Skip to main content

Prognostic value of the ratio of maximum to minimum diameter of primary tumor in metastatic clear cell renal cell carcinoma

Abstract

Background

Several models and markers were developed and found to predict outcome of advanced renal cell carcinoma. This study aimed to evaluate the prognostic value of the ratio of maximum to minimum tumor diameter (ROD) in metastatic clear cell renal cell carcinoma (mccRCC).

Methods

Patients with mccRCC (n = 213) treated with sunitinib from January 2008 to December 2018 were identified. Cutoff value for ROD was determined using receiver operating characteristic. Patients with different ROD scores were grouped and evaluated. Survival outcomes were estimated by Kaplan–Meier method.

Results

The optimal ROD cutoff value of 1.34 was determined for progression free survival (PFS) and overall survival (OS). Patients in ROD ≥ 1.34 group had shorter PFS (9.6 versus 17.7 months, p < 0.001) and OS (25.5 versus 32.6 months, p < 0.001) than patients in ROD < 1.34 group. After adjustment for other factors, multivariate analysis showed ROD ≥ 1.34 was an independent prognostic factor for PFS (p < 0.001) and OS (p = 0.006). Patients in ROD ≥ 1.34 group presented higher proportions of pT3/4 stage (89.2% versus 10.8%, p = 0.021), WHO/ISUP grade III/IV (72.0% versus 28.0%, p = 0.010), tumor necrosis (71.0% versus 29.0%, p = 0.039), sarcomatoid differentiation (79.1% versus 20.9%, p = 0.007), poor MSKCC risk score (78.4% versus 21.6%, p < 0.001) and poor IMDC risk score (74.4% versus 25.6%, p < 0.001) than ROD < 1.34 group.

Conclusion

Primary tumor with higher ROD was an independently prognostic factor for both PFS and OS in patients with mccRCC who received targeted therapy. Higher ROD was also associated with high pT stage, high WHO/ISUP grade, sarcomatoid features, tumor necrosis, poor MSKCC and IMDC risk score.

Peer Review reports

Background

Renal cell carcinoma (RCC) accounts for approximately 3% of malignant tumors in adults and 20% to 30% of them are diagnosed as advanced diseases with poor prognosis [1, 2]. Targeted therapy has been the standard metastatic RCC treatment since 2005 [3, 4]. Several risk score models, such as the Memorial Sloan Kettering Cancer Center (MSKCC) system and the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) criteria, have been found and validated to predict the prognosis of different patients with metastatic RCC [5, 6]. However, MSKCC and IMDC classification mostly focus on the state of the performance status and laboratory values while the primary tumor status has not been involved in.

Several studies have reported primary tumor size may be associated with prognosis of metastatic RCC [7, 8]. It is still controversial and cannot completely reflect the role of primary tumor. The ratio of maximum to minimum tumor diameter (ROD), a specific primary tumor feature, has not been mentioned. Previously, we have investigated the utility of ROD in predicting pathologic subtypes of RCC before surgery and found its association with adverse pathological factors [9]. We used ROD to quantify tumor irregularity. The more irregular the tumor, the higher the degree of malignancy was discovered. To further demonstrate the prognostic value of the ROD, in this study, we retrospectively analyzed the records of clear cell RCC (ccRCC) patients treated by surgery and followed sunitinib due to subsequent or simultaneous metastasis.

Patients and methods

Patient selection

This is a retrospective study focusing on patients with metastatic ccRCC (mccRCC), which was approved by the Cancer Hospital Chinese Academy of Medical Sciences of Ethics Committee (ID: NCC2016YJC-08). Patient consent for treatment and follow-up was included in each medical record. Patients diagnosed as ccRCC after surgery and treated with sunitinib for metastasis between January 2008 and December 2018 were collected. Metastasis was confirmed by imaging examination. Sunitinib was initially administered 50 mg once daily, on a 4/2 schedule. Patients received other systematic therapies after sunitinib treatment failure, including pazopanib, everolimus, axitinib, immune checkpoint inhibitors, or other free second-line therapy trials. Clinicopathological features, such as age, gender, Karnofsky performance status, presenting symptom, tumor size, World Health Organization/International Society of Urologic Pathologists (WHO/ISUP) grade, tumor necrosis, sarcomatoid differentiation, MSKCC and IMDC criteria were used to evaluate.

