Attention: Restrictions on use of AUA, AUAER, and UCF content in third party applications, including artificial intelligence technologies, such as large language models and generative AI.
You are prohibited from using or uploading content you accessed through this website into external applications, bots, software, or websites, including those using artificial intelligence technologies and infrastructure, including deep learning, machine learning and large language models and generative AI.

Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow Up (2021)

Using AUA Guidelines

This AUA guideline is provided free of use to the general public for academic and research purposes. However, any person or company accessing AUA guidelines for promotional or commercial use must obtain a licensed copy. To obtain the licensable copy of this guideline, please contact Keith Price at kprice@auanet.org.

To cite this guideline:
Campell SC, Clark PE, Chang SS et al: Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow-Up: AUA Guideline Part I. J Urol 2021; 206: 199.
Campbell SC, Uzzo RG, Karam JA, et al: Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow-up: AUA Guideline: Part II. J Urol 2021; 206: 209.

Guideline as it appears in The Journal of Urology® Part I [pdf] Part II [pdf]
Unabridged version of this Guideline [pdf]
RCC Follow-up Algorithm [pdf]
RCC Active Surveillance Algorithm [pdf]
RCC Evaluation and Intervention Algorithm [pdf]

Panel Members

Steven Campbell, MD, PhD; Robert G. Uzzo, MD; Mohamad E. Allaf, MD; Jay Todd Bishoff, MD; Eric B. Bass, MD, MPH; Jeffrey A. Cadeddu, MD; Anthony Chang, MD; Sam S. Chang, MD; Peter E. Clark, MD; Jonathan A. Coleman, MD; Philipp Dahm, MD; Brian J. Davis, MD, PhD; Ithaar H. Derweesh, MD; Mireya Diaz, PhD; Sherri M. Donat , MD; Leo Giambarresi, PhD; Debra A. Gervais, MD; S. Duke Herrell III, MD; Susan Hilton, MD; Susie L. Hu, MD; Eric Jonasch, MD; Jose A. Karam, MD; Brian R. Lane, MD, PhD; Bradley C. Leibovich, MD, FACS; Daniel Wei Lin,MD; Philip M. Pierorazio, MD; Victor Edward Reuter, MD; Lesley Souter, PhD

Executive Summary

Purpose

This AUA Guidelines focuses on the evaluation and management of clinically localized sporadic renal masses suspicious for renal cell carcinoma (RCC) in adults, including solid enhancing renal tumors and Bosniak 3 and 4 complex cystic renal masses. Some patients with clinically localized renal masses may present with findings suggesting aggressive tumor biology or may be upstaged on exploration or final pathology. Management considerations pertinent to the urologist in such patients will also be discussed. The follow-up of renal cancer patients after intervention is also addressed, including recommendations for periodic clinical follow-up and abdominal and chest imaging. Practice patterns regarding such tumors vary considerably, and the literature regarding evaluation, management, and surveillance has been rapidly evolving. Notable examples include controversies about the role of renal mass biopsy (RMB) and concerns regarding overutilization of radical nephrectomy (RN). Please also refer to the associated Renal Mass and Localized Renal Cancer Treatment and Follow-up after Intervention algorithms.

Methodology

The systematic review utilized in the creation of this guideline was completed in part through the Agency for Healthcare Research and Quality (AHRQ) and through additional supplementation that further addressed additional key questions and more recently published literature. A research librarian experienced in conducting literature searches for comparative effectiveness reviews searched in MEDLINE®, Embase®, the Cochrane Library, the Database of Abstracts of Reviews of Effects, the Health Technology Assessment Database, and the UK National Health Service Economic Evaluation database to capture both published and gray literature published from January 1, 1997 through May 1, 2015. A supplemental search was conducted adding additional literature published through August 2015, and a final update search was conducted through July 2016.

A systematic review was conducted in 2013 to identify published articles relevant to key questions specified by the Panel related to kidney neoplasms and their follow-up (imaging, renal function, markers, biopsy, and prognosis). This search covered English-language articles published between January 1999 and 2011. An updated query was later conducted to include studies published through August 2012.

In January of 2021, the Renal Mass and Localized Renal Cancer guideline underwent an additional amendment based on a current literature search. This literature search retrieved additional studies published between July 2016 to October 2020 using the same Key Questions and search criteria from the Renal Mass and Localized Renal Cancer guideline. Nineteen studies were identified from this search to provide data relevant to the management and treatment of Renal Mass. In addition, the Follow-Up for Clinically Localized Renal Neoplasms guideline published in 2013 was merged with the Renal Mass and Localized Renal Cancer guideline. Although the systematic search for follow-up interventions was not updated to 2020, the panel members conducted a comprehensive review of all evidence published since the original guideline. The language of many statements has been refined for clarity. For all evidence-based statements, supporting studies were identified only in the original systematic review and the evidence strength was not altered.

When sufficient evidence existed, the body of evidence for a particular treatment was assigned a strength rating of A (high), B (moderate) or C (low) for support of Strong, Moderate, or Conditional Recommendations. In the absence of sufficient evidence, additional information is provided as Clinical Principles and Expert Opinions.

Initial Evaluation and Diagnosis

Evaluation

  1. In patients with a solid or complex cystic renal mass, clinicians should obtain high quality, multiphase, cross-sectional abdominal imaging to optimally characterize and clinically stage the renal mass. Characterization of the renal mass should include assessment of tumor complexity, degree of contrast enhancement (where applicable), and presence or absence of fat. (Clinical Principle)
  2. In patients with suspected renal malignancy, clinicians should obtain a comprehensive metabolic panel, complete blood count, and urinalysis. Metastatic evaluation should include chest imaging to evaluate for possible thoracic metastases. (Clinical Principle)
  3. For patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians should assign chronic kidney disease (CKD) stage based on glomerular filtration rate (GFR) and degree of proteinuria. (Expert Opinion)

Counseling

  1. In patients with a solid or Bosniak 3/4 complex cystic renal mass, a urologist should lead the counseling process and should consider all management strategies. A multidisciplinary team should be included when necessary. (Expert Opinion)
  2. Clinicians should provide counseling that includes current perspectives about tumor biology and a patient-specific risk assessment inclusive of sex, tumor size/complexity, histology (when obtained), and imaging characteristics. For cT1a tumors, the low oncologic risk of many small renal masses should be reviewed. (Clinical Principle)
  3. During counseling of patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians must review the most common and serious urologic and non-urologic morbidities of each treatment pathway and the importance of patient age, comorbidities/frailty, and life expectancy. (Clinical Principle)
  4. Clinicians should review the importance of renal functional recovery related to renal mass management, including the risks of progressive CKD, potential short- or long-term need for renal replacement therapy, and long-term overall survival considerations. (Clinical Principle)
  5. Clinicians should consider referral to nephrology in patients with a high risk of CKD progression, including those with estimated glomerular filtration rate (eGFR) less than 45 mL/min/1.73m2, confirmed proteinuria, diabetics with preexisting CKD, or whenever eGFR is expected to be less than 30 mL/min/1.73m2 after intervention. (Expert Opinion)
  6. Clinicians should recommend genetic counseling for any of the following: all patients ≤ 46 years of age with renal malignancy, those with multifocal or bilateral renal masses, or whenever 1) the personal or family history suggests a familial renal neoplastic syndrome; 2) there is a first-or second-degree relative with a history of renal malignancy or a known clinical or genetic diagnosis of a familial renal neoplastic syndrome (even if kidney cancer has not been observed); or 3) the patient’s pathology demonstrates histologic findings suggestive of such a syndrome. (Expert Opinion)

Renal Mass Biopsy (RMB)

  1. When considering the utility of RMB, patients should be counseled regarding rationale, positive and negative predictive values, potential risks and non-diagnostic rates of RMB. (Moderate Recommendation; Evidence Level: Grade C)
  2. Clinicians should consider RMB when a mass is suspected to be hematologic, metastatic, inflammatory, or infectious. (Clinical Principle)
  3. In the setting of a solid renal mass, RMB should be obtained on a utility-based approach whenever it may influence management. RMB is not required for 1) young or healthy patients who are unwilling to accept the uncertainties associated with RMB; or 2) older or frail patients who will be managed conservatively independent of RMB findings. (Expert Opinion)
  4. For patients with a solid renal mass who elect RMB, multiple core biopsies should be performed and are preferred over fine needle aspiration (FNA). (Moderate Recommendation; Evidence Level: Grade C)

Management

Partial nephrectomy (PN) and nephron-sparing approaches

  1. Clinicians should prioritize PN for the management of the cT1a renal mass when intervention is indicated. In this setting, PN minimizes the risk of CKD or CKD progression and is associated with favorable oncologic outcomes, including excellent local control. (Moderate Recommendation; Evidence Level: Grade B)
  2. Clinicians should prioritize nephron-sparing approaches for patients with solid or Bosniak 3/4 complex cystic renal masses and an anatomic or functionally solitary kidney, bilateral tumors, known familial RCC, preexisting CKD, or proteinuria. (Moderate Recommendation; Evidence Level: Grade C)
  3. Nephron-sparing approaches should be considered for patients with solid or Bosniak 3/4 complex cystic renal masses who are young, have multifocal masses, or comorbidities that are likely to impact renal function in the future, including but not limited to moderate to severe hypertension, diabetes mellitus, recurrent urolithiasis, or morbid obesity. (Moderate Recommendation; Evidence Level: Grade C)
  4. In patients who elect PN, clinicians should prioritize preservation of renal function by optimizing nephron mass preservation and avoiding prolonged warm ischemia. (Expert Opinion)
  5. For patients undergoing PN, clinicians should prioritize negative surgical margins. The extent of normal parenchyma removed should be determined by surgeon discretion taking into account the clinical situation and tumor characteristics, including growth pattern, and interface with normal tissue. Tumor enucleation should be considered in patients with familial RCC, multifocal disease, or severe CKD to optimize parenchymal mass preservation. (Expert Opinion)

Radical Nephrectomy (RN)

  1. Clinicians should consider RN for patients with a solid or Bosniak 3/4 complex cystic renal mass whenever increased oncologic potential is suggested by tumor size, RMB (if obtained), and/or imaging. (Moderate Recommendation; Evidence Level: Grade B) In this setting, RN is preferred if all of the following criteria are met: 1) high tumor complexity and PN would be challenging even in experienced hands; 2) no preexisting CKD or proteinuria; and 3) normal contralateral kidney and new baseline eGFR will likely be greater than 45 mL/min/1.73m2 even if RN is performed. If all of these criteria are not met, PN should be considered unless there are overriding concerns about the safety or oncologic efficacy of PN. (Expert Opinion)

Surgical Principles

  1. For patients who are undergoing surgical excision of a renal mass with clinically concerning regional lymphadenopathy, clinicians should perform a lymph node dissection including all clinically positive nodes for staging purposes. (Expert Opinion)
  2. For patients who are undergoing surgical excision of a renal mass, clinicians should perform adrenalectomy if imaging and/or intraoperative findings suggest metastasis or direct invasion of the adrenal gland. (Clinical Principle)
  3. In patients undergoing surgical excision of a renal mass, a minimally invasive approach should be considered when it would not compromise oncologic, functional, and perioperative outcomes. (Expert Opinion)

Other Considerations

  1. Pathologic evaluation of the adjacent renal parenchyma should be performed and recorded after PN or RN to assess for possible intrinsic renal disease, particularly for patients with CKD or risk factors for developing CKD. (Clinical Principle)
  2. Clinicians should consider referral to medical oncology whenever there is concern for potential clinical metastasis or incompletely resected disease (macroscopic positive margin or gross residual disease). Patients with high-risk or locally advanced, fully resected renal cancers should be counselled about the risks/benefits of adjuvant therapy and encouraged to participate in adjuvant clinical trials, facilitated by medical oncology consultation when needed. (Clinical Principle)

Thermal Ablation (TA)

  1. Clinicians should consider TA as an alternate approach for the management of cT1a solid renal masses <3 cm in size. For patients who elect TA, a percutaneous technique is preferred over a surgical approach whenever feasible to minimize morbidity. (Moderate Recommendation; Evidence Level: Grade C)
  2. Both radiofrequency ablation (RFA) and cryoablation may be offered as options for patients who elect TA. (Conditional Recommendation; Evidence Level: Grade C)
  3. A RMB should be performed prior to (preferred) or at the time of ablation to provide pathologic diagnosis and guide subsequent surveillance. (Expert Opinion)
  4. Counseling about TA should include information regarding an increased likelihood of tumor persistence or local recurrence after primary TA relative to surgical excision, which may be addressed with repeat ablation if further intervention is elected. (Strong Recommendation; Evidence Level: Grade B)

Active Surveillance (AS)

  1. For patients with a solid renal mass <2cm, or those that are complex but predominantly cystic, clinicians may elect AS with potential for delayed intervention for initial management. (Conditional Recommendation; Evidence Level: Grade C)
  2. For patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians should prioritize AS/expectant management when the anticipated risk of intervention or competing risks of death outweigh the potential oncologic benefits of active treatment. In asymptomatic patients, the panel recommends periodic clinical surveillance and/or imaging based on shared decision making. (Clinical Principle)
  3. For patients with a solid or Bosniak 3/4 complex cystic renal mass in whom the risk/benefit analysis for treatment is equivocal and who prefer AS, clinicians should consider RMB (if the mass is solid or has solid components) for further oncologic risk stratification. Repeat cross-sectional imaging should be obtained approximately 3-6 months later to assess for interval growth. Periodic clinical/imaging surveillance can then be based on growth rate and shared decision-making with intervention recommended if substantial interval growth is observed or if other clinical/imaging findings suggest that the risk/benefit analysis is no longer equivocal or favorable for continued AS. (Expert Opinion)
  4. For patients with a solid or Bosniak 3/4 complex cystic renal mass in whom the anticipated oncologic benefits of intervention outweigh the risks of treatment and competing risks of death, clinicians should recommend intervention. AS with potential for delayed intervention may be pursued only if the patient understands and is willing to accept the associated oncologic risks. In this setting, clinicians should encourage RMB (if the mass is predominantly solid) for additional risk stratification. If the patient continues to prefer AS, close clinical and cross-sectional imaging surveillance with periodic reassessment and counseling should be recommended. (Moderate Recommendation; Evidence Level: Grade C)

Follow-Up after Intervention

General Prinicples

  1. Clinicians coordinating follow-up for patients who have undergone intervention for a renal mass should discuss the implications of stage, grade, and histology including the risks of recurrence and possible sequelae of treatment. Patients with pathologically-proven benign renal masses should undergo occasional clinical evaluation and laboratory testing for sequelae of treatment but most do not require routine periodic imaging. (Expert Opinion)
  2. Patients with treated malignant renal masses should undergo periodic medical history, physical examination, laboratory studies, and imaging directed at detecting signs and symptoms of metastatic spread and/or local recurrence as well as evaluation for possible sequelae of treatment. (Clinical Principle)
  3. Patients with treated malignant renal masses should have periodic laboratory testing including serum creatinine, eGFR, and urinalysis. Other laboratory evaluations (e.g., complete blood count, lactate dehydrogenase, liver function tests, alkaline phosphatase and calcium level) may be obtained at the discretion of the clinician or if advanced disease is suspected. (Expert Opinion)
  4. Patients undergoing follow-up for treated renal masses with progressive renal insufficiency or proteinuria should be referred to nephrology. (Expert Opinion)
  5. Patients undergoing follow-up for treated malignant renal masses should only undergo bone scan if one or more of the following is present: clinical symptoms such as bone pain, elevated alkaline phosphatase, or radiographic findings suggestive of a bony neoplasm. (Moderate Recommendation; Evidence Level: Grade C)
  6. Patients undergoing follow-up for treated malignant renal masses with acute neurological signs or symptoms should undergo prompt magnetic resonance imaging (MRI) or computed tomography (CT) scanning of the brain and/or spine. (Strong Recommendation; Evidence Level: Grade A)
  7. For patients undergoing follow-up for treated malignant renal masses, additional site-specific imaging can be ordered as warranted by clinical symptoms suggestive of recurrence or metastatic spread. Positron emission tomography (PET) scan should not be obtained routinely but may be considered selectively. (Moderate Recommendation; Evidence Level: Grade C)
  8. Patients with findings suggestive of metastatic renal malignancy should be evaluated to define the extent of disease and referred to medical oncology. Surgical resection or ablative therapies should be considered in select patients with isolated or oligo-metastatic disease. (Expert Opinion)
  9. Patients with findings suggesting a new renal primary or local recurrence of renal malignancy should undergo metastatic evaluation including chest and abdominal imaging. If the new primary or recurrence is isolated to the ipsilateral kidney and/or retroperitoneum, a urologist should be involved in the decision-making process, and surgical resection or ablative therapies may be considered. (Expert Opinion)

Follow-up after Surgery

  1. 42. Clinicians should classify patients who have been managed with surgery (PN or RN) for a malignant renal mass into one of the following risk groups for follow-up:
    Low Risk (LR):pT1 and Grade 1/2
    Intermediate Risk (IR):pT1 and Grade 3/4, or pT2 any Grade
    High Risk (HR):pT3 any Grade
    Very High Risk (VHR):pT4 or pN1, or sarcomatoid/rhabdoid dedeifferentiation, or macroscopic positive margin

    If final microscopic surgical margins are positive for cancer, the risk category should be considered at least one level higher, and increased clinical vigilance should be exercised. (Expert Opinion)

  2. Patients managed with surgery (PN or RN) for a renal malignancy should undergo abdominal imaging according to Table 1, with CT or MRI pre- and post-intravenous contrast preferred. (Moderate Recommendation; Evidence Strength: Grade C). After 2 years, abdominal ultrasound (US) alternating with cross-sectional imaging may be considered in the LR and IR groups at physician discretion. After 5 years, informed/shared decision-making should dictate further abdominal imaging. (Expert Opinion)
  3. Patients managed with surgery (PN or RN) for a renal malignancy should undergo chest imaging (chest x-ray [CXR] for LR and IR; CT chest preferred for HR and VHR) according to Table 1. (Moderate Recommendation; Evidence Strength: Grade C). After 5 years, informed/shared decision-making discussion should dictate further chest imaging and CXR may be utilized instead of chest CT for HR and VHR (Expert Opinion)
Table 1: Recommended follow-up schedule after surgery for renal cancer (in months)*
Risk3691218243036486072-8496-120
LRXXXXXX
IRXXXXXXXX
HRXXXXXXXXXX
VHRXXXXXXXXXXXX
*Follow-up timeline is approximate and allows flexibility to accommodate reasonable patient, caregiver, and institutional needs. Each follow-up visit should include relevant history, physical examination, laboratory testing, and abdominal and chest imaging. Overall, 30% of renal cancer recurrences after surgery are diagnosed beyond 60 months.1 Informed/shared decision-making should guide surveillance decisions beyond 60 months.

Follow-up after TA

  1. Patients undergoing ablative procedures with biopsy that confirmed malignancy or was non-diagnostic should undergo pre- and post-contrast cross-sectional abdominal imaging within 6 months (if not contraindicated). Subsequent follow-up should be according to the recommendations for the IR postoperative protocol (Table 1). (Expert Opinion)

Introduction

Purpose

This AUA Guidelines focuses on the evaluation and management of clinically localized sporadic renal masses suspicious for RCC in adults, including solid enhancing renal tumors and Bosniak 3 and 4 complex cystic renal masses. Some patients with clinically localized renal masses may present with findings suggesting aggressive tumor biology or may be upstaged on exploration or final pathology. Management considerations pertinent to the urologist in such patients will also be discussed. The follow-up of renal cancer patients after intervention is also addressed including recommendations for periodic clinical follow-up and abdominal and chest imaging. Practice patterns regarding such tumors vary considerably and the literature regarding evaluation, management, and surveillance has been rapidly evolving. Notable examples include controversies about the role of RMB and concerns about overutilization of RN.

