Open access peer-reviewed chapter

Perspective Chapter: An Update on Renal Cell Carcinoma

Written By

Jindong Chen

Submitted: 04 May 2023 Reviewed: 24 July 2023 Published: 09 August 2023

DOI: 10.5772/intechopen.112633

From the Edited Volume

Renal Cell Carcinoma - Recent Advances, New Perspectives and Applications

Edited by Jindong Chen

Chapter metrics overview

47 Chapter Downloads

View Full Metrics

Abstract

Incidence and mortality of renal cell carcinoma (RCC) significantly vary worldwide. While RCC incidence has been increasing, its mortality rate has been decreasing. Smoking, obesity, hypertension, chronic kidney disease (CKD), ethnicity, location, and other environmental factors are reported to be associated with RCC. With the use of the improved diagnostic methods, including ultrasound, contrast-enhanced ultrasound (CEUS), computed tomography (CT) scan, magnetic resonance imaging (MRI), and positron emission tomography (PET)/CT scan, the detection rate of RCC has significantly increased over the past decade. We have witnessed innovation in surgical techniques and robotic platforms with integration of imaging approaches, and urologists are now able to maximize functional and oncologic outcomes in nephron preservation and complication-free recovery. Thus, the paradigm in the surgical treatment of RCC has transformed and will continue to change in the future. In addition, targeted therapy, immunotherapy, and combination therapy are adopted to treat patients with advanced RCC. In recent years, the combination of immune checkpoint inhibition and antiangiogenic therapy is a very attractive combined therapeutic strategy for advanced/metastatic RCCs. Biomarkers, including epigenetic markers for RCC, have been increasing, which will be helpful to discover new therapeutic targets and related inhibitors for the treatment of advanced RCC.

Keywords

  • kidney cancer
  • renal cell carcinoma
  • RCC
  • genetics
  • targeted therapy
  • immunotherapy
  • combination therapy

1. Introduction

Kidney cancer has been known to have distinct histological types with different genetic profiles. The most common type is renal cell carcinoma (RCC), representing more than 90% of all kidney cancers [1]. RCC is consisted of a few subtypes. The most common subtype is clear cell renal cell carcinoma (ccRCC), followed by papillary RCC (pRCC, type I and type II), chromophobe RCC (cRCC), collecting duct carcinoma (CDC), etc. Kidney cancer is the sixth most common malignant disease in men and tenth in women. It accounts for 5% of male and 3% female malignancies [2, 3]. The RCC incidence is constantly rising as well. In urology, kidney cancer is the third most common malignant disease. While kidney cancer occurs mostly in North American and European, the incidence of renal cancer is relatively lower in Asian [4]. Based on the global cancer statistics in 2020 [5, 6], there were an estimated 431,300 people diagnosed with kidney cancer and approximately 179,400 deaths worldwide in 2020. It was recently estimated that there were 81,800 new kidney cancer cases and 14,890 deaths in the United States in 2023 [7]. Diagnosis of RCC at early stages is crucial in treating patients and improving survival rates. With the application of improved diagnostic techniques such as cross-section abdominal imaging [8], the detection rate of RCC has increased in recent years [9]. Choosing the best therapeutic strategy is essential for improving the outcome of patients with RCC.

Advertisement

2. Epidemiology of RCC

RCC is the most lethal urogenital cancer, accounting for 2–3% of all cancers. The incidence of RCC is continuously rising worldwide, and it is higher in developed countries compared to developing countries, and higher in men than women (male to female ratio is 1.5:1). The mortality rate of RCC is 30–40% and is higher in men compared to women [3]. The majority of RCC patients are aged over 60 years. Since the advance in diagnostic methods and public consciousness of the importance of periodic health screening, the number of RCC patients diagnosed in the early stages keeps increasing. Thus, the incidence of RCC has been rising as well in the past three decades [9]. Even though, the mortality rate of RCC has been constantly decreasing, especially in developed countries, due to early therapy and progress of the therapeutic strategies [10]. However, despite the improvement in disease control and increased survival rate, metastases are often observed in many RCC patients [11]. As reported, 30–50% of patients with local RCC progress to metastasis. In addition, metastasis was observed in 20–30% of the RCC patients at the early stage of diagnosis, and nearly 40% of the patients with localized RCC tumors presented distant metastases even after surgery. The most common distant metastatic sites are the lungs, bones (vertebrae, proximal part of the femur, pelvic bones), lymphatic nodes brain, liver, the opposite kidney, and suprarenal glands [12]. The patients with RCC metastases suffer from pain, nerve compression, hypercalcemia, and pathological fractures. The occurrence of metastases often worsens the patients’ prognosis [13]. The median survival for patients with RCC metastases is approximately 8 months and the mortality rate is 50% during the first year, and 10% for 5 years survival [10, 13, 14].