Radiological assessment

Patients had contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI) examination within 2 weeks before surgery for primary tumor diagnosis and clinical staging. CT scans were undertaken every 4–8 week since taking sunitinib, and the Response Evaluation Criteria in Solid Tumors (RECIST) criteria version 1.1 were used to evaluate [10]. The best response of treatment, including complete response (CR), partial response (PR), stable disease (SD) and progression disease (PD) was recorded.

The diameters of the tumor were measured including coronal, sagittal and axial view, including a maximum diameter, a sub maximum diameter and a minimum diameter in clinical staging before surgery (Fig. 1). Tumor’s three diameters were recorded separately by 2 independent radiologists. The ratio of the maximum diameter to the minimum diameter (ROD) was calculated to quantify tumor irregularity.

Fig. 1
figure 1

Example of measuring the three diameters: one maximum, one sub maximum, and one minimum

Statistical analysis

Chi-square test, Fisher’s exact test or Student’s t-test were used to analyze the relationship of different groups. The optimal cutoff value of ROD in prediction of survival outcomes was determined by Receiver Operating Characteristic (ROC) curve analysis. To investigate the survival outcome of patients with mccRCC, progression-free survival (PFS) and overall survival (OS) from the initiation of sunitinib were determined using the Kaplan–Meier method and were analyzed using the log-rank test. Cox proportional hazards regression models were used to assess the significant factors unadjusted and adjusted for covariates. Statistical analysis was performed using SPSS 23.0 software, and differences were considered statistically significant if p values were < 0.05.

Results

Patients’ characteristics

A total of 213 patients with a median age of 55.0 years (range, 17–76 years) were identified. Patients included 139 (65.3%) males and 74 (34.7%) females. Median primary tumor size measured in CT or MRI was 6.1 cm (range, 1.1–15.6 cm). The number of patients in cT1/2, cT3/4 stage was 199 (93.4%), 14 (6.6%), respectively. All patients were received nephrectomy before systemic therapy. Patients consisted of 96 (45.1%) cases of synchronous RCC and 117 (54.9%) cases of metachronous RCC. The number of patients in pT1/2, pT3/4 stage was 188 (88.3%), 25 (11.7%), respectively. The most common site of metastasis was pulmonary in 164 cases (77.0%), followed by lymph node metastasis in 86 cases (40.4%). Other metastases included bone, adrenal, liver, brain, spleen, and pancreas. There were 152 patients (71.4%) harboring more than 2 metastatic organs. Twenty-two patients (10.3%) had responders including CR (n = 5, 2.3%) and PR (n = 17, 8.0%) during the treatment of sunitinib. Patients’ characteristics were summarized in Table 1.

Table 1 Correlation between ROD and the clinicopathological features of patients with metastatic renal cell carcinoma

The association between ROD and clinicopathological features

ROC curves were constructed to determine the appropriate cutoff point of ROD (Additional file 1: fig S1). The most discriminative ROD cutoff value of 1.34 was selected for both PFS (sensitivity = 79.3%, specificity = 74.0%; Area Under Curve, AUC = 0.810, 95% confidence interval, CI:0.752–0.869, p < 0.001) and OS (sensitivity = 84.9%, specificity = 83.8%; AUC = 0.932, 95% CI:0.899–0.966, p < 0.001). The patients were divided into two groups including ROD < 1.34 group (n = 83) and ROD ≥ 1.34 group (n = 130). Comparison between ROD and the Clinicopathological features were showed in Table 1. Patients in ROD ≥ 1.34 group presented higher proportions of pT3/4 stage (89.2% versus 10.8%, p = 0.021), WHO/ISUP grade III/IV (72.0% versus 28.0%, p = 0.010), tumor necrosis (71.0% versus 29.0%, p = 0.039), sarcomatoid differentiation (79.1% versus 20.9%, p = 0.007), poor MSKCC risk score (78.4% versus 21.6%, p < 0.001) and poor IMDC risk score (74.4% versus 25.6%, p < 0.001) than ROD < 1.34 group.

