[Skip to Navigation]
Sign In
Visual Abstract. Effect of Doxorubicin + Olaratumab vs Doxorubicin + Placebo on Survival in Advanced Soft Tissue Sarcomas
Effect of Doxorubicin + Olaratumab vs Doxorubicin + Placebo on Survival in Advanced Soft Tissue Sarcomas
Figure 1.  Flow of Patient Disposition in the Total Soft Tissue Sarcoma Population
Flow of Patient Disposition in the Total Soft Tissue Sarcoma Population

PDGFR indicates platelet-derived growth factor receptor.

aIndividuals assessed for study eligibility were excluded prior to randomization for any reason other than those prespecified.

bA total of 106 patients received 1 dose or more of olaratumab monotherapy.

cStudy treatment in either group was discontinued for any reason other than those prespecified.

dThe 245 patients who received postdiscontinuation follow-up includes the 1 patient who did not receive intervention due to worsening clinical status preceding death.

eThe 236 patients who received postdiscontinuation follow-up includes the 2 patients who did not receive intervention due to worsening clinical status preceding death.

Figure 2.  Overall Survival and Progression-Free Survival in the Total Soft Tissue Sarcoma (STS) and Leiomyosarcoma (LMS) Populations
Overall Survival and Progression-Free Survival in the Total Soft Tissue Sarcoma (STS) and Leiomyosarcoma (LMS) Populations

The curves stop when the number of patients was less than 15%. Tick marks on the curves denote censored observations. In the doxorubicin + placebo and doxorubicin + olaratumab treatment groups, respectively, the median overall survival (interquartile range [IQR]) was 19.7 months (IQR, 9.9-35.5) vs 20.4 months (IQR,10.4-not estimated) in the total STS population and 21.9 months (IQR, 10.8-32.3) vs 21.6 months (IQR, 11.2-not estimated) in the LMS population; the median progression-free survival was 6.8 months (IQR, 2.6-10.7) vs 5.4 months (IQR, 1.4-10.2) in the total STS population and 6.9 months (IQR, 3.1-9.8) vs 4.3 months (IQR, 1.4-11.1) in the LMS population. The median length of follow-up was 31 months in both treatment groups.

Figure 3.  Overall Survival in Prespecified Subgroups in the Total Soft Tissue Sarcoma (STS) Population
Overall Survival in Prespecified Subgroups in the Total Soft Tissue Sarcoma (STS) Population

ECOG indicates Eastern Cooperative Oncology Group; PDGFR, platelet-derived growth factor receptor.

Hazard ratios and 95% CIs (Wald) were estimated using stratified Cox model for overall and unstratified Cox models for subgroups.

Table 1.  Baseline Patient Characteristics of the Total Soft Tissue Sarcoma Population
Baseline Patient Characteristics of the Total Soft Tissue Sarcoma Population
Table 2.  Adverse Events by Treatment Occurring in ≥5% of Patients in the Doxorubicin Plus Olaratumab Group
Adverse Events by Treatment Occurring in ≥5% of Patients in the Doxorubicin Plus Olaratumab Group
1.
Lahat  G, Tuvin  D, Wei  C,  et al.  New perspectives for staging and prognosis in soft tissue sarcoma.   Ann Surg Oncol. 2008;15(10):2739-2748. doi:10.1245/s10434-008-9970-6 PubMedGoogle ScholarCrossref
2.
Sbaraglia  M, Dei Tos  AP.  The pathology of soft tissue sarcomas.   Radiol Med. 2019;124(4):266-281. doi:10.1007/s11547-018-0882-7PubMedGoogle ScholarCrossref
3.
Seddon  B, Strauss  SJ, Whelan  J,  et al.  Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomised controlled phase 3 trial.   Lancet Oncol. 2017;18(10):1397-1410. doi:10.1016/S1470-2045(17)30622-8 PubMedGoogle ScholarCrossref
4.
Judson  I, Verweij  J, Gelderblom  H,  et al; European Organisation and Treatment of Cancer Soft Tissue and Bone Sarcoma Group.  Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: a randomised controlled phase 3 trial.   Lancet Oncol. 2014;15(4):415-423. doi:10.1016/S1470-2045(14)70063-4 PubMedGoogle ScholarCrossref
5.
Ryan  CW, Merimsky  O, Agulnik  M,  et al.  PICASSO III: a phase iii, placebo-controlled study of doxorubicin with or without palifosfamide in patients with metastatic soft tissue sarcoma.   J Clin Oncol. 2016;34(32):3898-3905. doi:10.1200/JCO.2016.67.6684 PubMedGoogle ScholarCrossref
6.
Tap  WD, Papai  Z, Van Tine  BA,  et al.  Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial.   Lancet Oncol. 2017;18(8):1089-1103. doi:10.1016/S1470-2045(17)30381-9 PubMedGoogle ScholarCrossref
7.
Papadopoulos  N, Lennartsson  J.  The PDGF/PDGFR pathway as a drug target.   Mol Aspects Med. 2018;62:75-88. doi:10.1016/j.mam.2017.11.007 PubMedGoogle ScholarCrossref
8.
Lartruvo. Highlights of prescribing information. Indianapolis, IN: Eli Lilly and Co; 2018. Accessed October 15, 2019. http://pi.lilly.com/us/lartruvo-uspi.pdf
9.
Tap  WD, Jones  RL, Van Tine  BA,  et al.  Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase 1b and randomised phase 2 trial.   Lancet. 2016;388(10043):488-497. doi:10.1016/S0140-6736(16)30587-6 PubMedGoogle ScholarCrossref
10.
Eisenhauer  EA, Therasse  P, Bogaerts  J,  et al.  New Response Evaluation Criteria in Solid Tumours: revised RECIST guideline (version 1.1).   Eur J Cancer. 2009;45(2):228-247. doi:10.1016/j.ejca.2008.10.026 PubMedGoogle ScholarCrossref
11.
Jones  RL, Mo  G, Baldwin  JR,  et al.  Exposure-response relationship of olaratumab for survival outcomes and safety when combined with doxorubicin in patients with soft tissue sarcoma.   Cancer Chemother Pharmacol. 2019;83(1):191-199. doi:10.1007/s00280-018-3723-4 PubMedGoogle ScholarCrossref
12.
Holzer  TR, O’Neill Reising  L, Credille  KM, Schade  AE, Oakley  GJ.  Variability in platelet-derived growth factor receptor alpha antibody specificity may impact clinical utility of immunohistochemistry assays.   J Histochem Cytochem. 2016;64(12):785-810. doi:10.1369/0022155416673979 PubMedGoogle ScholarCrossref
13.
Maurer  W Betz  F.  Multiple testing in group sequential trials using graphical approaches.   Stat Biopharm Res. 2013;5(4):311-320. doi:10.1080/19466315.2013.807748 Google ScholarCrossref
14.
Cox  DR.  Regression models and life-tables.   J R Stat Soc Series B Stat Methodol. 1972;34(2):187-220. doi:10.1111/j.2517-6161.1972.tb00899.x Google Scholar
15.
Schoenfeld  D.  Partial residuals for the proportional hazards regression model.   Biometrika. 1982;69:239-241. doi:10.1093/biomet/69.1.239 Google ScholarCrossref
16.
Jones  RL, Wagner  A, Kawai  A,  et al.  1671O Assessment of cardiotoxicity (CT) associated with doxorubicin (dox) in patients (pts) with advanced soft tissue sarcoma (STS) in a phase III randomized trial.   Ann Oncol. 2019;30(Suppl 5). doi:10.1093/annonc/mdz283.004Google Scholar
17.
Croswell  JM, Ransohoff  DF, Kramer  BS.  Principles of cancer screening: lessons from history and study design issues.   Semin Oncol. 2010;37(3):202-215. doi:10.1053/j.seminoncol.2010.05.006 PubMedGoogle ScholarCrossref
18.
Kramer  BS, Croswell  JM.  Cancer screening: the clash of science and intuition.   Annu Rev Med. 2009;60:125-137. doi:10.1146/annurev.med.60.101107.134802 PubMedGoogle ScholarCrossref
Original Investigation
April 7, 2020