Methodolgy

Systematic Review RM

The systematic review utilized in the creation of this guideline was completed in part through the AHRQ and through additional supplementation that further addressed additional key questions and more recently published literature. A research librarian experienced in conducting literature searches for comparative effectiveness reviews searched in MEDLINE®, Embase ®, the Cochrane Library, the Database of Abstracts of Reviews of Effects, the Health Technology Assessment Database, and the UK National Health Service Economic Evaluation database to capture both published and gray literature published from January 1, 1997 through May 1, 2015. A supplemental search was conducted adding additional literature published through August 2015, and a final update search was conducted through July 2016.

Systematic Review FRC

A systematic review was conducted to identify published articles relevant to key questions specified by the Panel related to kidney neoplasms and their follow-up (imaging, renal function, markers, biopsy, and prognosis). This search covered articles in English published between January 1999 and 2011. An updated query was later conducted to include studies published through August 2012. Study designs consisting of clinical trials (randomized or not), observational studies (cohort, case-control, case series) and systematic reviews were included. All other study types were excluded. Studies with full-text publication available were included, but studies in abstract form only were excluded.

Combination of Guidelines

In January of 2021, the Renal Mass and Localized Renal Cancer guideline underwent an additional amendment based on current literature. The updated literature search retrieved additional studies published between July 2016 to October 2020 using the same search strategy from the Renal Mass and Localized Renal Cancer guideline. Following study selection using the original PICO criteria, as reporting data relevant to the management and treatment of Renal Mass. In addition, the Follow-Up for Clinically Localized Renal Neoplasms guideline published in 2013 was merged with the Renal Mass and Localized Renal Cancer guideline. Although the systematic search for follow-up interventions was not updated to 2020, the panel members conducted a comprehensive review of all evidence published since the original guideline. The language of many statements has been refined for clarity. For all evidence-based statements, supporting studies were identified only in the original systematic review and the evidence strength was not altered.

Assessment of Risk-of-Bias of Individual Studies

Citations identified by the systematic search were screened independently by two reviewers using predefined PICO criteria. One reviewer completed data abstraction and a second reviewer checked abstraction for accuracy. Two reviewers independently assessed risk of bias for individual studies. The Cochrane Collaboration’s tool was used for assessing the risk of bias of randomized controlled trials (RCTs).2 For nonrandomized studies of treatment interventions, the reviewers used the Risk of Bias in Non-Randomized Studies – of Intervention (ROBINS-I). For diagnostic studies, we used the quality assessment tool for diagnostic accuracy studies (QUADAS -2).3,4 Differences between reviewers were resolved through consensus.

Determination of Evidence Strength

The categorization of evidence strength is conceptually distinct from the quality of individual studies. Evidence strength refers to the body of evidence available for a particular question and includes not only individual study quality but consideration of study design, consistency of findings across studies, adequacy of sample sizes, and generalizability of samples, settings, and treatments for the purposes of the guideline. The AUA categorizes body of evidence strength as Grade A (well-conducted and highly-generalizable RCTs or exceptionally strong observational studies with consistent findings), Grade B (RCTs with some weaknesses of procedure or generalizability or moderately strong observational studies with consistent findings), or Grade C (RCTs with serious deficiencies of procedure or generalizability or extremely small sample sizes or observational studies that are inconsistent, have small sample sizes, or have other problems that potentially confound interpretation of data). By definition, Grade A evidence is evidence about which the Panel has a high level of certainty, Grade B evidence is evidence about which the Panel has a moderate level of certainty, and Grade C evidence is evidence about which the Panel has a low level of certainty.5

AUA Nomenclature: Linking Statement Type to Evidence Strength

The AUA nomenclature system explicitly links statement type to body of evidence strength, level of certainty, magnitude of benefit or risk/burdens, and the Panel’s judgment regarding the balance between benefits and risks/burdens (Table 2). Strong Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is substantial. Moderate Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is moderate. Conditional Recommendations are non-directive statements used when the evidence indicates that there is no apparent net benefit or harm or when the balance between benefits and risks/burden is unclear. All three statement types may be supported by any body of evidence strength grade. Body of evidence strength Grade A in support of a Strong or Moderate Recommendation indicates that the statement can be applied to most patients in most circumstances and that future research is unlikely to change confidence. Body of evidence strength Grade B in support of a Strong or Moderate Recommendation indicates that the statement can be applied to most patients in most circumstances but that better evidence could change confidence. Body of evidence strength Grade C in support of a Strong or Moderate Recommendation indicates that the statement can be applied to most patients in most circumstances but that better evidence is likely to change confidence. Body of evidence strength Grade C is only rarely used in support of a Strong Recommendation. Conditional Recommendations can also be supported by any evidence strength. When body of evidence strength is Grade A, the statement indicates that benefits and risks/burdens appear balanced, the best action depends on patient circumstances, and future research is unlikely to change confidence. When body of evidence strength Grade B is used, benefits and risks/burdens appear balanced, the best action also depends on individual patient circumstances and better evidence could change confidence. When body of evidence strength Grade C is used, there is uncertainty regarding the balance between benefits and risks/burdens, alternative strategies may be equally reasonable, and better evidence is likely to change confidence.

Where gaps in the evidence existed, the Panel provides guidance in the form of Clinical Principles or Expert Opinion with consensus achieved using a modified Delphi technique if differences of opinion emerged.6 A Clinical Principle is a statement about a component of clinical care that is widely agreed upon by urologists or other clinicians for which there may or may not be evidence in the medical literature. Expert Opinion refers to a statement, achieved by consensus of the Panel, that is based on members' clinical training, experience, knowledge, and judgment for which there is no evidence.

Table 2: AUA Nomenclature Linking Statement Type to Level of Certainty, Magnitude, of Benefits or Risk/Burden, and Body of Evidence Strength
Evidence Strength A
(High Certainty)
Evidence Strength B
(Moderate Certainty)
Evidence Strength C
(Low Certainty)

Strong Recommendation

(Net benefit or harm substantial)

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) is substantial

Applies to most patients in most circumstances and future research is unlikely to change confidence

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) is substantial

Applies to most patients in most circumstances and future research could change confidence

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) appears substantial

Applies to most patients in most circumstances and future research is likely to change confidence

(rarely used to support a Strong Recommendation)

Moderate Recommendation

(Net benefit or harm moderate)

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) is moderate

Applies to most patients in most circumstances and future research is unlikely to change confidence

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) is moderate

Applies to most patients in most circumstances and future research could change confidence

Benefits > Risks/Burdens (or vice versa)

Net benefit (or net harm) appears moderate

Applies to most patients in most circumstances and future research is likely to change confidence

Conditional Recommendation

(No apparent net benefit or harm)

Benefits = Risks/Burdens (or vice versa)

Best action depends on individual patient circumstances

Future research unlikely to change confidence

Benefits = Risks/Burdens (or vice versa)

Best action appears to depend on individual patient circumstances

Better evidence could change confidence

Balance between Benefits and Risks/Burdens unclear

Alternative strategies may be equally reasonable

Better evidence likely to change confidence

Clinical PrincipleA statement about a component of clinical care that is widely agreed upon by urologists or other clinicians for which there may or may not be evidence in the medical literature
Expert OpinionA statement, achieved by consensus of the Panel, that is based on members' clinical training, experience, knowledge, and judgment for which there may or may not be evidence in the medical literature

Process

The Renal Mass and Localized Renal Cancer Panel was created in 2014 by the American Urological Association Education and Research, Inc. (AUA). The Practice Guidelines Committee (PGC) of the AUA selected the Panel Chair who in turn appointed the Vice Chair. In a collaborative process, additional Panel members, including additional members of the College of American Pathologists (CAP), Society of Urologic Oncology (SUO), American College of Radiology (ACR), American Society of Nephrology (ASN), Endourological Society, and Society of Interventional Radiology (SIR) with specific expertise in this area, were then nominated and approved by the PGC. The AUA conducted a thorough peer review process. The draft guidelines document was distributed to 124 peer reviewers, 54 of which submitted comments. The Panel reviewed and discussed all submitted comments and revised the draft as needed. Once finalized, the guideline was submitted for approval to the PGC and Science and Quality Council. Then it was submitted to the AUA, CAP, SUO, ACR, ASN, Endourological Society, and SIR Board of Directors for final approval. Panel members received no remuneration for their work.

A Renal Mass amendment panel consisting of five members was created in August 2020 to conduct an update to the Renal Mass and Localized Renal Cancer guideline. They were also tasked with integrating the Follow-Up for Clinically Localized Renal Neoplasms guideline from 2013 into the Renal Mass and Localized Renal Cancer guideline from 2017 to create one cohesive document on management and follow-up on renal mass. The panel consisted of members from both the Follow-Up for Clinically Localized Renal Neoplasms guideline and the Renal Mass and Localized Renal Cancer guideline with an additional new member who has not previously served on one of these panels. The AUA conducted a thorough peer review process. The draft guidelines document was distributed to 75 peer reviewers, 21 of which submitted comments. The Panel reviewed and discussed all submitted comments and revised the draft as needed. Once finalized, the guideline was submitted for approval to the PGC and Science and Quality Council and the AUA Board of Directors for final approval. Panel members received no remuneration for their work.

Background

Renal masses are a biologically heterogeneous group of tumors ranging from benign masses to cancers that can be indolent or aggressive.7,8 The true incidence of renal masses (including benign masses) is unknown. However, benign masses comprise approximately 15-20 percent of surgically resected tumors < 4 cm and allow estimations of benign incidence based on kidney cancer statistics.7,9,10 The vast majority (greater than 90%) of kidney cancers in the United States are renal cortical tumors known as RCC.

Epidemiology: United States

It is estimated there will be over 73,000 new cases of kidney cancer in the United States in 2020.11,12 The incidence of kidney cancer has been increasing steadily since the 1970’s in part due to more prevalent use of axial imaging (CT and MRI).13 In the United States, over the past decade, the incidence of kidney cancer continues to increase but at a much smaller increment, approximately 1% per year. The greatest increase in incidence has been in small, clinically localized renal masses which now represent at least 40 percent of incident tumors.14,15

The overall survival rate for all stages of renal cancer is approximately 74%, leaving an estimated 400,000 kidney cancer survivors in the United States as of 2013.11 However, approximately 14,800 men and women will die of kidney cancer in 2020.10 The mortality from kidney cancer has been steadily decreasing, approximately 1% per year, since 2004.16,17 Reasons for this decrease are multifactorial.

Kidney cancer is more common in men than women, and more common in African Americans, American Indian and Alaska Native populations than Caucasians.18 The median age at diagnosis is 64 years old, although kidney cancer can present at any age.19

Epidemiology: Global and International Considerations

Over 300,000 men and women are diagnosed with kidney cancer around the world each year and approximately 150,000 patients will die of disease.20 The incidence of kidney cancer varies dramatically around the world with the developed countries having the highest rates.21 Incidence rates have increased in both sexes and are most notable in the elderly population (greater than 75 years of age). Mortality rates have been stable in most countries but have been decreasing by 1 to 3 percent in Western and Northern Europe, the United States, and Australia. The improved mortality globally and in the US is attributed to decreased smoking rates, improved therapies, and access to medical care. The decrease in mortality has been faster in women than in men and overall mortality rates remain higher in men than women.

Etiology

There are a number of established and putative risk factors for RCC. Smoking is a well-established risk factor, accounting for 20 percent of incident cases and increasing the risk of RCC by 50 percent in men and 20 percent in women.22,23 Obesity is associated with 30% of incident cases of RCC and each 5 kg/m2 increase in body mass index increases the risk of RCC by 24 percent in men and 34 percent in women.23-25 Interestingly, an “obesity paradox” exists in kidney cancer – where obese patients are more likely to develop RCC, but these tumors are more likely to be low-grade, early stage tumors.25-27 Hypertension is also associated with increased risk of RCC.23,28,29 The role of CKD as a risk factor is controversial; however, patients on maintenance dialysis are also reported to have an increased risk of RCC.30 The data regarding environmental and occupational exposures are inconsistent with the exception of chlorinated solvents.23,31

Moderate alcohol intake,32,33 consumption of fruits and (cruciferous) vegetables,2,3,34,35 and a diet rich in fatty fish are believed to reduce the risk of RCC. Other studies suggest that non-steroidal anti-inflammatory agents and dietary factors do not play a role in the etiology of RCC.5,23,37

Hereditary and Familial RCC

Family history is associated with an increased risk of RCC and a number of familial RCC syndromes are now well-established, accounting for approximately 4-6% of cases of RCC overall.38 These syndromes include von Hippel-Lindau (VHL), hereditary papillary renal carcinoma (HPRC), Birt Hogg-Dubé (BHD), hereditary leiomyomatosis RCC (HLRCC), succinate dehydrogenase deficiency RCC, tuberous sclerosis, BAP-1 tumor predisposition syndrome, and PTEN hamartoma tumor syndrome (Cowden syndrome). Most of these syndromes have associated tumors or benign findings in other organ systems. RCC in these syndromes tends to be earlier in onset and multifocal and management should prioritize nephron-sparing approaches, including tumor enucleation when feasible to optimize preservation of parenchymal mass. For most of these syndromes, tumors can be observed if less than 3 cm as the risk of metastases remains low in this setting.39 HLRCC and succinate dehydrogenase deficiency RCC are the exception as tumors in these syndromes are often very aggressive and a proactive approach to evaluation and management should be pursued. Genetic counseling should also be strongly recommended for patients suspected of having familial RCC, as it may allow for more intensive evaluation of the patient for RCC and associated manifestations and identification of blood relatives that may be at syndromic risk.

Major Pathological Subtypes

Renal tumors are classified based on cell of origin and morphologic appearance with renal adenocarcinoma being the most common malignant tumor. Major sub-classifications of RCC include clear cell, papillary, chromophobe, collecting duct and unclassified RCC.40 A number of uncommon or rare subtypes exist including but not limited to acquired cystic disease-associated RCC, clear cell (tubulo) papillary, and renal medullary carcinoma, which is an aggressive variant typically seen in patients with sickle cell trait. The most common benign tumors of the kidney include oncocytoma and angiomyolipoma (AML). An abbreviated version of the 2016 World Health Organization classification of renal neoplasms is detailed in Table 3.41

Table 3: Modified 2016 World Health Organization classification of renal neoplasms with focus on adult neoplasms.41
Renal cell tumors
Clear cell RCC
Multilocular cystic renal neoplasm of low malignant potential
Papillary RCC
HLRCC
Chromophobe RCC
Collecting duct carcinoma
Renal medullary carcinoma
MiT Family translocation carcinomas
Succinate dehydrogenase (SDH) deficient RCC
Mucinous tubular and spindle cell carcinoma
Tubulocystic RCC
Acquired cystic disease associated RCC
Clear cell papillary RCC
RCC, unclassified
Benign renal tumors
Papillary adenoma
Oncocytoma
AML
Metanephric adenoma and other metanephric tumors
Adult cystic nephroma
Mixed epithelial stromal tumors
Juxtaglomerular cell tumor
Mescenchymal tumors
Leiomyosarcoma (including renal vein) and other sarcomas
Leiomyoma and other benign mesenchymal tumors
Others
Adults Wilms tumor
Primitive neuroectodermal tumor
Metastatic tumors, lymphoma, leukemia

Presentation and Diagnosis

Presentation

The “classic triad” of symptoms associated with a malignant renal mass include hematuria, flank pain and abdominal mass. Symptoms associated with RCC are often a result of local tumor growth, hemorrhage, paraneoplastic symptoms, or metastatic disease and are uncommon in patients with clinically localized disease. In fact, less than 5 percent of patients in contemporary series present with these symptoms and greater than 50 percent of renal masses are diagnosed incidentally during an evaluation for unrelated signs or symptoms.42,43

Diagnosis

Physical examination has a limited role in the diagnosis of clinically localized disease. However, physical examination may have value in distinguishing the signs and symptoms of advanced disease. For instance, paraneoplastic syndromes (i.e. hypertension, polycythemia, hypercalcemia) are present in approximately 10-20 percent of patients with metastatic RCC.6,7,44 Importantly, physical examination of patients with localized disease may occasionally reveal unsuspected adenopathy, varicocele or medical conditions that influence management decisions including body habitus, prior abdominal scars, stigmata of CKD, etc. In addition, careful physical examination may also reveal findings suggestive of familial disease, such as dermatologic lesions.

Laboratory Evaluation

There are no biomarkers or routine laboratory tests used to diagnose renal malignancies. As such, laboratory tests are useful in the assessment of renal function (GFR) and for completeness of metastatic evaluation. Routine laboratory tests for renal mass evaluation include complete metabolic panel, complete blood count, and urinalysis.

Imaging Techniques

Pre and post contrast-enhanced axial imaging, either CT or MRI, is the ideal imaging technique for the diagnosis and staging of clinically localized renal masses. Masses initially diagnosed by US or intravenous pyelography should be confirmed with pre/post contrast-enhanced imaging. Depending on tumor size, 20 to 30 percent of clinically localized renal masses may be benign.7,10 Patient and tumor characteristics can indicate populations more or less likely to harbor benign or malignant disease. For instance, women with smaller tumors have a higher likelihood of having benign tumors.9,45,46 However, with the exception of fat-containing AML, none of the current imaging modalities can reliably distinguish between benign and malignant tumors or between indolent and aggressive tumor biology.

Contrast-enhanced abdominal imaging (CT or MRI) best characterizes the mass, provides information regarding renal morphology (of the affected and unaffected kidney), assesses extrarenal tumor spread (venous invasion or regional lymphadenopathy) and evaluates the adrenal glands and other abdominal organs for visceral metastases. Based on the most recent consensus statement from the ACR and the National Kidney Foundation, patients with acute kidney injury or CKD and GFR less than 30 mL/min/1.73m2 who are not undergoing renal replacement therapy should receive intravenous normal saline prophylaxis prior to receiving iodinated contrast media.47 Patients with GFR of 30-44 mL/min/1.73m2 may be considered for intravenous fluid prophylaxis per individual physician discretion based on the patient’s risk factor for renal injury. However, MRI with second generation gadolinium-based intravenous contrast is now a safer option in many patients with severe CKD, as outlined below.

The association of gadolinium-based MRI contrast agents with the development of nephrogenic systemic fibrosis – a devastating and potentially fatal condition has been a concern for many years. More recently, however, with newer group II and III gadolinium-based contrast media the risk is felt to be lower than previously perceived. The most recent consensus statement from the ACR and the National Kidney Foundation suggests that such agents can be given to patients with a GFR under 30 mL/min/1.73m2.48 A recent systematic review of the risks of NSF in patients with CKD 4 and 5 noted that the risks of NSF using group II gadolinium-based agents was less than 0.07%. Current ACR guidelines on the use of contrast media state that patients need not be screened for renal function prior to receiving group II gadolinium-based agents. Non-contrast CT, MRI (with diffusion weighted images) and US (with Doppler and with or without microbubbles) can also be used to characterize renal masses in patients who cannot receive conventional intravenous contrast.47

In general, solid renal masses that enhance greater than 15-20 HU with intravenous contrast and do not exhibit fat density should be considered suspicious for RCC. Approximately 5-10% of AML’s are fat poor and difficult to identify on imaging. Fat poor AML’s often demonstrate suggestive features such as high attenuation on unenhanced CT, homogeneous enhancement on CT, or hypointensity on T2-weighted MR, but the diagnosis remains difficult. Complex cystic renal masses that have thickened irregular walls or septa in which measurable enhancement is present are classified as Bosniak 3. Approximately 50% of such lesions prove to be malignant on final pathology. Bosniak 4 complex cystic lesions are very suspicious for malignancy as they contain enhancing nodular soft tissue components and about 75-90% of such lesions prove to be RCC on final pathology. This guideline focuses primarily on the evaluation and management of clinically localized sporadic renal masses suspicious for RCC in adults, including solid enhancing renal tumors and Bosniak 3 and 4 cystic renal masses.

