Introduction: We evaluated the effectiveness and safety profile of the tyrosine kinase inhibitor sunitinib in patients with advanced or metastatic renal cell carcinoma (a/mRCC) in a real-world setting. Methods: We analyzed data of adult a/mRCC patients treated with sunitinib. Data were derived from the German non-interventional post-approval multicenter STAR-TOR registry (NCT00700258). Progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were evaluated using descriptive statistics and survival analyses for the entire cohort and patient subgroups. Results: A total of 116 study sites recruited 702 patients treated with sunitinib (73.1% male; median age 68.0 years; median Karnofsky index 90%) between November 2010 and May 2020. The most frequent histological subtype was clear cell RCC (81.6%). Sunitinib was administered as first-line treatment in 83.5%, as second line in 11.7%, and as third line or beyond in 4.8% of the patients. Drug-related AEs and serious AEs were reported in 66.3% and 13.9% of the patients, respectively (most common AE: gastrointestinal disorders; 39.7% of all patients). Conclusions: This study adds further real-world evidence of the persisting relevance of sunitinib for patients with a/mRCC who cannot receive or tolerate immune checkpoint inhibitors. The study population includes a high proportion of patients with unfavorable MSKCC poor-risk score, but shows still good PFS and OS results, while the drug demonstrates a favorable safety profile. The STAR-TOR registry is also registered in the database of US library of medicine (NCT00700258).

Treatment recommendations for advanced or metastatic renal cell carcinoma (a/mRCC) are divided into two paths based on patient characteristics. The suitable patients receive combination therapies with immune checkpoint inhibitors (ICIs), whereas for the remainder sunitinib and pazopanib are recommended plus cabozantinib in case of International Metastatic RCC Database Consortium (IMDC) intermediate and poor-risk status [1]. Of these therapeutic options, sunitinib is the most extensively studied drug and as such was regarded as the gold standard in first-line a/mRCC treatment for over 10 years. Therefore, sunitinib was the reference medication in most of the studies which assessed first-line ICI combination therapies in the last several years.

Sunitinib has been EMA-approved in July 2006 for pretreated patients with a/mRCC after failure of interferon-α or interleukin-2 therapy. The phase II registrational study demonstrated an overall response rate of 34% and a progression-free survival (PFS) of 8.3 months [2]. In the pivotal phase III trial, sunitinib was compared to interferon-α and demonstrated a striking PFS of 11 months for sunitinib versus 5 months for interferon-α (p < 0.001) [3]. Thus, in 2007 the marketing authorization of sunitinib was amended to include a/mRCC patients regardless of line of therapy.

Even though nowadays this study must be considered as a mere historic event, the survival benefit for patients that sunitinib brought to the treatment landscape was tremendous while treatment-related adverse events (AEs) seemed to be moderate. Not only was sunitinib intensely examined in prospective clinical trials but there was additional real-world evidence generated over the years. Beginning with the publication of the expanded access trial in 2009 which included 4,543 patients, several observational studies were to follow [4]. Nevertheless, the STAR-TOR registry represents one of the largest real-world data sets describing not only effectivity but also tolerability data for the use of sunitinib in the routine clinical setting.

Our data originate from the prospective STAR-TOR registry maintained by Pfizer Pharma GmbH, Berlin, Germany, and were collected in accordance with the Declaration of Helsinki in its most recent version. The STAR-TOR registry received prior approval by the Ethics Committee of Münster University Hospital in 2007 (Nr.2007-484-f-S) and is also registered in the database of the US library of medicine (NCT00700258). The registry evaluates the treatment with (a) axitinib, (b) sunitinib, or (c) temsirolimus in a routine clinical setting for its effectiveness, tolerability, and safety. Target diseases are a/mRCC, recurrent or refractory mantle cell lymphoma, and gastrointestinal stromal tumors. Additional details regarding the STAR-TOR registry have been published previously [5‒9].

Study Cohort

The here-presented analyses include adult patients with histologically confirmed a/mRCC treated with sunitinib monotherapy at the STAR-TOR study centers between November 2010 and May 2020. All patients gave written informed consent for enrollment in the STAR-TOR registry. All study centers were located in Germany.

Sunitinib Administration

The standard dosage of the orally administered sunitinib was 50 mg daily for 28 days, followed by a 14-day break (4/2 schedule). Treatment lines of sunitinib included any line, of which up to six lines of systemic therapy were documented.

Variables

The analyses included demographic variables and tumor variables. The MSKCC score was defined as the risk categorizing tool in the statistical analysis plan of the registry. As documentation of laboratory values was optional in this non-interventional study, the MSKCC score could not be calculated for every patient [10].

Serum lactate dehydrogenase (LDH) levels were measured by local laboratories. The MSKCC score for treatment-naïve patients defines a LDH concentration of 1.5 times the upper level of normal as a risk factor. Accordingly, “high LDH” was assumed when the serum LDH level exceeded 300 U/L [11].

Outcomes

This study evaluates three primary outcome measures: PFS, overall survival (OS), and AEs. Patients had their follow-up visits corresponding to the clinical routine at the participating sites in intervals of 8–12 weeks. At each follow-up visit, treatment status, response, tumor staging, laboratory values, and AEs were documented. All participating study centers received random monitoring visits ensuring correct patient assessment and data entry.

Treatment response was assessed according to RECIST 1.1 [12]. If RECIST evaluations were not routinely performed at the study sites, a physician’s assessment was accepted instead. Best response was determined by a physician at the final visit and entered in the case report form.

Definition for OS was time from the baseline visit at a participating STAR-TOR center until death from any cause. If documentation of death was not available, the subject was censored with the latest contact data. PFS was defined as the time from the baseline visit to documentation of disease progression or death.

AEs were reported by the study sites according to NCI-CTCAE criteria and the Medical Dictionary for Regulatory Activities (MedDRA) standard [13, 14]. For better comprehensibility of the analysis, AEs were grouped according to NCI-CTCAE standards.

Statistical Analyses

Categorial data are described as absolute frequencies and percentages. Continuous data are reported as median and range.

PFS and OS are visualized using Kaplan-Meier plots. The method of comparison for predefined subgroup analyses was log rank tests which were applied for treatment line, histologic subtype (clear cell RCC [ccRCC] vs. non-ccRCC), patient age (≤65 vs. >65 years), gender, MSKCC prognosis group (favorable vs. intermediate vs. poor), BMI (<25 vs. 25–30 vs. >30), and LDH levels (≤300 vs. >300).

Most performed analyses rely on univariate models as missing values, and patients with incomplete data would have demanded imputation of missing values or patient exclusion upon multivariate analyses what may have distorted our results. Possible correlations between nephrectomy and MSKCC prognosis group on PFS and OS were investigated using Cox regression models.

All statistical analyses were conducted using SAS software (version 9.4). The provided tests are two-sided and have to be interpreted purely exploratively.

The statistical analyses were performed by Dr. Thomas Fischer (Winicker Norimed GmbH, Nuremberg, Germany).

Study Cohort

A total of 702 patients from 116 centers were included, of whom 659 completed their study participation until the final examination. Mean patients’ age was 68.0 years. A total of 513 patients were male (73.2%) with a median Karnofsky performance status score of 90%. Predominant tumor histology was ccRCC (81.6%). Risk score according to the MSKCC criteria was favorable in 12.6% of the patients, intermediate in 62.2%, and poor in 25.2%. In 85.3% of the patients, nephrectomy was performed, achieving a R0 status in 72.2% of the cases. For n = 568 patients, time point of nephrectomy was available: n = 309 RCC patients underwent nephrectomy before the diagnosis of metastases, n = 259 patients underwent nephrectomy after being diagnosed with metastatic disease, and n = 103 patients did not undergo any nephrectomy. All patients had metastatic disease upon inclusion in the registry. Table 1 details the characteristics of the study cohort.

Table 1.