The evaluation of ROD in clinical outcomes

After 32.0 months (range, 2.6–125.8 months) median follow-up, patients in ROD ≥ 1.34 group experienced significantly shorter PFS (median, 9.6 versus 17.7 months, p < 0.001) and OS (median, 25.5 versus 32.6 months, p < 0.001) than patients in ROD < 1.34 group (Fig. 2).

Fig. 2
figure 2

Patients with ROD ≥ 1.34 had shorter progression free survival (A) and overall survival (B) than patients with ROD < 1.34

The univariate survival analysis revealed that ROD ≥ 1.34 (p < 0.001), WHO/ISUP grade (p < 0.001), tumor necrosis (p = 0.032), sarcomatoid differentiation (p < 0.001), MSKCC score (p < 0.001) and IMDC score (p < 0.001) appeared as significant prognostic factors for PFS (Table 2). After adjustment for WHO/ISUP grade (p < 0.001), tumor necrosis (p = 0.118), sarcomatoid differentiation (p = 0.035), MSKCC score (p = 0.008) and IMDC score (p < 0.001), the multivariate Cox regression analysis revealed that ROD ≥ 1.34 (adjusted hazard ratio, HR = 3.068, 95% CI:2.102–4.478, p < 0.001) was an independent prognostic factor for poor PFS (Table 3). In addition, presenting symptom (p = 0.007), ROD ≥ 1.34 (p < 0.001), WHO/ISUP grade (p < 0.001), tumor necrosis (p = 0.007), sarcomatoid differentiation (p = 0.016), MSKCC score (p < 0.001) and IMDC score (p < 0.001) were prognostic factors for OS (Table 2). After adjustment for presenting symptom (p = 0.009), WHO/ISUP grade (p < 0.001), tumor necrosis (p < 0.001), sarcomatoid differentiation (p = 0.621), MSKCC score (p = 0.002) and IMDC score (p < 0.001), ROD ≥ 1.34 (adjusted HR = 1.774, 95% CI:1.175–2.676, p = 0.006) was still an independent prognostic factors for OS (Table 3).

Table 2 Univariate Cox regression analysis of clinical factors in patients with metastatic renal cell carcinoma
Table 3 Independent prognostic factors analyzed using a multivariable Cox model in patients with metastatic renal cell carcinoma

Discussion

From published papers, intratumor heterogeneity is a key factor contributing to the survival of cancer, therapeutic failure, and drug resistance 11, 12]. The scale of heterogeneity within a tumor has also been found and proved in RCC [13, 14]. Studies have showed that intratumor heterogeneity may contribute to the polyclonal growth pattern of tumors [15]. In addition, polyclonal growth commonly accompanies with corresponding change of tumor microenvironment, which is also thought to play an important role for tumor growth and progression and to be involved in the treatment outcome of targeted therapy [16]. We considered that the intratumor heterogeneity evolved to the asymmetric tumor morphology. From the clinical practice, we observed that the ROD could be used to quantify the tumor irregularity. The higher the intratumor heterogeneity, the more irregular tumor and the higher ROD would achieve, which was likely to reveal the efficacy of later treatment.

The ROD, a specific feature of primary tumor, has not been investigated before. In this study, the most optimal ROD cutoff value of 1.34 was determined. Patients in ROD ≥ 1.34 group accounted for 61.0% (130/213). And we were not surprised to find that more cases of metastatic renal cell carcinoma were in the group with larger ROD. This finding suggested a new hypothesis that the stronger tumor heterogeneity have more chances of metastasis. In this study, patients of pT1/2 accounted for 88.3% which was more than pT3/4. Patients of pT1/2 increased because of the popularity of health examination. Although patients of pT3/4 were more easily to occur metastasis, pT1/2 had large base population which resulted in pT1/2 had a high proportion in metastatic patients. Another reason was that all patients enrolled were able to receive nephrectomy and unresectable T3/4 patients were excluded.