In patients with RCC or suspicion of RCC, complete staging is typically finalized with chest radiography (CXR) or chest CT. Chest CT scan should be obtained selectively, primarily for patients with pulmonary symptoms or abnormal CXR, or for patients with high-risk disease.49,50 Bone scans should be reserved primarily for patients with bone pain or elevated alkaline phosphatase and brain imaging for those with neurologic symptoms.51-53 PET scan has a very limited role in the routine evaluation or staging of RCC.

Renal Mass Biopsy

RMB currently has an adjunctive role in the diagnosis and risk stratification of patients with renal masses suspicious for renal cancer. Biopsy, or FNA, was traditionally reserved for patients suspected of having metastasis of another primary to the kidney, abscess, or lymphoma, or when needed to establish a pathologic diagnosis of RCC in occasional patients presenting with disseminated metastases or unresectable primary tumors. The role of RMB for clinically localized RCC has evolved considerably over the past few decades with substantial variance in practice patterns.

Tumor Characteristics

Staging

Kidney cancer is staged both clinically and pathologically using the staging system outlined by the American Joint Committee on Cancer (AJCC), also known as the tumor node metastases (TNM) classification.54 The AJCC TNM Staging System for Kidney Cancer is detailed in Table 4. Stage I and II tumors include cancers of any size that are confined to the kidney. This guideline statement identifies patients with renal masses suspicious for clinical stage I and II RCC, recognizing that a certain number of patients will be upstaged. Stage III tumors are either locally invasive (T3) or have involved lymph nodes (N1). Stage IV tumors have spread beyond the kidney into adjacent organs by direct invasion (T4) or distant metastases (M1). Prognosis is best predicted by stage with cancer-specific survival rates that approximate 85-90% for clinically localized (Stage I and II) RCC.

Table 4: The AJCC TNM Staging System for Kidney Cancer. 41

Primary Tumor (T), Regional Lymph Nodes (N) and Distant Metastases (M) are detailed in Table 4A; The Anatomic Stage/Prognostic Groups are detailed in Table 4B.

Table 4A.
Primary Tumor (T)
TXPrimary tumor cannot be assessed
T0No evidence of primary tumor.
T1Tumor ≤7 cm in greatest dimension, limited to the kidney.
T1aTumor ≤4 cm in greatest dimension, limited to the kidney.
T1bTumor >4 cm but not >7 cm in greatest dimension, limited to the kidney.
T2Tumor >7 cm in greatest dimension, limited to the kidney.
T2aTumor >7 cm but ≤10 cm in greatest dimension, limited to the kidney.
T2bTumor >10 cm, limited to the kidney.
T3Tumor extends into major veins or perinephric tissues but not into the ipsilateral adrenal gland and not beyond Gerota’s fascia.
T3aTumor extends into the renal vein or its segmental branches, or invades the pelvicaliceal system, or invades perirenal and/or renal sinus fat but not beyond Gerota’s fascia.
T3bTumor grossly extends into the vena cava below the diaphragm.
T3cTumor grossly extends into the vena cava above the diaphragm or invades the wall of the vena cava.
T4Tumor invades beyond Gerota’s fascia (including contiguous extension into the ipsilateral adrenal gland).
Regional Lymph Nodes (N)
NXRegional lymph nodes cannot be assessed.
N0No regional lymph node metastasis.
N1Metastases in regional lymph node(s).
Distant Metastasis (M)
M0No distant metastasis.
M1Distant metastasis.
Table 4b.
StageTNM
IT1N0M0
IIT2N0M0
IIIT1 or T2N1M0
T3N0 or N1M0
IVT4Any NM)
Any TAny NM1

Grading

Historically, a number of grading systems existed and evolved to describe tumor differentiation, cytologic aggressiveness, and prognosis of RCC based on nuclear size and irregularity. In 1982, the Fuhrman Grading system was described and became the most widely used grading system for RCC.55 In 2012, the International Society of Urological Pathology (ISUP) Grading System for RCC was proposed and was updated in 2016.41,56 The ISUP Grading System incorporates aspects of the Fuhrman Grading system but includes more objective criteria for nuclear characteristics. In addition, sarcomatoid and rhabdoid tumors, tumors with giant cells, and tumors with extreme nuclear pleomorphism are included within grade 4 tumors. Chromophobe RCC is no longer graded in the ISUP system. In general, higher grade is associated with larger tumor size and more aggressive tumors.57,58

Other Prognostic indicators and Nomograms

Other factors for prognostic consideration include tumor size, necrosis, sarcomatoid features, collecting system invasion, patient symptoms, signs of paraneoplastic syndromes, and performance status. Tumor size is important for risk stratification regarding the likelihood of malignancy and more aggressive pathology.7,10,46 Various algorithms including the UCLA Integrated Staging System (UISS),59,60 Stage, Size, Grade and Necrosis (SSIGN) score,61-63 and other nomograms8,9,64 incorporate a variety of pathological and patient characteristics in an effort to improve prognostication.

Other Clinical and Biological Indicators

A number of molecular studies and markers have been proposed for diagnostic and prognostic purposes in RCC. The AHRQ Systematic Review identified a number of biomarkers and laboratory tests that may have diagnostic or prognostic utility in the renal cancer literature.65 However, these studies were often univariable in design and therefore excluded from analysis due to a failure to include clinical variables or suboptimal methodology to validate the ultimate value of the tests. Therefore, the AHRQ report identified clinical and biological indicators as a major research gap in the renal cancer literature.66

Of note, urine aquaporin-1 and perilipin-2 were identified as emerging biomarkers with potential for the diagnosis of RCC.10,11,67,68 Carbonic anhydrase-9 (CAIX) expression is governed by the transcription factor hypoxia-inducible factor-1α (HIF-1α), a well-known component of the VHL pathway of clear cell RCC.69 While CAIX expression on primary tumors is a prognostic factor, especially in patients with metastatic RCC, high and homogenous levels of CAIX expression prevent risk stratification and clinical utility beyond the established clinical predictors of aggressive, clear cell RCC.70 Serum tests including C-reactive protein and platelet count may have prognostic roles, but further investigation is needed. New imaging modalities, including molecular imaging techniques using CAIX71-73 or 99m technetium-sestamibi74 single photon emission CT, may help to better differentiate between malignant and benign pathology. However, most markers and imaging modalities in this domain are best characterized as investigational.

Overview of Treatment Alternatives

A number of strategies exist for the management of sporadic renal masses suspicious for clinically localized renal cancer. Four strategies are considered standards of care and include AS, RN, PN, and TA.

Active Surveillance (AS)

A growing body of literature exists regarding AS for patients with clinically localized small renal masses (cT1a, ≤4cm). A number of retrospective studies and meta-analyses evaluate the safety of AS and quote the risk of metastatic progression while on AS to be less than 2 percent in well selected patients over the initial 3 years of AS.75-77 Two large prospective AS programs have been initiated that follow patients with serial imaging, and both report slow growth rates and extremely low rates of metastatic progression, albeit with relatively short follow-up.78-80 Both programs screen patients with an initial metastatic evaluation including serum laboratory evaluation and chest imaging. Patients are then evaluated every 3-6 months for two years and with extended imaging intervals beyond that. Rates of biopsy are variable with one group utilizing RMB in greater than 50 percent of the cohort and the other using biopsy in less than 10 percent of its patients. Further data with longer follow-up from these cohorts will help to inform the utility of AS in the small renal mass population, and should allow for more intelligent patient selection for AS. Of note, the Delayed Intervention and Surveillance for Small Renal Masses (DISSRM) Registry prospectively catalogues a contemporaneous cohort of patients undergoing AS and primary intervention and will offer data regarding comparative effectiveness.80

Radical Nephrectomy (RN)

RN was the mainstay of therapy for all renal masses for many decades. Historically, RN included the removal of the entire kidney including Gerota's/Zuckerkandel's fascia, regional lymph nodes and the adrenal gland. RN can be performed through an open incision or via minimally-invasive approaches (laparoscopic or robotic). Cancer-specific survival associated with RN is excellent; however, recent controversies regarding RN include its negative impact on renal function and historical overutilization for the management of stage I, especially T1a, tumors.

Partial Nephrectomy (PN)

PN is widely accepted as a nephron-sparing approach to the management of clinically localized RCC. Initially underutilized and predominantly performed in large academic centers,12,13,81 the management of clinically localized renal masses by PN has expanded with implementation of guideline statements and the expansion of robotic technology.14,15,82 PN can be performed through an open incision or via a minimally invasive approach, although the robotic approach has largely supplanted laparoscopic surgery as the preferred minimally invasive approach.83 The benefit of PN lies in the potential to preserve renal function but this is counterbalanced by an increased risk of urologic complications, although most are manageable and typically associated with good outcomes. Recent controversies surround modifiable and non-modifiable factors during surgery to improve renal functional outcomes, including parenchymal volume preservation, warm versus cold ischemia, and duration of ischemia.

Thermal Ablation (TA)

TA techniques were developed in an effort to improve patient procedural tolerance and reduce the potential for complications from PN, while still preserving renal function. A multitude of techniques/technologies have been investigated to ablate renal tumors; however, RFA and cryoablation have been most widely investigated and integrated into clinical practice. While the superiority of RFA or cryoablation remains controversial, it is generally accepted that oncologic outcomes are similar for both approaches.84-86 TA has traditionally been performed through a variety of approaches, including open, laparoscopic, and percutaneous. Concerns with the TA literature included relatively limited follow-up, lack of pre and post treatment biopsy to define malignancy and efficacy, and increased local recurrence rates relative to surgical excision. The latter require a longer period of surveillance (5 years) with cross-sectional imaging to monitor for late local recurrences.

Investigational Modalities

Other technologies including high-intensity focused US (HIFU), radiosurgery, microwave therapy, pulsed cavitational US, and laser thermal therapy remain investigational at this time.

Follow-up after Intervention

The prognosis of patients treated with surgery or thermal ablation for kidney cancer is primarily determined by tumor stage, with tumor size, grade, histology, with a variety of other contributing factors.61,62,87-93 Several algorithms and prognostic models have been published, yet a recent analysis of outcomes from a phase III randomized adjuvant clinical trial suggested that these models only marginally outperformed stage alone.94

Current surveillance and survivorship strategies for patients with RCC have incorporated clinical history, physical examination, relevant laboratory testing, and abdominal and chest imaging.181,387,95 This allows for assessment of potential complications or sequelae of intervention, functional recovery, and evaluation for common sites of recurrence, including those in the lungs, liver, adrenal glands, and other retroperitoneal sites. Cross-sectional imaging is generally preferred, particularly for high-risk patients, while abdominal ultrasound or chest radiography can be considered in lower-risk patients or as a potential alternative during long-term surveillance. Approximately thirty-percent of recurrences have been diagnosed after 5 years in some series, emphasizing the need to consider longer follow-up than advocated in most current surveillance protocols.1 Bone metastasis are only rarely identified during surveillance in the absence of bone pain, an elevated alkaline phosphatase, or radiographic findings suggesting a bony neoplasm, and bone scan can generally be reserved for these indications.96-99 Patients with acute neurological signs/symptoms should undergo prompt cross-sectional imaging of the brain and/or spine,370-372 but beyond this there is no role for routine neurologic imaging in surveillance of patients with localized renal cancer. Additional site-specific imaging should be ordered as warranted by clinical signs/symptoms suggestive of recurrence or metastatic spread. Current data do not support the use of PET scan in the routine surveillance of patients with renal cancer, and this test should only be considered selectively, such as for trouble-shooting when other tests are concerning but inconclusive.373

There are no prospective data to compare currently available surveillance strategies, resulting in substantial variability in the approach, modality, frequency, and duration of follow-up after intervention. The premise of early detection of tumor recurrence after primary intervention is that this approach will result in patient cure, improved survival, or appropriate palliation. In addition, surveillance allows the urologist to provide a measure of reassurance to the patient who is worried about cancer recurrence. Surveillance also offers the opportunity to monitor treatment effects and address survivorship issues that might arise. Taking all of these considerations into account, the Panel updated the follow-up strategies after intervention to strike a useful and measured balance.

Evaluation and Diagnosis

Evaluation

Guideline Statement 1

1. In patients with a solid or complex cystic renal mass, clinicians should obtain high quality, multiphase, cross-sectional abdominal imaging to optimally characterize and clinically stage the renal mass. Characterization of the renal mass should include assessment of tumor complexity, degree of contrast enhancement (where applicable), and presence or absence of fat. (Clinical Principle)

Discussion


Guideline Statement 2

2. In patients with suspected renal malignancy, clinicians should obtain a comprehensive metabolic panel, complete blood count, and urinalysis. Metastatic evaluation should include chest imaging to evaluate for possible thoracic metastases. (Clinical Principle)

Discussion


Guideline Statement 3

3. For patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians should assign chronic kidney disease (CKD) stage based on glomerular filtration rate (GFR) and degree of proteinuria. (Expert Opinion)

Discussion


Counseling

Guideline Statement 4

In patients with a solid or Bosniak 3/4 complex cystic renal mass, a urologist should lead the counseling process and should consider all management strategies. A multidisciplinary team should be included when necessary. (Expert Opinion)

Discussion


Guideline Statement 5

Clinicians should provide counseling that includes current perspectives about tumor biology and a patient-specific risk assessment inclusive of sex, tumor size/complexity, histology (when obtained), and imaging characteristics. For cT1a tumors, the low oncologic risk of many small renal masses should be reviewed. (Clinical Principle)

Discussion


Guideline Statement 6

During counseling of patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians must review the most common and serious urologic and non-urologic morbidities of each treatment pathway and the importance of patient age, comorbidities/frailty, and life expectancy. (Clinical Principle)

Discussion


Guideline Statement 7

Clinicians should review the importance of renal functional recovery related to renal mass management, including the risks of progressive CKD, potential short- or long-term need for renal replacement therapy, and long-term overall survival considerations. (Clinical Principle)

Discussion


Guideline Statement 8

Clinicians should consider referral to nephrology in patients with a high risk of CKD progression, including those with estimated glomerular filtration rate (eGFR) less than 45 mL/min/1.73m2, confirmed proteinuria, diabetics with preexisting CKD, or whenever eGFR is expected to be less than 30 mL/min/1.73m2 after intervention. (Expert Opinion)

Discussion


Guideline Statement 9

Clinicians should recommend genetic counseling for any of the following: all patients ≤ 46 years of age with renal malignancy, those with multifocal or bilateral renal masses, or whenever 1) the personal or family history suggests a familial renal neoplastic syndrome; 2) there is a first-or second-degree relative with a history of renal malignancy or a known clinical or genetic diagnosis of a familial renal neoplastic syndrome (even if kidney cancer has not been observed); or 3) the patient’s pathology demonstrates histologic findings suggestive of such a syndrome. (Expert Opinion)

Discussion


Renal Mass Biopsy (RMB)

Guideline Statement 10

When considering the utility of RMB, patients should be counseled regarding rationale, positive and negative predictive values, potential risks and non-diagnostic rates of RMB. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 11

Clinicians should consider RMB when a mass is suspected to be hematologic, metastatic, inflammatory, or infectious. (Clinical Principle)

Discussion


Guideline Statement 12

In the setting of a solid renal mass, RMB should be obtained on a utility-based approach whenever it may influence management. RMB is not required for 1) young or healthy patients who are unwilling to accept the uncertainties associated with RMB; or 2) older or frail patients who will be managed conservatively independent of RMB findings. (Expert Opinion)

Discussion


Guideline Statement 13

For patients with a solid renal mass who elect RMB, multiple core biopsies should be performed and are preferred over fine needle aspiration (FNA). (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Management

Partial nephrectomy (PN) and nephron-sparing approaches

Guideline Statement 14

Clinicians should prioritize PN for the management of the cT1a renal mass when intervention is indicated. In this setting, PN minimizes the risk of CKD or CKD progression and is associated with favorable oncologic outcomes, including excellent local control. (Moderate Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 15

Clinicians should prioritize nephron-sparing approaches for patients with solid or Bosniak 3/4 complex cystic renal masses and an anatomic or functionally solitary kidney, bilateral tumors, known familial RCC, preexisting CKD, or proteinuria. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 16

Nephron-sparing approaches should be considered for patients with solid or Bosniak 3/4 complex cystic renal masses who are young, have multifocal masses, or comorbidities that are likely to impact renal function in the future, including but not limited to moderate to severe hypertension, diabetes mellitus, recurrent urolithiasis, or morbid obesity. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 17

In patients who elect PN, clinicians should prioritize preservation of renal function by optimizing nephron mass preservation and avoiding prolonged warm ischemia. (Expert Opinion)

Discussion


Guideline Statement 18

For patients undergoing PN, clinicians should prioritize negative surgical margins. The extent of normal parenchyma removed should be determined by surgeon discretion taking into account the clinical situation and tumor characteristics, including growth pattern, and interface with normal tissue. Tumor enucleation should be considered in patients with familial RCC, multifocal disease, or severe CKD to optimize parenchymal mass preservation. (Expert Opinion)

Discussion


Radical Nephrectomy (RN)

Guideline Statement 19

Clinicians should consider RN for patients with a solid or Bosniak 3/4 complex cystic renal mass whenever increased oncologic potential is suggested by tumor size, RMB (if obtained), and/or imaging. (Moderate Recommendation; Evidence Level: Grade B) In this setting, RN is preferred if all of the following criteria are met: 1) high tumor complexity and PN would be challenging even in experienced hands; 2) no preexisting CKD or proteinuria; and 3) normal contralateral kidney and new baseline eGFR will likely be greater than 45 mL/min/1.73m2 even if RN is performed. If all of these criteria are not met, PN should be considered unless there are overriding concerns about the safety or oncologic efficacy of PN. (Expert Opinion)

Discussion


Surgical Principles

Guideline Statement 20

For patients who are undergoing surgical excision of a renal mass with clinically concerning regional lymphadenopathy, clinicians should perform a lymph node dissection including all clinically positive nodes for staging purposes. (Expert Opinion)

Discussion


Guideline Statement 21

For patients who are undergoing surgical excision of a renal mass, clinicians should perform adrenalectomy if imaging and/or intraoperative findings suggest metastasis or direct invasion of the adrenal gland. (Clinical Principle)

Discussion


Guideline Statement 22

In patients undergoing surgical excision of a renal mass, a minimally invasive approach should be considered when it would not compromise oncologic, functional, and perioperative outcomes. (Expert Opinion)

Discussion


Other Considerations

Guideline Statement 23

Pathologic evaluation of the adjacent renal parenchyma should be performed and recorded after PN or RN to assess for possible intrinsic renal disease, particularly for patients with CKD or risk factors for developing CKD. (Clinical Principle)

Discussion


Guideline Statement 24

Clinicians should consider referral to medical oncology whenever there is concern for potential clinical metastasis or incompletely resected disease (macroscopic positive margin or gross residual disease). Patients with high-risk or locally advanced, fully resected renal cancers should be counselled about the risks/benefits of adjuvant therapy and encouraged to participate in adjuvant clinical trials, facilitated by medical oncology consultation when needed. (Clinical Principle)

Discussion


Thermal Ablation (TA)

Guideline Statement 25

Clinicians should consider TA as an alternate approach for the management of cT1a solid renal masses <3 cm in size. For patients who elect TA, a percutaneous technique is preferred over a surgical approach whenever feasible to minimize morbidity. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 26

Both radiofrequency ablation (RFA) and cryoablation may be offered as options for patients who elect TA. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 27

A RMB should be performed prior to (preferred) or at the time of ablation to provide pathologic diagnosis and guide subsequent surveillance. (Expert Opinion)

Discussion


Guideline Statement 28

Counseling about TA should include information regarding an increased likelihood of tumor persistence or local recurrence after primary TA relative to surgical excision, which may be addressed with repeat ablation if further intervention is elected. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Active Surveillance

Introduction

The decision to embark on a course of AS or expectant management rather than treatment in the setting of a localized renal mass presumed to be a renal cancer requires thoughtful consideration by both the patient and the physician. In making the decision, an objective baseline evaluation of patient, tumor, and treatment-related factors should be undertaken (Figure 7). This should include formal decision-making tools whenever possible leading to a well communicated risk-benefit analysis unique to the individual patient’s circumstances.110,344-347 The shared decision-making process should be consistent with the patient’s inherent preferences and tolerance of uncertainty.348

Figure 7. Algorithm for AS or expectant management of localized renal masses suspicious for malignancy.

