Next Article in Journal
Physiotherapy in Prehabilitation for Bariatric Surgery—Analysis of Its Impact on Functional Capacity and Original Predictive Models of Functional Status Outcome
Next Article in Special Issue
Risk Stratification Using a Perioperative Nomogram for Predicting the Mortality of Bladder Cancer Patients Undergoing Radical Cystectomy
Previous Article in Journal
Digital Orthodontic Setups in Orthognathic Surgery: Evaluating Predictability and Precision of the Workflow in Surgical Planning
Previous Article in Special Issue
Lymphovascular Invasion Is a Predictor of Clinical Outcomes in Bladder Cancer Patients Treated with Radical Cystectomy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Decoding Treatment Failures in Metastatic Renal Cell Carcinoma: Predictors Across Immunotherapy and Targeted Therapies from a Retrospective Real-World Analysis

by
Sorin Saftescu
1,
Vlad-Norin Vornicu
1,
Dorel-Ionel Popovici
1,*,
Radu-Dumitru Dragomir
1,
Dana-Sonia Nagy
1,
Daniela-Lidia Sandu
1,
Ana Dulan
1,
Șerban-Mircea Negru
1 and
Alina-Gabriela Negru
2
1
Department of Oncology, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
2
Department of Cardiology, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(15), 5271; https://doi.org/10.3390/jcm14155271
Submission received: 13 June 2025 / Revised: 14 July 2025 / Accepted: 24 July 2025 / Published: 25 July 2025
(This article belongs to the Special Issue Advances and Perspectives in Cancer Diagnostics and Treatment)

Abstract

Background: Despite recent advances in the management of metastatic renal cell carcinoma (mRCC), real-world outcomes remain heterogeneous, and early treatment failure is common. Predictive biomarkers for time to treatment failure (TTF) outside clinical trials are poorly characterized. Objective: To identify clinical and laboratory predictors associated with early treatment failure in a real-world cohort of mRCC patients treated with immune checkpoint inhibitors (ICIs), tyrosine kinase inhibitors (TKIs), or combination regimens. Methods: We conducted a retrospective, single-center analysis of patients with metastatic non-urothelial RCC treated between 2018 and 2023. Cox proportional hazards regression was used to evaluate the association between baseline biological parameters and TTF for each treatment regimen. Results: Among 137 patients receiving first-line therapy, 50 received Ipilimumab + Nivolumab, 49 Sunitinib, and 17 Avelumab + Axitinib. For Ipilimumab + Nivolumab, elevated AST was significantly associated with shorter TTF. For Avelumab + Axitinib, shorter TTF was associated with lymph node metastases, low lymphocyte count, low creatinine, low BMI, and low hemoglobin. For Cabozantinib in subsequent lines, a higher platelet count, ALT, and presence of liver metastases were associated with shorter TTF. No statistically significant predictors were found for Nivolumab used in the second-line setting. Conclusions: Routine, accessible biomarkers such as AST, hemoglobin, lymphocyte count, and creatinine may serve as predictors of treatment failure in specific therapeutic contexts. These findings support risk-adapted strategies and individualized monitoring in real-world clinical practice, though further validation in larger cohorts is warranted.

1. Introduction

According to the European Cancer Information System (ECIS), in 2022, the incidence of renal cell carcinoma (RCC) in the EU-27 was estimated at 58,228 new cases in males (26.8 per 100,000, age-standardized rate [ASR]) and 37,493 in females (12.2 per 100,000 ASR), totaling 95,721 cases (20.3 per 100,000 ASR for both sexes). Projections for 2040 estimate a 15.19% increase in annual cases. The highest incidence in the EU is reported in Czechia (29.4 per 100,000) and the lowest in Cyprus (8.7 per 100,000). Historical data show a consistent rise in incidence, from 13.2 per 100,000 in 1976 to 20.3 in 2022 [1]. Similar trends are reported globally, with an average annual increase of 2–3% in most countries [2]. This increasing incidence is attributed to improved detection and lifestyle factors such as the rising prevalence of obesity [3], hypertension, and smoking—though smoking rates in the EU have decreased from 30.5% in 2010 to 26.4% in 2019 [4].
Key risk factors for RCC include obesity (RR = 1.35 in overweight patients; RR = 1.76 for BMI (body mass index) ≥ 30 kg/m2), accounting for ~26% of RCC cases [5], smoking (RR = 1.38) [6], hypertension (RR = 1.05 per +10 mmHg systolic, RR = 1.07 per +10 mmHg diastolic) [7], male sex (RR = 2) [8], diabetes (RR = 1.4) [8], chronic kidney disease (RR = 1.39) [9], and height: (RR = 1.3 per + 10 cm) [10]. Conversely, kidney stones, sodium intake, and fluid intake do not have statistically significant associations with RCC risk [11,12], and alcohol consumption may be protective (RR = 0.71) [13].
While only 3–5% of RCC cases are familial—with von Hippel–Lindau (VHL) disease, an autosomal dominant inherited disorder, accounting for approximately 1% of cases [14]—about 50–60% of sporadic cases harbor biallelic VHL inactivation. This inactivation is caused by mutations, loss of heterozygosity at 3p, or promoter hypermethylation. Overall, 90% of sporadic ccRCCs demonstrate some form of VHL inactivation [15,16].
Clear cell renal cell carcinoma (ccRCC) accounts for approximately 65–70% of adult renal carcinomas. Less common subtypes include papillary RCC (10–15%) and chromophobe RCC (5%) [17,18].
The current therapeutic landscape (NCCN Guidelines, Version 3, 2025, regimens category 1, 2A or 2B listed) is presented in Figure 1 [19].
Table 1 outlines the key clinical trials that reflect the historical evolution of therapeutic strategies in renal cell carcinoma.
When comparing IO–TKI to IO–IO combinations by efficacy trends, the progression-free survival (PFS) benefit of IO–TKI regimens appears greater. However, this advantage tends to diminish with longer follow-ups, and ongoing data maturation will clarify whether this trend significantly impacts long-term outcomes [28].
Real-world studies for mRCC indicate, for different cohorts (TKI, TKI+IO, IO–IO), a median duration of first-line treatment of 2.5–8.3 months and median survivals of between 39 and 42 months (Gu-Shun Lai et al., 2024), revealing a high proportion of cases with early treatment failure.
Real-world predictive factors identified included patients with bone or liver metastases, high C-reactive protein (CRP), paraneoplastic symptoms, or, without prior cytoreductive nephrectomy experience, shorter PFS on Ipilimumab + Nivolumab [29]. A large real-world analysis (n = 455) reported a median PFS of 13.5 months and a median OS of 51.5 months with Ipilimumab + Nivolumab. Predictors of response included male sex (OR 2.0, p = 0.017), prior nephrectomy (OR 2.1, p = 0.001), and lung metastasis (OR 2.4, p = 0.001). Better ECOG PS (0 or 1), IMDC intermediate risk, and completion of four induction cycles were also favorable, while antibiotic use before immunotherapy predicted a worse OS [30]. A smaller retrospective series (n = 48) suggested that normal or normalized CRP during treatment predicted longer PFS with Nivolumab + Ipilimumab [31]. High CRP (>1.27 mg/dL) also predicted shorter PFS in patients treated with Cabozantinib (n = 53), with a hazard ratio of 6.9 in univariate analysis, as was demonstrated in a real-world Japanese cohort study evaluating Cabozantinib outcomes (Iinuma et al., Biomedicines 2022, 10, 3172)
Over the past two decades, the management of metastatic renal cell carcinoma (mRCC) has evolved from a primary reliance on cytoreductive nephrectomy (CN) to a multimodal strategy increasingly centered on systemic therapies. Although observational and randomized studies have evaluated the role of CN in the era of targeted agents, optimal timing and patient selection remain matters of ongoing debate, highlighting the need for individualized, multidisciplinary treatment planning [32].