We found that the ROD ≥ 1.34 was also significantly associated with prognosis. Compared with patients in ROD ≥ 1.34 group, patients in ROD < 1.34 group achieved longer PFS (17.7 versus 9.6 months, p < 0.001) and OS (32.6 versus 25.5 months, p < 0.001). Variable pathology factors including WHO/ISUP grade, tumor necrosis, and sarcomatoid differentiation were also associated with outcome of RCC, which were confirmed in previous studies [17,18,19]. In this study, these features were also investigated. WHO/ISUP grade, sarcomatoid differentiation, MSKCC score and IMDC score were independent prognostic factors for PFS, and presenting symptom, WHO/ISUP grade, tumor necrosis, MSKCC score and IMDC score were independent prognostic factors for OS. Interestingly, we found that patients in ROD ≥ 1.34 group were more likely to present high WHO/ISUP grade, high pT stage, tumor necrosis, sarcomatoid differentiation, poor MSKCC risk score, and poor IMDC score. This showed that as an index to quantify tumor irregularity, ROD had a strong relationship with tumor malignancy. Besides above findings, we also found that ROD ≥ 1.34 was an independent prognostic factor for both poor PFS and OS.

Our study includes several limitations that need to be acknowledged. It was a retrospective study including limited cases, which still needed to accumulate data, and we will carry out prospective studies to confirm. In addition, we will further explore the possible molecular mechanism of poor outcome associated with irregularity of primary tumor.

Conclusions

Primary tumor with ROD ≥ 1.34 was an independently prognostic factor for both PFS and OS in patients with mccRCC who received targeted therapy and also associated with high pT stage, high WHO/ISUP grade, sarcomatoid features, tumor necrosis, poor MSKCC and IMDC risk score. Further prospective validation is required to confirm these findings.

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available due to individual privacy could be compromised but are available from the corresponding author on reasonable request.

Abbreviations

AUC:

Area under curve

CI:

Confidence interval

CR:

Complete response

CT:

Computed tomography

HR:

Hazard ratio

IMDC:

International metastatic renal cell carcinoma database consortium

mccRCC:

Metastatic clear cell renal cell carcinoma

mRCC:

Metastatic RCC

MRI:

Magnetic resonance imaging

MSKCC:

Memorial sloan kettering cancer center

NCC/CHCAMS:

National cancer center/cancer hospital, chinese academy of medical sciences

OS:

Overall survival

PD:

Progression disease

PFS:

Progression-free survival

PR:

Partial response

RCC:

Renal cell carcinoma

RECIST:

Response evaluation criteria in solid tumors

ROC:

Receiver operating characteristic

ROD:

Ratio of maximum to minimum tumor diameter

SD:

Stable disease

WHO/ISUP:

World health organization/international society of urologic pathologists

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA: Cancer J Clin. 2019;69(1):7–34.

    Google Scholar 

  2. Capitanio U, Bensalah K, Bex A, Boorjian SA, Bray F, Coleman J, et al. Epidemiology of renal cell carcinoma. Eur Urol. 2019;75(1):74–84.

    Article  Google Scholar 

  3. 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.

    Article  Google Scholar 

  4. 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 Offi J Am Soc Clin Oncol. 2009;27(22):3584–90.

    Article  CAS  Google Scholar 

  5. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Annal Oncol Off J Eur Soc Med Oncol. 2016;27(suppl 5):v58–68.

    Article  CAS  Google Scholar 

  6. Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol Off J Am Soc Clin Oncol. 2009;27(34):5794–9.