Advertisement

3. Risk factors associated with RCC

While age and gender are strongly associated with increased RCC incidence, hypertension, smoking, obesity, ethnicity, location, and history of using tobacco products increase the risk of RCC as well [3]. It was reported that smoking could increase the risk by about 54% in men and 22% in women. Smoking time and the daily number of cigarettes are directly associated with kidney cancer incidence for both men and women. In addition, some minor risk factors that may be related to RCC include acquired renal cystic disease, chronic kidney disease (CKD), end-stage renal disease (ESRD), exposure to cadmium and trichloroethylene, consumption of red and processed meat, chronic use of palliatives, type-2 diabetes, vitamin D level, viral hepatitis, increased triglycerides, decreased physical activity, and genetic syndromes [15]. Patients with renal insufficiency and in the terminal stage of the disease are four times more likely to develop kidney cancer. Measures of preventing RCC include smoking cessation, and reducing of the body mass index (BMI), etc.

Advertisement

4. Pathophysiology of RCC

Since RCC is a heterogeneous cancer that may stem from different type of renal cells, histological subtyping of RCC may exert a crucial impact on the choice of therapeutic strategies and prognosis. Based on the histological features, RCC is divided into 16 subtypes, including clear cell RCC, pRCC (type I and II), cRCC, collecting duct RCC, multilocular cystic RCC, medullary carcinoma, mucinous tubular, spindle cell carcinoma, neuroblastoma-associated RCC, Xp11.2 translocations—TFE3 carcinoma, hereditary cancer syndromes, and unclassified lesion [16]. Of them, clear cell RCC is the most common subtype of RCC and accounts for 75–85%. The second most common one is pRCC that constitutes 10–15%. cRCC ranked third and represents 3–5% of kidney cancers. Both clear cell carcinoma and pRCC are thought to stem from the proximal tubule, while chromophore RCC originates from the distal connecting tubules (DCT) and collecting duct system, especially the intercalated cells [17]. While most clear cell RCCs are sporadic, approximately 5% of the clear cell RCC is hereditary, and usually related to hereditary syndromes such as Von Hippel–Lindau disease (VHL) and tuberous sclerosis (TS). Clear cell RCC tends to metastasize to the lymph nodes, the lungs, the liver, and bones. Clear cell RCC has poorer prognosis compared to pRCC and cRCC. On the basis of histological and genetic differences, pRCC is further divided into two subtypes: type I and type II. The type I pRCC cannot be distinguished from type II through routine imaging techniques. Papillary type I can be detected at an early stage, and thus has a better prognosis compared to type II. cRCC is commonly observed in patients aged more than 60, and less aggressive compared to clear cell RCC. Therefore, cRCC has the best prognosis among all RCCs [4].

Advertisement

5. Genetics of RCC

The first categorization of kidney cancer based on molecular genetics was conducted by Heidelberg in 1997, and it was adopted in the WHO tumor classification of 2004, 2012 Vancouver ISUP [18], and the latest RCC classification of the WHO (2016) [16]. RCC has various genetic alterations, including gene mutations, epigenetic modifications, and chromosomal aberrations. While genetic alterations can cause both sporadic and hereditary types, patients with familial history (hereditary) represent only 3% of all kidney cancer cases. An increased number of RCC-related genes were identified in the past decades. Considering its high genetic heterogeneity, RCC can be caused by mutations in many genes and chromosomal abnormalities. Each subtype of RCC has its corresponding affected genes [19]. Clear cell RCC-related genes include VHL, PBRM1, BAP1, STED2, JARID1c, FLCN, and c-MYC. VHL mutations account for approximately 60% of clear cell RCC [20]. In addition, exome-sequencing has revealed that PBRM1, SETD2, BAP1, and JARID1c are associated with RCC as well (PBRM1, 40%, SETD2, 12%, BAP1,10%, and KDM5C,5%) [21]. Combinational mutations of the above genes were also detected in many clear cell RCCs. With the advent of new genetic tools, more RCC-related genes/biomarkers might be uncovered. pRCC is divided into two types: Type I and Type II. Although pRCC type I and type II are morphologically similar, they are cytologically different. Compare to pRCC II, pRCC type I has a low-grade tendency and better prognosis [22]. pRCC I is often caused by overexpression of the MET gene on chromosome 7q31, while pRCC II is less correlated to MET overexpression but associated with alterations in FH, CDKN2A, SETD2, BAP1, PBRM1, FLCN, NRF2-ARE, TERT, TFE3, and increased expression of NF2 [23, 24]. cRCC is a relatively rare type of RCC that originates from the distal convoluted tubule. cRCC is associated with mutations in PTEN, TP53, mTOR, c-kit, FAAH2, PDHB, PDXD1, ZNF765, PRKAG2, ARID1A, and ABHD3. Of them, PTEN mutation is the most common event in cRCC [25]. In addition, mutations in FLCN appear to cause all types of RCC in Flcn knockout mouse models [26, 27].