2. Materials and Methods

The objective of this study was to identify clinical and laboratory predictors associated with early treatment failure in patients with metastatic renal cell carcinoma (mRCC) treated with immune checkpoint inhibitors (ICIs), tyrosine kinase inhibitors (TKIs), or combination regimens in a real-world setting.
This retrospective single-center, observational cohort study was conducted at the OncoHelp Medical Center, Timișoara, Romania, a tertiary oncology referral center for patients with non-urothelial metastatic renal cell carcinoma (mRCC) treated in first and subsequent lines with ipilimumab + nivolumab, cabozantinib, sunitinib, pazopanib, nivolumab, or avelumab + axitinib at OncoHelp Hospital, Timisoara, Romania. The study fully complied with the ethical principles outlined in the Declaration of Helsinki, and ethical approval was obtained from the Ethics Committee of the OncoHelp Medical Center prior to study initiations (approval number 1178/2 May 2024).
Patients included in this retrospective study had histologically confirmed renal cell carcinoma with radiologically documented metastatic disease (M1), received at least one line of systemic therapy (ICI, TKI, or combinations) between February 2021 and July 2024, and had available baseline clinical and laboratory data; patients with non-metastatic disease, non-RCC histologies, or incomplete baseline records were excluded. No exclusion was applied based on comorbidities; the cohort included patients with various chronic conditions, such as cardiovascular, metabolic, or renal disorders.
Clinical and pathological data collected included age, sex, Eastern Cooperative Oncology Group (ECOG) performance status, histological subtype, prior treatment regimens, metastatic site locations, baseline complete blood count (CBC), and biochemistry at the start of each treatment line. We focused on routinely available clinical and laboratory parameters—such as AST, creatinine, lymphocyte count, and hemoglobin—given their widespread use in oncology practice, accessibility across care settings, and previous associations with prognosis or systemic treatment outcomes in solid tumors.
The diagnosis of renal cell carcinoma was confirmed in all patients through histopathological examination, based on the criteria outlined in the World Health Organization (WHO) Classification of Urinary and Male Genital Tumours, 5th edition (2022). All patients had radiological evidence of distant metastases (M1) documented prior to treatment initiation, confirmed by CT, MRI, or PET-CT according to institutional protocols.
The study analyzed 3576 treatment evaluations involving 137 unique patients diagnosed with ICD-10 C64 (renal cell carcinoma). For accurate treatment sequencing, 39 treatment lines initiated before the study period and 188 treatment lines initiated during the study period were identified and included in the analysis. Time to Treatment Failure (TTF) was defined as the time from the first to the last administration of a treatment line, with treatment discontinuation due to any cause (disease progression, toxicity, or death) considered as failure (Collett, D.—Modelling Survival Data in Medical Research, 3rd edition, CRC Press, 2015). The on-treatment status at the data cut-off was also recorded.
The primary objective was to identify predictive factors for prolonged time to treatment failure (TTF) for each therapeutic regimen. To achieve this, Cox proportional hazards models were constructed using Cox Proportional Hazards Survival Regression, based on baseline biological parameters obtained prior to the initiation of each treatment line.
No formal control group was included in this retrospective observational study. The analysis was descriptive and exploratory, focusing on identifying predictors of early treatment failure across different treatment cohorts. No blinding of participants or investigators was performed, as this was a retrospective observational study based on routinely collected real-world data.
No long-term clinical follow-up was conducted beyond treatment discontinuation, as the primary endpoint was time to treatment failure.
Cox proportional hazards regression analyses were performed using the online calculator available at https://statpages.info/prophaz.html, accessed on 22 March 2025, which implements the method described by Lee and Wang (2003). A two-tailed p-value of <0.05 was considered statistically significant. Given the small sample sizes in some subgroups and the potential interdependence between clinical and laboratory variables, we limited the analysis to univariate Cox models to avoid overfitting and unstable estimates. The proportional hazards assumption was not formally tested due to the exploratory nature of the analysis and the retrospective data structure. However, the short time-to-failure intervals and limited follow-up duration reduce the likelihood of major time-varying effects.
Patients who were still on treatment at the time of data cut-off or were lost to follow-up were censored at their last known treatment date. The Cox proportional hazards models accounted for right-censored data using standard partial likelihood estimation. No formal sensitivity analyses were conducted for censored observations due to the limited sample size.
Continuous variables were analyzed as continuous measures and, in selected cases, categorized based on clinical cut-offs (example: AST > 25). Categorization was applied to facilitate interpretation and enable analysis using the Cox proportional hazards model. For analytical purposes, the presence or absence of metastases in the liver, lymph nodes, lungs, or bones was treated as a binary variable (yes/no). The IMDC prognostic score was not applied due to missing data for key components (e.g., calcium, neutrophil count, performance status) in a significant proportion of cases.