Patient characteristics at baseline

n
Pts with final examination, n 659  
Age, years, median (range) 702 68.0 (23.0–87.7) 
Gender, male/female, % 697 73.2/26.8 
Risk assessment according to MSKCC score, % 460  
 Favorable risk  12.6 
 Intermediate risk  62.2 
 High risk  25.2 
Karnofsky performance status 690  
 Median (range)  90 (40–100) 
 100–90, %  51.3 
 ≤80, %  48.7 
Histological subtype*, % 702  
 Clear cell  81.6 
 Papillary  8.3 
 Chromophobe  2.1 
 Collecting duct  0.6 
 Unclassified and other  7.8 
Nephrectomy status, % 702  
 Total nephrectomy  76.6 
 Partial nephrectomy  8.7 
 No nephrectomy  14.7 
Date of nephrectomy available 568  
 Nephrectomy before diagnosis of metastatic disease  54.4 
 Nephrectomy after diagnosis of metastatic disease  45.6 
Primary tumor resection, % 601  
 R0  72.2 
 Incomplete  27.8 
Additional radiation therapy, % 453  
 Yes  10.6 
 No  89.4 
Number of metastatic sites, median (range) 702 2 (0–6) 
Location of metastases*, % 702  
 Lung  64.1 
 Lymph nodes  41.2 
 Bone  29.5 
 Liver  17.9 
 Adrenal gland  12.0 
 Contralateral kidney  7.1 
 Central nervous system  5.0 
 Other  26.8 
LDH level in serum, % 564  
 ≤300 U/L  81.9 
 >300 U/L  18.1 
BMI 654  
 <25  30.7 
 25–30  48.2 
 >30  21.1 
n
Pts with final examination, n 659  
Age, years, median (range) 702 68.0 (23.0–87.7) 
Gender, male/female, % 697 73.2/26.8 
Risk assessment according to MSKCC score, % 460  
 Favorable risk  12.6 
 Intermediate risk  62.2 
 High risk  25.2 
Karnofsky performance status 690  
 Median (range)  90 (40–100) 
 100–90, %  51.3 
 ≤80, %  48.7 
Histological subtype*, % 702  
 Clear cell  81.6 
 Papillary  8.3 
 Chromophobe  2.1 
 Collecting duct  0.6 
 Unclassified and other  7.8 
Nephrectomy status, % 702  
 Total nephrectomy  76.6 
 Partial nephrectomy  8.7 
 No nephrectomy  14.7 
Date of nephrectomy available 568  
 Nephrectomy before diagnosis of metastatic disease  54.4 
 Nephrectomy after diagnosis of metastatic disease  45.6 
Primary tumor resection, % 601  
 R0  72.2 
 Incomplete  27.8 
Additional radiation therapy, % 453  
 Yes  10.6 
 No  89.4 
Number of metastatic sites, median (range) 702 2 (0–6) 
Location of metastases*, % 702  
 Lung  64.1 
 Lymph nodes  41.2 
 Bone  29.5 
 Liver  17.9 
 Adrenal gland  12.0 
 Contralateral kidney  7.1 
 Central nervous system  5.0 
 Other  26.8 
LDH level in serum, % 564  
 ≤300 U/L  81.9 
 >300 U/L  18.1 
BMI 654  
 <25  30.7 
 25–30  48.2 
 >30  21.1 

*Multiple entries allowed.

LDH, lactate dehydrogenase.

Sunitinib Treatment

A total of 83.5% of the patients received sunitinib as first-line treatment, 11.7% as second line, and 4.8% as third line or beyond. Median treatment duration for all lines was 5.7 months. For 659 patients, data for treatment termination were available. Table 2 details the sunitinib treatment further.

Table 2.

Treatment characteristics

n
Sunitinib is used in, % 702  
 First line  83.5 
 Second line  11.7 
 Third line  3.1 
 Fourth line  1.0 
 Fifth line and beyond  0.7 
Treatment duration 642  
 Median, months (range)  5.7 (0–83.6) 
Duration of survival follow-up 367  
 Median, months (range)  9.1 (0–91.5) 
Dose modifications 576  
 Patients with dose changes, %  67.7 
Type of study end*, % 659  
 Documentation until patient’s death  14.7 
 Documentation discontinued  9.0 
 Therapy discontinued  76.3 
  Due to intolerability  22.5 
  Due to progression  75.7 
  Due to other reasons  3.6 
n
Sunitinib is used in, % 702  
 First line  83.5 
 Second line  11.7 
 Third line  3.1 
 Fourth line  1.0 
 Fifth line and beyond  0.7 
Treatment duration 642  
 Median, months (range)  5.7 (0–83.6) 
Duration of survival follow-up 367  
 Median, months (range)  9.1 (0–91.5) 
Dose modifications 576  
 Patients with dose changes, %  67.7 
Type of study end*, % 659  
 Documentation until patient’s death  14.7 
 Documentation discontinued  9.0 
 Therapy discontinued  76.3 
  Due to intolerability  22.5 
  Due to progression  75.7 
  Due to other reasons  3.6 

*More than one reason possible, for some pts no reason was given.

Sunitinib Treatment Response

Best response rates were reported for 669 patients with a clinical benefit rate of 63.8%. Complete response was reached in 6.0%, partial response in 24.8%, and stable disease in 33.0%, whereas 21.8% of the patients experienced disease progression. For 14.3% of the patients, no response was assessable. The median PFS for all patients was 6.7 months, and the median OS was 25.0 months.

Subgroup Analyses

Median survival was dependent on treatment line (1st vs. later line) with PFS 6.9 months (95% confidence interval [CI]: 6.1–8.3 months) versus 6.0 months (95% CI: 4.0–8.3 months, p = 0.29), and OS 25.6 months (95% CI: 22.2–28.9 months) versus 21.3 months (95% CI: 15.9–27.3 months, p = 0.11). Subgroup analysis results are displayed in Table 3. Median PFS and OS did not differ markedly between first, second, and third line and deteriorated clinically relevant not before sunitinib application in fourth and later lines.

Table 3.

Subgroup analyses of PFS and OS, months

PFSOS
nmonthsnmonths
Median survival according to BMI and LDH 
 All patients 702 6.7 702 25.0 
 BMI <25 201 4.8 201 20.9 
 BMI 25–30 315 7.6 315 27.2 
 BMI >30 138 11.1 138 27.8 
 Patients with LDH ≤300 U/La 462 7.7 462 27.3 
 Patients with LDH >300 U/La 102 4.4 102 9.8 
Median survival according to treatment line 
 All patients 702 6.7 702 25.0 
 First-line patients 586 6.9 586 25.6 
 Second-line patients 82 6.1 82 25.0 
 Third-line patients 22 6.9 22 24.3 
 ≥Fourth-line patients 12 4.9 12 8.6 
Median survival according to histology 
 All patients 702 6.7 702 25.0 
 Clear cell patients 573 7.4 573 25.7 
 All other histologies 129 5.0 129 20.9 
Median survival according to nephrectomy status 
 No nephrectomy 103 4.8 103 16.4 
 Nephrectomy before diagnosis of metastatic disease 309 8.0 309 28.0 
 Nephrectomy after diagnosis of metastatic disease 259 7.7 295 27.2 
PFSOS
nmonthsnmonths
Median survival according to BMI and LDH 
 All patients 702 6.7 702 25.0 
 BMI <25 201 4.8 201 20.9 
 BMI 25–30 315 7.6 315 27.2 
 BMI >30 138 11.1 138 27.8 
 Patients with LDH ≤300 U/La 462 7.7 462 27.3 
 Patients with LDH >300 U/La 102 4.4 102 9.8 
Median survival according to treatment line 
 All patients 702 6.7 702 25.0 
 First-line patients 586 6.9 586 25.6 
 Second-line patients 82 6.1 82 25.0 
 Third-line patients 22 6.9 22 24.3 
 ≥Fourth-line patients 12 4.9 12 8.6 
Median survival according to histology 
 All patients 702 6.7 702 25.0 
 Clear cell patients 573 7.4 573 25.7 
 All other histologies 129 5.0 129 20.9 
Median survival according to nephrectomy status 
 No nephrectomy 103 4.8 103 16.4 
 Nephrectomy before diagnosis of metastatic disease 309 8.0 309 28.0 
 Nephrectomy after diagnosis of metastatic disease 259 7.7 295 27.2 

BMI, body mass index; LDH, lactate dehydrogenase.

aExploratory analysis proved statistical significance with p < 0.0001.