High level data regarding the optimal frequency and preferred imaging modalities for renal mass surveillance are lacking. Therefore, at the time of the initial baseline assessment and during subsequent re-assessments, the clinician should estimate how to best achieve the goals of (1) preventing stage progression, (2) maintaining renal function and (3) avoiding the potential risks of treatment when it is unlikely to provide an oncologic or survival benefit. At the onset of AS, the clinician should request and evaluate prior abdominal imaging that may demonstrate the existence of the renal mass at an earlier time point to assess growth rate or changes in clinical stage. Next, patients placed on a program of non-intervention should be considered for either AS or expectant management (observation or watchful waiting) (Figure 7).

AS is most appropriate for patients in whom the anticipated net benefit of AS is modest to significant when compared to treatment. Excluded from this track are patients who are reasonable candidates for intervention if tumor size, infiltrative appearance, interval growth, or RMB suggest the potential for cancer progression, unless they are willing to accept the associated increase in oncologic risk (see statement 31 and 32 below). Patients with no prior imaging should have surveillance imaging initially every 3 to 6 months to assess for interval growth, substantial radiographic changes in the character of the lesion, or the presence of rare occult synchronous metastases in the setting of a small renal mass. The preferred modality is not well established in the literature, but initial imaging should preferably consist of contrast-enhanced cross-sectional imaging. Subsequent imaging may include the same or when appropriate an abdominal US can be substituted. Abdominal US (as opposed to retroperitoneal US), may have the additional benefit of a survey of the intraabdominal organs for progression. Differences in tumor dimension measurements between these different modalities may be significant and should be interpreted with caution when making treatment decisions.80 RMB can be considered for additional risk stratification for patients with solid masses on AS. For those with predominantly cystic lesions, RMB should be avoided.

It is recognized that not all patients on AS will require the same intensity of surveillance as their tumor biology, risk calculations and tradeoffs, and personal objectives may differ. Some patients may therefore require more intensive AS while others require less intensive AS. The decision as to the frequency and imaging modality must therefore be customized and informed by robust communication focusing on goals, risks and triggers for intervention. RMB can be a helpful adjunct to guide these clinical decisions (see statement 10). However, even when RMB suggests the tumor is benign, the predictive value of a core biopsy is imperfect due to tumor heterogeneity and the possibility of collision tumors.192,193 Currently there are insufficient data to recommend that all patients with benign RMBs can be advised that they no longer need follow-up imaging. Judicious surveillance in appropriate patients with benign appearing RMBs remains a prudent strategy.

Expectant management (observation) is appropriate in patients in whom treatment poses an unacceptably high periprocedural or renal functional risk than surveillance. In this setting, the use of abdominal US of the retroperitoneal and intraperitoneal organs can be performed more frequently than formal contrast based cross-sectional imaging to screen for stage progression which may trigger systemic or palliative therapy in the appropriately selected patient.

Regardless of the intensity of surveillance, chest imaging with plain radiography (CXR) is warranted annually or if intervention triggers are encountered or symptoms arise. The intensity of surveillance can be attenuated if the renal mass exhibits slow growth kinetics, is noted to be radiographically stable or if the patient’s medical condition deteriorates. In cases such as this, patients can cross over between AS and expected management (observation) based on changing risk profiles, performance status, absolute tumor size, tumor growth kinetics, stage progression or other recalibration triggers for possible intervention.349,350 While no level 1 data exist that define these triggers precisely, they should generally be based on changes in tumor-based risk (absolute size > 3cm, median growth rate in excess of 5mm/year, or stage migration) or patient-based risks (co-morbidities) with continual objective reassessments to include the use of RMB when appropriate.349,350 Published data demonstrate that in most instances, judicious delayed intervention for localized stage I renal masses remains effective.159,349-354

The key to successful AS of a localized renal mass remains thoughtful and recurrent reassessments and robust communication in partnership with the patient and his/her caregivers. Prospective trials, ideally randomized, of AS versus treatment, with improved reporting and more extended follow-up, should be prioritized to provide higher quality data about oncologic, functional and survival outcomes.

Guideline Statement 29

For patients with a solid renal mass < 2cm, or those that are complex but predominantly cystic, clinicians may elect AS with potential for delayed intervention for initial management. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 30

For patients with a solid or Bosniak 3/4 complex cystic renal mass, clinicians should prioritize AS/expectant management when the anticipated risk of intervention or competing risks of death outweigh the potential oncologic benefits of active treatment. In asymptomatic patients, the panel recommends periodic clinical surveillance and/or imaging based on shared decision making. (Clinical Principle)

Discussion


Guideline Statement 31

For patients with a solid or Bosniak 3/4 complex cystic renal mass in whom the risk/benefit analysis for treatment is equivocal and who prefer AS, clinicians should consider RMB (if the mass is solid or has solid components) for further oncologic risk stratification. Repeat cross-sectional imaging should be obtained approximately 3-6 months later to assess for interval growth. Periodic clinical/imaging surveillance can then be based on growth rate and shared decision-making with intervention recommended if substantial interval growth is observed or if other clinical/imaging findings suggest that the risk/benefit analysis is no longer equivocal or favorable for continued AS. (Expert Opinion)

Discussion


Guideline Statement 32

For patients with a solid or Bosniak 3/4 complex cystic renal mass in whom the anticipated oncologic benefits of intervention outweigh the risks of treatment and competing risks of death, clinicians should recommend intervention. AS with potential for delayed intervention may be pursued only if the patient understands and is willing to accept the associated oncologic risks. In this setting, clinicians should encourage RMB (if the mass is predominantly solid) for additional risk stratification. If the patient continues to prefer AS, close clinical and cross-sectional imaging surveillance with periodic reassessment and counseling should be recommended. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Follow-Up After Intervention

General Principles

Guideline Statement 33

Clinicians coordinating follow-up for patients who have undergone intervention for a renal mass should discuss the implications of stage, grade, and histology including the risks of recurrence and possible sequelae of treatment. Patients with pathologically-proven benign renal masses should undergo occasional clinical evaluation and laboratory testing for sequelae of treatment but most do not require routine periodic imaging. (Expert Opinion)

Discussion


Guideline Statement 34

Patients with treated malignant renal masses should undergo periodic medical history, physical examination, laboratory studies, and imaging directed at detecting signs and symptoms of metastatic spread and/or local recurrence as well as evaluation for possible sequelae of treatment. (Clinical Principle)

Discussion


Guideline Statement 35

Patients with treated malignant renal masses should have periodic laboratory testing including serum creatinine, eGFR, and urinalysis. Other laboratory evaluations (e.g., complete blood count, lactate dehydrogenase, liver function tests, alkaline phosphatase and calcium level) may be obtained at the discretion of the clinician or if advanced disease is suspected. (Expert Opinion)

Discussion


Guideline Statement 36

Patients undergoing follow-up for treated renal masses with progressive renal insufficiency or proteinuria should be referred to nephrology. (Expert Opinion)

Discussion


Guideline Statement 37

Patients undergoing follow-up for treated malignant renal masses should only undergo bone scan if one or more of the following is present: clinical symptoms such as bone pain, elevated alkaline phosphatase, or radiographic findings suggestive of a bony neoplasm. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 38

Patients undergoing follow-up for treated malignant renal masses with acute neurological signs or symptoms should undergo prompt magnetic resonance imaging (MRI) or computed tomography (CT) scanning of the brain and/or spine. (Strong Recommendation; Evidence Level: Grade A)

Discussion


Guideline Statement 39

For patients undergoing follow-up for treated malignant renal masses, additional site-specific imaging can be ordered as warranted by clinical symptoms suggestive of recurrence or metastatic spread. Positron emission tomography (PET) scan should not be obtained routinely but may be considered selectively. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 40

Patients with findings suggestive of metastatic renal malignancy should be evaluated to define the extent of disease and referred to medical oncology. Surgical resection or ablative therapies should be considered in select patients with isolated or oligo-metastatic disease. (Expert Opinion)

Discussion


Guideline Statement 41

Patients with findings suggesting a new renal primary or local recurrence of renal malignancy should undergo metastatic evaluation including chest and abdominal imaging. If the new primary or recurrence is isolated to the ipsilateral kidney and/or retroperitoneum, a urologist should be involved in the decision-making process, and surgical resection or ablative therapies may be considered. (Expert Opinion)

Discussion


Follow-up after Surgery

Guideline Statement 42

Clinicians should classify patients who have been managed with surgery (PN or RN) for a malignant renal mass into one of the following risk groups for follow-up:

Low Risk (LR)pT1 and Grade 1/2
Intermediate Risk (IR)pT1 and Grade 3/4, or pT2 any Grade
High Risk (HR)pT3 any Grade
Very High Risk (VHR)pT4 or pN1, or sarcomatoid/rhabdoid dedifferentiation, or macroscopic positive margin

If final microscopic surgical margins are positive for cancer, the risk category should be considered at least one level higher, and increased clinical vigilance should be exercised. (Expert Opinion)

Discussion


Guideline Statement 43

Patients managed with surgery (PN or RN) for a renal malignancy should undergo abdominal imaging according to Table 1, with CT or MRI pre- and post-intravenous contrast preferred. (Moderate Recommendation; Evidence Strength: Grade C). After 2 years, abdominal ultrasound (US) alternating with cross-sectional imaging may be considered in the LR and IR groups at physician discretion. After 5 years, informed/shared decision-making should dictate further abdominal imaging. (Expert Opinion)

Discussion


Guideline Statement 44

Patients managed with surgery (PN or RN) for a renal malignancy should undergo chest imaging (chest x-ray [CXR] for LR and IR; CT chest preferred for HR and VHR) according to Table 1. (Moderate Recommendation; Evidence Strength: Grade C). After 5 years, informed/shared decision-making discussion should dictate further chest imaging and CXR may be utilized instead of chest CT for HR and VHR (Expert Opinion)

Discussion


Follow-up after TA

Guideline Statement 45

Patients undergoing ablative procedures with biopsy that confirmed malignancy or was non-diagnostic should undergo pre- and post-contrast cross-sectional abdominal imaging within 6 months (if not contraindicated). Subsequent follow-up should be according to the recommendations for the IR postoperative protocol (Table 1). (Expert Opinion)

Discussion


Future Directions

The most promising routes to advance the field in localized renal cancer include (1) clinical trials, (2) collaborative quality initiatives, (3) novel diagnostics/biomarkers, and (4) improved technologies and systemic therapies. Each of these requires an unrelenting commitment to continuous clinical improvement and scientific investigation.

The management of localized renal cancer is an area for which there is a paucity of randomized clinical trials (RCT’s). Improving the strength of evidence will require an increased commitment to clinical trial design, conduct, and funding. Although our understanding of the nature and management of this disease continues to progress, without adequate engagement and support, our treatment paradigms will likely continue to be more art than science.

An appropriate companion to RCT’s is the development of collaborative quality initiatives (CQI’s).391 Within a CQI, participating hospitals and providers collect, share, and analyze data through clinical registries. CQI participants design and affect changes that improve outcomes of complex, highly technical areas of care.392 CQI registries allow for a more robust analysis of the link between processes and outcomes than can occur with retrospective single or multi-institutional studies; particularly as more sensitive and specific diagnostics/biomarkers are complemented by technologic advances. Scientific inquiry will continue to provide fundamental knowledge regarding the biological basis, inherent risks, and natural history of localized renal masses such that appropriate trade-offs can be made when considering optimal management.

Evaluation and Diagnosis

The localized renal mass remains primarily a radiographic diagnosis. The field of tumor radiomics, artificial intelligence and molecular imaging promises393 to improve our ability to discriminate tumor histology, grade394,395 and ultimately gene and protein expression with prognostic implications. The development of more sophisticated modeling of patient demographic features as recorded in the electronic medical record, such as age, gender, race, body mass index, comorbidities, exposure to tobacco, and other risk factors are being studied to contextualize and individualize management options. Finally, tumor markers detected in biopsy, blood, or urine should be studied to improve prognostic models for RCC. Efforts based on gene and protein expression have identified multiple promising markers that may one day distinguish between subtypes of malignant and benign renal tumors.396,397 Recent work through The Cancer Genome Atlas (TCGA) 398,399 to identify genomic markers for clear cell RCC400 papillary RCC401 and chromophobe RCC402 holds great clinical potential for more accurate diagnosis, prognostication, and surveillance of renal masses. The promise of measuring circulating tumor cells, or liquid tumor biopsies, for diagnosis and surveillance for recurrence and response to treatment is likely several years off, but could substantially transform care models.403-406

Counseling and Outcomes-based Research

As data emerge regarding variability in treatments performed for localized renal cancer, the impact of the individual physician-patient interaction becomes more evident. The quality of patient counseling can only be improved by providing high quality data, particularly from RCT’s. Given our current state of knowledge, translation of information from research studies and guidelines into practical materials for patients is not straight-forward. The development of decision aids for informed medical-decision making is ongoing.407,408 The appropriate application of data from large registries and implementation sciences to improve processes and standardization of care is an important initiative that must move forward. Increased quality of data, including improved assessment of tumor biology and prospective trials of management options, is greatly needed to facilitate more intelligent patient counseling.

Management

A major limitation of the literature supporting the current guidelines for management of localized renal cancer is the relatively low level of evidence. Prospective comparative trials, ideally randomized, comparing AS vs. active intervention (TA or excision) should be prioritized to provide higher quality data about oncologic and renal functional outcomes and to assess the treatment-related morbidities or limitations of each approach. With improved reporting and more extended follow-up, multi-institutional observational data will strengthen confidence in recommendations, but not nearly to the extent that clinical trials can provide.

Comparison of extirpative treatment modalities should include prospective evaluation of PN versus RN, prioritized in patients with a normal contralateral kidney and no preexisting CKD/albuminuria, with the goal of assessing the impact of new baseline functional status on overall survival, cardiovascular health, and subsequent renal stability on a longitudinal basis. Ideally, patients with tumors with increased oncologic potential (cT1b/T2) should be prioritized for such trials.259,409,410 Regarding nephron-sparing surgery, improved data comparing the relative merits and limitations of standard PN versus tumor enucleation should be sought, ideally through prospective evaluation incorporating improved reporting, and standard assessment of surgical margins.248

Multiple non-extirpative methods being actively investigated in the management of renal masses include stereotactic body radiation therapy (SBRT), HIFU, microwave ablation (MWA), and laser interstitial thermal therapy (LITT). These approaches differ in their mechanisms of action, invasiveness, reported outcomes and experience. Their use should be approached systematically and with caution, and they should be considered investigational at present. SBRT, also frequently referred to as stereotactic ablative radiotherapy (SABR), has been reported in a small number of series. SBRT involves relatively intense protocols (24 to 40 Gy) over one to five fractions and a high degree of spatial precision, offering the potential to be less invasive than surgical or conventional ablative approaches.411 Despite encouraging results, the current body of evidence is limited due to small patient numbers, short follow-up and inconsistent methods of reporting outcomes.411 Thus, SBRT in the management of localized renal masses at present remains investigational and should be primarily considered for patients who are medically inoperable and are not candidates for established TA approaches. Investigation through clinical trials should be prioritized.

Similarly, HIFU remains investigational in the management of renal masses, although it is currently used clinically to treat prostate cancer and uterine fibroids.412 HIFU relies on the use of a lens or focused transducer to deliver high-frequency sound waves to tissue, typically 1 to 5 MHz. HIFU may be administered in an entirely noninvasive means similar to extracorporeal lithotripsy, thus minimizing the risk of tumor seeding, urinary extravasation or hemorrhage.413 Initial clinical investigations have established the feasibility of transcutaneous HIFU; however, distinct regions of renal masses are frequently left untreated resulting in incomplete ablation.414-417

Similar to RFA, MWA delivers electromagnetic energy through flexible probes inserted into a target lesion. MWA produces target temperatures (>60° C) more rapidly than RFA, and, thus, appears to have significant potential as an ablative modality.418 LITT uses optical fibers that are inserted directly into the target tissue to deliver laser light that is converted into thermal energy. The most common laser type used in LITT is a neodymium: yttrium-aluminum-garnet (Nd:YAG) laser.419 Outcomes of clinical investigations are limited due to the small number of treated patients and short follow-up.420,421 Given the limited number of published studies involving HIFU, MWA and LITT and lack of long-term follow-up, appropriate use of these modalities in the management of small renal masses remains poorly defined. Larger prospective trials will be necessary to develop and assess optimal use, risks and morbidity.

Follow-up after Intervention

The proposed guidelines for follow-up after intervention for renal cancer attempt to provide a risk-based approach to surveillance and monitoring. Few high-quality studies currently exist to help formulate surveillance regimens, and many of the Panel’s recommendations are thus based primarily on expert opinion. Any cancer surveillance regimen is a balancing act that includes many variables such as the likelihood of disease recurrence at various sites, temporal considerations, the potential benefits of therapeutic interventions and effectiveness of these modalities based on timing of recurrence detection, improvements in diagnostic and initial interventions, patient characteristics, and the burden and cost of monitoring. As electronic medical records and quality and safety initiatives intensify, tracking outcomes of all patients will become increasingly codified and more usable for research purposes. These data can then also be used to inform the proper sequencing, timing, duration, and type of follow-up that improves patient outcomes with the most parsimonious monitoring.

Future research to make patient follow-up more efficient and effective could include one or many of these modalities: develop circulating biomarkers to supplement currently available imaging, develop novel functional imaging, conduct clinical trials to compare currently available imaging modalities, as well as clinical trials to guide the frequency of imaging/follow-up, similar to studies done in testicular cancer (MRC TE08)422, colon cancer (GILDA)423, and non-small cell lung cancer (IFCT-0302).424

Summary

In conclusion, improving the management of localized renal tumors will require a concerted effort among clinicians and allied fields to develop higher quality evidence and facilitate more precise estimations of relative risks and benefits of each therapeutic approach.

Tools and Resources

Abbreviations

Active SurveillanceAS
Agency for Healthcare Reserach and QualityAHRQ
American College of RadiologyACR
American Society of NephrologyASN
American Urological AssociationAUA
AngiomyolipomaAML
Birt Hogg-DubeBHD
Chest X-RayCXR
Chronic Kidney DiseaseCDK
College of American PathologistsCAP
Computed TomographyCT
End-Stage Renal DiseaseESRD
Estimated Glomerular filtration rateeGFR
Fine Needle AspirationFNA
Glomerular Filtration RateGFR
Hereditary Leiomyomatosis RCCHLRCC
Hereditary Papillary Renal CarcinomaHPRC
High RiskHR
High-Intensity Focused UltrasoundHIFU
Intermediate RiskIR
Laser Interstitial Thermal TherapyLITT
Low RiskLR
Magnetic Resonance ImagingMRI
Microwave AblationMWA
Partial NephrectomyPN
Positron Emission TomographyPET
Practice Guidelines CommitteePGC
Radical NephrectomyRN

References

  1. Stewart SB, Thompson RH, Psutka SP et al: Evaluation of the national comprehensive cancer network and american urological association renal cell carcinoma surveillance guidelines. J Clin Oncol 2014; 32: 4059.
  2. Higgins JDA: Assessing quality of included studies in Cochrane Reviews. The Cochrane Collaboration Methods Groups Newsletter 2007; 11.
  3. Whiting PF, Rutjes AW, Westwood ME et al: QUADAS-2: A revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 2011; 155: 529.
  4. Sterne JA, Hernan MA, Reeves BC, et al: ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016; 355: i4919.
  5. Faraday M, Hubbard H, Kosiak B et al: Staying at the cutting edge: a review and analysis of evidence reporting and grading; the recommendations of the American Urological Association. BJU Int 2009; 104: 294.
  6. Hsu C and Sandford BA: The Delphi technique: making sense of consensus. Practical Assessment, Research & Evaluation 2007; 12: 1.
  7. Kutikov A, Fossett LK, Ramchandani P et al: Incidence of benign pathologic findings at partial nephrectomy for solitary renal mass presumed to be renal cell carcinoma on preoperative imaging. Urology 2006; 68: 737.
  8. Thompson RH, Hill JR, Babayev Y et al: Metastatic renal cell carcinoma risk according to tumor size. J Urol 2009; 182: 41.
  9. Lane BR, Babineau D, Kattan MW et al: A preoperative prognostic nomogram for solid enhancing renal tumors 7 cm or less amenable to partial nephrectomy. J Urol 2007; 178: 429.
  10. Johnson DC, Vukina J, Smith AB et al: Preoperatively misclassified, surgically removed benign renal masses: a systematic review of surgical series and United States population level burden estimate. J Urol 2015; 193: 30.
  11. National Cancer Institute SEER: Stat Fact Sheets: Kidney and Renal Pelvis Cancer. Cancer Statistics 2016; http://seer.cancer.gov/statfacts/html/kidrp.html.
  12. Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin 2020; 70: 7.
  13. Mathew A, Devesa SS, Fraumeni JF et al: Global increases in kidney cancer incidence, 1973-1992. Eur J Cancer Prev 2002; 11: 171.
  14. Nguyen MM, Gill IS, Ellison LM: The evolving presentation of renal carcinoma in the United States: trends from the Surveillance, Epidemiology, and End Results program. J Urol 2006; 176: 2397.
  15. Kane CJ, Mallin K, Ritchey J et al: Renal cell cancer stage migration: analysis of the National Cancer Data Base. Cancer 2008; 113: 78.
  16. Smaldone MC, Egleston B, Hollingsworth JM et al: Understanding treatment disconnect and mortality trends in renal cell carcinoma using tumor registry data. Med Care 2016; Epub ahead of print.
  17. Hollingsworth JM, Miller DC, Daignault S et al: Rising incidence of small renal masses: a need to reassess treatment effect. JNCI 2006; 98: 1331.
  18. The American Cancer Society medical and editorial content team: What Are the Risk Factors for Kidney Cancer? American Cancer Society 2016; https://www.cancer.org/cancer/kidney-cancer/causes-risks-prevention/risk-factors.html#written_by.
  19. Campbell SC, Lane BR: Campbell-Walsh Urology, Malignant Renal Tumors. 11th ed: Elsevier 2016; 1314.
  20. Ferlay J, Soerjomataram I, Dikshit R et al: Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015; 136: E359.
  21. Znaor A, Lortet-Tieulent J, Laversanne M et al: Bray F. International variations and trends in renal cell carcinoma incidence and mortality. Eur Urol 2015; 67: 519.
  22. Hunt JD, van der Hel OL, McMillan GP et al: Renal cell carcinoma in relation to cigarette smoking: meta-analysis of 24 studies. Int J Cancer 2005; 114: 101.
  23. Lipworth L, Tarone RE, McLaughlin JK: The epidemiology of renal cell carcinoma. J Urol 2006; 176: 2353.
  24. Renehan AG, Tyson M, Egger M et al: Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008; 371: 569.
  25. Bjorge T, Tretli S, Engeland A: Relation of height and body mass index to renal cell carcinoma in two million Norwegian men and women. Am J Epidemiol 2004; 160: 1168.
  26. Hakimi AA, Furberg H, Zabor EC et al: An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma. J Natl Cancer Inst 2013; 105: 1862.
  27. Yang L, Drake BF, Colditz GA et al: Obesity and other cancers. JCO 2016; 34: 4231.
  28. Shapiro JA, Williams MA, Weiss NS et al: Hypertension, antihypertensive medication use, and risk of renal cell carcinoma. Am J Epidemiol 1999; 149: 521.
  29. Weikert S, Boeing H, Pischon T et al: Blood pressure and risk of renal cell carcinoma in the European prospective investigation into cancer and nutrition. Am J Epidemiol 2008; 167: 438.
  30. Stewart JH, Buccianti G, Agodoa L et al: Cancers of the kidney and urinary tract in patients on dialysis for end-stage renal disease: analysis of data from the United States, Europe, and Australia and New Zealand. JASN 2003; 14: 197.
  31. Guha N, Loomis D, Grosse Y et al: Carcinogenicity of trichloroethylene, tetrachloroethylene, some other chlorinated solvents, and their metabolites. The Lancet Oncology 2012; 13: 1192.
  32. Rashidkhani B, Akesson A, Lindblad P et al: Alcohol consumption and risk of renal cell carcinoma: a prospective study of Swedish women. Int J Cancer 2005; 117: 848.
  33. Greving JP, Lee JE, Wolk A et al: Alcoholic beverages and risk of renal cell cancer. Br J Cancer 2007; 97: 429.
  34. Lee JE, Mannisto S, Spiegelman D et al: Intakes of fruit, vegetables, and carotenoids and renal cell cancer risk: a pooled analysis of 13 prospective studies. Cancer Epidemiol Biomarkers Prev 2009; 18: 1730.
  35. Rashidkhani B, Lindblad P, Wolk A: Fruits, vegetables and risk of renal cell carcinoma: a prospective study of Swedish women. Int J Cancer 2005; 113: 451.
  36. Wolk A, Larsson SC, Johansson JE et al: Long-term fatty fish consumption and renal cell carcinoma incidence in women. Jama 2006; 296: 1371.
  37. McCredie M, Pommer W, McLaughlin JK et al: International renal-cell cancer study. II. Analgesics. Int J Cancer 1995; 60: 345.
  38. Linehan WM, Ricketts CJ: The metabolic basis of kidney cancer. Semin Cancer Biol 2013; 23: 46.
  39. Duffey BG, Choyke PL, Glenn G et al: The relationship between renal tumor size and metastases in patients with von Hippel-Lindau disease. J Urol 2004; 172: 63.
  40. Srigley JR, Delahunt B, Eble JN et al: The International Society of Urological Pathology (ISUP) Vancouver Classification of renal neoplasia. Am J Surg Pathol 2013; 37: 1469.
  41. Moch H, Humphrey PA, Ulbright TM et al: WHO classification of tumours of the urinary system and male genital organs. 4 ed: Lyon IARC; 2016.
  42. Jayson M, Sanders H: Increased incidence of serendipitously discovered renal cell carcinoma. Urology 1998; 51: 203.
  43. Luciani LG, Cestari R, Tallarigo C: Incidental renal cell carcinoma-age and stage characterization and clinical implications: study of 1092 patients (1982-1997). Urology 2000; 56: 58.
  44. Sufrin G, Chasan S, Golio A et al: Paraneoplastic and serologic syndromes of renal adenocarcinoma. Semin Urol 1989; 7: 158.
  45. Snyder ME, Bach A, Kattan MW et al: Incidence of benign lesions for clinically localized renal masses smaller than 7 cm in radiological diameter: influence of sex. J Urol 2006; 176: 2391.
  46. Ball MW, Gorin MA, Bhayani SB et al: Preoperative predictors of malignancy and unfavorable pathology for clinical T1a tumors treated with partial nephrectomy: A multi-institutional analysis. Urol Oncol 2015; 33: 112.
  47. Davenport MS, Perazella MA, Yee J et al: Use of intravenous iodinated contrast media in patients with kidney disease: Consensus statements from the american college of radiology and the national kidney foundation. Radiology 2020; 294: 660.
  48. Weinreb JC, Rodby RA, Yee J et al: Use of intravenous gadolinium-based contrast media in patients with kidney disease: Consensus statements from the American College of Radiology and the national kidney foundation. Radiology 2021; 298: 28.
  49. Lim DJ, Carter MF: Computerized tomography in the preoperative staging for pulmonary metastases in patients with renal cell carcinoma. J Urol 1993; 150: 1112.
  50. Mano R, Vertosick E, Sankin AI et al: Subcentimeter pulmonary nodules are not associated with disease progression in patients with renal cell carcinoma. J Urol 2015; 193: 776.
  51. Seaman E, Goluboff ET, Ross S et al: Association of radionuclide bone scan and serum alkaline phosphatase in patients with metastatic renal cell carcinoma. Urology 1996; 48: 692.
  52. Marshall ME, Pearson T, Simpson W et al: Low incidence of asymptomatic brain metastases in patients with renal cell carcinoma. Urology 1990; 36: 300.
  53. Koga S, Tsuda S, Nishikido M et al: The diagnostic value of bone scan in patients with renal cell carcinoma. J Urol 2001; 166: 2126.
  54. Amin MB, Greene FL, Edge SB et al: The eighth edition ajcc cancer staging manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging. CA Cancer J Clin 2017; 67: 93.
  55. Fuhrman SA, Lasky LC, Limas C: Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol 1982; 6: 655.
  56. Delahunt B, Cheville JC, Martignoni G et al: The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol 2013; 37: 1490.
  57. Bretheau D, Lechevallier E, de Fromont M et al: Prognostic value of nuclear grade of renal cell carcinoma. Cancer 1995; 76: 2543.
  58. Keegan KA, Schupp CW, Chamie K et al: Histopathology of surgically treated renal cell carcinoma: survival differences by subtype and stage. J Urol 2012; 188: 391.
  59. Zisman A, Pantuck AJ, Dorey F et al: Improved prognostication of renal cell carcinoma using an integrated staging system. J Clin Oncol 2001; 19: 1649.
  60. Patard JJ, Kim HL, Lam JS et al: Use of the University of California Los Angeles integrated staging system to predict survival in renal cell carcinoma: an international multicenter study. J Clin Oncol 2004; 22: 3316.
  61. Frank I, Blute ML, Cheville JC et al: An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score. J Urol 2002; 168: 2395.
  62. Leibovich BC, Blute ML, Cheville JC et al: Prediction of progression after radical nephrectomy for patients with clear cell renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer 2003; 97: 1663.
  63. Zigeuner R, Hutterer G, Chromecki T et al: External validation of the Mayo Clinic stage, size, grade, and necrosis (SSIGN) score for clear-cell renal cell carcinoma in a single European centre applying routine pathology. Eur Urol 2010; 57: 102.
  64. Karakiewicz PI, Suardi N, Capitanio U et al: A preoperative prognostic model for patients treated with nephrectomy for renal cell carcinoma. Eur Urol 2009; 55: 287.
  65. Pierorazio PM, Johnson MH, Patel HD et al: Management of renal masses and localized renal cancer. AHRQ Publication 16-EHC001-EF, 2016 #167.
  66. Patel HD, Iyoha E, Pierorazio PM et al: A Systematic Review of Research Gaps in the Evaluation and Management of Localized Renal Masses. Urology 2016; 98: 14.
  67. Morrissey JJ, Mobley J, Figenshau RS et al: Urine aquaporin 1 and perilipin 2 differentiate renal carcinomas from other imaged renal masses and bladder and prostate cancer. Mayo Clin Proc 2015; 90: 35.
  68. Song JB, Morrissey JJ, Mobley JM et al: Urinary aquaporin 1 and perilipin 2: Can these novel markers accurately characterize small renal masses and help guide patient management? Int J Urol 2019; 26: 260.
  69. Kaluz S, Kaluzova M, Liao SY et al: Transcriptional control of the tumor- and hypoxia-marker carbonic anhydrase 9: A one transcription factor (HIF-1) show? Biochim Biophys Acta 2009; 1795: 162.
  70. Stillebroer AB, Mulders PF, Boerman OC et al: Carbonic anhydrase IX in renal cell carcinoma: implications for prognosis, diagnosis, and therapy. Eur Urol 2010; 58: 75.
  71. Muselaers S, Mulders P, Oosterwijk E et al: Molecular imaging and carbonic anhydrase IX-targeted radioimmunotherapy in clear cell renal cell carcinoma. Immunotherapy 2013; 5: 489.
  72. Divgi CR, Pandit-Taskar N, Jungbluth AA et al: Preoperative characterization of clear-cell renal carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial. Lancet Oncol 2007; 8: 304.
  73. Divgi CR, Uzzo RG, Gatsonis C et al: Positron emission tomography/computed tomography identification of clear cell renal cell carcinoma: results from the REDECT trial. J Clin Oncol 2013; 31: 187.
  74. Gorin MA, Rowe SP, Baras AS et al: Prospective evaluation of (99m)Tc-sestamibi SPECT/CT for the diagnosis of renal oncocytomas and hybrid oncocytic/chromophobe tumors. Eur Urol 2016; 69: 413.
  75. Kunkle DA, Egleston BL, Uzzo RG. Excise, ablate or observe: the small renal mass dilemma--a meta-analysis and review. J Urol 2008; 179: 1227.
  76. Chawla SN, Crispen PL, Hanlon AL et al: The natural history of observed enhancing renal masses: meta-analysis and review of the world literature. J Urol 2006; 175: 425.
  77. Smaldone MC, Kutikov A, Egleston BL et al: Small renal masses progressing to metastases under active surveillance: A systematic review and pooled analysis. Cancer 2011; 118: 997.
  78. Jewett MA, Mattar K, Basiuk J et al: Active surveillance of small renal masses: progression patterns of early stage kidney cancer. Eur Urol 2011; 60: 39.
  79. Mason RJ, Abdolell M, Trottier G et al: Growth kinetics of renal masses: analysis of a prospective cohort of patients undergoing active surveillance. Eur Urol 2011; 59: 863.
  80. Pierorazio PM, Johnson MH, Ball MW et al: Five-year analysis of a multi-institutional prospective clinical trial of delayed intervention and surveillance for small renal masses: The DISSRM Registry. Eur Urol 2015; 68: 408.
  81. Patel SG, Penson DF, Pabla B et al: National trends in the use of partial nephrectomy: a rising tide that has not lifted all boats. J Urol 2012; 187: 816.
  82. Bjurlin MA, Walter D, Taksler GB et al: National trends in the utilization of partial nephrectomy before and after the establishment of AUA guidelines for the management of renal masses. Urology 2013; 82: 1283.
  83. Ghani KR, Sukumar S, Sammon JD et al: Practice patterns and outcomes of open and minimally invasive partial nephrectomy since the introduction of robotic partial nephrectomy: results from the nationwide inpatient sample. J Urol 2014; 191: 907.
  84. Kunkle DA, Uzzo RG: Cryoablation or radiofrequency ablation of the small renal mass : a meta-analysis. Cancer 2008; 113: 2671.
  85. El Dib R, Touma NJ, Kapoor A: Cryoablation vs radiofrequency ablation for the treatment of renal cell carcinoma: a meta-analysis of case series studies. BJU Int 2012; 110: 510.
  86. Pirasteh A, Snyder L, Boncher N et al: Cryoablation vs. radiofrequency ablation for small renal masses. Acad Radiol 2011; 18: 97.
  87. Kattan MW, Reuter V, Motzer RJ et al: A postoperative prognostic nomogram for renal cell carcinoma. J Urol 2001; 166: 63.
  88. Sorbellini M, Kattan MW, Snyder ME et al: A postoperative prognostic nomogram predicting recurrence for patients with conventional clear cell renal cell carcinoma. J Urol 2005; 173: 48.
  89. Zisman A, Pantuck AJ, Wieder J et al: Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma. J Clin Oncol 2002; 20: 4559.
  90. Karakiewicz PI, Briganti A, Chun FK et al: Multi-institutional validation of a new renal cancer-specific survival nomogram. J Clin Oncol 2007; 25: 1316.
  91. Cindolo L, de la Taille A, Messina G et al: A preoperative clinical prognostic model for non-metastatic renal cell carcinoma. BJU Int 2003; 92: 901.
  92. Cindolo L, de la Taille A, Messina G et al: A preoperative clinical prognostic model for non-metastatic renal cell carcinoma. BJU Int 2003; 92: 901.
  93. Yaycioglu O, Roberts WW, Chan T et al: Prognostic assessment of nonmetastatic renal cell carcinoma: A clinically based model. Urology 2001; 58: 141.
  94. Correa AF, Jegede O, Haas NB et al: Predicting renal cancer recurrence: Defining limitations of existing prognostic models with prospective trial-based validation. J Clin Oncol 2019; 37: 2062.
  95. Ljungberg B, Albiges L, Abu-Ghanem Y et al: European association of urology guidelines on renal cell carcinoma: The 2019 update. Eur Urol 2019; 75: 799.
  96. Blacher E, Johnson DE and Haynie TP: Value of routine radionuclide bone scans in renal cell carcinoma. Urology 1985; 26: 432.
  97. Lindner A, Goldman DG and deKernion JB: Cost effective analysis of prenephrectomy radioisotope scans in renal cell carcinoma. Urology 1983; 22: 127.
  98. Benson MA, Haaga JR and Resnick MI: Staging renal carcinoma. What is sufficient? Arch Surg 1989; 124: 71.
  99. Grünwald V, Eberhardt B, Bex A et al: An interdisciplinary consensus on the management of bone metastases from renal cell carcinoma. Nat Rev Urol 2018; 15: 511.
  100. Davenport MS, Hu EM, Smith AD et al: Reporting standards for the imaging-based diagnosis of renal masses on CT and MRI: a national survey of academic abdominal radiologists and urologists. Abdom Radiol 2017: 42; 1229.
  101. Berland LL, Silverman SG, Gore RM et al: Managing incidental findings on abdominal CT: white paper of the ACR incidental findings committee. J Am Coll Radiol 2010; 7: 754.
  102. Richmond L, Atri M, Sherman C et al: Renal cell carcinoma containing macroscopic fat on CT mimics angiomyolipoma due to bone metapolasia without macroscopic calcification. Br J Radiol 2010; 83: e179.
  103. Rofsky NM, Weinreb JC, Bosniak MA et al: Renal lesion characterization with gadolinium-enhanced MR imaging: efficacy and safety in patients with renal insufficiency. Radiology 1991; 180: 85.
  104. Schoots IG, Zaccai K, Hunink MG et al: Bosniak classification for complex renal cysts reevaluated: A systematic review. J Urol 2017; 198: 12.
  105. Silverman SG, Pedrosa I, Ellis JH et al: Bosniak classification of cystic renal masses, version 2019: An update proposal and needs assessment. Radiology 2019; 292: 475.
  106. Nicolau C, Buñesch L, Paño B et al: Prospective evaluation of CT indeterminate renal masses using US and contrast-enhanced ultrasound. Abdom Imaging 2015; 40: 542.
  107. Atri M, Tabatabaeifar L, Jang H-J et al: Accuracy of contrast-enhanced US for differentiating benign from malignant solid small renal masses. Radiology 2015; 276: 900.
  108. Kopp RP, Aganovic L, Palazzi KL et al: Differentiation of clear from non-clear cell renal cell carcinoma using CT washout formula. Can J Urol 2013; 20: 6790.
  109. Young JR, Margolis D, Sauk S et al: Clear cell renal cell carcinoma: discrimination from other renal cell carcinoma subtypes and oncocytoma at multiphasic multidetector CT. Radiology 2013; 267: 444.
  110. Kutikov A, Uzzo RG: The R.E.N.A.L. nephrometry score: a comprehensive standardized system for quantitating renal tumor size, location and depth. J Urol 2009; 182: 844.
  111. Ficarra V, Novara G, Secco S et al: Preoperative aspects and dimensions used for an anatomical (PADUA) classification of renal tumours in patients who are candidates for nephron-sparing surgery. Eur Urol 2009; 56: 786.
  112. Simmons MN, Ching CB, Samplaski MK et al: Kidney tumor location measurement using the C index method. J Urol 2010; 183: 1708.
  113. Tobert CM, Kahnoski RJ, Thompson DE et al: RENAL nephrometry score predicts surgery type independent of individual surgeon’s use of nephron-sparing surgery. Urology 2012; 80: 157.
  114. Stroup SP, Palazzi K, Kopp RP et al: RENAL nephrometry score is associated with operative approach for partial nephrectomy and urine leak. Urology 2012; 80: 151.
  115. Bruner B, Breau RH, Lohse CM et al: Renal nephrometry score is associated with urine leak after partial nephrectomy. BJU Int 2011; 108: 67.
  116. Kutikov A, Smaldone M, Egleston BL et al: Anatomic features of enhancing renal masses predict malignant and high-grade pathology: a preoperative nomogram using the RENAL Nephrometry score. Eur Uro 2011; 60: 241.
  117. Mehrazin R, Palazzi KL, Kopp RP et al: Impact of tumour morphology on renal function decline after partial nephrectomy. BJU Int 2013; 111: E374.
  118. Levey AS, Eckardt KU, Tsukamoto Y et al: Definition and classification of chronic kidney disease: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2005; 67: 2089.
  119. Hallan SI, Ritz E, Lydersen S et al: Combining GFR and albuminuria to classify CKD improves prediction of ESRD. J Am Soc Nephrol 2009; 20: 1069.
  120. Levey AS, Bosch JP, Lewis JB et al: A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of diet in renal disease study group. Ann Intern Med 1999; 130: 461.
  121. Levey AS, Stevens LA, Schmid CH et al: A new equation to estimate glomerular filtration rate. Ann Intern Med 2009; 150: 604.
  122. Barocas DA, Boorjian SA, Alvarez RD et al: Microhematuria: Aua/sufu guideline. J Urol 2020; 204: 778.
  123. Pinkham CA, Krause KJ: Liver function tests and mortality in a cohort of life insurance applicants. J Insur Med 2009; 41: 170.
  124. Guðmundsson E, Hellborg H, Lundstam S et al: Metastatic potential in renal cell carcinomas ≤7 cm: Swedish Kidney Cancer Quality Register data. Eur Urol 2011; 60: 975.
  125. Umbreit EC, Shimko MS, Childs MA et al: Metastatic potential of a renal mass according to original tumour size at presentation. BJU Int 2012; 109: 190.
  126. Barlow LJ, Korets R, Laudano M et al: Predicting renal functional outcomes after surgery for renal cortical tumours: a multifactorial analysis. BJU Int 2010; 106: 489.
  127. Kim SH, Lee SE, Hong SK et al: Incidence and risk factors of chronic kidney disease in Korean patients with t1a renal cell carcinoma before and after radical or partial nephrectomy. Jpn J Clin Oncol 2013; 43: 1243.
  128. Jeon HG, Jeong IG, Lee JW et al: Prognostic factors for chronic kidney disease after curative surgery in patients with small renal tumors. Urology 2009; 74: 1064.
  129. Huang WC, Levey AS, Serio AM et al: Chronic kidney disease after nephrectomy in patients with renal cortical tumours: a retrospective cohort study. Lancet Oncol 2006; 7: 735.
  130. Choi YS, Park YH, Kim YJ et al: Predictive factors for the development of chronic renal insufficiency after renal surgery: a multicenter study. Int Urol Nephrol 2014; 46: 681.
  131. Takagi T, Kondo T, Iizuka J et al: Postoperative renal function after partial nephrectomy for renal cell carcinoma in patients with pre-existing chronic kidney disease: a comparison with radical nephrectomy. Int J Urol 2011; 18: 472.
  132. Clark MA, Shikanov S, Raman JD et al: Chronic kidney disease before and after partial nephrectomy. J Urol 2011; 185: 43.
  133. Hung PH, Tsai HB, Hung KY et al: Increased risk of end-stage renal disease in patients with renal cell carcinoma: a 12-year nationwide follow-up study. Medicine (Baltimore) 2014; 93: e52.
  134. Go AS, Chertow GM, Fan D et al: Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med 2004; 351: 1296.
  135. Gerstein HC, Mann JF, Yi Q et al: Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. Jama 2001; 286: 421.
  136. Tourojman M, Kirmiz S, Boelkins B et al: Impact of Reduced Glomerular Filtration Rate and Proteinuria on Overall Survival of Patients with Renal Cancer. J Urol 2016; 195: 588.
  137. Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group. KDIGO 2012 Clinical Practice Guideline for the Evaluation and Management of Chronic Kidney Disease. Kidney Int Suppl 2013; 3: 1.
  138. Bachrach L, Negron E, Liu JS et al: Preoperative nuclear renal scan underestimates renal function after radical nephrectomy. Urology 2014; 84: 1402.
  139. Sankin A, Sfakianos JP, Schiff J et al: Assessing renal function after partial nephrectomy using renal nuclear scintigraphy and estimated glomerular filtration rate. Urology 2012; 80: 343.
  140. Patel SR, Abel EJ, Hedican SP et al: Ablation of small renal masses: practice patterns at academic institutions in the United States. J Endourol 2013; 27: 158.
  141. Menhadji AD, Nguyen V, Okhunov Z et al: Technique for office-based, ultrasonography-guided percutaneous biopsy of renal cortical neoplasms using a novel transducer for facilitated ultrasound targeting. BJU Int 2016; 117: 948.
  142. Bearrick EN, Packiam V, Bhindi B et al: Creation of a primary tumor tissue expression biomarker-augmented prognostic model for patients with metastatic renal cell carcinoma. Urol Oncol 2021; 39: 135.e1.
  143. Tosoian JJ, Feldman AS, Abbott MR et al: Biopsy cell cycle proliferation score predicts adverse surgical pathology in localized renal cell carcinoma. Eur Urol 2020; 78: 657.
  144. Bijol V, Mendez GP, Hurwitz S et al: Evaluation of the nonneoplastic pathology in tumor nephrectomy specimens: predicting the risk of progressive renal failure. Am J Surg Pathol 2006; 30: 575.
  145. Salvatore SP, Cha EK, Rosoff JS et al: Nonneoplastic renal cortical scarring at tumor nephrectomy predicts decline in kidney function. Arch Pathol Lab Med 2013; 137: 531.
  146. Henriksen KJ, Meehan SM, Chang A: Non-neoplastic renal diseases are often unrecognized in adult tumor nephrectomy specimens: a review of 246 cases. Am J Surg Pathol 2007; 31: 1703.
  147. Rini BI, Plimack ER, Takagi T et al: A Phase II Study of Pazopanib in Patients with Localized Renal Cell Carcinoma to Optimize Preservation of Renal Parenchyma. J Urol 2015; 194: 297.
  148. Karam JA, Devine CE, Urbauer DL et al: Phase 2 trial of neoadjuvant axitinib in patients with locally advanced nonmetastatic clear cell renal cell carcinoma. Eur Urol 2014; 66: 874.
  149. Bhindi B, Thompson RH, Lohse CM et al: The probability of aggressive versus indolent histology based on renal tumor size: Implications for surveillance and treatment. Eur Urol 2018; 74: 489.
  150. Frank I, Blute ML, Cheville JC et al: Solid renal tumors: an analysis of pathological features related to tumor size. J Urol 2003; 170: 2217.
  151. Pierorazio PM, Hyams ES, Tsai S et al: Multiphasic enhancement patterns of small renal masses (</=4 cm) on preoperative computed tomography: utility for distinguishing subtypes of renal cell carcinoma, angiomyolipoma, and oncocytoma. Urology 2013; 81: 1265.
  152. Chandrasekar T, Ahmad AE, Fadaak K et al: Natural history of complex renal cysts: Clinical evidence supporting active surveillance. J Urol 2018; 199: 633.
  153. Kashan M, Ghanaat M, Hötker AM et al: Cystic renal cell carcinoma: A report on outcomes of surgery and active surveillance in patients retrospectively identified on pretreatment imaging. J Urol 2018; 200: 275.
  154. Westerman ME, Cheville JC, Lohse CM et al: Long-term outcomes of patients with low grade cystic renal epithelial neoplasms. Urology 2019; 133: 145.
  155. Kutikov A, Egleston BL, Canter D et al: Competing risks of death in patients with localized renal cell carcinoma: a comorbidity based model. J Urol 2012; 188: 2077.
  156. Patel HD, Kates M, Pierorazio PM et al: Balancing cardiovascular (CV) and cancer death among patients with small renal masses: modification by CV risk. BJU Int 2015; 115: 58.
  157. Patel HD, Kates M, Pierorazio PM et al: Comorbidities and Causes of Death in the Management of Localized T1a Kidney Cancer. Int J Urol 2014;article in press.
  158. Bianchi M, Becker A, Abdollah F et al: Rates of open versus laparoscopic and partial versus radical nephrectomy for T1a renal cell carcinoma: a population-based evaluation. Int J Urol 2013; 20: 1064.
  159. McIntosh AG, Ristau BT, Ruth K et al: Active surveillance for localized renal masses: Tumor growth, delayed intervention rates, and >5-yr clinical outcomes. Eur Urol 2018; 74: 157.
  160. Sotimehin AE, Patel HD, Alam R et al: Selecting patients with small renal masses for active surveillance: A domain based score from a prospective cohort study. J Urol 2019; 201: 886.
  161. Mir MC, Capitanio U, Bertolo R et al: Role of active surveillance for localized small renal masses. Eur Urol Oncol 2018; 1: 177.
  162. Chow W-H, Dong LM, Devesa SS: Epidemiology and risk factors for kidney cancer. Nat Rev Urol 2010; 7: 245.
  163. Wu J, Suk-Ouichai C, Dong W et al: Analysis of survival for patients with chronic kidney disease primarily related to renal cancer surgery. BJU Int 2018; 121: 93.
  164. Lane BR, Demirjian S, Derweesh IH et al: Survival and functional stability in chronic kidney disease due to surgical removal of nephrons: Importance of the new baseline glomerular filtration rate. Eur Urol 2015; 68: 996.
  165. Lane BR, Campbell SC, Demirjian S et al: Surgically induced chronic kidney disease may be associated with a lower risk of progression and mortality than medical chronic kidney disease. J Urol 2013; 189: 1649.
  166. Li L, Lau WL, Rhee CM et al: Risk of chronic kidney disease after cancer nephrectomy. Nat Rev Nephrol 2014; 10: 135.
  167. Malcolm JB, Bagrodia A, Derweesh IH et al: Comparison of rates and risk factors for developing chronic renal insufficiency, proteinuria and metabolic acidosis after radical or partial nephrectomy. BJU Int 2009; 104: 476.
  168. Stiles KP, Moffatt MJ, Agodoa LY et al: Renal cell carcinoma as a cause of end-stage renal disease in the United States: Patient characteristics and survival. Kidney Int 2003; 64: 247.
  169. Jeon HG, Choo SH, Sung HH et al: Small tumour size is associated with new-onset chronic kidney disease after radical nephrectomy in patients with renal cell carcinoma. Eur J Cancer 2014; 50: 64.
  170. Cho A, Lee JE, Kwon G-Y et al: Post-operative acute kidney injury in patients with renal cell carcinoma is a potent risk factor for new-onset chronic kidney disease after radical nephrectomy. Nephrol Dial Transpl 2011; 26: 3496.
  171. Thadhani R, Pascual M, Bonventre JV: Acute Renal Failure. N Engl J Med 1996; 334: 1448.
  172. Jacobson HR: Chronic renal failure: pathophysiology. Lancet 1991; 338: 419.
  173. Nagata M, Kriz W: Glomerular damage after uninephrectomy in young rats. II. Mechanical stress on podocytes as a pathway to sclerosis. Kidney Int 1992; 42: 148.
  174. Romagnani P, Remuzzi G, Glassock R et al: Chronic kidney disease. Nat Rev Dis Primers 2017; 3: 17088.
  175. Webster AC, Nagler EV, Morton RL et al: Chronic kidney disease. Lancet 2017; 389: 1238.
  176. Huang WC, Donin NM, Levey AS et al: Chronic kidney disease and kidney cancer surgery: New perspectives. J Urol 2020; 203: 475.
  177. Linehan WM: Genetic basis of kidney cancer: Role of genomics for the development of disease-based therapeutics. Genome Res 2012; 22: 2089.
  178. Menko FH, Maher ER, Schmidt LS et al: Hereditary leiomyomatosis and renal cell cancer (HLRCC): renal cancer risk, surveillance and treatment. Fam Cancer 2014; 13: 637.
  179. Gudbjartsson T, Jónasdóttir TJ, Thoroddsen A et al: A population-based familial aggregation analysis indicates genetic contribution in a majority of renal cell carcinomas. Int J Cancer 2002; 100: 476.
  180. Hampel H, Bennett RL, Buchanan A et al: A practice guideline from the american college of medical genetics and genomics and the national society of genetic counselors: Referral indications for cancer predisposition assessment. Genet Med 2015; 17: 70.
  181. Motzer RJ, Jonasch E, Boyle S et al: Nccn guidelines insights: Kidney cancer, version 1.2021. J Natl Compr Canc Netw 2020; 18: 1160.
  182. Shuch B, Vourganti S, Ricketts CJ et al: Defining early-onset kidney cancer: implications for germline and somatic mutation testing and clinical management. J Clin Oncol 2014; 32: 431.
  183. Marconi L, Dabestani S, Lam TB et al: Systematic review and meta-analysis of diagnostic accuracy of percutaneous renal tumor biopsy. Euro Urology 2016; 69: 660.
  184. Kutikov A, Smaldone MC, Uzzo RG et al: Renal mass biopsy: always, sometimes, or never? Euro Urology 2016; 70: 403.
  185. Ingels A, Barret E, Sanchez-Salas R et al: Percutaneous renal biopsies for small renal masses: Complex tumors on nephrometry should be the first targets. Clinical Genitourinary Cancer 2016; 14: e457.
  186. Hoare D, Evans H, Richards H et al: Evaluating the role for renal biopsy in t1 and t2 renal masses: A single-centre study. Canadian Urological Association Journal 2018; 12: E226.
  187. Garbens A, Wallis CJD, Klaassen Z et al: Comprehensive assessment of the morbidity of renal mass biopsy: A population-based assessment of biopsy-related complications. Can Urol Assoc J 2020;
  188. Wang X, Lv Y, Xu Z et al: Accuracy and safety of ultrasound-guided percutaneous needle core biopsy of renal masses: A single center experience in china. Medicine 2018; 97: e0178.
  189. Alle N, Tan N, Huss J et al: Percutaneous image-guided core biopsy of solid renal masses: Analysis of safety, efficacy, pathologic interpretation, and clinical significance. Abdominal Radiology 2018; 43: 1813.
  190. Tong W, Lin X, Xu Y et al: The role of percutaneous fine needle aspiration biopsy in the management of small renal masses without chance of nephron-sparing surgery. Int Urol Nephrol 2020; 52: 2223.
  191. Prendeville S, Richard PO, Jewett MAS et al: Accuracy of renal tumour biopsy for the diagnosis and subtyping of papillary renal cell carcinoma: Analysis of paired biopsy and nephrectomy specimens with focus on discordant cases. J Clin Pathol 2019; 72: 363.
  192. Ginzburg S, Uzzo R, Al-Saleem T et al: Coexisting hybrid malignancy in a solitary sporadic solid benign renal mass: implications for treating patients following renal biopsy. J Urol 2014; 191: 296.
  193. Patel HD, Druskin SC, Rowe SP et al: Surgical histopathology for suspected oncocytomas on renal mass biopsy: a systematic review and meta-analysis. BJU Int 2017; Epub ahead of print.
  194. Huang H, Tamboli P, Karam JA et al: Secondary malignancies diagnosed using kidney needle core biopsies: a clinical and pathological study of 75 cases. Hum Pathol 2016; 52: 55.
  195. Adamy A, Von Bodman C, Ghoneim T et al: Solitary, isolated metastatic disease to kidney: Memorial Sloan-Kettering Cancer Center experience. BJU Int 2011; 108: 338.
  196. Surabhi VR, Menias C, Prasad SR et al: Neoplastic and non-neoplastic proliferative disorders of the perirenal space: cross-sectional imaging findings. Radiographics 2008; 28: 1005.
  197. Duveau A, Sayegh J, Beloncl F et al: Pseudotumours: an atypical presentation of renal sarcoidosis. QJM 2013; 106: 947.
  198. Dufour JF, Le Gallou T, Cordier JF et al: Urogenital manifestations in Wegener granulomatosis: a study of 11 cases and review of the literature. Medicine (Baltimore) 2012; 91: 67.
  199. Li JY, Yong TY, Coleman M et al: Bilateral renal inflammatory pseudotumour effectively treated with corticosteroid. Clin Exp Nephrol 2010; 14: 190.
  200. Patel HD, Johnson MH, Pierorazio PM et al: Diagnostic accuracy and risks of biopsy in the diagnosis of a renal mass suspicious for localized renal cell carcinoma: systematic review of the literature. J Urol 2016; 195: 1340.
  201. Richard PO, Jewett MA, Bhatt JR et al: Renal tumor biopsy for small renal masses: a single-center 13-year experience. Eur Urol 2015; 68: 1007.
  202. Aguilar Palacios D, Li J, Mahmood F et al: Partial nephrectomy for patients with severe chronic kidney disease-is it worthwhile? J Urol 2020; 204: 434.
  203. American Society of Cytopathology. ASC Rapid On-Site Evaluation (ROSE) Position Statement. Cytopahtology 2014; http://www.cytopathology.org/wp-content/uploads/2013/05/ASC-ROSE-Position-Final-Committee-Document.pdf.
  204. Pierorazio PM, Johnson MH, Patel HD et al: Management of renal masses and localized renal cancer: systematic review and meta-analysis. J Urol 2016; 196: 989.
  205. Van Poppel H, Da Pozzo L, Albrecht W et al: A prospective, randomized EORTC intergroup phase 3 study comparing the oncologic outcome of elective nephron-sparing surgery and radical nephrectomy for low-stage renal cell carcinoma. Eur Urol 2011; 59: 543.
  206. Wiklund F, Tretli S, Choueiri TK et al: Risk of bilateral renal cell cancer. J Clin Oncol 2009; 27: 3737.
  207. Kheterpal E and Taneja SS: Partial nephrectomy: Contemporary outcomes, candidate selection, and surgical approach. Urol Clin North Am 2012; 39: 199.
  208. Touijer K, Jacqmin D, Kavoussi LR et al: The expanding role of partial nephrectomy: A critical analysis of indications, results, and complications. Eur Urol 2010; 57: 214.
  209. Alam R, Patel HD, Osumah T et al: Comparative effectiveness of management options for patients with small renal masses: A prospective cohort study. BJU Int 2019; 123: 42.
  210. Park BK, Gong IH, Kang MY et al: Rfa versus robotic partial nephrectomy for t1a renal cell carcinoma: A propensity score-matched comparison of mid-term outcome. Eur Radiol 2018; 28: 2979.
  211. Andrews JR, Atwell T, Schmit G et al: Oncologic outcomes following partial nephrectomy and percutaneous ablation for ct1 renal masses. Eur Urol 2019; 76: 244.
  212. Talenfeld AD, Gennarelli RL, Elkin EB et al: Percutaneous ablation versus partial and radical nephrectomy for t1a renal cancer: A population-based analysis. Ann Intern Med 2018; 169: 69.
  213. Herring JC, Enquist EG, Chernoff A et al: Parenchymal sparing surgery in patients with hereditary renal cell carcinoma: 10-year experience. J Urol 2001; 165: 777.
  214. Campbell SC, Novick AC, Belldegrun A et al: Guideline for management of the clinical T1 renal mass. J Urol 2009; 182: 1271.
  215. Uzzo RG, Novick AC: Nephron sparing surgery for renal tumors: indications, techniques and outcomes. J Urol 2001; 166: 6.
  216. Nguyen CT, Campbell SC, Novick AC: Choice of operation for clinically localized renal tumor. Urol Clin North Am 2008; 35: 645.
  217. Fergany AF, Saad IR, Woo L et al: Open partial nephrectomy for tumor in a solitary kidney: experience with 400 cases. J Urol 2006; 175: 1630.
  218. Ghavamian R, Cheville JC, Lohse CM et al: Renal cell carcinoma in the solitary kidney: an analysis of complications and outcome after nephron sparing surgery. J Urol 2002; 168: 454.
  219. Lane BR, Demirjian S, Derweesh IH et al: Survival and functional stability in chronic kidney disease due to surgical removal of nephrons: importance of the new baseline glomerular filtration rate. Eur Urol 2015; 68: 996.
  220. Demirjian S, Lane BR, Derweesh IH et al: Chronic kidney disease due to surgical removal of nephrons: relative rates of progression and survival. J Urol 2014; 192: 1057.
  221. Lane BR, Russo P, Uzzo RG et al: Comparison of cold and warm ischemia during partial nephrectomy in 660 solitary kidneys reveals predominant role of nonmodifiable factors in determining ultimate renal function. J Urol 2011; 185: 421.
  222. Stevens PE, Levin A: Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med 2013; 158: 825.
  223. Scosyrev E, Messing EM, Sylvester R et al: Renal function after nephron-sparing surgery versus radical nephrectomy: results from EORTC randomized trial 30904. Eur Urol 2014; 65: 372.
  224. Antonelli A, Mari A, Longo N et al: Role of clinical and surgical factors for the prediction of immediate, early and late functional results, and its relationship with cardiovascular outcome after partial nephrectomy: Results from the prospective multicenter record 1 project. J Urol 2018; 199: 927.
  225. Rosen DC, Kannappan M, Kim Y et al: The impact of obesity in patients undergoing robotic partial nephrectomy. J Endourol 2019; 33: 431.
  226. Isharwal S, Ye W, Wang A et al: Impact of comorbidities on functional recovery from partial nephrectomy. J Urol 2018; 199: 1433.
  227. Volpe A, Blute ML, Ficarra V et al: Renal ischemia and function after partial nephrectomy: a collaborative review of the literature. Eur Urol 2015; 68: 61.
  228. Mir MC, Ercole C, Takagi T et al: Decline in renal function after partial nephrectomy: etiology and prevention. J Urol 2015; 193: 1889.
  229. Thompson RH, Lane BR, Lohse CM et al: Renal function after partial nephrectomy: effect of warm ischemia relative to quantity and quality of preserved kidney. Urology 2012; 79: 356.
  230. Wu J, Suk-Ouichai C, Dong W et al: Vascularized parenchymal mass preserved with partial nephrectomy: Functional impact and predictive factors. Eur Urol Oncol 2019; 2: 97.
  231. Dong W, Wu J, Suk-Ouichai C et al: Ischemia and functional recovery from partial nephrectomy: Refined perspectives. Eur Urol Focus 2018; 4: 572.
  232. Takagi T, Mir MC, Campbell RA et al: Predictors of precision of excision and reconstruction in partial nephrectomy. J Urol 2014; 192: 30.
  233. Zhang Z, Zhao J, Velet L et al: Functional recovery from extended warm ischemia associated with partial nephrectomy. Urology 2016; 87: 106.
  234. Zhang Z, Zhao J, Dong W et al: Acute kidney injury after partial nephrectomy: role of parenchymal mass reduction and ischemia and impact on subsequent functional recovery. Eur Urol 2016; 69: 745.
  235. Bravi CA, Larcher A, Capitanio U et al: Perioperative outcomes of open, laparoscopic, and robotic partial nephrectomy: A prospective multicenter observational study (the record 2 project). Eur Urol Focus 2019;
  236. Satkunasivam R, Tsai S, Syan S et al: Robotic unclamped "minimal-margin" partial nephrectomy: ongoing refinement of the anatomic zero-ischemia concept. Eur Urol 2015; 68: 705.
  237. Hung AJ, Cai J, Simmons MN et al: "Trifecta" in partial nephrectomy. J Urol 2013; 189: 36.
  238. Ng CK, Gill IS, Patil MB et al: Anatomic renal artery branch microdissection to facilitate zero-ischemia partial nephrectomy. Eur Urol 2012; 61: 67.
  239. Simone G, Gill IS, Mottrie A et al: Indications, techniques, outcomes, and limitations for minimally ischemic and off-clamp partial nephrectomy: A systematic review of the literature. Eur Urol 2015; 68: 632.
  240. Cacciamani GE, Medina LG, Gill TS et al: Impact of renal hilar control on outcomes of robotic partial nephrectomy: Systematic review and cumulative meta-analysis. Eur Urol Focus 2019; 5: 619.
  241. Khalifeh A, Kaouk JH, Bhayani S et al: Positive surgical margins in robot-assisted partial nephrectomy: a multi-institutional analysis of oncologic outcomes (leave no tumor behind). J Urol 2013; 190: 1674.
  242. Shah PH, Moreira DM, Okhunov Z et al: Positive surgical margins increase risk of recurrence after partial nephrectomy for high risk renal tumors. J Urol 2016; 196: 327.
  243. Walther MM, Thompson N, Linehan W: Enucleation procedures in patients with multiple hereditary renal tumors. World J Urol 1995; 13: 248.
  244. Minervini A, Tuccio A, Masieri L et al: Endoscopic robot-assisted simple enucleation (ERASE) for clinical T1 renal masses: description of the technique and early postoperative results. Surg Endosc 2015; 29: 1241.
  245. Laryngakis NA, Guzzo TJ: Tumor enucleation for small renal masses. Curr Opin Urol 2012; 22: 365.
  246. Minervini A, Campi R, Lane BR et al: Impact of resection technique on perioperative outcomes and surgical margins after partial nephrectomy for localized renal masses: A prospective multicenter study. J Urol 2020; 203: 496.
  247. Lu Q, Ji C, Zhao X et al: Histopathologic analysis of tumor bed and peritumoral pseudocapsule after in vitro tumor enucleation on radical nephrectomy specimen for clinical t1b renal cell carcinoma. Urol Oncol 2017; 35: 603.e15.
  248. Gupta GN, Boris RS, Zhang Z et al: Tumor enucleation for sporadic localized kidney cancer: opposing views. J Urol 2015; 194: 635.
  249. Jacob JM, Williamson SR, Gondim DD et al: Characteristics of the peritumoral pseudocapsule vary predictably with histologic subtype of T1 renal neoplasms. Urology 2015; 86: 956.
  250. Li QL, Guan HW, Zhang QP et al: Optimal margin in nephron-sparing surgery for renal cell carcinoma 4 cm or less. Eur Urol 2003; 44: 448.
  251. Xi W, Wang J, Liu L et al: Evaluation of tumor pseudocapsule status and its prognostic significance in renal cell carcinoma. J Urol 2018; 199: 915.
  252. Minervini A, di Cristofano C, Lapini A et al: Histopathologic analysis of peritumoral pseudocapsule and surgical margin status after tumor enucleation for renal cell carcinoma. Eur Urol 2009; 55: 1410.
  253. Bansal RK, Tanguay S, Finelli A et al: Positive surgical margins during partial nephrectomy for renal cell carcinoma: Results from canadian kidney cancer information system (ckcis) collaborative. Can Urol Assoc J 2017; 11: 182.
  254. Dash A, Vickers AJ, Schachter LR et al: Comparison of outcomes in elective partial vs radical nephrectomy for clear cell renal cell carcinoma of 4-7 cm. BJU Int 2006; 97: 939.
  255. Simmons MN, Weight CJ, Gill IS et al: Laparoscopic radical versus partial nephrectomy for tumors > 4 cm: intermediate-term oncologic and functional outcomes. Urology 2009; 73: 1077.
  256. Crépel M, Jeldres C, Perrotte P et al: Nephron-sparing surgery is equally effective to radical nephrectomy for T1BN0M0 renal cell carcinoma: a population-based assessment. Urology 2010; 75: 271.
  257. Badalato GM, Kates M, Wisnivesky JP et al: Survival after partial and radical nephrectomy for the treatment of stage T1bN0M0 renal cell carcinoma (RCC) in the USA: a propensity scoring approach. BJU Int 2012; 109: 1457.
  258. Kim SP, Thompson RH, Boorjian SA et al: Comparative effectiveness for survival and renal function of partial and radical nephrectomy for localized renal tumors: a systematic review and meta-analysis. J Urol 2012; 188: 51.
  259. Weight CJ, Miller DC, Campbell SC et al: The management of a cT1b renal tumor in the presence of a normal contralateral kidney. J Urol 2013; 189: 1198.
  260. Lee HJ, Liss MA, Derweesh IH: Outcomes of partial nephrectomy for clinical T1b and T2 renal tumors. Curr Opin Urol 2014; 24: 448.
  261. Tomaszewski JJ, Smaldone MC, Uzzo RG et al: Is radical nephrectomy a legitimate therapeutic option in patients with renal masses amenable to nephron-sparing surgery?. BJU Int 2015; 115: 357.
  262. Crispen PL, Boorjian SA, Lohse CM et al: Outcomes following partial nephrectomy by tumor size. J Urol 2008; 180: 1912.
  263. Simmons MN, Herts B, Campbell SC et al: Image-based approaches to the diagnosis and treatment of renal masses. AUA Update Series 2007; 26: 381.
  264. Ishigami K, Leite LV, Pakalniskis MG et al: Tumor grade of clear cell renal cell carcinoma assessed by contrast-enhanced computed tomography. Springer plus 2014; 26: 694.
  265. Crane A, Suk-Ouichai C, Campbell JA et al: Imprudent utilization of partial nephrectomy. Urology 2018; 117: 22.
  266. Simhan J, Smaldone MC, Tsai KJ et al: Objective measures of renal mass anatomic complexity predict rates of major complications following partial nephrectomy. Eur Urol 2011; 60: 724.
  267. Potretzke AM, Knight BA, Zargar H et al: Urinary fistula after robot-assisted partial nephrectomy: a multicentre analysis of 1791 patients. BJU Int 2016; 117: 131.
  268. Van Poppel H, Da Pozzao L, Albrect W et al: A prospective, randomized EORTC intergroup phase 3 study comparing the oncologic outcome of elective nephron-sparing surgery and radical nephrectomy for low-stage renal cell carcinoma. Eur Urol 2011; 59: 543.
  269. Breau RH, Kapoor A, Nash DM et al: Partial vs. Radical nephrectomy and the risk of all-cause mortality, cardiovascular, and nephrological outcomes. Can Urol Assoc J 2020; 14: 337.
  270. Streja E, Kalantar-Zadeh K, Molnar MZ et al: Radical versus partial nephrectomy, chronic kidney disease progression and mortality in us veterans. Nephrol Dial Transplant 2018; 33: 95.
  271. Kim SP, Campbell SC, Gill I et al: Collaborative review of risk benefit trade-offs between partial and radical nephrectomy in the management of anatomically complex renal masses. Eur Urol 2017; 72: 64.
  272. Ye Y, Tanaka H, Wang Y et al: Split renal function in patients with renal masses: Utility of parenchymal volume analysis vs nuclear renal scans. BJU Int 2020; 125: 686.
  273. Capitanio U, Becker F, Blute ML et al: Lymph node dissection in renal cell carcinoma. Eur Urol 2011; 60: 1212.
  274. Bekema HJ, MacLennan S, Imamura M et al: Systematic review of adrenalectomy and lymph node dissection in locally advanced renal cell carcinoma. Eur Urol 2013; 64: 799.
  275. Blom JH, van Poppel H, Maréchal JM et al: Radical nephrectomy with and without lymph-node dissection: final results of European Organization for Research and Treatment of Cancer (EORTC) randomized phase 3 trial 30881. Eur Urol 2009; 55: 28.
  276. Gershman B, Moreira D, Thompson H et al: Radical nephrectomy with or without lymph node dissection for nonmetastatic renal cell carcinoma: a propensity score-based analysis. Eur Urol 2017; 71: 560.
  277. Bhindi B, Wallis CJD, Boorjian SA et al: The role of lymph node dissection in the management of renal cell carcinoma: A systematic review and meta-analysis. BJU Int 2018; 121: 684.
  278. John NT, Blum KA and Hakimi AA: Role of lymph node dissection in renal cell cancer. Urol Oncol 2019; 37: 187.
  279. Gershman B, Thompson RH, Boorjian SA et al: Radical nephrectomy with or without lymph node dissection for high risk nonmetastatic renal cell carcinoma: A multi-institutional analysis. J Urol 2018; 199: 1143.
  280. Weight CJ, Mulders PF, Pantuck AJ et al: The role of adrenalectomy in renal cancer. Euro Urology Focus 2016; 1: 251.
  281. Lane BR, Tiong HY, Campbell SC et al: Management of the adrenal gland during partial nephrectomy. J Urol 2009; 181: 2430.
  282. Bratslavsky G, Linehan WM: Surgery: Routine adrenalectomy in renal cancer--an antiquated practice. Nat Rev Urol 2011; 8: 534.
  283. Kerbl K, Clayman RV, McDougall EM et al: Laparoscopic nephrectomy: the Washington University experience. Br J Urol 1994; 73: 231.
  284. Dunn MD, Portis AJ, Shalhav AL et al: Laparoscopic versus open radical nephrectomy: a 9-year experience. J Urol 2000; 164: 1153.
  285. Tan H-J, Wolf JS, Ye Z et al: Population-level comparative effectiveness of laparoscopic versus open radical nephrectomy for patients with kidney cancer. Cancer 2011; 117: 4184.
  286. Mullins JK, Feng T, Pierorazio PM et al: Comparative analysis of minimally invasive partial nephrectomy techniques in the treatment of localized renal tumors. Urology 2012; 80: 316.
  287. Xia L, Wang X, Xu T et al: Systematic review and meta-analysis of comparative studies reporting perioperative outcomes of robot-assisted partial nephrectomy versus open partial nephrectomy. J Endourol 2016; Epub ahead of print.
  288. Wu Z, Li M, Liu B et al: Robotic versus open partial nephrectomy: a systematic review and meta-analysis. PloS one 2014; 9: e94878.
  289. Luciani LG, Porpiglia F, Cai T et al: Operative safety and oncologic outcome of laparoscopic radical nephrectomy for renal cell carcinoma >7 cm: a multicenter study of 222 patients. Urology 2013; 81: 1239.
  290. Hillyer SP, Bhayani SB, Allaf ME et al: Robotic partial nephrectomy for solitary kidney: a multi-institutional analysis. Urology 2013; 81: 93.
  291. Pierorazio PM, Hyams ES, Lin BM et al: Laparoscopic radical nephrectomy for large renal masses: critical assessment of perioperative and oncologic outcomes of stage T2a and T2b tumors. Urology 2012; 79: 570.
  292. Abaza R, Shabsigh A, Castle E et al: Multi-Institutional experience with robotic nephrectomy with inferior vena cava tumor thrombectomy. J Urol 2016; 195: 865.
  293. Kundavaram C, Abreu AL, Chopra S et al. Advances in robotic vena cava tumor thrombectomy: intracaval balloon occlusion, patch grafting, and vena cavoscopy. Eur Urol 2016; 70: 884.
  294. Auffenberg GB, Curry M, Gennarelli R et al: Comparison of cancer specific outcomes following minimally invasive and open surgical resection of early stage kidney cancer from a national cancer registry. J Urol 2020; 203: 1094.
  295. Peyronnet B, Seisen T, Oger E et al: Comparison of 1800 robotic and open partial nephrectomies for renal tumors. Ann Surg Oncol 2016; 23: 4277.
  296. Cacciamani GE, Medina LG, Gill T et al: Impact of surgical factors on robotic partial nephrectomy outcomes: Comprehensive systematic review and meta-analysis. J Urol 2018; 200: 258.
  297. Acar C, Bilen C, Bayazit Y et al: Quality of life survey following laparoscopic and open radical nephrectomy. Urol J 2014; 11: 1944.
  298. Hyams E, Pierorazio P, Mullins JK et al: A comparative cost analysis of robot-assisted versus traditional laparoscopic partial nephrectomy. J Endourol 2012; 26: 843.
  299. Laydner H, Isac W, Autorino R et al: Single institutional cost analysis of 325 robotic, laparoscopic, and open partial nephrectomies. Urology 2013; 81: 533.
  300. Hughes D, Camp C, O'Hara J et al: Health resource use after robot-assisted surgery vs open and conventional laparoscopic techniques in oncology: analysis of English secondary care data for radical prostatectomy and partial nephrectomy. BJU Int 2016; 117: 940.
  301. Castle SM, Gorbatiy V, Avallone MA et al: Cost comparison of nephron-sparing treatments for cT1a renal masses. Urol Oncol 2013; 31: 1327.
  302. Mano R, Schulman A, Hakimi AA et al: Cost comparison of open and robotic partial nephrectomy using a short postoperative pathway. Urology 2015; 85: 596.
  303. Algaba F, Delahunt B, Berney DM et al: Handling and reporting of nephrectomy specimens for adult renal tumours: a survey by the European Network of Uropathology. J Clin Pathol 2012; 65: 106.
  304. Srigley JR, Amin MB, Campbell SC et al: Protocol for the examination of specimens from patients with invasive carcinoma of renal tubular origin. College of American Pathologists 2017; http://www.cap.org/web/oracle/webcenter/portalapp/pagehierarchy/cancer_protocol_templates.jspx?_adf.ctrl-state=15bj4qi2ja_4&_afrLoop=1300546090297309#!.
  305. Choueiri TK and Motzer RJ: Systemic therapy for metastatic renal-cell carcinoma. N Engl J Med 2017; 376: 354.
  306. Monteiro FSM, Soares A, Debiasi M et al: First-line treatment of metastatic renal cell carcinoma in the immuno-oncology era: Systematic review and network meta-analysis. Clin Genitourin Cancer 2020; 18: 244.
  307. Chowdhury N and Drake CG: Kidney cancer: An overview of current therapeutic approaches. Urol Clin North Am 2020; 47: 419.
  308. Heng DY, Xie W, Regan MM et al: External validation and comparison with other models of the international metastatic renal-cell carcinoma database consortium prognostic model: A population-based study. Lancet Oncol 2013; 14: 141.
  309. Wood E, Donin N and Shuch B: Adjuvant therapy for localized high-risk renal cell carcinoma. Urol Clin North Am 2020; 47: 345.
  310. Correa AF, Jegede O, Haas NB et al: Predicting renal cancer recurrence: Defining limitations of existing prognostic models with prospective trial-based validation. J Clin Oncol 2019; 37: 2062.
  311. Agrawal S, Haas NB, Bagheri M et al: Eligibility and radiologic assessment for adjuvant clinical trials in kidney cancer. JAMA Oncol 2019;
  312. Administration FaD: Fda approves sunitinib malate for adjuvant treatment of renal cell carcinoma. 2018. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-sunitinib-malate-adjuvant-treatment-renal-cell-carcinoma. 01/27/2021.
  313. Ravaud A, Motzer RJ, Pandha HS et al: Adjuvant sunitinib in high-risk renal-cell carcinoma after nephrectomy. N Engl J Med 2016; 375: 2246.