3. Results

The study included 137 patients, consisting of 95 men (69.3%) and 42 women (30.7%), receiving a total of 227 lines of treatment: 137 first-line treatments, 65 second-line treatments, and 25 third- or fourth-line treatments. The longest ongoing treatments were initiated up to 11 years ago (2013), predominantly in patients treated with sunitinib. Of the 227 treatment lines, 47 were ongoing at the time of data cut-off, while 180 were completed due to progression, intolerance, or death. The duration of the completed treatment lines ranged from 14 days (single administration) to 4017 days of treatment.
Table 2 provides the average age of patients receiving different systemic therapies, stratified by treatment line and regimen. The overall average age was slightly lower in second- and later-line settings (59.8 years) compared to first-line treatments (60.8 years), with Everolimus being administered to the youngest subgroup (average 45.3 years) and Axitinib + Avelumab to the oldest (65.9 years).
Sex Distribution by Treatment Line: first-line treatments included 93 male patients out of 137 (67.8%) and second to fourth-line treatments included 48 male patients out of 64 (75.0%). The difference did not reach statistical significance in a Z-test (p = 0.27).
Treatment Outcomes (TTF):
Table 3, Table 4, Table 5 and Table 6 present the average, median, and range of time to treatment failure (TTF) for each treatment line and medication evaluated.
Table 3 summarizes time to treatment failure (TTF) outcomes for each first-line regimen used in the cohort. Among first-line therapies, Sunitinib and Pazopanib were associated with the longest median and average TTFs, while Ipilimumab + Nivolumab and Avelumab + Axitinib showed shorter durations. The shortest TTF was observed with Tivozanib and Temsirolimus, which were rarely used.
Table 4 presents time to treatment failure (TTF) data for second-line therapies in patients with mRCC. Nivolumab and Axitinib showed the longest average TTFs among regimens used more frequently, while Cabozantinib was associated with notably shorter treatment durations. Rarely used combinations, such as Nivolumab + Axitinib, showed prolonged TTF in isolated cases.
Table 5 shows time to treatment failure (TTF) for third-line therapies administered in the cohort. Among agents used more than once, Nivolumab had the longest average TTF, while Cabozantinib demonstrated a shorter treatment duration. Everolimus showed moderate performance in two patients, and data for Axitinib were insufficient for interpretation.
Table 6 presents time to treatment failure (TTF) for fourth-line therapies in a limited subset of patients. Nivolumab was the only agent administered in this setting, with a median and average TTF of 611 days (20.3 months), suggesting prolonged benefit in selected patients despite advanced treatment lines.
Predictive Factors Analysis:
Clear cell RCC demonstrated better TTF compared to non-clear cell histologies (RR = 0.66, p = 0.0367).
Table 7 shows the results of univariate Cox regression analysis for baseline predictors of time to treatment failure (TTF) in patients receiving Ipilimumab + Nivolumab. Among the variables analyzed, elevated AST (either as a continuous variable or dichotomized at >25 U/L) was significantly associated with shorter TTF (RR = 1.01, 95% CI: 1.0024–1.024, p = 0.017; RR = 2.56, 95% CI: 1.15–5.70, p = 0.021). Similarly, the combined value of AST + ALT was a significant predictor (p = 0.022).
Other variables demonstrated trends toward significance: low hemoglobin (HR = 0.86, p = 0.070) and elevated ALT (HR = 1.01, p = 0.075) both showed a borderline association with earlier treatment failure.
No statistically significant associations were observed for neutrophils, BMI, calcium, or metastatic burden (including liver, bone, lymph node, or lung involvement). Parameters such as lymphocyte count, creatinine, and total bilirubin were also not predictive of TTF in this subgroup.
Table 8 presents the results of univariate Cox regression analysis for baseline factors associated with time to treatment failure (TTF) in patients treated with Axitinib + Avelumab.
Several variables were significantly associated with shorter TTF in this subgroup. The presence of lymph node metastases (M1 lym) was a strong predictor of early treatment failure (RR = 11.98, 95% CI: 1.657–86.63, p = 0.014). Additionally, low lymphocyte count, low creatinine, low body mass index (BMI), and low hemoglobin were all significantly correlated with reduced TTF (p between 0.025 and 0.041), suggesting a potential link with immune, nutritional, or functional reserve.
Two variables showed borderline significance—elevated platelet count (p = 0.061) and higher neutrophil count (p = 0.081)—both suggesting a possible inflammatory component associated with reduced treatment durability. Other parameters, including age, AST, ALT, bilirubin, calcium levels, and presence of metastases at other sites (lung, bone, liver), did not show a statistically significant association with treatment outcomes.
Table 9 presents the results of univariate Cox regression for patients treated with Cabozantinib in second or third line following prior immunotherapy-based regimens.
Several baseline parameters were significantly or borderline associated with shorter time to treatment failure (TTF). Among these, higher platelet count (RR = 1.005, p = 0.0223), presence of liver metastases (RR = 2.90, 95% CI: 1.088–7.74, p = 0.0332), and elevated ALT (TGP) (RR = 1.025, p = 0.0484) were statistically significant predictors of early treatment discontinuation.
Additionally, neutrophil count (p = 0.0512), total calcium (p = 0.067), serum potassium (p = 0.0676), and AST (TGO) (p = 0.0768) all showed borderline significance, suggesting a possible role of systemic inflammation, electrolyte imbalance, or hepatic involvement in early Cabozantinib failure.
Other parameters, such as BMI, hemoglobin, creatinine, or metastatic distribution (bone, lung, lymph nodes), did not reach statistical significance. These findings suggest that patients with inflammatory or liver-related baseline abnormalities may experience limited benefit from Cabozantinib in later treatment lines.
Although Nivolumab was administered to a substantial number of patients in the subsequent lines setting, no statistically significant predictive factors for TTF were identified in this group (Table 10). This absence of association may reflect limitations related to sample size and statistical power, or a relatively low variability in baseline clinical and laboratory parameters. Additionally, the heterogeneity of prior therapies and patient selection might have further diluted potential signals, making it challenging to isolate robust predictors in this real-world cohort.
We produced an exploratory Cox model comparing L1 Ipilimumab + Nivolumab vs. Avelumab + Axitinib. While this comparison contrasts different IMDC risk categories, it did not reach statistical significance (RR = 0.842, favoring Ipi + Nivo, but the confidence interval is 0.417–1.69, p = 0.63); it nonetheless adds interpretive value regarding treatment duration. Direct comparisons between older TKI regimens (e.g., Sunitinib, Pazopanib) were avoided due to survivorship bias, as many patients in the TKI groups were initiated on therapy in earlier years and represent a preselected population of long-term responders.
Figure 2 Kaplan–Meier analysis of time to treatment failure (TTF) in patients receiving Ipilimumab + Nivolumab versus Avelumab + Axitinib as first-line therapy.
Treatment allocation was not randomized; Ipilimumab + Nivolumab was preferentially prescribed to patients with intermediate or poor IMDC risk profiles, in accordance with guideline-based clinical practice. The survival curves reflect real-world treatment selection and associated outcomes rather than strictly comparable patient populations.