Patients with ccRCC had longer median survival as patients with non-clear cell histologies (PFS: 7.4 vs. 5.0 months, p = 0.12; OS: 25.7 vs. 20.9 months, p = 0.14). Small subgroup numbers did not allow for further differentiation by histology. There was no clinically relevant difference in PFS between younger and older patients (≤65 years vs. >65 years: PFS: 6.8 vs. 6.7 months, p = 0.83; OS: 26.3 vs. 24.7 months, p = 0.92).

Similarly, no clinically relevant gender-specific differences were observed comparing male to female patients: PFS: 6.9 versus 6.2 months, p = 0.64; OS: 25.7 versus 22.8 months, p = 0.41. Worse prognosis according to MSKCC risk score was associated with shorter median PFS and OS (PFS from favorable to poor prognosis: 13.8, 6.5, 3.8 months, p < 0.0001; OS from favorable to poor prognosis: 49.3, 26.9, and 7.7 months, p < 0.0001).

Patients with BMI <25 had shorter PFS and OS than patients with BMI 25–30 and >30 (PFS: 4.8, 7.6, and 11.0 months, p = 0.002; OS: 20.9, 27.2, 27.8 months, p = 0.02). Upon stratification for baseline LDH serum levels at a cut-off of 300 U/L, PFS and OS markedly differed between patients with high versus low LDH (PFS: 4.4 vs. 7.7 months, p < 0.001, OS: 9.8 vs. 27.3 months, p < 0.001).

Figure 1 depicts the corresponding Kaplan-Meier curves. In addition, we stratified the patients by the time point of nephrectomy: of n = 568 patients with known nephrectomy status, median PFS and OS were compared for patients with nephrectomy before the diagnosis of metastatic disease versus for patients with already known metastatic disease at the time point of surgery versus patients without nephrectomy. Median PFS was 8.0 versus 7.7 versus 4.8 months and median OS 28.0 versus 27.7 versus 16.4 months, respectively. On multivariate analyses, the PFS and OS differences showed no statistical significance after adjusting for the MSKCC score (p > 0.05). No statistically significant interaction was evident between the MSKCC score and the nephrectomy status (p = 0.0.645 for PFS and p = 0.827 for OS, respectively). Given the low sample size of the MSKCC subgroups, no further stratifications were possible. Online supplementary Figure A (available at https://doi.org/10.1159/000536563) shows the corresponding Kaplan-Meier curves.

Fig. 1.

Kaplan-Meier curves for stratified survival analyses with PFS and OS for MSKCC Risk Score (a), BMI (b), and LDH level (c).

Fig. 1.

Kaplan-Meier curves for stratified survival analyses with PFS and OS for MSKCC Risk Score (a), BMI (b), and LDH level (c).

Close modal

Adverse Events

Of 702 patients, 464 (66.3%) experienced at least one treatment-related AE. CTCAE grade III/IV events were reported for 97 (13.8%) patients. Table 4 details all AEs which were reported with a frequency of ≥2%, according to the MedDRA [13].

Table 4.

Treatment-related side effects in a total of 702 patients, incidence ≥2%

TotalGrade 3/4
Patients with AE, n (%) 464 (66.1) 97 (13.8) 
Gastrointestinal disorders, % 39.7 4.0 
General disorders, % 29.5 3.8 
Skin and subcutaneous tissue disorders, % 23.6 0.7 
Blood and lymphatic system disorders, % 19.8 2.4 
Nervous system disorders, % 18.7 0.9 
Investigations, % 12.1 0.1 
Metabolism and nutrition disorders, % 11.4 1.7 
Vascular disorders, % 11.4 1.1 
Respiratory, thoracic, and mediastinal disorders, % 7.7 1.4 
Musculoskeletal and connective tissue disorders, % 5.4 0.3 
Infections and infestations, % 4.8 2.0 
Endocrine disorders, % 4.3 0.0 
Eye disorders, % 2.6 0.0 
Renal and urinary tract disorders, % 2.0 1.0 
TotalGrade 3/4
Patients with AE, n (%) 464 (66.1) 97 (13.8) 
Gastrointestinal disorders, % 39.7 4.0 
General disorders, % 29.5 3.8 
Skin and subcutaneous tissue disorders, % 23.6 0.7 
Blood and lymphatic system disorders, % 19.8 2.4 
Nervous system disorders, % 18.7 0.9 
Investigations, % 12.1 0.1 
Metabolism and nutrition disorders, % 11.4 1.7 
Vascular disorders, % 11.4 1.1 
Respiratory, thoracic, and mediastinal disorders, % 7.7 1.4 
Musculoskeletal and connective tissue disorders, % 5.4 0.3 
Infections and infestations, % 4.8 2.0 
Endocrine disorders, % 4.3 0.0 
Eye disorders, % 2.6 0.0 
Renal and urinary tract disorders, % 2.0 1.0 

Sunitinib was the standard therapeutic option in a/mRCC before the onset of ICI combination therapies and is even nowadays a well-established option for treatment for ICI unsuitable patients. Many trials investigated the effectiveness and safety of sunitinib, either at the beginning of the TKI era comparing it to previous options such as interferon-α or later on as the standard therapeutic option in modern combination therapy studies [3, 15]. Over time, sunitinib was compared to other TKIs in few phase II and phase III trials [16‒19]. Additionally, many real-world data sets have been published. The largest was the expanded access trial comprising 4,543 patients published by Gore et al. [4] in 2009, followed by numerous studies which were summarized by Moran et al. [20] in 2019. After 2019, to our knowledge, seven new real-world data sets were published.

As the number of publications directly evaluating sunitinib is large, Table 5 provides a comprehensive overview of the literature. Our study provides additional evidence and presents real-world data from the non-interventional post-approval multicenter STAR-TOR registry showing an overall OS of 25.0 months and a PFS of 6.7 months. Subgroup analyses confirmed worse prognosis score according to MSKCC, low BMI, and high LDH levels as negative predictive markers for sunitinib effectiveness. With respect to drug safety, AEs were observed in 66.3% of the patients (grade III/IV in 13.8%) with the most common AEs being gastrointestinal disorders (39.7% of all patients).

Table 5.