Effect of Doxorubicin Plus Olaratumab vs Doxorubicin Plus Placebo on Survival in Patients With Advanced Soft Tissue Sarcomas: The ANNOUNCE Randomized Clinical Trial

Author Affiliations
  • 1Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
  • 2Department of Medicine, Weill Cornell Medical College, New York, New York
  • 3Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
  • 4University Hospitals Leuven, Department of General Medical Oncology, Leuven Cancer Institute, Leuven, Belgium
  • 5Medical Oncology Department in University Hospital Virgen del Rocio and Institute of Biomedicine of Sevilla (IBIS) (HUVR, CSIC, University of Sevilla), Sevilla, Spain
  • 6University of Copenhagen, Copenhagen, Denmark
  • 7Stanford University, Stanford, California
  • 8Division of Medical Oncology, Center for Immuno-oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
  • 9National Yang-Ming University School of Medicine, Taipei, Taiwan
  • 10Princess Margaret Cancer Center, Toronto, Ontario, Canada
  • 11Virginia Cancer Specialists, Fairfax
  • 12National Cancer Center Hospital, Tokyo, Japan
  • 13Institut Gustave Roussy, Villejuif, France
  • 14Department of Internal Medicine, Washington University in St Louis School of Medicine, St Louis, Missouri
  • 15National Cancer Center Hospital East, Kashiwa, Japan
  • 16Sungkyunkwan University Samsung Medical Center, Seoul, Republic of Korea
  • 17Blokhin Cancer Research Center, Moscow, Russian Federation
  • 18Magyar Honvedseg Eu Kp, Budapest, Hungary
  • 19Eli Lilly and Company, Indianapolis, Indiana
  • 20Eli Lilly and Company, Windlesham, United Kingdom
  • 21Sarcoma Unit, Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
JAMA. 2020;323(13):1266-1276. doi:10.1001/jama.2020.1707
Key Points

Question  In patients with advanced soft tissue sarcoma (STS), does the addition of olaratumab to doxorubicin improve overall survival?

Findings  In this randomized clinical trial of 509 adults with advanced STS, there was no significant difference in overall survival between patients who received doxorubicin plus olaratumab and patients who received doxorubicin plus placebo in the total STS and leiomyosarcoma populations (total STS: hazard ratio, 1.05; median, 20.4 vs 19.7 months; leiomyosarcoma: hazard ratio, 0.95; median, 21.6 vs 21.9 months).

Meaning  There was no significant difference in overall survival with the addition of olaratumab to doxorubicin in patients with advanced STS.

Abstract

Importance  Patients with advanced soft tissue sarcoma (STS) have a median overall survival of less than 2 years. In a phase 2 study, an overall survival benefit in this population was observed with the addition of olaratumab to doxorubicin over doxorubicin alone.

Objective  To determine the efficacy of doxorubicin plus olaratumab in patients with advanced/metastatic STS.

Design, Setting, and Participants  ANNOUNCE was a confirmatory, phase 3, double-blind, randomized trial conducted at 110 sites in 25 countries from September 2015 to December 2018; the final date of follow-up was December 5, 2018. Eligible patients were anthracycline-naive adults with unresectable locally advanced or metastatic STS, an Eastern Cooperative Oncology Group performance status of 0 to 1, and cardiac ejection fraction of 50% or greater.

Interventions  Patients were randomized 1:1 to receive doxorubicin, 75 mg/m2 (day 1), combined with olaratumab (n = 258), 20 mg/kg in cycle 1 and 15 mg/kg in subsequent cycles, or placebo (n = 251) on days 1 and 8 for up to 8 21-day cycles, followed by olaratumab/placebo monotherapy.

Main Outcomes and Measures  Dual primary end points were overall survival with doxorubicin plus olaratumab vs doxorubicin plus placebo in total STS and leiomyosarcoma (LMS) populations.

Results  Among the 509 patients randomized (mean age, 56.9 years; 58.2% women; 46.0% with LMS), all were included in the primary analysis and had a median length of follow-up of 31 months. No statistically significant difference in overall survival was observed between the doxorubicin plus olaratumab group vs the doxorubicin plus placebo group in either population (total STS: hazard ratio, 1.05 [95% CI, 0.84-1.30], P = .69, median overall survival, 20.4 months vs 19.7 months; LMS: hazard ratio, 0.95 [95% CI, 0.69-1.31], P = .76, median overall survival, 21.6 months vs 21.9 months). Adverse events of grade 3 or greater reported in 15% or more of total patients with STS were neutropenia (46.3% vs 49.0%), leukopenia (23.3% vs 23.7%), and febrile neutropenia (17.5% vs 16.5%).

Conclusions and Relevance  In this phase 3 clinical trial of patients with advanced STS, treatment with doxorubicin plus olaratumab vs doxorubicin plus placebo resulted in no significant difference in overall survival. The findings did not confirm the overall survival benefit observed in the phase 2 trial.

Trial Registration  ClinicalTrials.gov Identifier: NCT02451943

Introduction

Soft tissue sarcomas (STSs) are rare, heterogeneous malignancies of connective tissues that arise from mesenchymal precursors most often in the extremities and comprise about 1% of cancers in adults.1,2 The most common of the more than 50 biologically distinct histologies identified are leiomyosarcoma (LMS), liposarcoma (LPS), and undifferentiated pleomorphic sarcoma (UPS).1,2 Standard first-line systemic therapy for advanced disease includes doxorubicin alone or in combination. Recent first-line phase 3 trials comparing doxorubicin monotherapy with gemcitabine plus docetaxel3 or doxorubicin plus ifosfamide,4 palifosfamide,5 or evofosfamide6 did not improve historic median overall survival of about 14 to 19 months and 2-year survival rates of 20% to 30%. Consequently, there is a need for new, effective treatments.

Platelet-derived growth factor receptors (PDGFRs) are homodimers and heterodimers of alpha and beta isoforms. The PDGF-PDGFR signaling pathway is active in mesenchymal stem cell differentiation.7 Dysfunction of this pathway has been observed in STS.7 Olaratumab, a recombinant, fully human IgG subclass 1 monoclonal antibody, binds to PDGFR-α and blocks downstream signaling induced by ligands PDGF-AA, -BB, and -CC.8 In a randomized, open-label, phase 2 trial, up to 8 cycles of doxorubicin plus olaratumab (doxorubicin + olaratumab) followed by olaratumab monotherapy improved median progression-free survival (PFS) by 2.5 months and median overall survival by 11.8 months, vs single-agent doxorubicin, in patients with advanced STS.9 In the LMS subgroup, median overall survival was 15.1 months longer for doxorubicin + olaratumab over doxorubicin alone.9 Based on these results, olaratumab + doxorubicin received regulatory approvals in many regions worldwide, including the United States, Europe, Canada, Korea, and Brazil. Most approvals were granted under the condition that the sponsor conduct a confirmatory phase 3 study.