4. Discussion

The findings of this study must be interpreted in light of its retrospective design, non-randomized structure, and the relatively small sample size, all of which may limit the generalizability of the results.
In our analysis, clear cell RCC demonstrated better TTF compared to non-clear cell histologies (RR = 0.66, p = 0.0367). This is consistent with a 2017 cohort study of 1943 patients, which reported survival differences primarily between papillary type I RCC and clear cell RCC, but not broadly across all non-clear cell subtypes [33].
Regarding Ipilimumab + Nivolumab, we identified elevated AST (TGO) levels—both as a continuous variable and when exceeding 25 U/L—as statistically significant negative predictive factors for shorter TTF. The combined transaminase score (AST + ALT) further supported this association. Interestingly, BMI did not emerge as a favorable predictor in our analysis, which contrasts with the “obesity paradox” reported in meta-analyses of RCC patients receiving immunotherapy (OS HR = 0.77 for overweight/obese patients) [34]. Another retrospective study of 126 patients with mRCC treated with Nivolumab found a significant positive association between BMI and OS (OS = 40.6 months for BMI ≥ 25 vs. 9.4 months for BMI < 25) [35]. Elevated AST may reflect a more aggressive disease phenotype or occult liver metastases. This observation is supported by prior studies in other cancer types, such as the Gustave Roussy Immune (GRIm) score in early breast cancer, where elevated AST is considered a negative prognostic factor [36].
For Avelumab + Axitinib, the presence of lymph node metastases emerged as a strong negative predictive factor for shorter TTF, while higher absolute lymphocyte counts were associated with longer TTF [37]. A modest association between higher baseline creatinine levels and longer TTF was also observed; although not definitively explained, this finding may relate to differences in muscle mass or nutritional status, which have previously been explored in the context of immune checkpoint inhibitor outcomes [38]. However, this interpretation remains speculative and should be validated in larger studies.
Consistent with the obesity paradox, both higher BMI and higher hemoglobin levels were positively associated with longer TTF in this subgroup. While hemoglobin is already part of the IMDC prognostic model, our findings suggest that it may also have predictive value for TTF in Avelumab + Axitinib treatment.
In the Cabozantinib used in subsequent lines subgroup, negative predictive factors for shorter TTF included elevated ALT, platelet counts, and the presence of liver metastases (RR = 2.904, p = 0.0332). These findings align with other real-world evidence identifying poor-risk features such as liver metastases and inflammatory markers as associated with worse outcomes. Notably, previous studies have highlighted higher BMI, prior nephrectomy, and favorable IMDC scores as positive prognostic factors for overall survival in Cabozantinib-treated patients [39], though they have not consistently addressed the impact of liver metastases on TTF specifically.
Higher baseline potassium levels show a tendency for shorter TTF (RR = 7.36, p = 0.0676), although this finding is based on a small sample size (n = 8) with no abnormal potassium values recorded. This observation warrants further validation.
Although we were unable to demonstrate statistically significant predictive markers for response to nivolumab treatment, in the same second line, a Nivolumab real-world Dutch study (Verhaart et al. 2021) discovered early increases in eosinophil count and neutrophil decreases to be associated with improved TTF (p ≤ 0.014), illustrating the value of hematologic dynamics as predictive signals. While our study focused on baseline, easily measurable biomarkers (e.g., AST, lymphocytes, platelets), these findings collectively underscore the need for both baseline and on-treatment markers to optimize early identification of treatment failure in routine practice.
Given the exploratory nature of the study and its real-world design, the sample size was adequate to identify potential predictive signals, though limited cohort sizes for some treatment groups warrant cautious interpretation of the results.
This study has several limitations. First, its retrospective and single-center design may limit the generalizability of the findings. The absence of a control group and the lack of blinding may introduce bias in both treatment selection and outcome interpretation. Additionally, the relatively small sample size in some treatment subgroups reduced statistical power. Time to treatment failure (TTF) was used as a pragmatic endpoint in the real-world setting; however, it may be influenced by non-clinical factors such as physician judgment or patient preference. Despite these limitations, the study provides real-world insights into early treatment failure in mRCC and identifies clinically accessible predictors that may help inform therapeutic decisions and risk stratification in routine oncology practice.
By identifying routine and accessible clinical and laboratory predictors—specific to each regimen—that are associated with shorter time to treatment failure, we aimed to help clinicians optimize treatment sequencing strategies, tailor monitoring intensity, and enhance patient counseling in daily oncology practice.

5. Conclusions

In this real-world, single-center, retrospective cohort of patients with non-urothelial metastatic RCC, we observed a progressive loss of patient numbers with each successive treatment line, emphasizing the ongoing clinical challenge of optimizing treatment sequencing for these patients. We identified several negative predictive factors for shorter time to treatment failure (TTF), which differed depending on the treatment received. For patients treated with Ipilimumab plus Nivolumab, elevated AST was associated with shorter TTF. In those receiving Avelumab plus Axitinib, the presence of lymph node metastases, low lymphocyte count, low creatinine, low body mass index, and low hemoglobin was predictive of poorer outcomes. For patients treated with Cabozantinib in subsequent lines, elevated ALT, platelets, and the presence of liver metastases were associated with shorter TTF. No statistically significant predictive factors for TTF were identified in subsequent lines of Nivolumab treatment.
These findings contribute to the growing body of evidence supporting the use of biological and clinical markers to individualize treatment strategies in mRCC. However, the hypothesis-generating nature of these results requires future validation in larger, prospectively designed studies.

Author Contributions

Conceptualization, S.S., V.-N.V. and D.-I.P.; methodology, S.S., D.-I.P., V.-N.V. and R.-D.D.; validation, S.S., D.-I.P. and Ș.-M.N.; formal analysis, S.S., D.-I.P., V.-N.V., R.-D.D.; investigation, D.-I.P. and V.-N.V.; resources, A.D., V.-N.V. and R.-D.D.; data curation, S.S. and D.-I.P.; writing—original draft preparation, S.S. and V.-N.V.; writing—review and editing, D.-I.P. and R.-D.D.; visualization, Ș.-M.N., D.-I.P. and V.-N.V.; supervision, Ș.-M.N., D.-L.S., D.-S.N., A.D., A.-G.N.; project administration, D.-I.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. We would like to thank ‘Victor Babeş’ University of Medicine and Pharmacy Timisoara for their support in covering the publication costs of this research paper.

Institutional Review Board Statement

The study protocol was conducted according to the principles of the Declaration of Helsinki after the approval of the OncoHelp Association Timisoara Ethical Committee (approval number 1178/2 May 2024).

Informed Consent Statement

All patients gave their informed consent for the procedure. All patients provided written informed consent for participation in the study and data collection.