    Article  CAS  Google Scholar 

  7. DiNatale RG, Xie W, Becerra MF, Silagy AW, Attalla K, Sanchez A, et al. The association between small primary tumor size and prognosis in metastatic renal cell carcinoma: insights from two independent cohorts of patients who underwent cytoreductive nephrectomy. Eur Urol Oncol. 2020;3(1):47–56.

    Article  Google Scholar 

  8. Thompson RH, Hill JR, Babayev Y, Cronin A, Kaag M, Kundu S, et al. Metastatic renal cell carcinoma risk according to tumor size. J Urol. 2009;182(1):41–5.

    Article  Google Scholar 

  9. Jiang W, Wang D, Shi H, Shang B, Wen L, Zhang L, et al. Ratio of maximum to minimum tumor diameter can predict the pathology type of renal cell carcinoma before surgery. Tumori. 2021;107(1):64–70.

    Article  CAS  Google Scholar 

  10. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.

    Article  CAS  Google Scholar 

  11. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015;27(1):15–26.

    Article  CAS  Google Scholar 

  12. Greaves M. Evolutionary determinants of cancer. Cancer Discov. 2015;5(8):806–20.

    Article  CAS  Google Scholar 

  13. Martinez P, Birkbak NJ, Gerlinger M, McGranahan N, Burrell RA, Rowan AJ, et al. Parallel evolution of tumour subclones mimics diversity between tumours. J Pathol. 2013;230(4):356–64.

    Article  CAS  Google Scholar 

  14. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225–33.

    Article  CAS  Google Scholar 

  15. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613–28.

    Article  CAS  Google Scholar 

  16. Olson OC, Joyce JA. Microenvironment-mediated resistance to anticancer therapies. Cell Res. 2013;23(2):179–81.

    Article  CAS  Google Scholar 

  17. Zhang L, Zha Z, Qu W, Zhao H, Yuan J, Feng Y, et al. Tumor necrosis as a prognostic variable for the clinical outcome in patients with renal cell carcinoma: a systematic review and meta-analysis. BMC Cancer. 2018;18(1):870.

    Article  Google Scholar 

  18. Chrom P, Stec R, Semeniuk-Wojtas A, Bodnar L, Spencer NJ, Szczylik C. Fuhrman grade and neutrophil-to-lymphocyte ratio influence on survival in patients with metastatic renal cell carcinoma treated with first-line tyrosine kinase inhibitors. Clin Genitourin Cancer. 2016;14(5):457–64.

    Article  Google Scholar 

  19. Kim T, Zargar-Shoshtari K, Dhillon J, Lin HY, Yue B, Fishman M, et al. Using percentage of sarcomatoid differentiation as a prognostic factor in renal cell carcinoma. Clin Genitourin Cancer. 2015;13(3):225–30.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by Beijing hope run special fund of cancer foundation of China (LC2018L02).

Author information

Authors and Affiliations

Authors

Contributions

All authors listed in this manuscript contributed significantly to the study. Shi HZ and Cao CZ contributed to writing the manuscript. Wen L contributed to clinical data collection and analysis. Zhang LY and Zhang J contributed to reviewing CT or MRI data. Ma JH contributed to conception of the study. Li CL and Shou JZ contributed to the study design and reviewed the manuscript for critical revisions. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jianzhong Shou or Changling Li.

Ethics declarations

Ethics approval and consent to participate

The research study was reviewed and approved by the Cancer Hospital Chinese Academy of Medical Sciences of Ethics Committee (ID: NCC2016YJC-08). Patient details were anonymized before analysis. Patient written informed consent for treatment and follow-up was included in each medical record. All methods were carried out in accordance with relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1:

 Fig. S1. Receiver operating characteristics curves were used to determine the appropriate cut-off point of ROD predicting survival. (A) progession free survival and (B) overall survival.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, H., Cao, C., Wen, L. et al. Prognostic value of the ratio of maximum to minimum diameter of primary tumor in metastatic clear cell renal cell carcinoma. BMC Urol 22, 95 (2022). https://doi.org/10.1186/s12894-022-01047-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12894-022-01047-y

Keywords