Sunitinib in clinical trials and real-world data sets – an overview

Author, publication yearStudy typePts receiving sunitinib, nStudy drugs/comparisonsRisk score usedRisk stratification, %Percentage of ccRCC pts, %ORR, %PFS [months], medianOS [months], medianAE rate
fav.int.poorCRPRSDPDall, %grade 3/4, %
STAR-TOR: this data set Observational 702 Sunitinib or axitinib or temsirolimus MSKCC 12.6 62.2 25.2 81.6 6.0 24.8 33.0 21.8 6.7 25.0 66.3 13.9 
Motzer et al. [15] (2009) Ph III RCT 375 Sunitinib versus IFN-α MSKCC 38 56 100 44 40 11.0 26.4 N/R1 N/R 
Motzer et al. [45] (2012) Ph II RCT 1462 Sunitinib only MSKCC 25 60 15 100 32 43 N/R 8.5 23.1 N/R N/R 
Motzer et al. [16] (2013) Ph III RCT 548 Sunitinib versus pazopanib MSKCC (IMDC) 27 (25) 59 (26) 9 (17) 100 <1 24 44 19 9.5 29.3 99 57/173 
Miyake et al. [21] (2014) Observational 102 Sunitinib only MSKCC 25.5 50.9 23.6 96 28 65 15 7.8 33.2 100 N/R 
Motzer et al. [46] (2014) Ph II RCT 233 Everolimus followed by sunitinib versus sunitinib followed by everolimus MSKCC 30 56 14 85 25 52 14 10.7 32.0 N/R N/R 
Schnadig et al. [27] (2014) Observational 134 Sunitinib only MSKCC (IMDC) 11.2 (4.5) 28.4 (56.7) 8.2 (23.1) 82.8 2.2 14.2 18.7 N/R 7.5 15.5 85.1 N/R 
Eichelberg et al. [17] (2015) Ph III RCT 183 Sorafenib followed by sunitinib versus sunitinib followed by sorafenib MSKCC 45 51 0.5 84 3.4 26 35 N/R 8.5 30.2 N/R 58.5 
Gore et al. [30] (2015) Expanded access trial 4,543 Sunitinib only MSKCC 20 33 26 88 14 45 19 9.4 18.7 95 N/R 
Pan et al. [47] (2015) Observational 50 Sunitinib only MSKCC 20 56 24 84.6 10 28 58 9.4 N/R N/R N/R 
Tan et al. [29] (2015) Observational 1114 Sunitinib only MSKCC (IMDC) 11.3 (13.4) 46.4 (51.3) 42.3 (35.3) 82.7 31.44 34.34 34.34 7.94 21.84 N/R 58.6 
Ruiz-Morales et al. [48] (2016) Observational 6,519 Sunitinib versus pazopanib IMDC 23 57 20 90 N/R N/R N/R N/R 8.4 22.3 N/R N/R 
Rini et al. [49] (2016) Ph III RCT 135 Sunitinib versus pembrolizumab plus axitinib IMDC 26 71 100 N/R N/R N/R N/R 15.1 87 41 
Choueiri et al. [18] (2017) Ph II RCT 78 Sunitinib versus cabozantinib IMDC 80.9 19.1 100 11.5 42.3 25.6 5.6 21.8 98.6 68.1 
Noize et al. [22] (2017) Observational 302 Sunitinib only N/R N/R N/R N/R 83.1 2.3 28.8 40.1 18.5 8.4 23.6 97 58.3 
Lalani et al. [50] (2017) Observational 577 Sunitinib or pazopanib IMDC 22 58 20 83.7 N/R N/R N/R N/R 11.0 31.7 N/R N/R 
Zhang et al. [51] (2017) Observational 362 Sunitinib versus sorafenib MSKCC (IMDC) 41.7 (39.8) 53.0 (51.7) 5.2 (8.6) 91.4 1.1 19.9 67.4 11.6 10.0 24.0 N/R N/R 
Motzer et al. [52] (2018) Ph III RCT 546 Nivolumab plus ipilimumab versus sunitinib IMDC 23 61 16 100 25 45 17 8.4 26 97 63 
Poprach et al. [53] (2019) Observational 1,0166 Multiple TKIs MSKCC 27.8 64.7 7.6 95.3 5.1 23.0 38.2 22.2 10.8 31.9 30.1 7.7 
Choueiri et al. [54] (2019) Ph III RCT 328 Nivolumab plus cabozantinib versus sunitinib IMDC 22 57.3 20.7 100 4.6 22.6 42.1 13.7 8.3 99.1 70.6 
Motzer et al. [28] (2019) Ph III RCT 444 Sunitinib versus avelumab plus axitinib MSKCC (IMDC) 22.5 (21.6) 66.0 (62.2) 10.1 (16.0) 100 1.8 23.9 45.5 18.7 8.4 37.87 99.3 71.5 
Rini et al. [55] (2019) Ph III RCT 429 Pembrolizumab plus axitinib versus sunitinib IMDC 30.5 57.3 12.1 100 1.9 33.8 39.4 17.0 11.1 99.5 70.6 
Savard et al. [23] (2020) Retrospective database study 1,769 Sunitinib only IMDC 18 58 24 100 3.7 28.7 38.2 29.4 8.1 28.6 N/R N/R 
Schmidinger et al. [26] (2020) Observational 467 Sunitinib only MSKCC (IMDC) 16.6 (15.7) 29.1 (25.2) 7.5 (10.3) N/R N/R N/R N/R N/R 10.4 34.0 N/R N/R 
Choueiri et al. [56] (2021) Ph III RCT 328 Sunitinib versus nivolumab plus cabozantinib IMDC 22.0 57.3 20.7 100 4.6 22.6 42.1 13.7 8.3 34.38 99.1 70.6 
Costello et al. [57] (2021) Observational 75 Sunitinib or pazopanib IMDC 14.7 65.3 20.0 78,5 N/R N/R N/R N/R 5.69 26.2 N/R N/R 
Motzer et al. [58] (2021) Ph III RCT 357 Sunitinib versus lenvatinib plus pembrolizumab versus lenvatinib plus everolimus IMDC 27.2 63.9 100 4.2 31.9 38.1 14.0 9.2 98.5 71.8 
Escudier et al. [59] (2022) Observational 3,172 Sunitinib or pazopanib or others N/R N/R N/R N/R N/R N/R N/R N/R N/R N/R 21 N/R N/R 
Nikic et al. [25] (2022) Observational 673 Sunitinib or pazopanib MSKCC 32.6 67.4 N/R 100 3.4 21.7 51.8 8.9 14 17 N/R 27.3 
van de Laar et al. [24] (2022) Observational 34 Sunitinib or pazopanib or nivolumab plus ipilimumab or others IMDC 38 53 56 N/R N/R N/R N/R 6.9 28.1 N/R N/R 
Author, publication yearStudy typePts receiving sunitinib, nStudy drugs/comparisonsRisk score usedRisk stratification, %Percentage of ccRCC pts, %ORR, %PFS [months], medianOS [months], medianAE rate
fav.int.poorCRPRSDPDall, %grade 3/4, %
STAR-TOR: this data set Observational 702 Sunitinib or axitinib or temsirolimus MSKCC 12.6 62.2 25.2 81.6 6.0 24.8 33.0 21.8 6.7 25.0 66.3 13.9 
Motzer et al. [15] (2009) Ph III RCT 375 Sunitinib versus IFN-α MSKCC 38 56 100 44 40 11.0 26.4 N/R1 N/R 
Motzer et al. [45] (2012) Ph II RCT 1462 Sunitinib only MSKCC 25 60 15 100 32 43 N/R 8.5 23.1 N/R N/R 
Motzer et al. [16] (2013) Ph III RCT 548 Sunitinib versus pazopanib MSKCC (IMDC) 27 (25) 59 (26) 9 (17) 100 <1 24 44 19 9.5 29.3 99 57/173 
Miyake et al. [21] (2014) Observational 102 Sunitinib only MSKCC 25.5 50.9 23.6 96 28 65 15 7.8 33.2 100 N/R 
Motzer et al. [46] (2014) Ph II RCT 233 Everolimus followed by sunitinib versus sunitinib followed by everolimus MSKCC 30 56 14 85 25 52 14 10.7 32.0 N/R N/R 
Schnadig et al. [27] (2014) Observational 134 Sunitinib only MSKCC (IMDC) 11.2 (4.5) 28.4 (56.7) 8.2 (23.1) 82.8 2.2 14.2 18.7 N/R 7.5 15.5 85.1 N/R 
Eichelberg et al. [17] (2015) Ph III RCT 183 Sorafenib followed by sunitinib versus sunitinib followed by sorafenib MSKCC 45 51 0.5 84 3.4 26 35 N/R 8.5 30.2 N/R 58.5 
Gore et al. [30] (2015) Expanded access trial 4,543 Sunitinib only MSKCC 20 33 26 88 14 45 19 9.4 18.7 95 N/R 
Pan et al. [47] (2015) Observational 50 Sunitinib only MSKCC 20 56 24 84.6 10 28 58 9.4 N/R N/R N/R 
Tan et al. [29] (2015) Observational 1114 Sunitinib only MSKCC (IMDC) 11.3 (13.4) 46.4 (51.3) 42.3 (35.3) 82.7 31.44 34.34 34.34 7.94 21.84 N/R 58.6 
Ruiz-Morales et al. [48] (2016) Observational 6,519 Sunitinib versus pazopanib IMDC 23 57 20 90 N/R N/R N/R N/R 8.4 22.3 N/R N/R 
Rini et al. [49] (2016) Ph III RCT 135 Sunitinib versus pembrolizumab plus axitinib IMDC 26 71 100 N/R N/R N/R N/R 15.1 87 41 
Choueiri et al. [18] (2017) Ph II RCT 78 Sunitinib versus cabozantinib IMDC 80.9 19.1 100 11.5 42.3 25.6 5.6 21.8 98.6 68.1 
Noize et al. [22] (2017) Observational 302 Sunitinib only N/R N/R N/R N/R 83.1 2.3 28.8 40.1 18.5 8.4 23.6 97 58.3 
Lalani et al. [50] (2017) Observational 577 Sunitinib or pazopanib IMDC 22 58 20 83.7 N/R N/R N/R N/R 11.0 31.7 N/R N/R 
Zhang et al. [51] (2017) Observational 362 Sunitinib versus sorafenib MSKCC (IMDC) 41.7 (39.8) 53.0 (51.7) 5.2 (8.6) 91.4 1.1 19.9 67.4 11.6 10.0 24.0 N/R N/R 
Motzer et al. [52] (2018) Ph III RCT 546 Nivolumab plus ipilimumab versus sunitinib IMDC 23 61 16 100 25 45 17 8.4 26 97 63 
Poprach et al. [53] (2019) Observational 1,0166 Multiple TKIs MSKCC 27.8 64.7 7.6 95.3 5.1 23.0 38.2 22.2 10.8 31.9 30.1 7.7 
Choueiri et al. [54] (2019) Ph III RCT 328 Nivolumab plus cabozantinib versus sunitinib IMDC 22 57.3 20.7 100 4.6 22.6 42.1 13.7 8.3 99.1 70.6 
Motzer et al. [28] (2019) Ph III RCT 444 Sunitinib versus avelumab plus axitinib MSKCC (IMDC) 22.5 (21.6) 66.0 (62.2) 10.1 (16.0) 100 1.8 23.9 45.5 18.7 8.4 37.87 99.3 71.5 
Rini et al. [55] (2019) Ph III RCT 429 Pembrolizumab plus axitinib versus sunitinib IMDC 30.5 57.3 12.1 100 1.9 33.8 39.4 17.0 11.1 99.5 70.6 
Savard et al. [23] (2020) Retrospective database study 1,769 Sunitinib only IMDC 18 58 24 100 3.7 28.7 38.2 29.4 8.1 28.6 N/R N/R 
Schmidinger et al. [26] (2020) Observational 467 Sunitinib only MSKCC (IMDC) 16.6 (15.7) 29.1 (25.2) 7.5 (10.3) N/R N/R N/R N/R N/R 10.4 34.0 N/R N/R 
Choueiri et al. [56] (2021) Ph III RCT 328 Sunitinib versus nivolumab plus cabozantinib IMDC 22.0 57.3 20.7 100 4.6 22.6 42.1 13.7 8.3 34.38 99.1 70.6 
Costello et al. [57] (2021) Observational 75 Sunitinib or pazopanib IMDC 14.7 65.3 20.0 78,5 N/R N/R N/R N/R 5.69 26.2 N/R N/R 
Motzer et al. [58] (2021) Ph III RCT 357 Sunitinib versus lenvatinib plus pembrolizumab versus lenvatinib plus everolimus IMDC 27.2 63.9 100 4.2 31.9 38.1 14.0 9.2 98.5 71.8 
Escudier et al. [59] (2022) Observational 3,172 Sunitinib or pazopanib or others N/R N/R N/R N/R N/R N/R N/R N/R N/R N/R 21 N/R N/R 
Nikic et al. [25] (2022) Observational 673 Sunitinib or pazopanib MSKCC 32.6 67.4 N/R 100 3.4 21.7 51.8 8.9 14 17 N/R 27.3 
van de Laar et al. [24] (2022) Observational 34 Sunitinib or pazopanib or nivolumab plus ipilimumab or others IMDC 38 53 56 N/R N/R N/R N/R 6.9 28.1 N/R N/R 

AE, adverse event; ccRCC, clear cell renal cell carcinoma; CR, complete remission; N/R, not reported; ORR, overall response rate; OS, overall survival; PD, progressive disease; PFS, progression free survival; PR, partial remission; RCT, randomized controlled trial; SD, stable disease.

1Not reported.

2Schedule 4/2 patients.

3grade 3 and grade 4, respectively.

437.5 mg/d sunitinib for 4 weeks, then 2 weeks off, only first-line pts.

5Not reached.

6Only pts <70 years.

7Haanen et al., J Clin Oncol 39, no.15_suppl (May 20, 2021) 4574.

8Powles et al., J Clin Oncol 40, no. 6_suppl (Feb 20, 2022) 350.

9Time to treatment discontinuation.

Survival Endpoints

Comparing the STAR-TOR real-world data to the first phase III trial published by Motzer et al. [3, 15] in 2007 and 2009, it strives the reader’s attention that the clinical efficacy is inferior with a PFS of 6.7 months (OS 25 months) in the STAR-TOR registry compared to 11 months (OS 26.4 months) published by Motzer et al. [3, 15]. A similar, albeit not as pronounced trend can be observed for almost all subsequent sunitinib trials: PFS usually ranged from 8 to 11 months. We performed an explorative analysis according to histology which may explain the detrimental effect of non-clear cell histologies for this finding: clear cell a/mRCC patients yielded a PFS of 7.4 months compared to 5.0 months for all other histologies. These 7.4 months (7.7 months for 477 first-line ccRCCs) do not markedly differ from the results of other published real-world data [21‒24]. Only Nikic et al. [25] and Schmidinger et al. [26] reported relevantly larger PFS of 10.4 and 14 months. For OS, time ranged from 15.5 to up to 37.8 months [27, 28]. Here, our analyses yielded a survival of 25 months that might be situated close to the mean. According to our subgroup analysis, clear cell a/mRCC patients had a median OS of 25.7 months compared to non-ccRCC patients with 20.9 months. Furthermore, the reason for the short PFS in our study might be due to the large proportion of patients with poor risk of disease which was 25% compared to 6% in the studies by Motzer et al. [3, 15]. Similarly, most of the other studies derived their data from participants with better prognosis scores. Yet, studies with proportions of poor-risk patients comparable to the STAR-TOR data (around 25%) showed comparable OS and PFS [21, 23, 24, 29, 30].

It is widely accepted and seems to be common knowledge in oncology that the therapeutic efficacy of antineoplastic agents decreases with treatment line. This was exemplarily shown in the SWITCH trial which compared the sequential treatment of a/mRCC patients with sunitinib followed by sorafenib and vice versa [17]. While sunitinib in this trial in first line yielded a median PFS of 8.5 months, the PFS deteriorated to 5.4 months when used in second-line after first-line sorafenib. For sorafenib, this effect was even worse. Another example for this widely known effect was the GERCOR study published by Tournigand et al. [31] in 2004 for the sequential treatment in colorectal cancer. Therefore, we found it even more surprising that we could not observe this phenomenon in our data set. As is shown in Table 3 and was outlined in the Results part of our publication, we observed no relevant deterioration of clinical efficacy of sunitinib until the fourth treatment line.

Subgroup Analyses

Our analyses showed that the survival results are very different according to histologic subtype with ccRCC reaching a numerically longer survival compared to non-ccRCC. This corresponds to findings of the meta-analysis by Vera-Badillo et al. [32]. Similarly, Noize et al. [22] showed better survival for ccRCC patients upon treatment with sunitinib. A prospective trial published by Tannir et al. [33] in 2012 showed a median PFS of 2.7 months in a mixed group of non-clear cell a/mRCC patients, whereas Armstrong et al. [34] showed in the ASPEN trial a median PFS of 8.3 months (OS 31.5 months) for sunitinib-treated patients. Later, Tannir et al. [35] detected in 2016 a median PFS of 6.1 months (OS not reached at the time of publication) for sunitinib in the ESPN trial. Obviously, the treatment effect in non-clear cell a/mRCC depends very much on the variety of treated histological subtypes. The STAR-TOR non-clear cell cohort with predominantly papillary histology yielded a median PFS of 5.0 months and a median OS of 20.9 months compared relatively well in this context.

With respect to BMI, our data confirm low BMI (<25) to be a negative predictor for OS and PFS. This has been shown previously for TKI treatment by several studies [36‒39]. In correspondence to these observations, it has been shown that patients with BMI <25 seem to experience more dose-limiting toxicities [40].

Similarly, MSKCC risk score as a well-established predictor of survival has been clearly confirmed by our data [21, 41]. Our study confirmed patients’ serum LDH levels were identified as a prognostic marker for OS and PFS with lower LDH levels being associated with better survival. This has been described by Motzer et al. [42] for sunitinib. In addition, high serum LDH levels are well known to be associated with poor survival in other solid tumor entities [43].

Lastly, in our study population, patients with different nephrectomy statuses had different survival times: whereas median PFS and OS were very similar for patients with nephrectomy before the diagnosis of metastatic disease and patients with already known metastatic disease at the time point of surgery, shorter survival times were detected for patients without nephrectomy. This effect seemed to be at least partially driven by the MSKCC risk score, with a higher proportion of poor-risk MSKCC patients not receiving nephrectomy. As our findings are based on retrospective cohort study data, the intention of the nephrectomy such as a “cytoreductive” surgery is impossible to determine. Comparisons to studies specifically evaluating cytoreductive nephrectomy should be made with caution, but we believe that our results reinforce the claim for further evidence [44].

Adverse Events

Regarding sunitinib-related AEs, the real-world registry character of our study must be taken into account. On one hand, the event rates (in total 66.1%, grade 3/4 13.8%) of the three predominant types of AEs (gastrointestinal disorders, general disorders, and skin and subcutaneous tissue disorders) are also high in other real-world data sets: Miyake et al. [21] reported even 100% AEs of all grades with similar symptoms but mentioning hypothyroidism and hypertension as being equally frequent. Grade 3/4 AEs were reported as follows: thrombocytopenia 53.6%, leucopenia 20.9%, fatigue 20%, hand-foot syndrome 13.6%, and arterial hypertension 10.9%.

Yet, the STAR-TOR data seem to underestimate grade 3/4 AEs: Nikic et al. [25] report grade 3/4 AEs in 27.3% of the patients, whereas Noize et al. [22] described grade 3/4 AEs in 58.3% of the patients. Even these rates of real-world data are often much lower than AE rates reported by randomized controlled trials. As shown by Table 5, the rates of grade 3/4 AEs usually range between almost 60 and around 70%. The reason for these discrepancies might be the high prevalence of already pretreated hypertension in a real-world setting, masking sunitinib side effects.

Limitations

Several limitations must be considered when interpreting our results. First, several variables suffer from missing data. Particularly, MSKCC score was not available in 30.2% of the patients and therefore may have hampered our statistical testing. Missing data are a common issue in real-world studies: Schmidinger et al. [26] similarly report missingness in 49% of the patients. Van Laar reported missing performance status in 41% of the patients [24]. Several here-discussed real-world studies did not even transparently report missing data [21, 23, 25]. Nevertheless, it must be repeated that missingness at random cannot be assumed and that some of the results might be distorted by data unavailability. However, the monitoring of participating STAR-TOR centers may have reduced the clinically relevant bias.

Second, our study and many phase IV observational studies are prone to some extent of reporting bias. Even though all STAR-TOR study sites were supposed to use RECIST 1.1. criteria, some of the outcome measures may lack precision. Similarly, some physicians might have omitted using CTCAE for documentation of AEs. Also, in both cases the random monitoring visits of the study centers should have diminished the extent of bias.

Nevertheless, the STAR-TOR data on sunitinib treatment must be considered as an important step toward complete understanding for treatment patterns and response in clinical routine. The study size together with the prospective data collection and the possibility of subgroup analyses are unique. Its study population with a high proportion of patients with unfavorable MSKCC poor-risk score paves the way for comparisons yet to be published real-world data from the ICI combination era. It therefore will facilitate the development of clinical decision models evaluating the patients’ fitness for ICI therapy.

This prospective real-world data study adds further evidence to the persisting relevance of sunitinib in treatment of patients with a/mRCC who cannot receive or tolerate ICIs. The study population includes a high proportion of patients with unfavorable MSKCC poor-risk score, but shows still reasonably good PFS and OS results, while the drug demonstrates a favorable safety profile. Subgroup analyses confirmed worse prognosis score according to MSKCC, low BMI, and high LDH levels to be negative predictive markers for sunitinib effectiveness, what should be considered for clinical decision models.

The STAR-TOR registry received prior approval by the Ethics Committee of Münster University Hospital in 2007 (Nr.2007-484-f-S) and was registered in the US library of medicine database (NCT00700258). The study received prior ethical approval (Ethics Committee of Münster University Hospital in 2007 [Nr.2007-484-f-S]). Written informed consent was obtained from all participants (or their parent/legal guardian/next of kin) to participate in the study. All the research was conducted ethically in accordance with the World Medical Association Declaration of Helsinki.

Michael Woike is an employee of Pfizer Germany. Annemarie Uhlig has received honoraria, travel, and accommodations by Pfizer and APOGEPHA. Katrin Schlack received fees as speaker, consultant, or advisor and travel expenses from APOGEPHA, BMS, Eisai, EUSA Pharma, Ipsen, Merck Healthcare, MSD, Novartis, and Pfizer. Johannes Uhlig, Thomas Fischer, Lutz Trojan, Lothar Bergmann, Martin Bögemann, Peter J. Goebell, Michael Rink, Mathias Reichert, Marianne Leitsmann, and Arne Strauß have no conflicts of interest to disclose.

The STAR-TOR registry was funded by Pfizer Pharma GmbH, Berlin, Germany.

Conceptualization of the research project: A.U., J.U., M.W., M.L., and A.S. Design of the methodology and contribution to data analysis: A.U., J.U., M.W., and T.F. Data collection: L.T., L.B., M.B., P.J.G., M.Ri., M.Re., K.S., M.L., and A.S. Data curation: M.W. and T.F. Visualization: T.F. and A.U. Writing of the manuscript: A.U., J.U., M.W., M.L., M.Ri., and M.Re. Reviewing and editing: T.F., L.T., L.B., M.B., P.J.G., M.R., K.S., M.Ri., and M.Re.

The data that support the findings of this study are not publicly available for company data restriction regulations. Pfizer is the owner of these data and does not disclose patient-related information. Requests regarding the availability of data that support the findings of this study may be directed to Pfizer Pharma GmbH, Friedrichstrasse 110, 10117 Berlin, Germany.