Data Availability Statement

The data generated or analyzed during this study are included in this published article or are available from the corresponding author upon reasonable request.

Acknowledgments

The authors thank the editorial staff and peer reviewers for their valuable feedback.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

The following abbreviations are used in this manuscript:
mRCCMetastatic renal cell carcinoma
ccRCCClear cell renal cell carcinoma
IMDCInternational mRCC Database Consortium
ECOGEastern Cooperative Oncology Group
BMIBody Mass Index
ASTAspartate Aminotransaminase
ALTAlanine Aminotransaminase
CRPC-reactive protein
CBCComplete blood count
TTFTime to Treatment Failure
PFSProgression-free survival
OSOverall survival
M1 ossBone metastases
M1 hepHepatic metastases
M1 pulLung metastases
M1 lymLymph node metastases
SDStandard deviation
NNumber

References

  1. Homepage|ECIS–European Cancer Information System. Available online: https://ecis.jrc.ec.europa.eu (accessed on 2 June 2024).
  2. Znaor, A.; Lortet-Tieulent, J.; Laversanne, M.; Jemal, A.; Bray, F. International variations and trends in renal cell carcinoma incidence and mortality. Eur. Urol. 2015, 67, 519–530. [Google Scholar] [CrossRef]
  3. World Health Organization. WHO European Regional Obesity Report 2022. 2022. Available online: https://iris.who.int/bitstream/handle/10665/353747/9789289057738-eng.pdf?sequence=1 (accessed on 2 June 2024).
  4. MACROTRENDS. European Union Smoking Rate 2000–2025. Available online: https://www.macrotrends.net/global-metrics/countries/EUU/european-union/smoking-rate-statistics (accessed on 28 February 2025).
  5. Liu, X.; Sun, Q.; Hou, H.; Zhu, K.; Wang, Q.; Liu, H.; Zhang, Q.; Ji, L.; Li, D. The association between BMI and kidney cancer risk: An updated dose-response meta-analysis in accordance with PRISMA guideline. Medicine 2018, 97, e12860. [Google Scholar] [CrossRef] [PubMed]
  6. Hunt, J.D.; van der Hel, O.L.; McMillan, G.P.; Boffetta, P.; Brennan, P. Renal cell carcinoma in relation to cigarette smoking: Meta-analysis of 24 studies. Int. J. Cancer 2005, 114, 101–108. [Google Scholar] [CrossRef] [PubMed]
  7. Seretis, A.; Cividini, S.; Markozannes, G.; Tseretopoulou, X.; Lopez, D.S.; Ntzani, E.E.; Tsilidis, K.K. Association between blood pressure and risk of cancer development: A systematic review and meta-analysis of observational studies. Sci. Rep. 2019, 9, 8565. [Google Scholar] [CrossRef] [PubMed]
  8. Bonilla-Sanchez, A.; Rojas-Munoz, J.; Garcia-Perdomo, H.A. Association Between Diabetes and the Risk of Kidney Cancer: Systematic Review and Meta-Analysis. Clin. Diabetes 2022, 40, 270–282. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Lowrance, W.T.; Ordoñez, J.; Udaltsova, N.; Russo, P.; Go, A.S. CKD and the Risk of Incident Cancer. J. Am. Soc. Nephrol. 2014, 25, 2327–2334. [Google Scholar] [CrossRef]
  10. Scelo, G.; Larose, T.L. Epidemiology and Risk Factors for Kidney Cancer. J. Clin. Oncol. 2018, 36, 3574–3581. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  11. Cheungpasitporn, W.; Thongprayoon, C.; O’Corragain, O.A.; Edmonds, P.; Ungprasert, P.; Kittanamongkolchai, W.; Erickson, S. The risk of kidney cancer in patients with kidney stones: A systematic review and meta-analysis. QJM 2015, 108, 205–212. [Google Scholar] [CrossRef]
  12. Deckers, I.; van den Brandt, P.; van Engeland, M.; Soetekouw, P.M.M.B.; Baldewijns, M.M.L.L.; A Goldbohm, R.; Schouten, L.J. Long-term dietary sodium, potassium and fluid intake; exploring potential novel risk factors for renal cell cancer in the Netherlands Cohort Study on diet and cancer. Br. J. Cancer 2014, 110, 797–801. [Google Scholar] [CrossRef]
  13. Song, D.Y.; Song, S.; Song, Y.; Lee, J.E. Alcohol intake and renal cell cancer risk: A meta-analysis. Br. J. Cancer 2012, 106, 1881–1890. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Maher, E.R. Hereditary renal cell carcinoma syndromes: Diagnosis, surveillance and management. World J. Urol. 2018, 36, 1891–1898. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013, 499, 43–49. [Google Scholar] [CrossRef] [PubMed]
  16. Choueiri, T.K.; Kaelin, W.G., Jr. Targeting the HIF2–VEGF axis in renal cell carcinoma. Nat. Med. 2020, 26, 1519–1530. [Google Scholar] [CrossRef] [PubMed]
  17. Popescu, M.C.; Tretiakova, M. Renal Cell Carcinoma Overview. PathologyOutlines.Com Website. Available online: https://www.pathologyoutlines.com/topic/kidneytumormalignantrcc.html (accessed on 22 March 2025).
  18. Moch, H.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Gill, A.J.; Hartmann, A.; Menon, S.; Raspollini, M.R.; Rubin, M.A.; Srigley, J.R.; et al. The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur. Urol. 2022, 82, 458–468. [Google Scholar] [CrossRef]
  19. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Kidney Cancer (Version 3.2025). 2025. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1440 (accessed on 14 June 2025).
  20. Motzer, R.J.; Hutson, T.E.; Tomczak, P.; Michaelson, M.D.; Bukowski, R.M.; Rixe, O.; Oudard, S.; Negrier, S.; Szczylik, C.; Kim, S.T.; et al. Sunitinib versus Interferon Alfa in Metastatic Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 115–124. [Google Scholar] [CrossRef]
  21. Sternberg, C.N.; Davis, I.D.; Mardiak, J.; Szczylik, C.; Lee, E.; Wagstaff, J.; Barrios, C.H.; Salman, P.; Gladkov, O.A.; Kavina, A.; et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: Results of a randomized phase III trial. J. Clin. Oncol. 2010, 28, 1061–1068. [Google Scholar] [CrossRef] [PubMed]
  22. Motzer, R.J.; Escudier, B.; McDermott, D.F.; George, S.; Hammers, H.J.; Srinivas, S.; Tykodi, S.S.; Sosman, J.A.; Procopio, G.; Plimack, E.R.; et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1803–1813. [Google Scholar] [CrossRef]
  23. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
  24. Choueiri, T.K.; Halabi, S.; Sanford, B.L.; Hahn, O.; Michaelson, M.D.; Walsh, M.K.; Feldman, D.R.; Olencki, T.; Picus, J.; Small, E.J.; et al. Cabozantinib versus Sunitinib as Initial Targeted Therapy for Patients with Metastatic Renal Cell Carcinoma of Intermediate or Poor Risk (CABOSUN): A Randomised, Open-Label, Phase 2 Trial. Lancet Oncol. 2016, 17, 917–927. [Google Scholar] [CrossRef]
  25. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. Lancet Oncol. 2019, 20, 1370–1380. [Google Scholar] [CrossRef]