The phase 3 ANNOUNCE trial was a randomized, placebo-controlled study of doxorubicin with or without olaratumab in patients with locally advanced or metastatic STS that aimed to confirm the results of the phase 2 trial.

Methods
Trial Oversight

This trial was conducted at 110 sites in 25 countries from September 2015 to December 2018. The protocol was approved by the institutional review board at each participating center, and the study was performed in accordance with the International Conference on Harmonization Guidelines for Good Clinical Practice and the Declaration of Helsinki. All patients provided written informed consent prior to any trial procedures. Interim safety analyses were performed with regular frequency by an independent data monitoring committee. One blinded interim efficacy analysis, without formal stopping rules due to the confirmatory nature of the trial, was performed. The complete trial protocol is available in Supplement 1.

Patients

Patients aged 18 years or older were eligible if they had locally advanced or metastatic STS not amenable to curative treatment. Patients with grade I LPS were eligible only at the time of radiologic progression or with histological evidence of aggressive transformation. Patients were required to have evaluable disease by Response Evaluation Criteria in Solid Tumours (RECIST) 1.110; an Eastern Cooperative Oncology Group performance status (ECOG PS) of 0 or 1; no prior anthracycline treatment; available tumor tissue for central review; adequate hematologic, organ, and coagulation function; a cardiac ejection fraction of 50% or greater; a negative pregnancy test result; agreed to use appropriate contraceptive methods; and a life expectancy of at least 3 months. Histological review of submitted tissue samples was centrally performed but not required prior to randomization.

Patients were ineligible if they had Kaposi sarcoma or gastrointestinal stromal tumor, a prolonged QTcB (>450 milliseconds for males and >470 milliseconds for females calculated by the Bazett formula: QTc = QT [heart rate/60]1/2 = QT [duration of ventricular cardiac cycle]–1/2), untreated central nervous system metastases, prior radiotherapy to the mediastinum/pericardium or whole pelvis, other active cancer, planned elective or cancer surgery during study treatment, uncontrolled intercurrent illnesses, known allergy to any study treatments, or if breastfeeding.

Trial Design and Interventions

Eligible patients were randomized 1:1 by Interactive Web Response System to receive doxorubicin + olaratumab or doxorubicin plus placebo (doxorubicin + placebo). Randomization was stratified by number of prior systemic therapies for advanced or metastatic disease (none vs ≥1), histology (LMS vs LPS vs UPS vs other subtypes), ECOG PS (0 vs 1), and geographic region (North America vs Europe vs rest of world). Race/ethnicity data, as self-reported by participants through fixed categories on a study case report form, were collected for description of the study population. Patients, medical staff, investigators, and the sponsor were blinded to treatment. Treatment consisted of 21-day cycles of olaratumab or placebo (days 1 and 8) in combination with doxorubicin 75 mg/m2 (day 1) for up to 8 cycles, followed by olaratumab or placebo monotherapy. The olaratumab dose was 20 mg/kg in cycle 1 and 15 mg/kg in subsequent cycles. The olaratumab loading dose was selected to achieve steady-state olaratumab serum levels within the first cycle and to minimize the number of patients with trough concentration at the end of cycle 1 (Cmin1) below the level associated with efficacy in the phase 2 trial.11 Premedication with antihistamines and corticosteroids for olaratumab/placebo was recommended at study open and subsequently required by protocol amendment due to the risk of severe infusion-related reactions. The cardioprotectant dexrazoxane was allowed at investigator discretion prior to doxorubicin administration starting at cycle 1 and recommended starting with cycle 5.

Cardiac evaluation by echocardiogram or multigated acquisition scan was performed after 4, 6, and 8 cycles, then every 3 months for the first year, every 6 months for the second year, and annually thereafter. Tumor assessments were performed by computed tomography or magnetic resonance imaging scan every 6 weeks using RECIST 1.1 until tumor progression. Patients continued treatment until progressive disease, unacceptable toxicity, patient/physician decision, or significant nonadherence. If doxorubicin was discontinued for toxicity or reasons other than disease progression prior to cycle 8, blinded olaratumab/placebo monotherapy was permitted to continue at that time.

End Points and Assessments

Dual primary end points of overall survival (time from randomization to death from any cause) compared doxorubicin + olaratumab vs doxorubicin + placebo in (1) all randomized patients (total STS population) and in (2) the subset of randomized patients with LMS.

Secondary objectives tested in type I error–controlled fashion were PFS (determined by investigator assessment according to RECIST 1.1)10 and objective response rate (ORR), defined as the proportion of patients achieving best overall response of complete response plus partial response. Supportive prespecified secondary objectives are described in eTable 1 in Supplement 2. Those reported here are disease control rate, defined as the proportion of patients achieving best overall response of complete response plus partial response plus stable disease; adverse events (AEs); pharmacokinetics; and patient-reported outcomes assessed by European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 version 3 (EORTC-QLQ-C30) and modified Brief Pain Inventory–Short Form (mBPI-sf) scales. A high score on the 100-point scales of the EORTC-QLQ-C30 represents a high/healthy level of functioning, a high quality of life, or a high level of symptomology/problems. A change of 10 points or more in these categories was considered clinically meaningful. On the 10-point mBPI-sf scale, 0 was the best possible pain score. Time to first worst pain score was defined as an increase from baseline of 2 points or more with no change in analgesic use or an increase from baseline of 1 point or more with an increase in analgesic use of 1 level or more.

For exploratory analyses, tumor tissue was assessed by immunohistochemistry for expression of PDGFR-α12 and PDGFR-β to test for correlation to overall survival and PFS in prespecified (PDGFR-α) and post hoc (PDGFR-β) analyses. Other prespecified exploratory analyses included exposure to doxorubicin, olaratumab, and dexrazoxane and postdiscontinuation therapy received by patients.

Statistical Analysis

Primary and secondary efficacy analyses were performed on an intention-to-treat basis in all patients in the group to which they were randomized, regardless of actual treatment received. Statistical tests for both primary end points of overall survival and key secondary end points of PFS and ORR were conducted according to the graphical method of Maurer and Bretz13 to control the overall type I error rate at 0.025 (1-sided). The overall α was split between the dual primary end points of overall survival in the total STS population (α = .02) and LMS subpopulation (α = .005). A total of 322 overall survival events in the total STS population provided an 80% statistical power at a 1-sided .02 significance level, assuming the true hazard ratio (HR) for overall survival was 0.723. Assumptions regarding effect size were made with input from investigators and global sarcoma experts. Assuming a 30% censoring rate, a sample size of 460 randomized patients was required. The study was designed to be positive if either or both primary end points were met.

For the primary and secondary efficacy analyses conducted using the time-to-event methodology, if an event of interest was not observed, censoring rules were applied (refer to the statistical analysis plan in Supplement 1 for censoring rules). For response analyses (ORR and disease control rate), patients with missing data were considered nonresponders. For health outcomes/quality-of-life analyses, patients with a missing baseline (cycle 1) assessment or without at least 1 subsequent assessment during the study period were excluded. Patients with a baseline worst pain score of 8 or more or missing baseline score were excluded from mBPI-sf analysis.

The overall survival and PFS analyses were based on the stratified log-rank test. The overall survival and PFS curves, medians with 95% CIs and survival rates at various time points for each treatment group, were estimated using the Kaplan-Meier method. The HRs were estimated using a stratified Cox proportional hazards model.14 Unstratified models were used for subgroup analyses related to patient demographics or baseline disease characteristics. The proportional hazards assumption was checked using graphical diagnostics based on the Schoenfeld residuals15 and plots of the log-negative-log of the estimated survival density function vs the log of time. Preplanned subgroup analyses for overall survival are listed in eTable 2 in Supplement 2. Statistical analyses were performed using SAS version 9.1.2 software (SAS Institute).

Results
Patients

Between September 2015 and July 2016, 624 patients provided informed consent to enroll in the trial (Figure 1 [total STS]; eFigure 1 in Supplement 2 [LMS]). Of the patients who were screened for study participation, 115 were ineligible and 509 were randomized to doxorubicin + olaratumab (n = 258) or doxorubicin + placebo (n = 251). One patient randomized to the doxorubicin + olaratumab group and 2 patients randomized to the doxorubicin + placebo group did not receive any study treatment. Baseline characteristics of the total STS population are presented in Table 1. Demographics were well balanced between treatments except for a higher percentage of men randomized to the doxorubicin + olaratumab group than to the doxorubicin + placebo group (44.2% vs 39.4%). Extent of disease was well balanced, with 83.7% of patients in the doxorubicin + olaratumab group and 82.1% of patients in the doxorubicin + placebo group having metastatic disease at randomization. Only 73 patients (28.3%) in the doxorubicin + olaratumab group and 69 patients (27.5%) in the doxorubicin + placebo group received prior systemic therapy. Of these 142 patients, 78 (54.9%) had 1 prior treatment regimen with gemcitabine and docetaxel. Tumor histology and grade were both well balanced between treatments (eTable 3 in Supplement 2). Most patients had LMS or LPS. Twenty-two unique histologies comprised the “other” category. For LMS, the uterus was the primary tumor site in both treatment groups (46 patients [38.7%] in the doxorubicin + olaratumab group, 48 patients [41.7%] in the doxorubicin + placebo group).

Efficacy

No statistically significant difference in overall survival was observed between treatments in the total STS population, with a median overall survival of 20.4 months vs 19.7 months for the doxorubicin + olaratumab (n = 258) and doxorubicin + placebo (n = 251) groups, respectively (HR, 1.05 [95% CI, 0.84-1.30]; P = .69) (Figure 2A). The median length of follow-up was 31 months. Similarly, no survival benefit in the doxorubicin + olaratumab group was observed in the LMS population, with a median overall survival of 21.6 months vs 21.9 months for the doxorubicin + olaratumab (n = 119) and doxorubicin + placebo (n = 115) groups (HR, 0.95 [95% CI, 0.69-1.31]; P = .76) (Figure 2B). Prespecified subgroup analyses did not reveal improvement in overall survival in the doxorubicin + olaratumab group in any notable clinical subgroup (Figure 3).

For the doxorubicin + olaratumab and doxorubicin + placebo groups, respectively, median PFS was 5.4 months vs 6.8 months in the total STS population (Figure 2C) and 4.3 months vs 6.9 months in the LMS population (Figure 2D).

Between the doxorubicin + olaratumab and doxorubicin + placebo groups, respectively, the ORR in the total STS population was 14% (36/258) vs 18.3% (46/251) and in the LMS population was 13.4% (16/119) vs 22.6% (26/115). Disease control rate in the total STS population was 67.4% (174/258) vs 75.7% (190/251) and in the LMS population was 63.0% (75/119) vs 82.6% (95/115). Response rate data are included in eTables 4 and 5 in Supplement 2.

Postdiscontinuation therapies in the total STS population were similar in both treatment groups, apart from more doxorubicin + placebo patients who received radiation after discontinuation from study treatment. The most common postdiscontinuation therapies are included in eTable 6 in Supplement 2.

Consistent with a shorter PFS in the doxorubicin + olaratumab group, doxorubicin exposure was lower in the doxorubicin + olaratumab group than in the doxorubicin + placebo group (median cycles, 6 vs 7 [range, 1-8 in both groups]; median weeks, 18 vs 23; median cumulative dose, 409 mg/m2 vs 483 mg/m2), as was olaratumab or placebo exposure (median cycles, 6 vs 8 [range, 1-53 vs 1-51]; median weeks, 19 vs 26; median cumulative dose, 186 mg/kg vs 250 mg/kg). Olaratumab serum concentrations were comparable with those seen in prior trials. Due to loading dose design, the median Cmin1 was 142.34, and was higher than the Cmin1 from previous olaratumab trials,8 placing patients in a Cmin1 range previously associated with efficacy.

Tumor tissue for PDGFR status was available for analysis in 91% of patients (n = 462 for PDGFR-α, n = 464 for PDGFR-β). Exploratory Cox regression analyses of overall survival dichotomized by PDGFR-α and PDGFR-β tumor status found no association between PDGFR-α or PDGFR-β expression and olaratumab response in the total STS population (eTable 7 and eFigure 2 in Supplement 2 [overall survival by PDGFR-α tumor status, PDGFR-β tumor status not shown]). In the doxorubicin + olaratumab and doxorubicin + placebo groups, respectively, median overall survival by PDGFR tumor status was 17.2 months vs 19.1 months for PDGFR-α positive, 23.6 months vs 21.9 months for PDGFR-α negative, 18.8 months vs 19.9 months for PDGFR-β positive, and 28.3 months vs 20.6 months for PDGFR-β negative. For patients with tumors that were both PDGFR-α and PDGFR-β negative, median overall survival was 28.5 months vs 20.6 months in the doxorubicin + olaratumab and doxorubicin + placebo groups, respectively.

Quality of Life

Of the 398 patients (78.2%) evaluated with EORTC-QLQ-C30 in the total STS population, the median time to first worsening of the Global Health Status was 1.5 months in the doxorubicin + olaratumab group vs 1.8 months in the doxorubicin + placebo group. Of the 417 patients (81.9%) evaluated with mBPI-sf in the total STS population, the median time to first worsening of worst pain score was 7.7 months in the doxorubicin + olaratumab group vs 8.1 months in the doxorubicin + placebo group.

Adverse Events

A total of 506 treated patients (257 in the doxorubicin + olaratumab group, 249 in the doxorubicin + placebo group) were included in the analysis of AEs. Overall, AEs in both treatment groups were consistent with known toxicities of doxorubicin; the only AEs of grade 3 or greater occurring in 10% or more of patients in either group were hematologic toxicities. Table 2 shows all AEs by treatment group that occurred in 5% or more of doxorubicin + olaratumab–treated patients. Febrile neutropenia was the only AE reported as “serious” (as determined by investigator, regardless of grade) that occurred in 5% or more of patients (n = 33 in both groups [12.8% olaratumab, 13.3% placebo]). Infusion-related reactions of immediate hypersensitivity (defined by Standardized Medical Dictionary for Regulatory Activities Queries of anaphylaxis, angioedema, or hypersensitivity) occurred more frequently in doxorubicin + olaratumab–treated patients (any grade, 11.7%; grade ≥3, 2.3%) than in doxorubicin + placebo–treated patients (any grade, 7.2%; grade ≥3, 0.8%).

An assessment of cardiotoxicity associated with doxorubicin treatment in this trial has been presented.16 Cardiac dysfunction of grade 3 or greater was similar in both groups (5 patients [1.9%] in the doxorubicin + olaratumab group, 6 patients [2.4%] in the doxorubicin + placebo group). Median follow-up for cardiac events was 28 weeks. Dexrazoxane was received by 63.0% of doxorubicin + olaratumab–treated patients (initiated at cycle 1 in 39.5%) vs 65.1% of doxorubicin + placebo–treated patients (initiated at cycle 1 in 25.9%). The AEs of cardiac dysfunction and decreased left ventricular ejection fraction are shown in eTable 8 in Supplement 2.

Discussion

This trial failed to demonstrate an overall survival benefit of doxorubicin + olaratumab compared with doxorubicin + placebo in patients with advanced STS. Both treatment groups showed similar toxicity rates, with only infusion-related reactions occurring more frequently in olaratumab-treated patients. These findings considerably differed from the results of the randomized phase 2 trial9 that demonstrated substantial improvement in overall survival with higher but acceptable toxicity rates in the doxorubicin + olaratumab group compared with doxorubicin alone.

No single reason has been identified for the differential outcomes between the phase 2 and 3 trials. No discrepancies in trial conduct or data integrity were noted. If olaratumab has some activity in STS, it is therefore likely that the disparate results were due to the cumulative effect of multiple contributing factors within and between these studies. Possible explanations include inherent differences in design elements between the phase 2 and 3 trials, respectively: open label vs blinded with placebo control, primary end point of PFS vs overall survival, and enrollment limited to the United States vs globally. If olaratumab has no activity in STS, the outcome of the phase 2 trial might be attributed to patients randomized to single-agent doxorubicin who crossed over to olaratumab monotherapy after documented disease progression, possibly creating a negative bias for survival. These patients could have experienced a more prolonged period of progression, through 2 clinical trial events, before moving on to other possibly effective agents. In contrast, patients with disease progression in the doxorubicin + olaratumab group of the blinded phase 3 trial could have moved off study to an agent with known efficacy because crossover of placebo-treated patients was not allowed.

Another potential explanation for the disparate findings between the phase 2 and 3 trials may be the inclusion of differing patient populations. The heterogeneity of STS histologies may have led to differences in outcomes, not only due to unique biology with differential sensitivity to systemic therapy, but also differences in the quality, number, and efficacy of available poststudy treatments, which could affect overall survival. Even a defined histology such as LMS has significant heterogeneity in underlying biology and clinical behavior, encompassing a spectrum of relatively indolent to extremely aggressive diseases, leading to unapparent, or not easily accounted for, but potentially meaningful differences in populations between the trials. Additionally, all patients in the phase 2 study had metastatic disease at enrollment, whereas 17.1% of patients in this trial had locally advanced, nonmetastatic disease. Factors related to enrolled histologies, choice of end point, extent of disease, and clinical behavior are some of the common explanations for why promising phase 2 data are not replicated in phase 3 STS trials; these and others are likely to be causes for the varied outcomes seen in the case of olaratumab in combination with doxorubicin.

This trial had the longest median overall survival in any recent first-line randomized phase 3 STS trial.3-6 Along with these trials,3-6 this study provides additional prospective evidence of the current improvement in median overall survival in advanced STS potentially due to improved multidisciplinary and supportive care and the availability of additional lines of effective systemic therapies for certain sarcoma subtypes. Earlier detection and initiation of treatment of metastatic disease could lead to improvement in measured survival, either through access to more effective therapies or potentially from lead time bias.17,18

Given that doxorubicin is a common denominator in the aforenoted studies3-6 showing longer overall survival for patients with STS, specific consideration should be given to its effect on survival. Supportive care advances, such as dexrazoxane and granulocyte colony-stimulating factor, both of which were received by most patients in this trial, may contribute to clinicians’ willingness to continue doxorubicin beyond the traditional cumulative limit of 450 mg/m2. In this trial, treatment with up to 8 cycles (600 mg/m2) of doxorubicin was allowed and was administered to 90 patients (17.9%), yet grade 3 or more cardiac AEs were uncommon. The relationship between higher doses of doxorubicin and improved overall survival of the entire cohort should be carefully examined in the context of cardiac data as it continues to emerge.

Despite the complexities of conducting studies in rare diseases such as STS, this trial clearly demonstrated that trials can rapidly accrue, enrolling more than 500 patients in 10 months. However, its negative results in conjunction with the previously noted recent negative STS trials3-6 raises the question of whether performing trials recruiting all STS histologies is appropriate or rather if histology-specific or biomarker-driven trials will be required. This trial shows the importance of confirmatory phase 3 trials to validate results from smaller trials and exemplifies the role of accelerated/conditional approvals to provide access to promising medicines for patients with unmet medical needs until the potential of the medicine can be validated, provided there is an acceptable risk profile and plan for rapid validation. Although this study did not show any benefit from the addition of olaratumab to doxorubicin, it similarly did not demonstrate significant increases in drug-related toxicity and confirmed the incremental increase in survival for patients with STS treated with doxorubicin-based therapy. STSs remain an area of unmet medical need and a fertile ground for scientific discovery and drug development, and this trial provides several findings that can be applied to future clinical trials.

Limitations

This study has limitations. First, the mechanism of action of olaratumab is not completely understood. The lack of association between PDGFR-α or PDGFR-β expression and overall survival shown in exploratory analyses presented here and in the phase 2 trial9 are worth noting as possible features of the mechanism of action of olaratumab that could have affected outcomes. These findings, although exploratory, emphasize the need for a better understanding of STS biologies to develop effective treatments.

Second, higher olaratumab doses were not tested prior to this trial; therefore, the dosing regimen studied may not have been maximized.

Conclusions

In this phase 3 clinical trial of patients with advanced STS, treatment with doxorubicin + olaratumab vs doxorubicin + placebo resulted in no significant difference in overall survival. The findings did not confirm the overall survival benefit observed in the phase 2 trial.

Back to top
Article Information

Corresponding Author: William D. Tap, MD, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (tapw@mskcc.org).

Accepted for Publication: February 4, 2020.

Author Contributions: Drs Tap and Wagner had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Both contributed equally to this article.

Concept and design: Tap, Wagner, Schöffski, Krarup-Hansen, Ganjoo, Fedenko, Soldatenkova, Shahir, Mo, Jones.

Acquisition, analysis, or interpretation of data: Tap, Wagner, Schöffski, Martin-Broto, Krarup-Hansen, Ganjoo, Yen, Abdul Razak, Spira, Kawai, Le Cesne, Van Tine, Naito, Park, Pápai, Soldatenkova, Shahir, Mo, Wright, Jones.

Drafting of the manuscript: Tap, Wagner, Krarup-Hansen, Spira, Fedenko, Pápai, Shahir, Mo, Wright, Jones.

Critical revision of the manuscript for important intellectual content: Tap, Wagner, Schöffski, Martin-Broto, Krarup-Hansen, Ganjoo, Yen, Abdul Razak, Spira, Kawai, Le Cesne, Van Tine, Naito, Park, Soldatenkova, Shahir, Mo, Wright, Jones.

Statistical analysis: Soldatenkova, Mo, Wright, Jones.

Administrative, technical, or material support: Tap, Schöffski, Ganjoo, Yen, Naito, Park, Shahir, Mo, Wright, Jones.

Supervision: Tap, Martin-Broto, Krarup-Hansen, Kawai, Van Tine, Mo, Wright, Jones.

Conflict of Interest Disclosures: Dr Tap reported receiving a standard budget from Eli Lilly and Company for site participation for this trial. Apart from the submitted work, Dr Tap reported receiving personal fees from Eli Lilly and Company, EMD Serono, Novartis, Eisai, Janssen, Immune Design, Adaptimmune, Daiichi Sankyo, Blueprint, Loxo, GlaxoSmithKline, Agios Pharmaceuticals, and NanoCarrier. In addition, Dr Tap reported having a patent pending to MSKCC/SKI for Companion Diagnostic for CDK4 inhibitors 14/854,329; participating on an advisory board for and having stock ownership of both Certis Oncology Solutions and Atropos Therapeutics; and consulting for Daiichi Sankyo’s FDA Oncologic Drugs Advisory Committee meeting on pexidartinib. All research at Memorial Sloan Kettering is supported in part by a grant from the National Institutes of Health, National Cancer Institute (No. P30 CA008748). Dr Wagner reported receiving grants and personal fees from Eli Lilly and Company for participation in this trial. Apart from the submitted work, Dr Wagner reported receiving personal fees and grants from Daiichi Sankyo and Five Prime Therapeutics; personal fees from Novartis, Deciphera, NanoCarrier, and Epizyne; and grants from Karyopharm, Aadi Bioscience, Plexxikon, and Celldex. Dr Schöffski reported grants to Leuven Cancer Institute from Eli Lilly and Company for participation in this trial. Apart from the submitted work, Dr Schöffski reported payments to Leuven Cancer Institute for consulting or advisory role from Eli Lilly and Company, Plexxikon, Eisai, Loxo, Blueprint Medicines, Ellipses Pharma, Deciphera, Merck, Servier, Genmab, Adaptimmune, Intellisphere, and Transgene and research funding to Leuven Cancer Institute from Eli Lilly and Company, Blueprint Medicines, Boehringer Ingelheim, CoBioRes NV, Eisai, Exelixis, G1 Therapeutics, Novartis, PharmaMar, and Plexxikon. Dr Martin-Broto reported receiving personal fees for consulting or an advisory role from Eli Lilly and Company, PharmaMar, Bayer, Roche, and Daichii; for speakers’ bureaus from PharmaMar; and for research from Novartis, PharmaMar, Eisai, Pfizer, and Bristol-Myers Squibb outside of this work. Dr Ganjoo reported receiving personal fees from Eli Lilly and Company for advisory board participation for this trial. Apart from the submitted work, Dr Ganjoo reported receiving personal fees from Daiichi Sankyo for advisory board participation. Dr Yen reported receiving grants and personal fees, in addition to grants to Taipei Veterans General Hospital and National Yang-Ming University School of Medicine, from Eli Lilly and Company for participation in this trial. Apart from the submitted work, Dr Yen reported receiving grants from Eli Lilly and Company, ONO, TLC, Eisai, and EffPha for clinical trial participation; personal fees from Merck, Deciphera, and Athenex; and personal fees for travel, accommodations, and other meeting expenses from Merck, Roche, ONO, and Eisai. Dr Abdul Razak reported receiving personal fees, in addition to grants to Princess Margaret Cancer Center, from Eli Lilly and Company for participation in this trial. Apart from the submitted work, Dr Abdul Razak reported providing expert testimony to Eli Lilly and Company. Dr Spira reported grants and provision of writing assistance, medicines, equipment, or administrative support to Virginia Cancer Specialists from Eli Lilly and Company for participation in this trial. Dr Spira also reported receiving grants from Eli Lilly during the conduct of the study and grants from Bristol-Myers Squibb, CytomX, Novartis, Roche, Trovagene, Arch, Merck, AbbVie, LAM Pharmaceuticals, Arcus, Loxo, and Amgen; grants, personal fees, and nonfinancial support from Takeda; and grants and personal fees from AstraZeneca outside the submitted work. Dr Kawai reported receiving personal fees for travel to meetings and participation in review activities from Eli Lilly and Company for this trial. Dr Kawai also reported receiving personal fees from Novartis and Eisai outside the submitted work. Dr Le Cesne reported receiving personal fees from Eli Lilly and Company for participation in this trial. Apart from the submitted work, Dr Le Cesne reported receiving personal fees for board membership and lectures from PharmaMar and for lectures from Pfizer, Eisai, and Bayer. Dr Van Tine reported receiving personal fees for travel expenses and study support for this trial from Eli Lilly and Company. Apart from the submitted work, Dr Van Tine reported receiving personal fees for consulting from Eli Lilly and Company, Epizyme, CytRx, Janssen, Bayer, Adaptimmune, Immune Design, Daiichi Sankyo, Plexxikon, and Pfizer; grants to Washington University in St Louis School of Medicine from Pfizer, Merck, and Tracon; personal fees for lectures from Eli Lilly and Company, Janssen, Caris, and Adaptimmune; and travel and research expenses from GlaxoSmithKline. Dr Naito reported receiving personal fees for advisory board participation for this trial from Eli Lilly and Company. Apart from the submitted work, Dr Naito reported receiving personal fees for advisory board participation from Daiichi Sankyo; research grants from Roche; personal fees for participation on speakers’ bureaus from Eli Lilly and Company, Pfizer, Chugai, Eisai, Merck Serono, Taiho, AstraZeneca, Nihon Kayaku, Fuji RI, Roche, Novartis, and Bayer; and personal fees from Mieji Seika. Dr Fedenko reported receiving personal fees for advisory board participation for this trial from Eli Lilly and Company. Apart from the submitted work, Dr Fedenko reported receiving personal fees for participation on speakers’ bureaus from Eli Lilly and Company, Eisai, Merck Serono, AstraZeneca, Roche, and Novartis. Ms Soldatenkova and Drs Shahir, Mo, and Wright are employees and stockholders of Eli Lilly and Company. In addition, Dr Mo reported receiving payment for a patent for dosing regimen assigned to ImClone LCC, a wholly owned subsidiary of Eli Lilly and Company. Dr Jones reported receiving personal fees for consulting apart from the submitted work from Eli Lilly and Company, Adaptimmune, Blueprint, Clinigen, Eisai, Epizyme, Daiichi Sankyo, Deciphera, Immunedesign, Merck, and PharmaMar and personal fees from UpToDate, as well as serving as an unpaid consultant for Tracon. No other disclosures were reported.

Funding/Support: All funding for the trial was provided by the sponsor, Eli Lilly and Company.

Role of the Funder/Sponsor: Eli Lilly and Company, in collaboration with academic authors, had a role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, and approval of the manuscript; and decision to submit the manuscript for publication. The sponsor did not have the right to veto publication. The decision regarding to which journal the paper was to be submitted was collaboratively made between the lead authors and the sponsor.

Group Information: Principal investigators: Argentina: Guillermo Lerzo, MD (CENIT Centro de Neurociencias, Investigacion, Y Tratamiento, Buenos Aires); Maria Alejandra Bartoli, MD, Sandra Ostoich, MD (Hospital Provincial del Centenario, Santa Fe); Gabriela Cinat, MD (Instituto Alexander Fleming, Buenos Aires); Australia: Martin Tattersall, MD (University of Sydney Lifehouse, Camperdown); Austria: Thomas Brodowicz, MD, PhD (Allgemeines Krankenhaus, Wien); Belgium: Patrick Schöffski, MD, PhD, MPH (University Hospitals Leuven, Leuven); Lore Lapeire, MD (Universitair Ziekenhuis Gent, Gent); Filomena Mazzeo, MD (Cliniques Universita Saint-Luc, Brussels); Brazil: Andreia Cristina de Melo, MD (INCA—Instituto Nacional Street, Rio de Janeiro); Rodrigo Munhoz, MD (Inst do Cancer do Estado, Sao Paulo); Canada: Albiruni R. Abdul Razak, MRCPI (Princess Margaret Hospital, Toronto, Ontario, Canada); Donald Morris, MD (Tom Baker Cancer Centre, Calgary, Alberta); Margaret Knowling, MD, Amirrtha Srikanthan, MD, Christine Simmons, MD (British Columbia Cancer Agency, Vancouver); Thierry Alcindor, MD, MSc (Royal Victoria Hospital, Montreal, Quebec); Denmark: Anders Krarup-Hansen, MD, PhD (Herlev Hospital, Herlev); Finland: Paula Lindholm, MD (Turku Univ Central Hospital, Turku); Kaisa Sunela, MD, PhD (Tampere University Hosp TAYS, Tampere); France: Jean-Yves Blay, MD, PhD (Centre Leon Berard, Lyon); Christine Chevreau, MD (Institut Claudius Regaud, Toulouse); Florence Duffaud, MD (CHU Hospital d'enfants Timone, Marseille); Nicolas Isambert, MD (Centre Georges Francois Lecler, Dijon); Axel Le Cesne, MD (Gustave Roussy, Villejuif); Sophie Piperno-Neumann, MD (Institut Curie, Paris); Maud Toulmonde, MD (Institut Bergonie, Bordeaux); Germany: Peter Reichardt, MD, PhD (HELIOS Klinikum Berlin-Buch, Berlin); Lars Lindner, MD, PhD (Klinikum der Univ München, München); Sebastian Bauer, MD, PhD (Universitätsklinikum Essen, Essen); Peter Hohenberger, MD, PhD (Klinikum Mannheim GmbH, Heidelberg); Hans-Georg Kopp, MD, Barbara Hermes, MD (Universitätsklinikum Tübingen, Tübingen); Hungary: Zsuzsa Pápai, MD, PhD (Magyar Honvedseg Egeszsegugyi Kozpont, Budapest); Israel: Ofer Merimsky, MD (Tel Aviv Sourasky Medical Cent, Tel Aviv); Daniela Katz, MD, Aviad Zick, MD (Hadassah Medical Center, Jerusalem); Rony Weitzen, MD (Sheba Medical Center, Tel Hashomer); Italy: Massimo Aglietta, MD (Fondazione Istituto del Cancro, Torino); Paolo Giovanni Casali, MD, PhD (Istituto Nazionale dei Tumori, Milano); Armando Santoro, MD (Istituto Clinico Humanitas, Milano); Héctor Soto Parra, MD (Universita degli Studi Catania, Catania); Japan: Hiroaki Hiraga, MD (National Hospital Organization Hokkaido, Sapporo); Keiichi Fujiwara, MD (Saitama Medical University, Saitama); Yoichi Naito, MD (National Cancer Center Hospital East, Chiba); Akira Kawai, MD, PhD (National Cancer Center Hospital, Tokyo); Shunji Takahashi, MD (The Cancer Institute Hospital, Tokyo); Yuichi Ando, MD (Nagoya University Hospital, Nagoya); Taroh Satoh, MD (Osaka University Hospital, Osaka); Takaaki Tanaka, MD, Kazuya Oshima, MD (Osaka International Cancer Institute, Osaka); Takafumi Ueda, MD, PhD (National Osaka Hospital, Osaka); Toshiyuki Kunisada, MD (Okayama University Hospital, Okayama); Ryohei Yokoyama, MD (Kyushu Cancer Center, Fukuoka); Republic of Korea: Jin Hee Ahn, MD (Asan Medical Center, Seoul); JiEun Lee, MD (Seoul St Mary’s Hospital, Seoul); Tak Yun, MD (National Cancer Center, Goyang-si); Se Hoon Park, MD (Samsung Medical Center, Seoul); Sun Young Rha, MD (Severance Hospital Yonsei University Health System, Seoul); Mexico: Joaquin Reinoso, MD (Consultorio Dr Reinoso, Monterrey); Alejandro Molina Alavez, MD (Centro de Investgacion Clínica Oncologia, Merida); Paulina Gonzalez Cisneros, MD (Ce Al Esp Reu In del Potosi SC, San Luis Potosi); Jennifer Ramirez Puente, MD (Hospital Angeles de Tijuana, Tijuana); Francisco Ramirez Godinez, MD (Hospital Civil De Guadalajara, Guadalajara); Netherlands: A. K. L. Reyners, MD (Univ Medisch Centrum Groningen, Groningen); Stefan Sleijfer, MD, PhD (Erasmus Medisch Centrum, Rotterdam); Ingeborg Vriens, MD (Univ Medisch Centrum Maastrich, Maastricht); Ingrid Desar, MD (Univ Medisch Centrum St Radbou, Nijmegen); A. J. Hans Gelderblom, MD, PhD (Leids Universitair Medisch, Leiden); Poland: Piotr Rutkowski, MD, PhD (Centrum Onkologii-Instytut, Warszawa); Russia: Mamed Aliev, MD, Alexander Fedenko, MD (Blokhin Cancer Research Center, Moscow); Vladimir Moiseyenko, MD (St-Petersburg Scientific Center, St Petersburg); Guzel Mukhametshina, MD (Kazan Oncology Dispensary, Kazan); Spain: Javier Martin-Broto, MD, PhD (Hospital Virgen del Rocio, Sevilla); Jose Antonio Lopez-Martin, MD, PhD (Hospital 12 de Octubre, Madrid); Francisco Javier Garcia del Muro Solans, MD, PhD (Hospital Duran I Reynals, Barcelona); Claudia Valverde, MD, PhD (Hospital Vall d'Hebron, Barcelona); Antonio Casado, MD (Hospital Clinico San Carlos, Madrid); Roberto Díaz Beveridge, MD (Hospital Universitario La Fe, Valencia); Sweden: Mikael Eriksson, MD, PhD (Skånes Universitetssjukhus, Lund); Switzerland: Attila Kollar, MD (Inselspital Bern, Bern); Christian Rothermundt, MD (Kantonsspital St Gallen, Sankt Gallen); Taiwan: Ruey-Long Hong, MD (National Taiwan Univ Hospital, Taipei City); Chuech-Chuan Yen, MD (Taipei Veterans General Hosp, Taipei City); Chi-Ting Liau, MD (Chang Gung Mem Hosp–Linkou, Taoyuan Hsien); United Kingdom: Nasim Ali, MD (Clatterbridge Cancer Centre, Merseyside); Palma Dileo, MD (Univ College Hospital–London, London); Robin L. Jones, MD, MBBS, BSc, MRCP (Royal Marsden NHS Trust, London); Michael Leahy, MD (Christie NHS Foundation Trust, Manchester); Robin Young, MD (Weston Park Hospital Cancer Research Centre, Sheffield); United States: Victor Villalobos, MD, PhD (University of Colorado Cancer, Aurora); Warren Chow, MD (City of Hope National Medical Center, Duarte, California); Arun Singh, MD (UCLA Med Center, Los Angeles, California); Brian A. Van Tine, MD, PhD (Washington University Medical Center, Saint Louis, Missouri); John Hainsworth, MD (Tennessee Oncology PLLC, Nashville); Suzanne Partridge, MD (Oncology Hematology Care Inc, Cincinnati, Ohio); Andrew J. Wagner, MD, PhD (Dana Farber Cancer Institute, Boston, Massachusetts); Mihaela Druta, MD (Moffitt Cancer Center, Tampa, Florida); Kirsten Leu, MD (Nebraska Methodist Cancer Center, Omaha); David Portnoy, MD (The West Clinic, Germantown, Tennessee); Vicki Keedy, MD (Vanderbilt University Medical Center, Nashville, Tennessee); John Ward, MD (Huntsman Cancer Institute, Salt Lake City, Utah); Stephan Kendall, MD (Utah Cancer Specialists, Salt Lake City); Richard Riedel, MD (Duke University Medical Center, Durham, North Carolina); Lara Davis, MD (Oregon Health and Science University, Portland); Vi Chiu, MD (University of New Mexico Cancer Center, Albuquerque); Gary Schwartz, MD (Columbia University Medical Center, New York, New York); Gina D’Amato, MD (Georgia Cancer Specialists, Atlanta); Lee Hartner, MD (Pennsylvania Oncology Hematology, Philadelphia); William D. Tap, MD (Memorial Sloan Kettering Cancer Center, New York, New York); Alexander Spira, MD, PhD, FACP (Fairfax N VA Hemat Oncology PC, Fairfax, Virginia); Melissa Burgess, MD (University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania); Kristen Ganjoo, MD (Stanford University, Stanford, California); Scott Schuetze, MD (University of Michigan, Ann Arbor); Steven Attia, MD (Mayo Clinic–Jacksonville, Jacksonville, Florida).

Meeting Presentation: This study was presented at the American Society of Clinical Oncology (ASCO) 55th Annual Meeting; June 2, 2019; Chicago, Illinois.

Data Sharing Statement: See Supplement 3.

Additional Contributions: We are indebted to the patients who participated in this study and their families. We are indebted to all the physicians, nurses, and study coordinators who cared for these patients. We thank Patrick Peterson, PhD, for assistance with statistical and clinical interpretation of data and Karen Paulsrud, RPh, for writing assistance; both received compensation for this work through their employment with Eli Lilly and Company.

References
1.
Lahat  G, Tuvin  D, Wei  C,  et al.  New perspectives for staging and prognosis in soft tissue sarcoma.   Ann Surg Oncol. 2008;15(10):2739-2748. doi:10.1245/s10434-008-9970-6 PubMedGoogle ScholarCrossref
2.
Sbaraglia  M, Dei Tos  AP.  The pathology of soft tissue sarcomas.   Radiol Med. 2019;124(4):266-281. doi:10.1007/s11547-018-0882-7PubMedGoogle ScholarCrossref
3.
Seddon  B, Strauss  SJ, Whelan  J,  et al.  Gemcitabine and docetaxel versus doxorubicin as first-line treatment in previously untreated advanced unresectable or metastatic soft-tissue sarcomas (GeDDiS): a randomised controlled phase 3 trial.   Lancet Oncol. 2017;18(10):1397-1410. doi:10.1016/S1470-2045(17)30622-8 PubMedGoogle ScholarCrossref
4.
Judson  I, Verweij  J, Gelderblom  H,  et al; European Organisation and Treatment of Cancer Soft Tissue and Bone Sarcoma Group.  Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: a randomised controlled phase 3 trial.   Lancet Oncol. 2014;15(4):415-423. doi:10.1016/S1470-2045(14)70063-4 PubMedGoogle ScholarCrossref
5.
Ryan  CW, Merimsky  O, Agulnik  M,  et al.  PICASSO III: a phase iii, placebo-controlled study of doxorubicin with or without palifosfamide in patients with metastatic soft tissue sarcoma.   J Clin Oncol. 2016;34(32):3898-3905. doi:10.1200/JCO.2016.67.6684 PubMedGoogle ScholarCrossref
6.
Tap  WD, Papai  Z, Van Tine  BA,  et al.  Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial.   Lancet Oncol. 2017;18(8):1089-1103. doi:10.1016/S1470-2045(17)30381-9 PubMedGoogle ScholarCrossref
7.
Papadopoulos  N, Lennartsson  J.  The PDGF/PDGFR pathway as a drug target.   Mol Aspects Med. 2018;62:75-88. doi:10.1016/j.mam.2017.11.007 PubMedGoogle ScholarCrossref
8.
Lartruvo. Highlights of prescribing information. Indianapolis, IN: Eli Lilly and Co; 2018. Accessed October 15, 2019. http://pi.lilly.com/us/lartruvo-uspi.pdf
9.
Tap  WD, Jones  RL, Van Tine  BA,  et al.  Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase 1b and randomised phase 2 trial.   Lancet. 2016;388(10043):488-497. doi:10.1016/S0140-6736(16)30587-6 PubMedGoogle ScholarCrossref
10.
Eisenhauer  EA, Therasse  P, Bogaerts  J,  et al.  New Response Evaluation Criteria in Solid Tumours: revised RECIST guideline (version 1.1).   Eur J Cancer. 2009;45(2):228-247. doi:10.1016/j.ejca.2008.10.026 PubMedGoogle ScholarCrossref
11.
Jones  RL, Mo  G, Baldwin  JR,  et al.  Exposure-response relationship of olaratumab for survival outcomes and safety when combined with doxorubicin in patients with soft tissue sarcoma.   Cancer Chemother Pharmacol. 2019;83(1):191-199. doi:10.1007/s00280-018-3723-4 PubMedGoogle ScholarCrossref
12.
Holzer  TR, O’Neill Reising  L, Credille  KM, Schade  AE, Oakley  GJ.  Variability in platelet-derived growth factor receptor alpha antibody specificity may impact clinical utility of immunohistochemistry assays.   J Histochem Cytochem. 2016;64(12):785-810. doi:10.1369/0022155416673979 PubMedGoogle ScholarCrossref
13.
Maurer  W Betz  F.  Multiple testing in group sequential trials using graphical approaches.   Stat Biopharm Res. 2013;5(4):311-320. doi:10.1080/19466315.2013.807748 Google ScholarCrossref
14.
Cox  DR.  Regression models and life-tables.   J R Stat Soc Series B Stat Methodol. 1972;34(2):187-220. doi:10.1111/j.2517-6161.1972.tb00899.x Google Scholar
15.
Schoenfeld  D.  Partial residuals for the proportional hazards regression model.   Biometrika. 1982;69:239-241. doi:10.1093/biomet/69.1.239 Google ScholarCrossref
16.
Jones  RL, Wagner  A, Kawai  A,  et al.  1671O Assessment of cardiotoxicity (CT) associated with doxorubicin (dox) in patients (pts) with advanced soft tissue sarcoma (STS) in a phase III randomized trial.   Ann Oncol. 2019;30(Suppl 5). doi:10.1093/annonc/mdz283.004Google Scholar
17.
Croswell  JM, Ransohoff  DF, Kramer  BS.  Principles of cancer screening: lessons from history and study design issues.   Semin Oncol. 2010;37(3):202-215. doi:10.1053/j.seminoncol.2010.05.006 PubMedGoogle ScholarCrossref
18.
Kramer  BS, Croswell  JM.  Cancer screening: the clash of science and intuition.   Annu Rev Med. 2009;60:125-137. doi:10.1146/annurev.med.60.101107.134802 PubMedGoogle ScholarCrossref
×