1

AbstractPurpose:We applied a method that analyzes tumor response, quantifying the rates of tumor growth (g)

and regression (d), using tumor measurements obtained while patients receive therapy. We used data from

the phase III trial comparing sunitinib and IFN-a in metastatic renal cell carcinoma (mRCC) patients.

Methods: The analysis used an equation that extracts d and g.

Results: For sunitinib, overall survival (OS) was strongly correlated with log g (Rsq¼ 0.44, P < 0.0001);

much less with log d (Rsq¼ 0.04; P¼ 0.0002). Themedian g of tumors in these patients (0.00082 per days;

log g¼�3.09)was about half that (P <0.001) of tumors in patients receiving IFN-a (0.0015 per day; log g¼�2.81). With IFN-a, the OS/log g correlation (Rsq ¼ 0.14) was weaker. Values of g from measurements

obtained by study investigators or central reviewwere highly correlated (Rsq¼ 0.80).No advantage resulted

in including data fromcentral review in regressions. Furthermore,g canbe estimated accurately fourmonths

before treatment discontinuation. Extrapolating g in a model that incorporates survival generates the

hypothesis that g increased after discontinuation of sunitinib but did not accelerate.

Conclusions: In patients with mRCC, sunitinib reduced tumor growth rate, g, more than did IFN-a.Correlating gwith OS confirms earlier analyses suggesting gmay be an important clinical trial endpoint, to

be explored prospectively and in individual patients. Clin Cancer Res; 18(8); 2374–81. �2012 AACR.

IntroductionIn 2010, approximately 570,000 people died of cancer in

theUnited States, most from chemotherapy-refractory solidtumors, including more than 13,000 from metastatic renalcell carcinoma (mRCC; ref. 1).With the advent of sorafenib,sunitinib, temsirolimus, bevacizumab with IFN, everoli-mus, pazopanib, and now axitinib therapy of mRCC hasimproved (2–7). However, these therapies are not curative,underscoring the need for alternative treatment strategiesand novel decision paradigms (8).

We developed a method to analyze tumor responses totherapy, quantifying the rate of tumor regression (decay, d)

and growth (g), using measurements obtained whilepatients receive therapy (9–12). The rate constants arederived using data collected in clinical trials. In respondingtumors, regression dominates from start of therapy untilnadir, growth dominating after nadir. If tumors do notrespond, growth dominates throughout. Previous analysesof phase II studies in mRCC used computed tomography(CT) measurements of tumors (9, 11); in prostate cancer,serumprostate-specific antigen (10, 12)wasused. For those,growth rate constants, g, correlated with overall survival(OS), while, surprisingly, regression rate constants, d, didnot.

Here, we used randomized phase III trial data comparingsunitinib with IFN-a in untreated mRCC (3, 13). Weillustrate the value of the growth rate constant as a clinicaltrial endpoint, reflecting the impact of sunitinib on tumorgrowth.

Materials and MethodsClinical trial and study design

The study, an international, multicenter, randomized,phase III trial, compared sunitinib (SUTENT, Pfizer), withIFN-a (3, 13). OS was calculated from randomization dateuntil date of death. Tumor measurements from CT scanswere recorded as the sum of longest diameters (LD) of

Authors' Affiliations: 1Medical Oncology Branch, National Cancer Insti-tute, NIH, Bethesda, Maryland; 2Silberman Institute of Life Sciences,Hebrew University, Jerusalem, Israel; 3Pfizer, Inc., La Jolla, California; and4Memorial Sloan-Kettering Cancer Center, New York, New York

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

This article was presented, in part, as a poster at the 2010 ASCO annualmeeting.

Corresponding Author: Medical Oncology Branch, NCI, NIH, Building 10,RM 12N226, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-402-1357; Fax: 301-402-1608; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-2275

�2012 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 18(8) April 15, 20122374

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

target lesions. Responses and progressions were assessedaccording to Response Evaluation Criteria in Solid Tumors(RECIST 1.0; ref. 14). Anonymized tumor measurementdata, enrollment and off-study dates, and date of death datawere provided in spreadsheet format by Pfizer, Inc.