  314. Olweny EO, Park SK, Tan YK et al: Radiofrequency ablation versus partial nephrectomy in patients with solitary clinical t1a renal cell carcinoma: Comparable oncologic outcomes at a minimum of 5 years of follow-up. Eur Urol 2012; 61: 1156.
  315. Thompson RH, Atwell T, Schmit G et al: Comparison of partial nephrectomy and percutaneous ablation for ct1 renal masses. Eur Urol 2015; 67: 252.
  316. Youn CS, Park JM, Lee JY et al: Comparison of laparoscopic radiofrequency ablation and open partial nephrectomy in patients with a small renal mass. Korean J Urol 2013; 54: 603.
  317. Tanagho YS, Roytman TM, Bhayani SB et al: Laparoscopic cryoablation of renal masses: single-center long-term experience. Urology 2012; 80: 307.
  318. Gervais DA, McGovern FJ, Arellano RS et al: Radiofrequency ablation of renal cell carcinoma: part 1, Indications, results, and role in patient management over a 6-year period and ablation of 100 tumors. AJR Am J Roentgenol 2005; 185: 64.
  319. Best SL, Park SK, Youssef RF et al: Long-term outcomes of renal tumor radio frequency ablation stratified by tumor diameter: size matters. J Urol 2012; 187: 1183.
  320. Sidana A, Aggarwal P, Feng Z et al: Complications of renal cryoablation: a single center experience. J Urol 2010; 184: 42.
  321. Lehman DS, Hruby GW, Phillips CK et al: First Prize (tie): Laparoscopic renal cryoablation: efficacy and complications for larger renal masses. J Endourol 2008; 22: 1123.
  322. Atwell TD, Carter RE, Schmit GD et al: Complications following 573 percutaneous renal radiofrequency and cryoablation procedures. J Vasc Interv Radiol 2012; 23: 48.
  323. Hinshaw JL, Shadid AM, Nakada SY et al: Comparison of percutaneous and laparoscopic cryoablation for the treatment of solid renal masses. AJR Am J Roentgenol 2008; 191: 1159.
  324. Leveillee RJ, Castle SM, Gorbatiy V et al: Oncologic outcomes using real-time peripheral thermometry-guided radiofrequency ablation of small renal masses. J Endourol 2013; 27: 480.
  325. Ramirez D, Ma YB, Bedir S et al: Laparoscopic radiofrequency ablation of small renal tumors: long-term oncologic outcomes. J Endourol 2014; 28: 330.
  326. Finley DS, Beck S, Box G et al: Percutaneous and laparoscopic cryoablation of small renal masses. J Urol 2008; 180: 492.
  327. Bandi G, Hedican S, Moon T et al: Comparison of postoperative pain, convalescence, and patient satisfaction after laparoscopic and percutaneous ablation of small renal masses. J Endourol 2008; 22: 963.
  328. Hegarty NJ, Gill IS, Desai MM et al: Probe-ablative nephron-sparing surgery: cryoablation versus radiofrequency ablation. Urology 2006; 68: 7.
  329. Atwell TD, Schmit GD, Boorjian SA et al: Percutaneous ablation of renal masses measuring 3.0 cm and smaller: comparative local control and complications after radiofrequency ablation and cryoablation. AJR 2013; 200: 461.
  330. Kunkle DA, Uzzo RG: Cryoablation or radiofrequency ablation of the small renal mass. Cancer 2008; 113: 2671.
  331. Hong K, Georgiades C: Radiofrequency ablation: mechanism of action and devices. J Vasc Interv Radiol 2010; 21:S179.
  332. Ahmed M, Brace CL, Lee FT et al: Principles of and advances in percutaneous ablation. Radiology 2011; 258: 351.
  333. Modabber M, Martin J, Athreya S: Thermal versus impedance-based ablation of renal cell carcinoma: a meta-analysis. Cardiovasc Intervent Radiol 2014; 37: 176.
  334. Littrup PJ, Jallad B, Vorugu V et al: Lethal isotherms of cryoablation in a phantom study: Effects of heat load, probe size and number. J Vasc Interv Radiol 2009; 20: 1343.
  335. Littrup PJ, Ahmed A, Aoun HD et al: CT-guided percutaneous cryotherapy of renal masses. J Vasc Interv Radiol 2007; 18: 383.
  336. Woolley ML, Schulsinger DA, Durand DB et al: Effect of freezing parameters (freeze cycle and thaw process) on tissue destruction following renal cryoablation. J Endourol 2002; 16: 519.
  337. Young JL , McCormick DW, Kolla SB et al: Are multiple cryoprobes additive or synergistic in renal cryotherapy? Urology 2012; 79: 484.
  338. Tan YK, Best SL, Olweny EO et al: Radiofrequency ablation of the incidental benign small renal tumor: outcomes and follow-up protocol. Urology 2012; 79: 827.
  339. Lay AH, Faddegon S, Olweny EO et al: Oncologic efficacy of radiofrequency ablation for small renal masses: histologic subtype matters. J Urol 2016; 196: 41.
  340. Widdershoven CV, Aarts BM, Zondervan PJ et al: Renal biopsies performed before versus during ablation of t1 renal tumors: Implications for prevention of overtreatment and follow-up. Abdom Radiol (NY) 2020;
  341. Wells SA, Wong VK, Wittmann TA et al: Renal mass biopsy and thermal ablation: Should biopsy be performed before or during the ablation procedure? Abdom Radiol (NY) 2017; 42: 1773.
  342. Jimenez J, Zhang Z, Zhao J et al: Surgical salvage of thermal ablation failure for renal cell carcinoma. J Urol 2016; 195: 594.
  343. Karam JA, Wood CG, Compton ZR et al: Salvage surgery after energy ablation for renal masses. BJU Int 2015; 115: 74.
  344. Nuttall M, van der Meulen J, Emberton M: Charlson scores based on ICD-10 administrative data were valid in assessing comorbidity in patients undergoing urological cancer surgery. J Clin Epidemiol 2006; 59: 265.
  345. Lascano D, Pak JS, Kates M et al: Validation of a frailty index in patients undergoing curative surgery for urologic malignancy and comparison with other risk stratification tools. Urol Oncol 2015; 33: 426.
  346. Klatte T, Ficarra V, Gratzke C et al: A literature review of renal surgical anatomy and surgical strategies for partial nephrectomy. Eur Urol 2015; 68: 980.
  347. Bilimoria KY, Liu Y, Paruch JL et al: Development and evaluation of the universal ACS NSQIP surgical risk calculator: a decision aid and informed consent tool for patients and surgeons. J Am Coll Surg 2013; 217: 833.
  348. Charles C, Gafni A, Whelan T: Decision-making in the physician-patient encounter: revisiting the shared treatment decision-making model. Soc Sci Med 1999; 49: 651.
  349. Crispen PL, Viterbo R, Boorjian SA et al: Natural history, growth kinetics, and outcomes of untreated clinically localized renal tumors under active surveillance. Cancer 2009; 115: 2844.
  350. Kunkle DA, Crispen PL, Chen DY et al: Enhancing renal masses with zero net growth during active surveillance. J Urol 2007; 177: 849.
  351. Crispen PL, Viterbo R, Fox EB et al: Delayed intervention of sporadic renal masses undergoing active surveillance. Cancer 2008; 112: 1051.
  352. Gupta M, Blute ML, Jr., Su LM et al: Delayed intervention of small renal masses on active surveillance. J Kidney Cancer VHL 2017; 4: 24.
  353. Whelan EA, Mason RJ, Himmelman JG et al: Extended duration of active surveillance of small renal masses: A prospective cohort study. J Urol 2019; 202: 57.
  354. Campi R, Sessa F, Corti F et al: Triggers for delayed intervention in patients with small renal masses undergoing active surveillance: A systematic review. Minerva Urol Nefrol 2020; 72: 389.
  355. Mehrazin R, Smaldone MC, Egleston B et al: Is anatomic complexity associated with renal tumor growth kinetics under active surveillance? Urol Oncol 2015; 33: 167.
  356. Lane BR, Abouassaly R, Gao T et al: Active treatment of localized renal tumors may not impact overall survival in patients aged 75 years or older. Cancer 2010; 116: 3119.
  357. Kawaguchi S, Fernandes KA, Finelli A et al: Most renal oncocytomas appear to grow: Observations of tumor kinetics with active surveillance. J Urol 2011; 186: 1218.
  358. Uzosike AC, Patel HD, Alam R et al: Growth kinetics of small renal masses on active surveillance: Variability and results from the dissrm registry. J Urol 2018; 199: 641.
  359. Finelli A, Cheung DC, Al-Matar A et al: Small renal mass surveillance: Histology-specific growth rates in a biopsy-characterized cohort. Eur Urol 2020; 78: 460.
  360. Richard PO, Jewett MA, Bhatt JR et al: Active surveillance for renal neoplasms with oncocytic features is safe. J Urol 2016; 195: 581.
  361. Crispen PL, Wong YN, Greenberg RE et al: Predicting growth of solid renal masses under active surveillance. Urol Oncol 2008; 26: 555.
  362. Surgeons ACo: Acs risk calculator 2021. https://riskcalculator.facs.org/RiskCalculator/. 01/27/2021.
  363. Motzer RJ, Bacik J, Mariani T et al: Treatment outcome and survival associated with metastatic renal cell carcinoma of non-clear-cell histology. J Clin Oncol 2002; 20: 2376.
  364. Heng DY, Xie W, Regan MM 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 2009; 27: 5794.
  365. Lee SE, Byun SS, Han JH et al: Prognostic significance of common preoperative laboratory variables in clear cell renal cell carcinoma. BJU Int 2006; 98: 1228.
  366. Bos SD, Piers DA and Mensink HJ: Routine bone scan and serum alkaline phosphatase for staging in patients with renal cell carcinoma is not cost-effective. Eur J Cancer 1995; 31a: 2422.
  367. Kriteman L and Sanders WH: Normal alkaline phosphatase levels in patients with bone metastases due to renal cell carcinoma. Urology 1998; 51: 397.
  368. Grünwald V, Eberhardt B, Bex A et al: An interdisciplinary consensus on the management of bone metastases from renal cell carcinoma. Nat Rev Urol 2018; 15: 511.
  369. Koga S, Tsuda S, Nishikido M et al: The diagnostic value of bone scan in patients with renal cell carcinoma. J Urol 2001; 166: 2126.
  370. Young RJ, Sills AK, Brem S et al: Neuroimaging of metastatic brain disease. Neurosurgery 2005; 57: S10.
  371. Brem S and Panattil JG: An era of rapid advancement: Diagnosis and treatment of metastatic brain cancer. Neurosurgery 2005; 57: S5.
  372. Suarez-Sarmiento A, Jr., Nguyen KA, Syed JS et al: Brain metastasis from renal-cell carcinoma: An institutional study. Clin Genitourin Cancer 2019; 17: e1163.
  373. Liu Y: The place of fdg pet/ct in renal cell carcinoma: Value and limitations. Front Oncol 2016; 6: 201.
  374. Fuccio C, Ceci F, Castellucci P et al: Restaging clear cell renal carcinoma with 18f-fdg pet/ct. Clin Nucl Med 2014; 39: e320.
  375. Alongi P, Picchio M, Zattoni F et al: Recurrent renal cell carcinoma: Clinical and prognostic value of fdg pet/ct. Eur J Nucl Med Mol Imaging 2016; 43: 464.
  376. Wang HY, Ding HJ, Chen JH et al: Meta-analysis of the diagnostic performance of [18f]fdg-pet and pet/ct in renal cell carcinoma. Cancer Imaging 2012; 12: 464.
  377. University R: Zirconium-89-girentuximab pet/ct imaging in renal cell carcinoma. 2016. https://www.clinicaltrials.gov/ct2/show/NCT02883153. 02/04/2021.
  378. Psutka SP and Master VA: Role of metastasis-directed treatment in kidney cancer. Cancer 2018; 124: 3641.
  379. Dabestani S, Beisland C, Stewart GD et al: Long-term outcomes of follow-up for initially localised clear cell renal cell carcinoma: Recur database analysis. Eur Urol Focus 2019; 5: 857.
  380. Borregales LD, Kim DY, Staller AL et al: Prognosticators and outcomes of patients with renal cell carcinoma and adjacent organ invasion treated with radical nephrectomy. Urol Oncol 2016; 34: 237.e19.
  381. Yu KJ, Keskin SK, Meissner MA et al: Renal cell carcinoma and pathologic nodal disease: Implications for american joint committee on cancer staging. Cancer 2018; 124: 4023.
  382. Merrill MM, Wood CG, Tannir NM et al: Clinically nonmetastatic renal cell carcinoma with sarcomatoid dedifferentiation: Natural history and outcomes after surgical resection with curative intent. Urol Oncol 2015; 33: 166.e21.
  383. Zhang BY, Thompson RH, Lohse CM et al: A novel prognostic model for patients with sarcomatoid renal cell carcinoma. BJU Int 2015; 115: 405.
  384. Merrill SB, Sohl BS, Hamirani A et al: Capturing renal cell carcinoma recurrences when asymptomatic improves patient survival. Clin Genitourin Cancer 2019; 17: 132.
  385. Thomas AZ, Adibi M, Borregales LD et al: Surgical management of local retroperitoneal recurrence of renal cell carcinoma after radical nephrectomy. J Urol 2015; 194: 316.
  386. Beisland C, Guðbrandsdottir G, Reisæter LA et al: A prospective risk-stratified follow-up programme for radically treated renal cell carcinoma patients: Evaluation after eight years of clinical use. World J Urol 2016; 34: 1087.
  387. Donat SM, Diaz M, Bishoff JT et al: Follow-up for clinically localized renal neoplasms: Aua guideline. J Urol 2013; 190: 407.
  388. Levy DA, Slaton JW, Swanson DA et al: Stage specific guidelines for surveillance after radical nephrectomy for local renal cell carcinoma. J Urol 1998; 159: 1163.
  389. Karam JA and Wood CG: The role of surgery in advanced renal cell carcinoma: Cytoreductive nephrectomy and metastasectomy. Hematol Oncol Clin North Am 2011; 25: 753.
  390. Matsumoto ED, Watumull L, Johnson DB et al: The radiographic evolution of radio frequency ablated renal tumors. J Urol 2004; 172: 45.
  391. Ganz PA, Hassett MJ, Miller DC: Challenges and opportunities in delivering high-quality cancer care: A 2016 update. Am Soc Clin Oncol Educ Book 2016; 35: e294.
  392. Peabody H, Patel A, Johnson A et al: Development of a novel scoring system quantifies opportunities to reduce surgery for benign renal neoplasms: A retrospective quality improvement analysis within the music-kidney collaborative. J Urol 2020; 204: 1160.
  393. Lubner MG: Radiomics and artificial intelligence for renal mass characterization. Radiol Clin North Am 2020; 58: 995.
  394. Farber NJ, Wu Y, Zou L et al: Challenges in RCC imaging: renal insufficiency, post-operative surveillance, and the role of radiomics. Kidney Cancer J 2015; 13: 84.
  395. Gorin MA, Rowe SP, Allaf ME: Nuclear imaging of renal tumours: a step towards improved risk stratification. Nat Rev Urol 2015; 12: 445.
  396. Takahashi M, Rhodes DR, Furge KA et al: Gene expression profiling of clear cell renal cell carcinoma: gene identification and prognostic classification. Proc Natl Acad Sci USA 2001; 98: 9754.
  397. Durinck S, Stawiski EW, Pavia-Jimenez A et al: Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet 2015; 47: 13.
  398. Linehan WM and Ricketts CJ: The cancer genome atlas of renal cell carcinoma: Findings and clinical implications. Nat Rev Urol 2019; 16: 539.
  399. Linehan WM, Spellman PT, Ricketts CJ et al: Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med 2016; 374: 135.
  400. Cancer Genome Atlas Research Network: Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013; 499: 43.
  401. Cancer Genome Atlas Research Network, Linehan WM, Spellman PT et al: Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med 2016; 374: 135.
  402. Davis CF, Ricketts CJ, Wang M et al: The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 2014; 26: 319.
  403. Nel I, Gauler TC, Bublitz K et al: Circulating tumor cell composition in renal cell carcinoma. PLoS One 2016; 11: e0153018.
  404. Ellinger J, Gevensleben H, Muller SC et al: The emerging role of non-coding circulating RNA as a biomarker in renal cell carcinoma. Expert Rev Mol Diagn 2016: 16: 1059.
  405. Clark DJ, Dhanasekaran SM, Petralia F et al: Integrated proteogenomic characterization of clear cell renal cell carcinoma. Cell 2019; 179: 964.
  406. Gorin MA, Verdone JE, van der Toom E et al: Circulating tumour cells as biomarkers of prostate, bladder, and kidney cancer. Nat Rev Urol 2017; 14: 90.
  407. Lin GA, Fagerlin A: Shared decision making: state of the science. Circ Cardiovasc Qual Outcomes 2014; 7: 328.
  408. Witteman HO, Dansokho SC, Colquhoun H et al: User-centered design and the development of patient decision aids: protocol for a systematic review. Syst Rev 2015; 4: 11.
  409. Suk-Ouichai C, Tanaka H, Wang Y et al: Renal cancer surgery in patients without preexisting chronic kidney disease-is there a survival benefit for partial nephrectomy? J Urol 2019; 201: 1088.
  410. Gershman B, Thompson RH, Boorjian SA et al: Radical versus partial nephrectomy for ct1 renal cell carcinoma. Eur Urol 2018; 74: 825.
  411. Campbell SP, Song DY, Pierorazio PM et al: Stereotactic ablative radiotherapy for the treatment of clinically localized renal cell carcinoma. J Oncol 2015; 2015: 547143.
  412. Maloney E, Hwang JH: Emerging HIFU applications in cancer therapy. Int J Hyperthermia 2015; 31: 302.
  413. Lele PP, Pierce AD: The thermal hypothesis of the mechanism of ultrasonic focal destruction in organized tissues. Interactions of ultrasound and biological tissues - workshop proceedings. U.S.D.H.E.W. Publication (FDA); 1972.
  414. Vallancien G, Chartier-Kastler E, Harouni M et al: Focused extracorporeal pyrotherapy: experimental study and feasibility in man. Semin Urol 1993; 11: 7.
  415. Kohrmann KU, Michel MS, Gaa J et al: High intensity focused ultrasound as noninvasive therapy for multilocal renal cell carcinoma: case study and review of the literature. J Urol 2002; 167: 2397.
  416. Marberger M, Schatzl G, Cranston D et al: Extracorporeal ablation of renal tumours with high-intensity focused ultrasound. BJU Int 2005; 95: Suppl 2:52.
  417. Hacker A, Michel MS, Marlinghaus E et al: Extracorporeally induced ablation of renal tissue by high-intensity focused ultrasound. BJU Int 2006; 97: 779.
  418. Moore C, Salas N, Zaias J et al: Effects of microwave ablation of the kidney. J Endourol 2010; 24: 439.
  419. Stafford RJ, Fuentes D, Elliott AA et al: Laser-induced thermal therapy for tumor ablation. Crit Rev Biomed Eng 2010; 38: 79.
  420. Gettman MT, Lotan Y, Lindberg G et al: Laparoscopic interstitial laser coagulation of renal tissue with and without hilar occlusion in the porcine model. J Endourol 2002; 16: 565.
  421. Williams JC, Swischuk PN, Morrison PM et al: Laser-induced thermotherapy of renal cell carcinoma in man: dosimetry, ultrasound, and histopathologic correlation. Proc. SPIE 2000.
  422. Rustin GJ, Mead GM, Stenning SP et al: Randomized trial of two or five computed tomography scans in the surveillance of patients with stage i nonseminomatous germ cell tumors of the testis: Medical research council trial te08, isrctn56475197--the national cancer research institute testis cancer clinical studies group. J Clin Oncol 2007; 25: 1310.
  423. Rosati G, Ambrosini G, Barni S et al: A randomized trial of intensive versus minimal surveillance of patients with resected dukes b2-c colorectal carcinoma. Ann Oncol 2016; 27: 274.
  424. Liu W, Mu S, Yao J et al: Analytical and clinical validation of a next-generation sequencing-based circulating tumor DNA (ctDNA) assay assures its clinical application. Ann Oncol 2017; 28: v449-v452.