  26. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Oyervides Juárez, V.M.; Hsieh, J.J.; Basso, U.; Shah, A.Y.; et al. Nivolumab plus Cabozantinib versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2021, 384, 1305–1316. [Google Scholar] [CrossRef] [PubMed]
  27. Motzer, R.; Alekseev, B.; Rha, S.-Y.; Porta, C.; Eto, M.; Powles, T.; Grünwald, V.; Hutson, T.E.; Kopyltsov, E.; Méndez-Vidal, M.J.; et al. Lenvatinib Plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N. Engl. J. Med. 2021, 384, 1289–1300. [Google Scholar] [CrossRef] [PubMed]
  28. Hoeh, B.; Flammia, R.S.; Hohenhorst, L.; Sorce, G.; Panunzio, A.; Tappero, S.; Tian, Z.; Saad, F.; Gallucci, M.; Briganti, A.; et al. IO-IO vs. IO-TKI efficacy in metastatic kidney cancer patients: A structured systematic review over time. Semin. Oncol. 2022, 49, 394–399. [Google Scholar] [CrossRef] [PubMed]
  29. Ohba, K.; Nakanishi, H.; Kawada, K.; Nakamura, Y.; Mitsunari, K.; Matsuo, T.; Mochizuki, Y.; Imamura, R. Predictive factors of nivolumab plus ipilimumab treatment efficacy in metastatic renal cell carcinoma patients. Jpn. J. Clin. Oncol. 2024, 54, 827–832. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Kim, J.H.; Shin, S.J.; Bae, W.K. Real-world efficacy of first-line nivolumab plus ipilimumab and its practical predictive biomarkers in advanced renal cell carcinoma: First analysis from RENOIR study [KCSG GU22-13]. Ann. Oncol. 2024, 35, S1026. [Google Scholar] [CrossRef]
  31. Tachibana, H.; Nemoto, Y.; Ishihara, H.; Fukuda, H.; Yoshida, K.; Iizuka, J.; Hashimoto, Y.; Kondo, T.; Tanabe, K.; Takagi, T. Predictive Impact of Early Changes in Serum C-Reactive Protein Levels in Nivolumab Plus Ipilimumab Therapy for Metastatic Renal Cell Carcinoma. Clin. Genitourin. Cancer 2022, 20, e81–e88. [Google Scholar] [CrossRef] [PubMed]
  32. Napolitano, L.; Manfredi, C.; Cirillo, L.; Fusco, G.M.; Passaro, F.; Abate, M.; La Rocca, R.; Mastrangelo, F.; Spirito, L.; Pandolfo, S.D.; et al. Cytoreductive nephrectomy and metastatic renal cell carcinoma: State of the art and future perspectives. Medicina 2023, 59, 767. [Google Scholar] [CrossRef]
  33. Wagener, N.; Edelmann, D.; Benner, A.; Zigeuner, R.; Borgmann, H.; Wolff, I.; Krabbe, L.M.; Musquera, M.; Dell’oGlio, P.; Capitanio, U.; et al. Outcome of papillary versus clear cell renal cell carcinoma varies significantly in non-metastatic disease. PLoS ONE 2017, 12, e0184173. [Google Scholar] [CrossRef]
  34. Takemura, K.; Yonekura, S.; Downey, L.E.; Evangelopoulos, D.; Heng, D.Y. Impact of Body Mass Index on Survival Outcomes of Patients with Metastatic Renal Cell Carcinoma in the Immuno-oncology Era: A Systematic Review and Meta-analysis. Eur. Urol. Open Sci. 2022, 39, 62–71. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Çolak, R.; Gültürk, İ.; Dinç, G.; Akdağ, G.; Yıldırım, S.; Yılmaz, M.; Tural, D. The relationship between body mass index and survival in patients with renal cell carcinoma treated with nivolumab. J. Chemother. 2025, 23, 1–7. [Google Scholar] [CrossRef] [PubMed]
  36. Lien, Y.H. Looking for Sarcopenia Biomarkers. Am. J. Med. 2017, 130, 502–503. [Google Scholar] [CrossRef] [PubMed]
  37. Takenaka, Y.; Oya, R.; Takemoto, N.; Inohara, H. Predictive impact of sarcopenia in solid cancers treated with immune checkpoint inhibitors: A meta-analysis. J. Cachexia Sarcopenia Muscle 2021, 12, 1122–1135. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Albiges, L.; Fléchon, A.; Chevreau, C.; Topart, D.; Gravis, G.; Oudard, S.; Tourani, J.M.; Geoffrois, L.; Meriaux, E.; Thiery-Vuillemin, A.; et al. Real-world evidence of cabozantinib in patients with metastatic renal cell carcinoma: Results from the CABOREAL Early Access Program. Eur. J. Cancer 2021, 142, 102–111. [Google Scholar] [CrossRef] [PubMed]
  39. Santoni, M.; Massari, F.; Bracarda, S.; Procopio, G.; Milella, M.; De Giorgi, U.; Basso, U.; Aurilio, G.; Incorvaia, L.; Martignetti, A.; et al. Body mass index in patients treated with cabozantinib for advanced renal cell carcinoma: A new prognostic factor? Diagnostics 2021, 11, 138. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Figure 1. Axi = Axitinib, Pembro = Pembrolizumab, Cabo = Cabozantinib, Nivo = Nivolumab, Lenca = Lenvatinib, Ipi = Ipilimumab, Ave = Avelumab, Pazo = Pazopanib, Suni= Sunitinib. Preferred regimens in bold, category 1 in black, category 2A in gray, category 2B in blue.
Figure 1. Axi = Axitinib, Pembro = Pembrolizumab, Cabo = Cabozantinib, Nivo = Nivolumab, Lenca = Lenvatinib, Ipi = Ipilimumab, Ave = Avelumab, Pazo = Pazopanib, Suni= Sunitinib. Preferred regimens in bold, category 1 in black, category 2A in gray, category 2B in blue.
Jcm 14 05271 g001
Figure 2. Kaplan–Meier analysis of time to treatment failure (TTF) in patients receiving Ipilimumab + Nivolumab versus Avelumab + Axitinib as first-line therapy. (in blue Ipi + Nivo, in red Ave + Axi, duration in days).
Figure 2. Kaplan–Meier analysis of time to treatment failure (TTF) in patients receiving Ipilimumab + Nivolumab versus Avelumab + Axitinib as first-line therapy. (in blue Ipi + Nivo, in red Ave + Axi, duration in days).
Jcm 14 05271 g002
Table 1. Synthesis of main trials in kidney cancer treatment.
Table 1. Synthesis of main trials in kidney cancer treatment.
TreatmentComparatorYear of PublicationHR for PFS (95% CI)HR for OS (95% CI)% Grade ≥ 3 Adverse Reactions
Sunitinib,
L1 [20]
Interferon alfa2007HR = 0.42 (0.32–0.54)HR = 0.82 (0.66–1.00)~40%
Pazopanib,
L1 [21]
Placebo2010HR = 0.46 (0.34–0.62)HR = 0.91 (0.66–1.25)~30%
Nivolumab,
≥L2 [22]
Everolimus2015HR = 0.88 (0.75–1.03)HR = 0.73 (0.57–0.93)~20%
Ipilimumab + Nivolumab, L1 [23]Sunitinib2018HR = 0.82 (0.66–1.02)HR = 0.63 (0.44–0.89)~55%
Cabozantinib,
L1 [24]
Sunitinib2016HR = 0.66 (0.46–0.95)HR = 0.80 (0.53–1.21)~60%
Avelumab + Axitinib,
L1 [25]
Sunitinib2019HR = 0.61 (0.47–0.79)HR = 0.92 (0.70–1.21)~50%
Cabozantinib + Nivolumab, L1 [26]Sunitinib2021HR = 0.51 (0.41–0.64)HR = 0.66 (0.53–0.83)~55%
Lenvatinib + Pembrolizumab,
L1 [27]
Sunitinib2021HR = 0.39 (0.28–0.55)HR = 0.66 (0.49–0.88)~55%
Table 2. Average age of patients by treatment line and regimen.
Table 2. Average age of patients by treatment line and regimen.
Line of TreatmentAverage Age (Years)Number of Patients
CabozantinibL157.663
Ipilimumab + NivolumabL158.9450
PazopanibL160.1415
All medications L1L160.8137