Chromosomal aberration is also a common feature in RCC [28]. In clear cell RCC, the loss of the short arm of chromosome 3 is the most frequently occurred event, which is related to the loss of the VHL gene located on 3p [28, 29]. Other chromosomal aberrations include loss of chromosome Y, gaining 5q31, 8q, 4p, 14q, 9p, and trisomy of chromosome 7. Patients with gaining of 4p, 9p, and 14q have poor prognosis while gaining 5q31 is connected with prolonged survival in high-grade clear cell RCC. Deletion of the Y chromosome usually leads to clear cell RCC with distant metastasis. In addition, gaining chromosome 8q can cause metastasis of clear cell RCC, which may be associated with overexpression of C-MYC. For chromosomal abnormity, pRCC type I is also different from papillary type II. Trisomy of chromosomes (e.g., 3q, 7, 8, 12, 16, 17, 20) commonly appears in type I pRCC. In addition, loss of chromosome Y is an important feature of papillary type II in men. In contrast, gaining a chromosome 8q and losing 1p and 9p are frequently observed in RCC papillary type II. cRCC is also associated with chromosome aberrations, including loss of chromosomes 1, 2, 6, 10, 13, 17, and 21 [20]. Losing copies of chromosomes 1, 2, 6, 10, 13, and 17 are more commonly observed in the classical type of cRCC than the eosinophilic type [30]. In addition, gaining chromosome copies (e.g., 4, 7, 11, 12, 14q, and 18q) is related to cRCC as well [31].

Advertisement

6. Clinical diagnosis of RCC

Since small RCC masses are usually asymptomatic, the majority of RCCs are accidentally detected by routine imaging for various medical screening purposes [32]. Patients diagnosed with RCC based on the symptoms account for only 30%, and approximately 20–30% of the patients present metastasis at the time of diagnosis. Flank pain, hematuria, and abdominal mass are usually considered as the classic symptoms for RCC diagnosis, but these symptoms are observed in only 4–17% of the cases. Other symptoms for diagnosis include fever, abdominal pain, anemia-induced fatigue, weight loss, bone pain, and cough caused by metastasis of cancer cells or lower limb edema, peripheral lymphadenopathy (LAP), and varicocele caused by inferior vena cava (IVC) or renal thrombosis [33]. Once signs and symptoms appear, further laboratory investigation should be performed. The laboratory investigation includes renal function tests, complete blood cell count (CBC), liver function tests, urinary analysis, thyroid function tests, and examination of the level of calcium, lactate dehydrogenase, and alkaline phosphatase.

Various preclinical imaging modalities are valuable tools for detecting renal masses. These imaging modalities include ultrasound, CEUS, computed tomography (CT) scan, magnetic resonance imaging (MRI), positron emission tomography (PET)/CT, etc. Many renal masses and benign cystic kidney lesions can be easily detected with ultrasound [34]. In addition, as an inexpensive and accurate imaging approach, contrast-enhanced ultrasound (CEUS) has been adopted to evaluate indeterminate renal lesions, though it may not effectively discriminate between malignant and benign renal masses. For more accurately identifying malignant masses, CT scan or MRI is required [35]. In some cases, both CT and MRI should be performed by contrast because contrast absorption is a key factor in determining malignant masses [36]. For small lesions (1–2 cm) and in cases of renal insufficiency, pregnant women, and patients allergic to contrast material, MRI should be conducted [37]. The primary purpose of imaging is to inspect the characteristics of the affected mass, identify possible abdominal metastases, mass expansion, and venous involvement for staging. PET scan, is not a standard scan strategy, is very helpful in the diagnosis and follow-up of RCC. Thus, routine use of PET/CT is also recommended [38]. Other imaging approaches, including advanced MRI techniques or the combination use of iodine PET and CT, may be performed to determine renal masses [39]. In addition, biopsy and histopathology are required to carry out in suspected masses before further treatment.