Mathematical, data, and statistical analysesMathematical analysis. Our regression-growth equa-

tion is based on the assumption that change in tumorquantity during therapy, indicated by change in the sumof LDs, results from 2 independent component process-es (both following first order kinetics): an exponentialdecrease/regression, d, and an exponential growth/regrowth of the tumor, g. The equation is:

f ðtÞ ¼ exp ð�d � tÞ þ expðg � tÞ � 1 ðAÞ

where f(t) is the tumor quantity (sum of LDs) at time t indays, normalized to the sum of LDs at t ¼ 0, and d (decay,fraction per day) and g (growth, also per day) are thepertinent rate constants; exp is the base of the naturallogarithms.Theoretical curves depicting the separate components of

Eq. (A) and how these combine together to give the timedependence of f, the tumor quantity, appear in Fig. 1. Fordata showing a continuous decrease from start of treatment,g is eliminated:

f ðtÞ ¼ expð�d * tÞ ðBÞ

Similarly, when tumormeasurements show a continuousincrease, d is eliminated:

f ðtÞ ¼ expðg * tÞ ðCÞ

Data analysis. We attempted to fit each data set forwhich more than one data point was available. Curveswere fit and parameters estimated with the procedure

NLIN, the non-linear regression model, in SAS1 (1) soft-ware. The routine extracts parameters g and d with associ-ated Student t and P values.

Statistical analyses. Linear regressions used the polyno-mial linear routine of SigmaPlot 11.0 (Systat Software).Sample comparisons of data sets were on SigmaPlot 11.0with Student t test for normally distributed data, or theMann–Whitney rank-sum procedure for data that were notnormally distributed.We report the appropriate P values fora 2-sided assessment.

Figure 1. Time course of the decay and growth of a tumor. Plots for theregression/growth model using the sum of the LDs of target lesions in4 representative patients treated with either sunitinib or IFN-a. Thelines are curve-fits of a mathematical equation that describes in A andB the initial regression (dotted lines) and concomitant growth of thetumor (dashed lines). In all panels, the solid circles are the sum of theactual measured LDs of target lesions obtained in the clinical setting;the solid line represents the sum of the fraction of tumor that isregressing and the fraction that is growing. A and B, examplesshowing a transient decrease in the sum of the LDs followed by anincrease (growth). C, a case in which no growth is seen during thetreatment period. D, an example of a case in which there is noapparent tumor shrinkage.

Translational RelevanceSunitinib was the first agent in metastatic renal cell

cancer to show major clinical responses in a significantsubset of patients; the randomized trial against IFNmade it the first-line choice. However, survival analysiswas complicated by the use of sunitinib and similaragents after disease progression. The growth rate con-stant, g, is a clinical trial endpoint that could have aideddevelopment of sunitinib by documenting reducedtumor growth rate early in the clinical trial. Results ofthe analysis of g are highly suggestive that reduced g canbe equatedwith clinical benefit. A hypothesis-generatingmodel incorporating g also suggests that continuingtherapy after conventional measures of progression ina patient with marked reduction in g could increasesurvival. These analyses suggest that calculation of g canaid in both drug development and individual patientmanagement.

1The output for this paper was generated using Base SAS software�,Version [9.1.3] of the SAS System for Windows. Copyright � [2002—2003] SAS Institute Inc. SAS and all other SAS Institute Inc. product orservice names are registered trademarks of SAS Institute Inc., Cary, NC,USA.

Tumor Growth Rate Constants in the Sunitinib Pivotal Trial

www.aacrjournals.org Clin Cancer Res; 18(8) April 15, 2012 2375

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

ResultsAnalyzing the time course of tumor decay and growth

The data analyzed were obtained in the phase III regis-tration trial of patients with mRCC randomized to eithersunitinib or IFN-a (3, 13). There were 379 patients assignedto sunitinib and 377 to IFN-a. Tumor measurements wereassessed as the sum of the LDs of target lesions. Figure 1depicts 4 examples of on-study change in LD sum, depictedas solid circles. Solid lines are curve-fits of the equationsdescribing the sum of concomitant regression (decay) andgrowth fractions of the tumor, represented as dotted anddashed lines, respectively [Eqs. (A–C)]. Figure 1A and Bshow a decrease in LD sum followed by an increase. Figure1C depicts a case in which no growth is seen duringtreatment, whereas Fig. 1D shows a case with no apparenttumor shrinkage. We extracted g and d parameters, exclud-ing patients with no tumor assessment data, with onlybaseline CT scan data, or with only one follow-up assess-ment that differed by less than 1.2-fold, these last patientsnot meeting criteria for progressive disease—treatment dis-continued for toxicity or withdrawal of consent (3). Exclu-sions left 331 patients in the sunitinib arm and 268 in theIFN-a arm. A consort diagram showing patient numbers isin Supplementary Fig. S1. Setting the significance for thederived parameters at P < 0.1, we extracted acceptableg and/ordparameters in 319 (84%)of the patients assignedto sunitinib, and 240 (64%) of those assigned to IFN-a(differences in proportions, P ¼ 0.007, c2 test).

To test whether the difference in ability to extract a g valueindicated different patient populations, we compared OS,initial tumor quantity Q0, nadir fraction, and time to nadirfor patientswith acceptableg/dparameters (i.e., extracted atP < 0.1) with those for the whole population (Supplemen-tary Table S1). For both IFN-a and sunitinib arms, for eitherdeceased patients or those still living at study closure,neither OS, initial tumor quantity, nadir fraction, nor timeto nadir were different (every P > 0.25, Mann–Whitneyrank-sum) between those with acceptable g/d and thewhole population. In addition, Kaplan–Meier survivalsplots were not different between those with acceptableg/d parameters and those without (log rank: medians forsunitinib 130.6 and122.7weeks,P¼0.79, for IFN-a, 137.7,119.4, P ¼ 0.69). Thus, the cases with acceptable g/dparameters were representative of the whole population.

To validate the goodness of fit, we tabulated the Rsqbetween data points and fitted line for 20 cases selectedrandomly among those that had an acceptable g parameter.The median Rsq was 0.933, (25%–75%: 0.891–0.958),indicating acceptable agreement between fitting equationand tumor quantity values over the data set. In addition, themedian of the P values for all acceptable g/d values was P¼0.00003 (25%–75%:0.000–0.00192) for the sunitinib arm,andP¼0.000540, (25%–75%:0.00002–0.00830) for IFN2,again indicating thegoodnessof fit of our analytic approach.

Correlating growth and regression rate constants withOS in patients treated with sunitinib or IFN-a

Previous analyses (9–12) suggested the growth rateconstant, g, could be added to response rate and pro-gression-free survival (PFS) as a measure of efficacy. Here,the median g of tumors in patients receiving sunitinib(0.00082 per day; log g ¼ �3.09) was 45% lower thanthat (P < 0.001) of tumors in patients receiving IFN-a(0.0015 per day; log g ¼ �2.81).

Recognizing OS as the "gold standard" of drug efficacy,we assessed the correlation between g ord andOS. The finalanalysis of OS (13) was used, with 262 OS events [date ofdeath (DOD) documented] that yielded acceptable g/dparameters (including 226 with acceptable g). Figure 2 top,left shows the OS/log g correlation for the 131 sunitinibpatients having both a valid g (P < 0.1) and DOD. The righttop panel is a similar plot for the 95 patients treated withIFN-a. For sunitinib, log g and OS were significantly neg-atively correlated (Rsq¼ 0.44; P < 0.001), whereas the datafor IFN-a (Rsq ¼ 0.14; P < 0.001), were less stronglycorrelated. These g values were calculated using tumormeasurements obtained by study investigators. Regressionsusing measurements from central review were similar: forsunitinib, log g and OS correlated significantly negatively(Rsq ¼ 0.30; P < 0.001) in contrast to those for patientsrandomized to IFN-a (Rsq¼ 0.14; P < 0.0044). The bottompanels of Fig. 2 compare regressions of tumor nadir (themeasure used in response rate, defined as the ratio ofminimum sum of LDs to initial sum of LDs) and PFS orOS in the sunitinib arm. In prostate cancer (10), nadir depthand time to reach nadir were both surrogates of g, fastergrowth rates producing higher nadirs, shorter times tonadir, and, in turn, shorter PFS (10). Accordingly, lowercorrelations (Rsq ¼ 0.19) were found when nadir, ratherthan logg, is regressed onOS in this data set, the nadir beingmerely a surrogate of g. The regression of PFS with OS wascomparable with the results obtained with g (data notshown).

To test whether patients who died while on study wererepresentative of the whole population, Supplementary Fig.S2 shows log g versus OS for all patients for whom we hadvalid g parameters as solid circles, with the subset that haddied as red open circles. The data sets fall on a continuum,suggesting that deceased patients are indeed representativeof the whole population insofar as dependence of survivalon g is concerned.

A stepwise regression of OS on log g, log d, and initialtumor quantity (f0, sum of LDs) showed that only log gcontributed to the regression for patients randomized tosunitinib (P < 0.001), whereas both log g (P¼ 0.010) and f0(P ¼ 0.028) contributed significantly to OS in patientsrandomized to IFN-a (data not shown).

Comparing growth rate constants extracted from thetwo study arms

The 2 panels on Fig. 3 left, depict waterfall plots of tumorfraction after 12 treatment weeks as percentage of the sumof the LD at enrollment and show that sunitinib is more2Statistically higher at P < 0.001.

Stein et al.

Clin Cancer Res; 18(8) April 15, 2012 Clinical Cancer Research2376

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

effective than IFN-a. Because calculations of g and d use thetime elapsed between assessments, comparisons amongstudies can be made regardless of differences in assessmentprotocols. Figure 3 right, depicts dot plots of log g valuesderived in our previous studies in patients with mRCC,treated with a placebo, bevacizumab, or ixabepilone(9, 11, 15, 16) as well as the present 4 data sets (sunitinibor IFN-a, each measured by study investigators or centralreview). Confirming the waterfall plots, g values for suni-tinib-treated patients are significantly slower at P < 0.0001[investigators/central review log g values were: sunitinib ¼�3.09/�2.94; IFN-a ¼ �2.81/�2.78].

Comparing rate parameters derived from dataobtained by study investigators or by independentcentral reviewFigure 4 is a direct comparison of results obtained using

data from study investigators or central review. (Log gvalues calculated using the central review on the x axis,corresponding log g values using study investigator mea-surements on the y axis). Rsq for the plot is 0.80; theregression slope is 1.14 � 0.04, close to unity. Accord-ingly, no statistical advantage was gained by includingcentral review data.

g and d parameters can be extracted with accuracybefore the nadir is reachedThe 8 panels of Fig. 5, left, show successive values of the

sum of LDs from 1 patient, obtained over time with startingquantity arbitrarily set at 1. In eachplot, one additional timepoint is added. Applying Eq. (A) to the data in each plot, we

obtained intermediary values for g andd (mean� 95%CI),plotting the values in panels on the right. By the fourthreevaluation both g and d are obtained with accuracy; latervalues differ little. Thus, g can be accurately estimated longbefore tumor measurements show growth: the downwardtrajectory is "deviated upward" by the as yet "undetected"growing fraction. In similar analyses of 20 randomly chosenpatients, an accurate estimate of g could, in most cases, beobtained before the data showed a more than 20% rise inthe sum of LDs (not shown). For these 20 cases, the timebetween recording a g estimate and either amore than 20%rise in LD sumabove the nadir or the end of assessment, wasa median of 126 days (25%–75%: 84–169 days). Thus, 4months before treatment is discontinued, the tumor growthrate can be discerned.

The stability of the derived g parameter was examined,carrying out the same procedure on the entire data set.For the overwhelming majority of cases, g remainedconstant; only rarely was a final g value significantlyabove previously determined values. The first 8 cases ineach arm with valid g/d values, in which data collectionhad continued above 320 days, are depicted in Supple-mentary Fig. S3.

Estimating the time to death, had treatment continuedbeyond the arbitrary definition of progression

Patients (with known DOD) were divided into 5 groups(quintiles), of increasing g. The highest quintile comprisespatients with no apparent benefit from sunitinib,whose on-study g is likely similar to their off-treatment g. Substitutingmedian g and d values into Eq. (A), using DOD as (t), we

Figure 2. The growth rate constants(log g) correlate (negatively) with OSin patients treated with sunitinib orIFN-a. We extracted the rateconstants from the data, using themodel and then asked whether d or gcorrelates with OS. Top left,treatment with sunitinib. Top right,treatmentwith IFN-a.Weuse loggonthe y axis rather than g itself becausethe dependence of OS on g is ahyperbolic function, not a straightline. The growth rates (g) aresignificantly negatively correlatedwith OS (Rsq ¼ 0.44, P < 0.0001)for sunitinib and less so (Rsq ¼ 0.14,P¼ 0.0002) for IFN-a. The regressionrates (d) have no significantcorrelation with OS (Rsq ¼ 0.04 forsunitinib and IFN-a, both, not shown).Bottom, regressions of tumor nadir(surrogate of the growth rateconstant) with PFS and OS forsunitinib.

Tumor Growth Rate Constants in the Sunitinib Pivotal Trial

www.aacrjournals.org Clin Cancer Res; 18(8) April 15, 2012 2377

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

estimated the relative "tumor size" at time of death. For thehighest g quintile, the median relative tumor size (sum ofLDs, f) at DOD was 2.1-fold that of the median entry value(sum of LDs) for the whole population (for a sphere this

represents a 9.3-fold increase in tumor volume). Assuminggroups of patients die with similar tumor burdens (somereaching this faster than others), we used 2.1-fold the entryvalue as the tumor quantity at death. In Fig. 6, we usedmedian g and d values, in quintiles, for sunitinib-treatedpatients, to determine the time at which the projectedtumor growth curve intersects a final expected tumor quan-tity of 2.1-fold. For the slowest growing quintile (largestnegative log g value), in which most patients showedonly regression while on study, the model projects notumor growth, although median OS was 516 days (We didnot depict this quintile because the g values of nearly zerowould give a large error in tumor growth projections). Inthe next 2 quintiles (A and B), the projected OS based onthe on-study g and d values is considerably longer thanactual OS. Our projected tumor curve assumes a constantg until the 2.1-fold tumor quantity is reached, so theshorter actual OS implies that g increased after discontin-uation of drug. We hypothesize that patients with slow gvalues could have lived closer to the predicted OS hadtreatment continued beyond the conventional "20% abovenadir" threshold. In C, the second fastest growing quintile,predicted OS is 377 days, close to the 322 found, suggestingthe on-study g accurately predicts the g when therapyceases, perhaps indicating drug resistant tumor. For the

Figure 3. Left, waterfall plot depicting on the y axis, the percentage decrease at 12 weeks in sum of the LDs from baseline values [as opposed to the bestresponse assessment reported by Motzer and colleagues (3)]. Cases are stratified in terms of decreasing values of the change in the sum of LDs.Right, the y axis is the logarithm of the derived growth rate constant (log g). The horizontal lines in each set are themedian values and the 25%and 75% limits.The growth rate constants are shown from left to right for patients with RCC enrolled on the NCI placebo versus bevacizumab study (15), the NCI ixabepilonestudy (16), and the 2 arms of the sunitinib phase III study (3, 13). Using data from study investigators, the median g was 0.0015 per day and medianlog gwas�2.81 (25%–75%:�2.38 to�3.54) for IFN-a–treated patients comparedwith amediang of 0.00082 per day andmedian logg of�3.09 (25%–75%:�2.70 to �3.46) for the sunitinib-treated cohort, significantly different at P < 0.0001. The medians and spread of the g values are hardly different fordata on tumor sizes measured by central review (�2.78, 25%–75%: �2.44 to �3.23 for IFN-a and �2.94, 25%–75%: �2.66 to �3.16 for sunitinib). Thedifference between the median log g values for bevacizumab and for sunitinib almost reaches significance at P ¼ 0.053. The median log g values forixabepilone (�2.74) and IFN-a (�2.85) do not differ significantly. IR, data from central (independent) review; INV, data from study investigator measurements.

Regressoin

r = 0.892Rsq = 0.7956

Slope = 1.143 ± 0.40

Lo

g g

(in

vest

igat

or)

Log g (independent review)

−−1.6

−2.4

−3.2

−4.0

−3.6 −3.2 −2.8 −2.4

Line of identity

Figure 4. Tumor growth rate constants using 2 sources of sum of LDare similar. Tumor growth rate constants determined with radiologicalmeasurements obtained by either the study investigators (y axis)or central review (x axis) were highly correlated. The regression isy ¼ 1.11 (�0.04) � x þ 0.23 (þ 0.13) (Rsq ¼ 0.80, P < 0.0001, n ¼ 205).

Stein et al.

Clin Cancer Res; 18(8) April 15, 2012 Clinical Cancer Research2378

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

fastest quintile (D), the predicted OS of 170 days is, ofcourse, not different from the 174 days observed, as thisobserved OS was used to derive the value of 2.1, the fold-increase in the sum of LDs at death.

DiscussionIn this study, using data from the pivotal trial of sunitinib

versus IFN-a (3, 13), we show that analysis of data routinelygathered during a clinical trial can provide estimates of therates of tumor regression (d) and growth (g). The growthrate constant is an excellent "surrogate" for OS, correlatingsignificantlywithOS (P < 0.001);d correlates less well. Bothg andd can be extracted, and tumor response characterized,even before the nadir is reached. Sunitinib results in asignificantly slower rate of tumor growth (P < 0.0001) ascomparedwith IFN-a, the sunitinib rate being almost better(P ¼ 0.053) than that achieved with bevacizumab in asmaller randomized trial (15). The median g of tumors inpatients receiving sunitinib (0.00082 per day; log g ¼�3.09)was about half that (P < 0.001) of tumors in patientsreceiving IFN-a (0.0015 per day; log g ¼ �2.81). This isconsistent with the reported best response rates determinedby RECIST, 47% for sunitinib, compared with 12% for IFN-a (3, 13). The correlation of gwith OS observed in patientsrandomized to sunitinib is consistent with the strong cor-relation of gwithOS that has been observed in our previousanalyses (9–12; and in several additional histologies cur-rently under study3), but was less so in those randomizedto-

IFN-a. This may be due to responses in patients on the IFN-a arm who went on to receive sunitinib after diseaseprogression. Although more patients had g/d values deter-mined than had PFS scored (319 vs. 204 for sunitinib and240 vs. 186 for IFN-a), one caveat is worth noting. Patientson the IFN-a arm were more likely to have "unacceptable"g values (63 had only 2 evaluations with <1.2-fold differ-ence) than those on sunitinib (26 had only 2 evaluationswith <1.2-fold difference; Supplementary Fig. S1), poten-tially introducing the same type of bias that can occur inKaplan–Meier projections when patients are censoredbefore progression.

We envision the g value to be particularly useful as aclinical trial endpoint that could provide an independentanalysis of drug efficacy. Using SigmaPlot’s size function inits statistics package, and the SE of 0.0019 per day found forthe g values in the sunitinib arm, 100 cases in each of 2 armswould be needed in some future trial to find a decrease of50% in the mean of g with a power of 0.85 at P ¼ 0.05. Wealso compared tumor measurements determined by studyinvestigators and by central review and observed that,despite absolute differences in measurements, the rate ofchange of these measurements—that is, growth rate, g—isnearly identical, and their correlation with OS indistin-guishable. These analyses are in agreement with priorreports of overall concordance of independent and inves-tigator review and suggestions that such reviewdoes not addvalue (17, 18). As an efficacy endpoint, g could supportinvestigator assessments andhelp eliminate need for centralreview.

Despite the overall correlation of on-study g with OS inthe sunitinib arm, we could model a change in growth rate3Multiple myeloma, and breast and thyroid cancer (work in progress).

Figure 5. The g and d parameterscan be extracted from the data withgood accuracy even before the nadirvalue is reached. The 8 panels on theleft show successive data sets fromone patient, obtained over time. Thetumor quantity is plotted with thestarting quantity arbitrarily set at 1.The panels on the right plot the g andd parameters extracted using Eq. A,each with its SD. The g and d valuesobtained from the secondreassessment onward are plotted.By the fourth evaluation, bothg anddcan be obtained with accuracy; latervalues differ little. Note that a g valuecan be accurately extracted longbefore tumor measurements showgrowth: the downward trajectory is"deviated upward" by the growingfraction. The greater the growth rate,the greater the deviation, allowingone to calculate g long before actualgrowth is measured.

Tumor Growth Rate Constants in the Sunitinib Pivotal Trial

www.aacrjournals.org Clin Cancer Res; 18(8) April 15, 2012 2379

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

after study discontinuation in some patients who hadmarked slowing of growth while on study. Using the OSof enrolled patients and the derived g and d, we estimated arelative tumor size at death 2.1-fold that at the time ofenrollment (9.3-fold increase in tumor volume) in patientswho did not benefit from therapy. This value is similar tothat determined in a previous mRCC analysis (11.5-foldincrease in tumor volume). On the basis of growth ratesdetermined while on study, patients with slower on-study gvalues should have lived longer. The model suggests tumorgrowth rates increased after treatment was discontinued, toapproximate that of a placebo rate [faster than the rate ontherapy, but one that would not be characterized as "accel-eration" (19)]. We found no evidence that g accelerated astreatment continued. As shown in Fig. 5 and SupplementaryFig. S3, on-study g did not rise appreciably—indeed at thetime of treatment discontinuation, the value of g wascomparable with that obtained months earlier.

An important consequence of the survival projectionfollows: continuation of sunitinib treatment beyond studycriteria for progression might have extended survival sub-stantially in patients whose g values indicated substantialslowing of tumor growth. For example, for the 30% ofpatients with the slowest calculable g value, the modelpredicts survival to 1,365 days (to reach 2.1-relative size),while the actual median OS and 25% to 75% limit was 633(523–777) days. Because we lack curative therapies, usingthe g value to predict who might benefit from continuedtreatment, provided it is clinically feasible and tolerable,

offers the possibility of prolonging survival, in the absenceof good salvage or curative therapies—a hypothesis thatneeds to be prospectively tested.

Several caveats should be noted about the survivalanalysis. First, the survival modeling was carried out onlyin patients who had died, rather than the entire studypopulation; once the study was closed, collection ofsurvival data ceased. Thus, a survival benefit due toextended therapy may be only seen in a subgroup withmore advanced disease—the measured parameters inSupplementary Table S1 certainly suggest that the patientsin the trial population who had died had more advanceddisease, although the analysis of g in the 2 groups inSupplementary Fig. S2 suggests dependence of survival ongrowth rate was similar whether patients were living orhad died. Second, because many patients receiving IFNeventually received other therapies including sunitiniband other VEGF inhibitors, this may have reduced thesurvival difference between the 2 arms. In a similar way,the multiple lines of salvage therapy that are now avail-able for renal carcinoma could confound the interpreta-tion of a study aimed at proving the benefit of extendingsunitinib therapy in renal cancer.

Finally, we would observe that in mRCCwe have enteredthe era of "targeted therapies" leaving behind IL-2 and IFN-a, without understandingwhobenefited andwhy (20).Ouranalysis concurs with a recent review that concluded "gen-uine, if modest, effectiveness" of IFN-a in mRCC (21), inthat there were a number of patients in the IFN-a cohort

Figure 6. Predicted OS if therapywere continued after theconventional definition of diseaseprogression (nadir þ 20%).Entering the study's derivedmedianganddparameters intoEq.(A), the equation canbe solved for apredicted time of death whentumor quantity, f, reaches 210%the on-study value (this being thetumor quantity relative to t ¼ 0,estimated for the 20% of patientswith the fastest g values,i.e., fastest quintile). The panelsdepict the predicted time course oftumor quantity in patients treatedwith sunitinib divided into quintilesaccording to their on-study gvalues. The vertical line and grayarrow above indicate the actualdate of death, while the gray arrowto the right indicates the predictedOS had the treatment continuedbeyond the conventional 20%above nadir threshold.

Stein et al.

Clin Cancer Res; 18(8) April 15, 2012 Clinical Cancer Research2380

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

with very slow growth rates—although proof that thiscould be ascribed to IFN-a requires pretreatment tumormeasurements.In summary, we show that a tumor’s growth rate

constant can be reliably calculated from clinical datagathered during a randomized phase III trial and thatthis value can reliably predict the FDA gold standard, OS.We confirm the efficacy of sunitinib versus IFN-a andcompare it with other agents used in mRCC. A similaranalysis of data from other trials could reliably compareall existing therapies for mRCC. As regards individualpatients, we show that very early in treatment, long beforedisease progression is scored, indeed before tumor growthis observed, one can determine a growth rate constant.This knowledge enables a reliable estimate of the pre-dicted OS, which could be used in clinical trial designs tocontinue therapy past the arbitrary 20% progression in

those with very slow g values. These results validate ourprevious analyses of the growth rate constant in renal andprostate cancer and will hopefully stimulate additionalinvestigations of this novel clinical trial endpoint.

Disclosure of Potential Conflicts of InterestS.T. Kim: stock, Pfizer. R.J. Motzer received commercial research support

and is a consultant on the advisory board of Pfizer. No other potentialconflicts of interest were disclosed by the other authors.

Grant SupportThis work was supported by NIH Intramural Research Program.The costs of publication of this article were defrayed in part by the

payment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received September 2, 2011; revised January 8, 2012; accepted January 27,2012; published OnlineFirst February 17, 2012.

References1. American Cancer Society: Cancer facts & figures. Tables & figures;

2010. Available from: http://www.cancer.org/Research/CancerFacts-Figures/CancerFactsFigures/most-requested-tables-figures-2010

2. EscudierB, Eisen T, StadlerWM,SzczylikC,OudardS,SiebelsM, et al.Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med2007;356:125–34.

3. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM,Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cellcarcinoma. N Engl J Med 2007;356:115–24.

4. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al.Temsirolimus, interferon alfa, or both for advanced renal-cell carcino-ma. N Engl J Med 2007;356:2271–81.

5. EscudierB,PluzanskaA,Koralewski P,RavaudA,BracardaS,SzczylikC, et al. Bevacizumab plus interferon alfa-2a for treatment of meta-static renal cell carcinoma: a randomised, double-blind phase III trial.Lancet 2007;370:2103–11.

6. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S,et al. Efficacy of everolimus in advanced renal cell carcinoma: adouble-blind, randomised, placebo-controlled phase III trial. Lancet2008;372:449–56.

7. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E,Wagstaff J, et al.Pazopanib in locally advanced or metastatic renal cell carcinoma:results of a randomized phase III trial. J Clin Oncol 2010;28:1061–8.

8. Larkin J, GoreM. Is advanced renal cell carcinomabecoming a chronicdisease? Lancet 2010;376:574–5.

9. SteinWD, Yang J, Bates SE, Fojo T. Bevacizumab reduces the growthrate constants of renal carcinomas: a novel algorithm suggests earlydiscontinuation of bevacizumab resulted in a lack of survival advan-tage. Oncologist 2008;13:1055–62.

10. Stein WD, Figg WD, Dahut W, Stein AD, Hoshen MB, Price D, et al.Tumor growth rates derived from data for patients in a clinical trialcorrelate strongly with patient survival: a novel strategy for evaluationof clinical trial data. Oncologist 2008;13:1046–54.

11. Stein WD, Huang H, Menefee M, Edgerly M, Kotz H, Dwyer A, et al.Other paradigms: growth rate constants and tumor burden determinedusing computed tomography data correlate strongly with the overallsurvival of patients with renal cell carcinoma. Cancer J 2009;15:441–7.

12. Stein WD, Gulley J, Schlom J, Madan RA, Dahut W, Figg WD, et al.Tumor regression and growth rates determined in five intramural NCI

prostate cancer trials: the growth rate as an indicator of therapeuticefficacy. Clin Cancer Res 2011;17:907–14.

13. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM,Oudard S, et al. Overall survival and updated results for sunitinibcompared with IFN-alfa in patients with metastatic renal cell carcino-ma. J Clin Oncol 2009;27:3584–90.

14. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,Rubinstein L, et al. New guidelines to evaluate the response totreatment in solid tumors. European Organization for Research andTreatment of Cancer, National Cancer Institute of the United States,National Cancer Institute of Canada. J Natl Cancer Inst 2000;92:205–16.

15. Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ,Topalian SL, et al. A randomized trial of bevacizumab, an anti-vascularendothelial growth factor antibody, for metastatic renal cancer. N EnglJ Med 2003;349:427–34.

16. Huang H, Menefee M, Edgerly M, Zhuang S, Kotz H, Poruchynsky M,et al. A phase II clinical trial of ixabepilone (Ixempra; BMS-247550;NSC710428), an epothilone B analog, in patients with metastatic renal cellcarcinoma. Clin Cancer Res 2010;16:1634–41.

17. Dodd LE, EKorn EL, Freidlin B, JaffeCC, Rubinstein LV, Dancey J, et al.Blinded independent central review of progression-free survival inphase III clinical trials: important design element or unnecessaryexpense? J Clin Oncol 2008;26:3791–6.

18. Gray R, Bhattacharya S, Bowden C, Miller K, Comis RL. Independentreview of E2100: a phase III trial of bevacizumab plus paclitaxel versuspaclitaxel in women with metastatic breast cancer. J Clin Oncol2009;27:4966–72.

19. Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG,Kerbel RS. Accelerated metastasis after short-term treatment witha potent inhibitor of tumor angiogenesis. Cancer Cell 2009;15:232–9.

20. McDermott DF. Update on the application of interleukin-2 in thetreatment of renal cell carcinoma. Clin Cancer Res 2007;13:716s–720s.

21. Canil C, Hotte S, Mayhew LA, Waldron TS, Winquist E. IFN-alfa in thetreatment of patients with inoperable locally advanced or metastaticrenal cell carcinoma: a systematic review. Can Urol Assoc J 2010;4:201–8.

Tumor Growth Rate Constants in the Sunitinib Pivotal Trial

www.aacrjournals.org Clin Cancer Res; 18(8) April 15, 2012 2381

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275

2012;18:2374-2381. Published OnlineFirst February 17, 2012.Clin Cancer Res   Wilfred D. Stein, Julia Wilkerson, Sindy T. Kim, et al.   Regression and GrowthRenal Cell Carcinoma, Using a Method That Assesses Tumor

inαAnalyzing the Pivotal Trial That Compared Sunitinib and IFN-

  Updated version

  10.1158/1078-0432.CCR-11-2275doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2012/02/17/1078-0432.CCR-11-2275.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/18/8/2374.full#ref-list-1

This article cites 20 articles, 9 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/18/8/2374.full#related-urls

This article has been cited by 8 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/18/8/2374To request permission to re-use all or part of this article, use this link

on April 30, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst February 17, 2012; DOI: 10.1158/1078-0432.CCR-11-2275