SunitinibL163.8749
Axitinib + AvelumabL165.9417
EverolimusL2–L3–L445.33
CabozantinibL2–L3–L458.527
All medications L2–L4L2–L3–L459.890
AxitinibL2–L3–L460.912
NivolumabL2–L3–L461.746
Table 3. First-line treatments.
Table 3. First-line treatments.
First-Line TreatmentsNumber of Patients (n = 137)Median TTF
Days (Months)
Average TTF
Days (Months)
Range
Days (Months)
Ipilimumab + Nivolumab50178 (5.9)286 (9.5)21–1159 (0.7–38.6)
Sunitinib49334 (11.1)616 (20.5)28–4017 (0.9–133.9)
Avelumab + Axitinib17215 (7.2)248 (8.3)14–691 (0.5–23.0)
Pazopanib15324 (10.8)508 (16.9)62–1400 (2.1–46.7)
Cabozantinib3242 (8.1)214 (7.1)153–248 (5.1–8.3)
Tivozanib130 (1.0)
Temsirolimus2114 (3.8)231 (7.7)25-671 (0.83–22.3)
Table 4. Second-line treatments.
Table 4. Second-line treatments.
Line 2 TreatmentsNumber of Patients (n = 65)Median TTF
Days (Months)
Average TTF
Days (Months)
Range
Days (Months)
Nivolumab32277 (9.2)378 (12.6)16–1371 (0.5–45.7)
Cabozantinib1953 (1.8)105 (3.5)20–473 (0.7–15.8)
Axitinib10138 (4.6)392 (13.1)31–1584 (1.0–52.8)
Everolimus161 (2.0)
Sunitinib2185 (6.2)185 (6.2)32–337 (1.1–11.2)
Nivolumab + Axitinib1823 (27.4)
Table 5. Third-line treatments.
Table 5. Third-line treatments.
Line 3 TreatmentsNumber of Patients (n = 23)Median TTF
Days (Months)
Average TTF
Days (Months)
Range
Days (Months)
Nivolumab11122 (4.1)355 (11.8)32–1208 (1.1–40.3)
Cabozantinib8100 (3.3)89 (3.0)35–123 (1.2–4.1)
Axitinib2265 (8.8)265 (8.8)257–274 (8.5–9.1)
Everolimus2122 (4.1)122 (4.1)92–153 (3.1–5.1)
Table 6. Fourth-line treatments.
Table 6. Fourth-line treatments.
Line 4 TreatmentsNumber of Patients (n = 23)Median TTF
Days (Months)
Average TTF
Days (Months)
Range
Days (Months)
Nivolumab2611 (20.3)611 (20.3)117–1106 (3.9–36.8)
Table 7. Predictive factors analyzed for Ipilimumab + Nivolumab TTF.
Table 7. Predictive factors analyzed for Ipilimumab + Nivolumab TTF.
ParameterAverage±SDnRR95% Confidence Intervalp
AST24.627451.011.0024–1.0240.017
AST > 2524%452.561.15–5.700.021
AST + ALT54.461.7441.011.0009–1.01150.022
Hb11.792.05450.860.73–1.010.070
ALT29.638.1441.010.999–1.0150.075
Neutrophils6.242.94451.070.96–1.180.21
Total bilirubin0.520.18424.210.37–480.25
M1 lym0.840.366500.64130.274–1.1990.31
Leucocytes8.863.07451.060.94–1.180.34
Body mass index kg/m226.26.8330.960.89–1.040.35
Calcium (total)9.430.76391.250.72–2.150.43
M1 oss0.220.414500.7070.294–1.670.43
Age58.912.8500.990.96–1.010.59
M1 hep0.280.449500.81330.348–1.8970.63
Lymphocytes1.720.65450.890.511–1.540.67
Creatinine1.060.52441.120.55–2.310.74
Sex66% men501.130.52–2.420.75
M1 pul0.680.4665500.90560.433–1.8950.79
Thrombocytes338.9131450.99960.997–1.0030.80
(ALT = alanin amino transpherase, AST = aspartate aminotransferase, Hb = hemoglobin, M1 pul = lung metastases, M1 lym = distant lymph node metastases, M1 hep = liver metastases, M1 oss = bone metastases).
Table 8. Predictive factors analyzed for Axitinib + Avelumab TTF.
Table 8. Predictive factors analyzed for Axitinib + Avelumab TTF.
ParameterAverage±SDnRR95% Confidence Intervalp
M1 lym0.1170.3221711.981.657–86.630.014
Lymphocytes1.730.96150.3560.145–0.8770.025
Creatinine mg/dL1.140.31150.05730.004–0.870.039
BMI kg/m229.58.380.8190.677–0.9910.040
Hb g/dL13.32.9150.7380.551–0.9970.041
Thrombocytes283.497.5151.0080.999–1.0160.061
Neutrophils5.952.66151.310.967–1.7650.081
Sex76% men 170.3780.089–1.6100.189
M1 pul0.6470.4779170.41580.130–1.560.2098
M1 hep0.2350.424170.5790.122–2.7450.4919
Total bilirubin mg/dL0.540.15150.2610.005–14.080.509
M1 oss0.41180.4922171.40750.403–4.9160.5922
Leuccocytes8.422.64151.0660.772–1.4720.698
ALT UI/L17.310.8150.9860.918–1.0590.704
AST UI/L21.210.6150.9880.923–1.0570.725
Age65.98.4171.0070.919–1.1030.882
Total calcium mg/dL9.380.94121.0240.32–3.270.968
(ALT = alanin amino transpherase, AST = aspartate aminotransferase, Hb = hemoglobin, M1 pul = lung metastases, M1 lym = distant lymph node metastases, M1 hep = liver metastases, M1 oss = bone metastases).
Table 9. Predictive factors analyzed for Cabozantinib TTF, only subsequent lines L2–L3 after IO–IO or IO–TKI.
Table 9. Predictive factors analyzed for Cabozantinib TTF, only subsequent lines L2–L3 after IO–IO or IO–TKI.
ParameterAverage±SDnRR95% Confidence Intervalp
Thrombocytes318.07159.53141.0051.0007–1.0090.0223
M1 hep29.60% 272.9041.088–7.740.0332
TGP/ALT (U/L)31.6427.88141.02491.0002–1.050.0484
Neutrophils7.876.5141.0990.999–1.2090.0512
Calcium total (mg/dL)9.120.71132.6750.93–7.670.067
K+ mmol/L4.660.6487.630.863–67.50.0676
TGO/AST (U/L)46.7365.47141.00860.999–1.0180.0768
BMI kg/m229.927.2120.93130.854–1.0150.1056
Hemoglobin (g/dL)12.27%1.61140.8040.54–1.170.261
Leucocytes12.271.6140.80440.549–1.1770.2631
Lymphocytes1.6670.779140.5950.227–1.5530.2891
M1 oss77.70% 271.9720.552–7.0340.2954
Creatinine (mg/dL)1.10.22142.9790.299–29.610.3156
sex74% males 270.730.25–2.090.56
age (y)58.511.61271.01210.939–1.0570.586
Btotal bilirubin (mg/dL)76.90%0.42131.3310.19–9.170.771
ECOG95.00%0.75230.9170.47–1.770.799
M1 pul66.60% 271.1070.39–3.130.848
M1 lym66.60% 271.0730.402–2.860.887
(ALT = alanin amino transpherase, AST = aspartate aminotransferase, Hb = hemoglobin, M1 pul = lung metastases, M1 lym = distant lymph node metastases, M1 hep = liver metastases, M1 oss = bone metastases).
Table 10. Predictive factors analyzed for Nivolumab used in L2-L3, TTF.
Table 10. Predictive factors analyzed for Nivolumab used in L2-L3, TTF.
ParameterAverage±SDnRR95% Confidence Intervalp
Thrombocytes268124261.00320.999–1.0070.116
Calcium total, mg/dL9.520.72200.5760.258–1.2820.177
Bilirubin total mg/dL0.620.22244.660.459–47.40.193
Hb g/dL12.22.8260.8780.721–1.0700.199
Sex75% males281.880.630–5.6300.257
Neutrophils5.14.5261.04140.956–1.1340.352
Age years62.47.4281.0250.957–1.0970.481
TGO ASAT U/L23.414.5250.9890.958–1.0200.499
Lymphocytes1.760.83261.1620.721–1.8750.536
Leucocytes7.64.5261.0490.967–1.1380.541
TGP SLAT U/L17%8.8260.9870.931–1.0460.666
Creatinine mg/dL1.330.77251.0130.601–1.7090.959
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saftescu, S.; Vornicu, V.-N.; Popovici, D.-I.; Dragomir, R.-D.; Nagy, D.-S.; Sandu, D.-L.; Dulan, A.; Negru, Ș.-M.; Negru, A.-G. Decoding Treatment Failures in Metastatic Renal Cell Carcinoma: Predictors Across Immunotherapy and Targeted Therapies from a Retrospective Real-World Analysis. J. Clin. Med. 2025, 14, 5271. https://doi.org/10.3390/jcm14155271