Advertisement

7. Treatment and management of RCC

To date, surgery is still an essential treatment for RCC, and RE nephrectomy has been regarded as the standard of care for the management of renal tumors. For small renal lesions and surgery-induced chronic kidney disorders, active surveillance of nephron surgery and minimally invasive approaches have been adopted to limit the invasion and loss of kidney function in clinical [40]. RCC patients with severe comorbidities, elderly patients with small tumors ≤4 cm, or patients with a short life expectancy are recommended for active surveillance [41]. In some cases, active surveillance is also suggested to monitor the rate of the large tumor’s growth. In addition, imaging can be performed for active surveillance in the first half year, and then, every 6 months at 2–3 years, and later, annually [42]. In addition, for treating small randomly detected tumors, minimally invasive procedures should be used. Cryotherapy (CRYO) and radiofrequency ablations (RFA) were suggested for patients with only one kidney or for those unable to undergo major surgery.

7.1 Nephron preservation surgery

While radical nephrectomy is the removal of the entire kidney and is suitable for patients with larger renal tumors (T2, >7 cm), nephron-sparing partial nephrectomy only removes the small localized renal tumor (T1, <7 cm) and preserves the parenchyma. Thus, in most cases including locally advanced and metastatic diseases, nephron-sparing partial nephrectomy is recommended except for those that technically impossible to be removed due to their unfavorable location.

The advent and improvement of robot-assisted surgical approaches and robotic platforms have helped to close the technical gap and allow for greater adoption of the laparoscopic approach and nephron-sparing surgery, leading to a revolution for surgical management of RCC in recent decades [43, 44]. As the use of robotic platforms combined with imaging techniques in surgery, larger, higher stage, and more complex renal lesions can now be treated in a minimally invasive fashion, which leads to decreased morbidity, shortened patients’ hospitalization, and less side effects [45, 46]. Robot-assisted surgery has continued to evolve and has been playing an ever-expanding role in the treatment of RCC worldwide [45]. Laparoscopic partial nephrectomy and robot-assisted partial nephrectomy are becoming the standard treatment for patients eligible for nephron-sparing surgery. While the nephron-sparing surgery has been endorsed as the gold standard treatment of T1a tumors ≤4 cm in size and T1b lesions in the United States and Europe when technically possible with experienced surgeons [4147], robot-assisted partial nephrectomy has been adopted for treatment of cT2 RCC [48]. While new robotic platforms such as single-port (SP) robotic surgical system and multiport robotic surgical systems are currently under development, the popular da Vinci platform has significantly expanded the laparoscopic paradigm spectrum in the surgical treatment of RCC [49, 50, 51].

7.2 Cytokine treatment of metastatic disease

Before targeted therapy, patients with metastatic clear cell RCC were previously treated with immunosuppressive agents such as INFα and IL2, namely systemic therapy. In 1990s, systemic therapy with a high-dose IL2 turned into a commonly used modality for various cancer patients with metastasis. However, complete response was only observed in <10% of patients treated with a longer high dose of IL-2. It simultaneously caused severe toxic effects. Thus, high-dose IL2 treatment is not recommended for patients with metastatic RCC unless the patients are young and in very good conditions with low tumor volume [52].

Aldesleukin and INFα (along with Bevacizumab) are the only safe modulating drugs that are approved in selected metastatic RCC. INFα is adopted to treat RCC patients in various formulas with a response rate of 10–15% and a response time of 4 months. Patients who benefit from INF treatment were then subjected to nephrectomy. Although INFα treatment shows some efficacy in the treatment of patients with metastatic RCC, it is not suggested for treating RCC patients as a single drug.

7.3 Targeted therapy and immunotherapy of metastatic RCC

In the past decades, the most significant advance in the therapeutic strategy of metastatic RCC has been the development of treatments that specifically target the RCC-related biological pathways and related biomarkers [53, 54]. In the 1990s, cytokines, such as IFNα and IL-2, were used to treat metastatic clear cell RCC (Figure 1). In the 2000s, targeted therapies that target the VEGF/PDGFR/mTOR pathways replaced the cytokine therapies. In the targeted therapies, tyrosine kinase inhibitors (TKIs) are effective agents in the treatment of metastatic RCC, which was used as the first line and the second line treatment options. To date, five TKIs (e.g., cabozantinib, axitinib, pazopanib, sorafenib, and sunitinib) have been approved internationally for the targeted therapy of metastatic RCC (Figure 1). Cabozantinib targets the tyrosine-protein kinase receptor UFO and the MET receptor tyrosine kinase. Pazopanib targets the VEGFR, while axitinib inhibits VEGFR with improved specificity, sorafenib an multi-kinase inhibitor, inhibits RAF-1, B-RAF, VEGFR-2, VEGFR-3, PDGF-β, KIT, and FLT-3. Sunitinib targets VEGFR2(Flk-1) and PDGFR-β. Whereas, lenvatinib suppresses VEGFRs and fibroblast growth factor receptors [5556]. In addition, temsirolimus and everolimus, two mTOR inhibitors, have been approved for the treatment of advanced RCC as well. Bevacizumab, an anti-VEGF monoclonal antibody, is approved for the treatment of advanced RCC when it is used in combination with INF. In 2015, nivolumab, an anti-PD-1 immune checkpoint inhibitor that prevents signaling through programmed cell death 1, has been approved for the first immunotherapy agent of metastatic RCC [57]. Later, ipilimumab, an anti-CTLA-4 monoclonal antibody, in combination with nivolumab was approved for the first-line treatment of advanced RCC [20, 58]. Recently, increasing attention has been paid to the combination therapy. The latest combination therapy strategies such as axitinib–avelumab (target VEGF and immune checkpoints) [5960], axitinib–pembrolizumab (target VEGFR) in combination with PD-L1 and PD-1, respectively, have been used to treat metastatic RCC. To date, immune checkpoint inhibition plus antiangiogenic therapy constitutes a very promising combined therapeutic strategy for advanced/metastatic RCCs.

Figure 1.

Development of approved anti-RCC agents.

In addition, some epigenetic markers have been proposed as promising epigenetic RCC markers based on DNA methylation, ncRNA expression, and histone modification [61, 62, 63]. Many epigenetic markers and epigenetic modifiers are likely candidates for clinical use, but further validation is needed [63, 64]. The development of epigenetic therapies, either alone or in combination with antiangiogenic agents and/or immune-checkpoint inhibitors, is a hopeful therapeutic strategy for RCC.

Advertisement

8. Conclusion

RCC accounts for 4%of all malignant tumors. In urology, RCC is the third most common malignant tumor. Risk factors include positive family history, smoking, obesity, high blood pressure, etc. Although most cases of RCC are diagnosed accidentally, the improvement in diagnostic approaches has increased the detection rate over the past decades. Many RCC-related genes and predictive biomarkers are being identified and may further improve the diagnosis of RCC. In addition, choosing the best therapeutic strategy is critical to improve the outcome of RCC. The tumor size, the stage of the disorder, and the surgeon’s experience are the determinant impact factors in the choice of the optimal treatment approach. While the open surgery is still reserved for locally advanced diseases, it is gradually displaced by robotic-assisted partial nephrectomy. Although the optimal first-line treatment, including targeted therapy, immunotherapy, or combined therapy for metastatic RCC may differ by clinical risk group and efficacy endpoint, the first-line treatment landscape for metastatic RCC is rapidly evolving. Thus, knowledge of the latest advances in the diagnosis and management of RCC could assist the related researchers, physicians, and nephrologists, to better diagnose and treat RCC.

References

  1. 1. Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nature Reviews. Disease Primers. 2017;3:17009
  2. 2. Miller KD, Goding Sauer A, Ortiz AP, Fedewa SA, Pinheiro PS, Tortolero-Luna G, et al. Cancer statistics for hispanics/latinos, 2018. CA: A Cancer Journal for Clinicians. 2018;68(6):425-445
  3. 3. Bahadoram S, Davoodi M, Hassanzadeh S, Bahadoram M, Barahman M, Mafakher L. Renal cell carcinoma: An overview of the epidemiology, diagnosis, and treatment. Giornale Italiano di Nefrologia. 2022;39(3)
  4. 4. Capitanio U, Bensalah K, Bex A, Boorjian SA, Bray F, Coleman J, et al. Epidemiology of renal cell carcinoma. European Urology. 2019;75(1):74-84
  5. 5. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA: A Cancer Journal for Clinicians. 2020;70(1):7-30
  6. 6. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021;71(3):209-249
  7. 7. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA: A Cancer Journal for Clinicians. 2023;73(1):17-48
  8. 8. Kane CJ, Mallin K, Ritchey J, Cooperberg MR, Carroll PR. Renal cell cancer stage migration: analysis of the National Cancer Data Base. Cancer. 2008;113(1):78-83
  9. 9. Ljungberg B, Campbell SC, Choi HY, Jacqmin D, Lee JE, Weikert S, et al. The epidemiology of renal cell carcinoma. European Urology. 2011;60(4):615-621
  10. 10. Medina-Rico M, Ramos HL, Lobo M, Romo J, Prada JG. Epidemiology of renal cancer in developing countries: review of the literature. Canadian Urological Association Journal. 2018;12(3):E154-E162
  11. 11. Vasudev NS, Wilson M, Stewart GD, Adeyoju A, Cartledge J, Kimuli M, et al. Challenges of early renal cancer detection: symptom patterns and incidental diagnosis rate in a multicentre prospective UK cohort of patients presenting with suspected renal cancer. BMJ Open. 2020;10(5):e035938
  12. 12. Ather MH, Masood N, Siddiqui T. Current management of advanced and metastatic renal cell carcinoma. Urology Journal. 2010;7(1):1-9
  13. 13. Umer M, Mohib Y, Atif M, Nazim M. Skeletal metastasis in renal cell carcinoma: a review. Ann Med Surg (Lond). 2018;27:9-16
  14. 14. Dovalova D, Rybar L, El Falougy H, Kubikova E, Mifkovic A. Renal cell carcinoma - summarizing overview, biomarkers, metastases and new perspectives. Bratislavské Lekárske Listy. 2022;123(10):697-704
  15. 15. Choueiri TK, Je Y, Cho E. Analgesic use and the risk of kidney cancer: a meta-analysis of epidemiologic studies. International Journal of Cancer. 2014;134(2):384-396
  16. 16. Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO classification of tumours of the urinary system and male genital organs - Part A: renal, penile, and testicular tumours. European Urology. 2016;70(1):93-105
  17. 17. Warren AY, Harrison D. WHO/ISUP classification, grading and pathological staging of renal cell carcinoma: standards and controversies. World Journal of Urology. 2018;36(12):1913-1926
  18. 18. Delahunt B, Srigley JR, Montironi R, Egevad L. Advances in renal neoplasia: recommendations from the 2012 International Society of Urological Pathology Consensus Conference. Urology. 2014;83(5):969-974
  19. 19. Cairns P. Renal cell carcinoma. Cancer Biomarkers. 2010;9(1-6):461-473
  20. 20. Maher ER. Hereditary renal cell carcinoma syndromes: diagnosis, surveillance and management. World Journal of Urology. 2018;36(12):1891-1898
  21. 21. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469(7331):539-542
  22. 22. Klatte T, Rao PN, de Martino M, LaRochelle J, Shuch B, Zomorodian N, et al. Cytogenetic profile predicts prognosis of patients with clear cell renal cell carcinoma. Journal of Clinical Oncology. 2009;27(5):746-753
  23. 23. Tovar EA, Graveel CR. MET in human cancer: germline and somatic mutations. Annals of Translational Medicine. 2017;5(10):205
  24. 24. Furge KA, Chen J, Koeman J, Swiatek P, Dykema K, Lucin K, et al. Detection of DNA copy number changes and oncogenic signaling abnormalities from gene expression data reveals MYC activation in high-grade papillary renal cell carcinoma. Cancer Research. 2007;67(7):3171-3176
  25. 25. Durinck S, Stawiski EW, Pavia-Jimenez A, Modrusan Z, Kapur P, Jaiswal BS, et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nature Genetics. 2015;47(1):13-21
  26. 26. Chen J, Futami K, Petillo D, Peng J, Wang P, Knol J, et al. Deficiency of FLCN in mouse kidney led to development of polycystic kidneys and renal neoplasia. PLoS One. 2008;3(10):e3581
  27. 27. Chen J, Huang D, Rubera I, Futami K, Wang P, Zickert P, et al. Disruption of tubular Flcn expression as a mouse model for renal tumor induction. Kidney International. 2015;88(5):1057-1069
  28. 28. Quddus MB, Pratt N, Nabi G. Chromosomal aberrations in renal cell carcinoma: An overview with implications for clinical practice. Urology Annals. 2019;11(1):6-14
  29. 29. Chen J, Lui WO, Vos MD, Clark GJ, Takahashi M, Schoumans J, et al. The t(1;3) breakpoint-spanning genes LSAMP and NORE1 are involved in clear cell renal cell carcinomas. Cancer Cell. 2003;4(5):405-413
  30. 30. Ohashi R, Schraml P, Angori S, Batavia AA, Rupp NJ, Ohe C, et al. Classic chromophobe renal cell carcinoma incur a larger number of chromosomal losses than seen in the eosinophilic subtype. Cancers (Basel). 2019;11(10):1492
  31. 31. Tan MH, Wong CF, Tan HL, Yang XJ, Ditlev J, Matsuda D, et al. Genomic expression and single-nucleotide polymorphism profiling discriminates chromophobe renal cell carcinoma and oncocytoma. BMC Cancer. 2010;10:196
  32. 32. Tsui KH, Shvarts O, Smith RB, Figlin R, de Kernion JB, Belldegrun A. Renal cell carcinoma: prognostic significance of incidentally detected tumors. The Journal of Urology. 2000;163(2):426-430
  33. 33. Mota M, Bezerra ROF, Garcia MRT. Practical approach to primary retroperitoneal masses in adults. Radiologia Brasileira. 2018;51(6):391-400
  34. 34. Helenon O, Crosnier A, Verkarre V, Merran S, Mejean A, Correas JM. Simple and complex renal cysts in adults: classification system for renal cystic masses. Diagnostic and Interventional Imaging. 2018;99(4):189-218
  35. 35. Agnello F, Albano D, Micci G, Di Buono G, Agrusa A, Salvaggio G, et al. CT and MR imaging of cystic renal lesions. Insights Into Imaging. 2020;11(1):5
  36. 36. Johnson PT, Horton KM, Fishman EK. Optimizing detectability of renal pathology with MDCT: protocols, pearls, and pitfalls. AJR. American Journal of Roentgenology. 2010;194(4):1001-1012
  37. 37. Kim JH, Sun HY, Hwang J, Hong SS, Cho YJ, Doo SW, et al. Diagnostic accuracy of contrast-enhanced computed tomography and contrast-enhanced magnetic resonance imaging of small renal masses in real practice: sensitivity and specificity according to subjective radiologic interpretation. World Journal of Surgical Oncology. 2016;14(1):260
  38. 38. Farolfi A, Koschel S, Murphy DG, Fanti S. PET imaging in urology: a rapidly growing successful collaboration. Current Opinion in Urology. 2020;30(5):623-627
  39. 39. Courcier J, de la Taille A, Nourieh M, Leguerney I, Lassau N. Ingels A: Carbonic anhydrase IX in renal cell carcinoma, implications for disease management. International Journal of Molecular Sciences. 2020;21(19):7146
  40. 40. Hora M, Eret V, Travnicek I, Prochazkova K, Pitra T, Dolejsova O, et al. Surgical treatment of kidney tumors - contemporary trends in clinical practice. Central European Journal of Urology. 2016;69(4):341-346
  41. 41. Campbell S, Uzzo RG, Allaf ME, Bass EB, Cadeddu JA, Chang A, et al. Renal mass and localized renal cancer: AUA guideline. The Journal of Urology. 2017;198(3):520-529
  42. 42. Sebastia C, Corominas D, Musquera M, Pano B, Ajami T, Nicolau C. Active surveillance of small renal masses. Insights Into Imaging. 2020;11(1):63
  43. 43. Potretzke AM, Weaver J, Benway BM. Review of robot-assisted partial nephrectomy in modern practice. Journal of Kidney Cancer and VHL. 2015;2(2):30-44
  44. 44. Phung MC, Lee BR. Recent advancements of robotic surgery for kidney cancer. Asian Journal of Endoscopic Surgery. 2018;11(4):300-307
  45. 45. Ghani KR, Sukumar S, Sammon JD, Rogers CG, Trinh QD, Menon M. Practice patterns and outcomes of open and minimally invasive partial nephrectomy since the introduction of robotic partial nephrectomy: results from the nationwide inpatient sample. The Journal of Urology. 2014;191(4):907-912
  46. 46. Li M, Cheng L, Zhang H, Ma L, Wang Y, Niu W, et al. Laparoscopic and robotic-assisted partial nephrectomy: an overview of hot issues. Urologia Internationalis. 2020;104(9-10):669-677
  47. 47. Ljungberg B, Bensalah K, Canfield S, Dabestani S, Hofmann F, Hora M, et al. EAU guidelines on renal cell carcinoma: 2014 update. European Urology. 2015;67(5):913-924
  48. 48. Suek T, Davaro F, Raza SJ, Hamilton Z. Robotic surgery for cT2 kidney cancer: analysis of the National Cancer Database. Journal of Robotic Surgery. 2022;16(3):723-729
  49. 49. Carbonara U, Amparore D, Borregales LD, Calio A, Ciccarese C, Diana P, et al. Single-port robotic partial nephrectomy: impact on perioperative outcomes and hospital stay. Therapeutic Advances in Urology. 2023;15:17562872231172834
  50. 50. Salkowski M, Checcucci E, Chow AK, Rogers CC, Adbollah F, Liatsikos E, et al. New multiport robotic surgical systems: a comprehensive literature review of clinical outcomes in urology. Therapeutic Advances in Urology. 2023;15:17562872231177781
  51. 51. Valero R, Sawczyn G, Garisto J, Yau R, Kaouk J. Multiquadrant combined robotic radical prostatectomy and left partial nephrectomy: a combined procedure by a single approach. Actas Urológicas Españolas (English Edition). 2020;44(2):119-124
  52. 52. McDermott DF, Regan MM, Clark JI, Flaherty LE, Weiss GR, Logan TF, et al. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. Journal of Clinical Oncology. 2005;23(1):133-141
  53. 53. Dizman N, Philip EJ, Pal SK. Genomic profiling in renal cell carcinoma. Nature Reviews. Nephrology. 2020;16(8):435-451
  54. 54. Osawa T, Takeuchi A, Kojima T, Shinohara N, Eto M, Nishiyama H. Overview of current and future systemic therapy for metastatic renal cell carcinoma. Japanese Journal of Clinical Oncology. 2019;49(5):395-403
  55. 55. Choueiri TK, Escudier B, Powles T, Mainwaring PN, Rini BI, Donskov F, et al. Cabozantinib versus everolimus in advanced renal-cell carcinoma. The New England Journal of Medicine. 2015;373(19):1814-1823
  56. 56. Motzer RJ, Hutson TE, Glen H, Michaelson MD, Molina A, Eisen T, et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. The Lancet Oncology. 2015;16(15):1473-1482
  57. 57. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. The New England Journal of Medicine. 2015;373(19):1803-1813
  58. 58. Jonasch E. NCCN guidelines updates: management of metastatic kidney cancer. Journal of the National Comprehensive Cancer Network. 2019;17(5.5):587-589
  59. 59. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. The New England Journal of Medicine. 2019;380(12):1103-1115
  60. 60. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. The New England Journal of Medicine. 2019;380(12):1116-1127
  61. 61. Angulo JC, Manini C, Lopez JI, Pueyo A, Colas B, Ropero S. The role of epigenetics in the progression of clear cell renal cell carcinoma and the basis for future epigenetic treatments. Cancers (Basel). 2021;13(9):2071
  62. 62. Xie L, Wu S, He R, Li S, Lai X, Wang Z. Identification of epigenetic dysregulation gene markers and immune landscape in kidney renal clear cell carcinoma by comprehensive genomic analysis. Frontiers in Immunology. 2022;13:901662
  63. 63. Burlibasa L, Nicu AT, Chifiriuc MC, Medar C, Petrescu A, Jinga V, et al. H3 histone methylation landscape in male urogenital cancers: from molecular mechanisms to epigenetic biomarkers and therapeutic targets. Frontiers in Cell and Development Biology. 2023;11:1181764
  64. 64. El Khoury LY, Fu S, Hlady RA, Wagner RT, Wang L, Eckel-Passow JE, et al. Identification of DNA methylation signatures associated with poor outcome in lower-risk Stage, Size, Grade and Necrosis (SSIGN) score clear cell renal cell cancer. Clinical Epigenetics. 2021;13(1):12

Written By

Jindong Chen

Submitted: 04 May 2023 Reviewed: 24 July 2023 Published: 09 August 2023