1.
Ljungberg
B
,
Albiges
L
,
Abu-Ghanem
Y
,
Bedke
J
,
Capitanio
U
,
Dabestani
S
, et al
.
European association of urology guidelines on renal cell carcinoma: the 2022 update
.
Eur Urol
.
2022
;
82
(
4
):
399
410
. .
2.
Motzer
RJ
,
Rini
BI
,
Bukowski
RM
,
Curti
BD
,
George
DJ
,
Hudes
GR
, et al
.
Sunitinib in patients with metastatic renal cell carcinoma
.
JAMA
.
2006
;
295
(
21
):
2516
24
. .
3.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Rixe
O
, et al
.
Sunitinib versus interferon alfa in metastatic renal-cell carcinoma
.
N Engl J Med
.
2007
;
356
(
2
):
115
24
. .
4.
Gore
ME
,
Szczylik
C
,
Porta
C
,
Bracarda
S
,
Bjarnason
GA
,
Oudard
S
, et al
.
Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial
.
Lancet Oncol
.
2009
;
10
(
8
):
757
63
. .
5.
Boegemann
M
,
Hubbe
M
,
Thomaidou
D
,
Blackburn
S
,
Bent-Ennakhil
N
,
Wood
R
, et al
.
Sunitinib treatment modification in first-line metastatic renal cell carcinoma: analysis of the STAR-TOR registry
.
Anticancer Res
.
2018
;
38
(
11
):
6413
22
. .
6.
Schrader
AJ
,
Seseke
S
,
Keil
C
,
Herrmann
E
,
Goebell
PJ
,
Weikert
S
, et al
.
Temsirolimus in daily use: results of a prospective multicentre noninterventional study of patients with metastatic kidney cancer
.
Eur Urol
.
2014
;
66
(
2
):
275
81
. .
7.
Boegemann
M
,
Goebell
PJ
,
Woike
M
,
Buncke
J
,
Schlack
K
,
Schrader
AJ
.
Assessment of prognosis by established prognosis scores and physicians’ judgement in mRCC patients: an analysis of the STAR-TOR registry
.
Transl Androl Urol
.
2021
;
10
:
4062
74
. .
8.
Uhlig
A
,
Uhlig
J
,
Trojan
L
,
Woike
M
,
Leitsmann
M
,
Strauß
A
.
Toxicities of axitinib, sunitinib and temsirolimus: implications for progression-free and overall survival in metastatic renal cell cancer
.
Future Oncol
.
2021
;
17
(
1
):
45
56
. .
9.
Strauss
A
,
Schmid
M
,
Rink
M
,
Moran
M
,
Bernhardt
S
,
Hubbe
M
, et al
.
Real-world outcomes in patients with metastatic renal cell carcinoma according to risk factors: the STAR-TOR registry
.
Future Oncol
.
2021
;
17
(
18
):
2325
38
. .
10.
Motzer
RJ
,
Mazumdar
M
,
Bacik
J
,
Berg
W
,
Amsterdam
A
,
Ferrara
J
.
Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma
.
J Clin Oncol
.
1999
;
17
(
8
):
2530
40
. .
11.
Motzer
RJ
,
Bacik
J
,
Schwartz
LH
,
Reuter
V
,
Russo
P
,
Marion
S
, et al
.
Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma
.
J Clin Oncol
.
2004
;
22
(
3
):
454
63
. .
12.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
.
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
.
2009
;
45
(
2
):
228
47
. .
13.
Brown
EG
,
Wood
L
,
Wood
S
.
The medical dictionary for regulatory activities (MedDRA)
.
Drug Saf
.
1999
;
20
(
2
):
109
17
. .
14.
Health UDo
,
Services
H
.
Common terminology criteria for adverse events. Version 5.0
. Published November 27, 2017. In:
2020
.
15.
Motzer
RJ
,
Hutson
TE
,
Tomczak
P
,
Michaelson
MD
,
Bukowski
RM
,
Oudard
S
, et al
.
Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma
.
J Clin Oncol
.
2009
;
27
(
22
):
3584
90
. .
16.
Motzer
RJ
,
Hutson
TE
,
Cella
D
,
Reeves
J
,
Hawkins
R
,
Guo
J
, et al
.
Pazopanib versus sunitinib in metastatic renal-cell carcinoma
.
N Engl J Med
.
2013
;
369
(
8
):
722
31
. .
17.
Eichelberg
C
,
Vervenne
WL
,
De Santis
M
,
Fischer von Weikersthal
L
,
Goebell
PJ
,
Lerchenmüller
C
, et al
.
SWITCH: a randomised, sequential, open-label study to evaluate the efficacy and safety of sorafenib-sunitinib versus sunitinib-sorafenib in the treatment of metastatic renal cell cancer
.
Eur Urol
.
2015
;
68
(
5
):
837
47
. .
18.
Choueiri
TK
,
Halabi
S
,
Sanford
BL
,
Hahn
O
,
Michaelson
MD
,
Walsh
MK
, et al
.
Cabozantinib versus sunitinib as initial targeted therapy for patients with metastatic renal cell carcinoma of poor or intermediate risk: the alliance A031203 CABOSUN trial
.
J Clin Oncol
.
2017
;
35
(
6
):
591
7
. .
19.
Escudier
B
,
Porta
C
,
Bono
P
,
Powles
T
,
Eisen
T
,
Sternberg
CN
, et al
.
Randomized, controlled, double-blind, cross-over trial assessing treatment preference for pazopanib versus sunitinib in patients with metastatic renal cell carcinoma: PISCES Study
.
J Clin Oncol
.
2014
;
32
(
14
):
1412
8
. .
20.
Moran
M
,
Nickens
D
,
Adcock
K
,
Bennetts
M
,
Desscan
A
,
Charnley
N
, et al
.
Sunitinib for metastatic renal cell carcinoma: a systematic review and meta-analysis of real-world and clinical trials data
.
Target Oncol
.
2019
;
14
(
4
):
405
16
. .
21.
Miyake
H
,
Miyazaki
A
,
Harada
K
,
Fujisawa
M
.
Assessment of efficacy, safety and quality of life of 110 patients treated with sunitinib as first-line therapy for metastatic renal cell carcinoma: experience in real-world clinical practice in Japan
.
Med Oncol
.
2014
;
31
(
6
):
978
. .
22.
Noize
P
,
Grelaud
A
,
Bay
JO
,
Chevreau
C
,
Gross-Goupil
M
,
Culine
S
, et al
.
Real-life patterns of use, safety and effectiveness of sunitinib in first-line therapy of metastatic renal cell carcinoma: the SANTORIN cohort study
.
Pharmacoepidemiol Drug Saf
.
2017
;
26
(
12
):
1561
9
. .
23.
Savard
MF
,
Wells
JC
,
Graham
J
,
Dudani
S
,
Steinharter
JA
,
McGregor
BA
, et al
.
Real-world assessment of clinical outcomes among first-line sunitinib patients with clear cell metastatic renal cell carcinoma (mRCC) by the international mRCC database Consortium risk group
.
Oncologist
.
2020
;
25
(
5
):
422
30
. .
24.
van Laar
SA
,
Gombert-Handoko
KB
,
Groenwold
RHH
,
van der Hulle
T
,
Visser
LE
,
Houtsma
D
, et al
.
Real-world metastatic renal cell carcinoma treatment patterns and clinical outcomes in The Netherlands
.
Front Pharmacol
.
2022
;
13
:
803935
. .
25.
Nikic
P
,
Babovic
N
,
Dzamic
Z
,
Salma
S
,
Stojanovic
V
,
Matkovic
S
, et al
.
Real world overall survival of patients with metastatic renal cell carcinoma treated with only available sunitinib and pazopanib in first-line setting
.
Front Oncol
.
2022
;
12
:
892156
. .
26.
Schmidinger
M
,
Porta
C
,
Oudard
S
,
Denechere
G
,
Brault
Y
,
Serfass
L
, et al
.
Real-world experience with sunitinib treatment in patients with metastatic renal cell carcinoma: clinical outcome according to risk score
.
Clin Genitourin Cancer
.
2020
;
18
(
5
):
e588
97
. .
27.
Schnadig
ID
,
Hutson
TE
,
Chung
H
,
Dhanda
R
,
Halm
M
,
Forsyth
M
, et al
.
Dosing patterns, toxicity, and outcomes in patients treated with first-line sunitinib for advanced renal cell carcinoma in community-based practices
.
Clin Genitourin Cancer
.
2014
;
12
(
6
):
413
21
. .
28.
Motzer
RJ
,
Penkov
K
,
Haanen
J
,
Rini
B
,
Albiges
L
,
Campbell
MT
, et al
.
Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
.
2019
;
380
(
12
):
1103
15
. .
29.
Tan
HS
,
Li
H
,
Hong
YW
,
Toh
CK
,
Wong
A
,
Lopes
G
, et al
.
Efficacy and safety of an attenuated-dose sunitinib regimen in metastatic renal cell carcinoma: results from a prospective registry in Singapore
.
Clin Genitourin Cancer
.
2015
;
13
(
4
):
e285
95
. .
30.
Gore
ME
,
Szczylik
C
,
Porta
C
,
Bracarda
S
,
Bjarnason
GA
,
Oudard
S
, et al
.
Final results from the large sunitinib global expanded-access trial in metastatic renal cell carcinoma
.
Br J Cancer
.
2015
;
113
(
1
):
12
9
. .
31.
Tournigand
C
,
André
T
,
Achille
E
,
Lledo
G
,
Flesh
M
,
Mery-Mignard
D
, et al
.
FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study
.
J Clin Oncol
.
2004
;
22
(
2
):
229
37
. .
32.
Vera-Badillo
FE
,
Templeton
AJ
,
Duran
I
,
Ocana
A
,
de Gouveia
P
,
Aneja
P
, et al
.
Systemic therapy for non–clear cell renal cell carcinomas: a systematic review and meta-analysis
.
Eur Urol
.
2015
;
67
(
4
):
740
9
. .
33.
Tannir
NM
,
Plimack
E
,
Ng
C
,
Tamboli
P
,
Bekele
NB
,
Xiao
L
, et al
.
A phase 2 trial of sunitinib in patients with advanced non-clear cell renal cell carcinoma
.
Eur Urol
.
2012
;
62
(
6
):
1013
9
. .
34.
Armstrong
AJ
,
Halabi
S
,
Eisen
T
,
Broderick
S
,
Stadler
WM
,
Jones
RJ
, et al
.
Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (ASPEN): a multicentre, open-label, randomised phase 2 trial
.
Lancet Oncol
.
2016
;
17
(
3
):
378
88
. .
35.
Tannir
NM
,
Jonasch
E
,
Albiges
L
,
Altinmakas
E
,
Ng
CS
,
Matin
SF
, et al
.
Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (ESPN): a randomized multicenter phase 2 trial
.
Eur Urol
.
2016
;
69
(
5
):
866
74
. .
36.
Goebell
PJ
,
Müller
L
,
Hübner
A
,
Harich
HD
,
Boller
E
,
Koska
M
, et al
.
Body mass index as independent predictor of overall survival in patients with advanced renal cell carcinoma at start of systemic treatment—analyses from the German clinical RCC-Registry
.
Urol Oncol
.
2018
;
36
(
10
):
470.e1
e9
. .
37.
Albiges
L
,
Hakimi
AA
,
Xie
W
,
McKay
RR
,
Simantov
R
,
Lin
X
, et al
.
Body mass index and metastatic renal cell carcinoma: clinical and biological correlations
.
J Clin Oncol
.
2016
;
34
(
30
):
3655
63
. .
38.
Takemoto
K
,
Sekino
Y
,
Kitano
H
,
Ikeda
K
,
Goto
K
,
Hieda
K
, et al
.
Impact of low and high body mass index on predicting therapeutic efficacy and prognosis in patients with metastatic renal cell carcinoma treated with first-line tyrosine kinase inhibitors
.
Int J Urol
.
2023
;
30
(
3
):
319
27
. .
39.
Steffens
S
,
Grünwald
V
,
Ringe
KI
,
Seidel
C
,
Eggers
H
,
Schrader
M
, et al
.
Does obesity influence the prognosis of metastatic renal cell carcinoma in patients treated with vascular endothelial growth factor-targeted therapy
.
Oncologist
.
2011
;
16
(
11
):
1565
71
. .
40.
Huillard
O
,
Mir
O
,
Peyromaure
M
,
Tlemsani
C
,
Giroux
J
,
Boudou-Rouquette
P
, et al
.
Sarcopenia and body mass index predict sunitinib-induced early dose-limiting toxicities in renal cancer patients
.
Br J Cancer
.
2013
;
108
(
5
):
1034
41
. .
41.
Fiala
O
,
Finek
J
,
Poprach
A
,
Melichar
B
,
Kopecký
J
,
Zemanova
M
, et al
.
Outcomes according to MSKCC risk score with focus on the intermediate-risk group in metastatic renal cell carcinoma patients treated with first-line sunitinib: a retrospective analysis of 2,390 patients
.
Cancers
.
2020
;
12
(
4
):
808
. .
42.
Motzer
RJ
,
Bukowski
RM
,
Figlin
RA
,
Hutson
TE
,
Michaelson
MD
,
Kim
ST
, et al
.
Prognostic nomogram for sunitinib in patients with metastatic renal cell carcinoma
.
Cancer
.
2008
;
113
(
7
):
1552
8
. .
43.
Petrelli
F
,
Cabiddu
M
,
Coinu
A
,
Borgonovo
K
,
Ghilardi
M
,
Lonati
V
, et al
.
Prognostic role of lactate dehydrogenase in solid tumors: a systematic review and meta-analysis of 76 studies
.
Acta Oncol
.
2015
;
54
(
7
):
961
70
. .
44.
Méjean
A
,
Ravaud
A
,
Thezenas
S
,
Colas
S
,
Beauval
JB
,
Bensalah
K
, et al
.
Sunitinib alone or after nephrectomy in metastatic renal-cell carcinoma
.
N Engl J Med
.
2018
;
379
(
5
):
417
27
. .
45.
Motzer
RJ
,
Hutson
TE
,
Olsen
MR
,
Hudes
GR
,
Burke
JM
,
Edenfield
WJ
, et al
.
Randomized phase II trial of sunitinib on an intermittent versus continuous dosing schedule as first-line therapy for advanced renal cell carcinoma
.
J Clin Oncol
.
2012
;
30
(
12
):
1371
7
. .
46.
Motzer
RJ
,
Barrios
CH
,
Kim
TM
,
Falcon
S
,
Cosgriff
T
,
Harker
WG
, et al
.
Phase II randomized trial comparing sequential first-line everolimus and second-line sunitinib versus first-line sunitinib and second-line everolimus in patients with metastatic renal cell carcinoma
.
J Clin Oncol
.
2014
;
32
(
25
):
2765
72
. .
47.
Pan
X
,
Huang
H
,
Huang
Y
,
Liu
B
,
Cui
X
,
Gan
S
, et al
.
Sunitinib dosing schedule 2/1 improves tolerability, efficacy, and health-related quality of life in Chinese patients with metastatic renal cell carcinoma
.
Urol Oncol
.
2015
;
33
(
6
):
268.e9
2.68E15
. .
48.
Ruiz-Morales
JM
,
Swierkowski
M
,
Wells
JC
,
Fraccon
AP
,
Pasini
F
,
Donskov
F
, et al
.
First-line sunitinib versus pazopanib in metastatic renal cell carcinoma: results from the international metastatic renal cell carcinoma database consortium
.
Eur J Cancer
.
2016
;
65
:
102
8
. .
49.
Rini
BI
,
Dorff
TB
,
Elson
P
,
Rodriguez
CS
,
Shepard
D
,
Wood
L
, et al
.
Active surveillance in metastatic renal-cell carcinoma: a prospective, phase 2 trial
.
Lancet Oncol
.
2016
;
17
(
9
):
1317
24
. .
50.
Lalani
AKA
,
Li
H
,
Heng
DYC
,
Wood
L
,
Kalirai
A
,
Bjarnason
GA
, et al
.
First-line sunitinib or pazopanib in metastatic renal cell carcinoma: the Canadian experience
.
Can Urol Assoc J
.
2017
;
11
(
3–4
):
112
7
. .
51.
Zhang
HL
,
Sheng
XN
,
Li
XS
,
Wang
HK
,
Chi
ZH
,
He
ZS
, et al
.
Sorafenib versus sunitinib as first-line treatment agents in Chinese patients with metastatic renal cell carcinoma: the largest multicenter retrospective analysis of survival and prognostic factors
.
BMC Cancer
.
2017
;
17
(
1
):
16
. .
52.
Motzer
RJ
,
Tannir
NM
,
McDermott
DF
,
Arén Frontera
O
,
Melichar
B
,
Choueiri
TK
, et al
.
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
.
2018
;
378
(
14
):
1277
90
. .
53.
Poprach
A
,
Rumanova
K
,
Lakomý
R
,
Chloupková
R
,
Stanik
M
,
Pokrivcak
T
, et al
.
Tyrosine kinase inhibitors in the first-line treatment for metastatic nonclear cell renal carcinoma: a retrospective analysis of a national database
.
Urol Oncol
.
2019
;
37
(
4
):
294.e1
294.e8
. .
54.
Choueiri
TK
,
Powles
T
,
Burotto
M
,
Escudier
B
,
Bourlon
MT
,
Zurawski
B
, et al
.
Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
.
2021
;
384
(
9
):
829
41
. .
55.
Rini
BI
,
Plimack
ER
,
Stus
V
,
Gafanov
R
,
Hawkins
R
,
Nosov
D
, et al
.
Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
.
2019
;
380
(
12
):
1116
27
. .
56.
Choueiri
TK
,
Powles
T
,
Burotto
M
,
Escudier
B
,
Bourlon
MT
,
Zurawski
B
, et al
.
Nivolumab plus cabozantinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
.
2021
;
384
(
9
):
829
41
. .
57.
Costello
BA
,
Bhavsar
NA
,
Zakharia
Y
,
Pal
SK
,
Vaishampayan
U
,
Jim
H
, et al
.
A prospective multicenter evaluation of initial treatment choice in metastatic renal cell carcinoma prior to the immunotherapy era: the MaRCC registry experience
.
Clin Genitourin Cancer
.
2022
;
20
:
1
10
. .
58.
Motzer
R
,
Alekseev
B
,
Rha
S-Y
,
Porta
C
,
Eto
M
,
Powles
T
, et al
.
Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma
.
N Engl J Med
.
2021
;
384
(
14
):
1289
300
. .
59.
Escudier
B
,
de Zélicourt
M
,
Bourouina
R
,
Nevoret
C
,
Thiery-Vuillemin
A
.
Management and health resource use of patients with metastatic renal cell carcinoma treated with systemic therapy over 2014-2017 in France: a national real-world study
.
Clin Genitourin Cancer
.
2022
;
20
(
6
):
533
42
. .