AMA Style

Saftescu S, Vornicu V-N, Popovici D-I, Dragomir R-D, Nagy D-S, Sandu D-L, Dulan A, Negru Ș-M, Negru A-G. Decoding Treatment Failures in Metastatic Renal Cell Carcinoma: Predictors Across Immunotherapy and Targeted Therapies from a Retrospective Real-World Analysis. Journal of Clinical Medicine. 2025; 14(15):5271. https://doi.org/10.3390/jcm14155271

Chicago/Turabian Style

Saftescu, Sorin, Vlad-Norin Vornicu, Dorel-Ionel Popovici, Radu-Dumitru Dragomir, Dana-Sonia Nagy, Daniela-Lidia Sandu, Ana Dulan, Șerban-Mircea Negru, and Alina-Gabriela Negru. 2025. "Decoding Treatment Failures in Metastatic Renal Cell Carcinoma: Predictors Across Immunotherapy and Targeted Therapies from a Retrospective Real-World Analysis" Journal of Clinical Medicine 14, no. 15: 5271. https://doi.org/10.3390/jcm14155271

APA Style

Saftescu, S., Vornicu, V.-N., Popovici, D.-I., Dragomir, R.-D., Nagy, D.-S., Sandu, D.-L., Dulan, A., Negru, Ș.-M., & Negru, A.-G. (2025). Decoding Treatment Failures in Metastatic Renal Cell Carcinoma: Predictors Across Immunotherapy and Targeted Therapies from a Retrospective Real-World Analysis. Journal of Clinical Medicine, 14(15), 5271. https://doi.org/10.3390/jcm14155271

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop