Next Article in Journal
Role of a Structured Physical Activity Pathway in Improving Functional Disability, Pain and Quality of Life in a Case of Breast and Gynecological Cancer Survivorship
Next Article in Special Issue
Fast Tac Metabolizers at Risk—It is Time for a C/D Ratio Calculation
Previous Article in Journal
EGFR and KRAS Mutations in the Non-Tumoral Lung. Prognosis in Patients with Adenocarcinoma
Previous Article in Special Issue
Subarachnoid Hemorrhage in Hospitalized Renal Transplant Recipients with Autosomal Dominant Polycystic Kidney Disease: A Nationwide Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Incidence and Mortality of Renal Cell Carcinoma after Kidney Transplantation: A Meta-Analysis

by
Api Chewcharat
1,2,
Charat Thongprayoon
3,*,
Tarun Bathini
4,
Narothama Reddy Aeddula
5,
Boonphiphop Boonpheng
6,
Wisit Kaewput
7,
Kanramon Watthanasuntorn
8,
Ploypin Lertjitbanjong
8,
Konika Sharma
8,
Aldo Torres-Ortiz
9,
Napat Leeaphorn
10,
Michael A. Mao
11,
Nadeen J. Khoury
12 and
Wisit Cheungpasitporn
9
1
Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
2
Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10300, Thailand
3
Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA
4
Department of Internal Medicine, University of Arizona, Tucson, AZ 85721, USA
5
Division of Nephrology, Department of Medicine, Deaconess Health System, Evansville, IN 47747, USA
6
Department of Internal Medicine, East Tennessee State University, Johnson City, TN 37614, USA
7
Department of Military and Community Medicine, Phramongkutklao College of Medicine, Bangkok 10400, Thailand
8
Department of Internal Medicine, Bassett Medical Center, Cooperstown, NY 13326, USA
9
Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
10
Department of Nephrology, Department of Medicine, Saint Luke’s Health System, Kansas City, MO 64111, USA
11
Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL 32224, USA
12
Department of Nephrology, Department of Medicine, Henry Ford Hospital, Detroit, MI 48202, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2019, 8(4), 530; https://doi.org/10.3390/jcm8040530
Submission received: 31 March 2019 / Revised: 13 April 2019 / Accepted: 15 April 2019 / Published: 17 April 2019
(This article belongs to the Special Issue Recent Advances and Clinical Outcomes of Kidney Transplantation)

Abstract

:
Background: The incidence and mortality of renal cell carcinoma (RCC) after kidney transplantation (KTx) remain unclear. This study’s aims were (1) to investigate the pooled incidence/incidence trends, and (2) to assess the mortality/mortality trends in KTx patients with RCC. Methods: A literature search was conducted using the MEDLINE, EMBASE and Cochrane databases from inception through October 2018. Studies that reported the incidence or mortality of RCC among kidney transplant recipients were included. The pooled incidence and 95% CI were calculated using a random-effect model. The protocol for this meta-analysis is registered with PROSPERO; no. CRD42018108994. Results: A total of 22 observational studies with a total of 320,190 KTx patients were enrolled. Overall, the pooled estimated incidence of RCC after KTx was 0.7% (95% CI: 0.5–0.8%, I2 = 93%). While the pooled estimated incidence of de novo RCC in the native kidney was 0.7% (95% CI: 0.6–0.9%, I2 = 88%), the pooled estimated incidence of RCC in the allograft kidney was 0.2% (95% CI: 0.1–0.4%, I2 = 64%). The pooled estimated mortality rate in KTx recipients with RCC was 15.0% (95% CI: 7.4–28.1%, I2 = 80%) at a mean follow-up time of 42 months after RCC diagnosis. While meta-regression analysis showed a significant negative correlation between year of study and incidence of de novo RCC post-KTx (slopes = −0.05, p = 0.01), there were no significant correlations between the year of study and mortality of patients with RCC (p = 0.50). Egger’s regression asymmetry test was performed and showed no publication bias in all analyses. Conclusions: The overall estimated incidence of RCC after KTX was 0.7%. Although there has been a potential decrease in the incidence of RCC post-KTx, mortality in KTx patients with RCC has not decreased over time.

1. Introduction

Kidney transplantation (KTx) is the renal replacement therapy of choice for the majority of patients with end-stage renal disease (ESRD) and it significantly improves survival and quality of life [1,2]. The long-term mortality rate is 48% to 82% lower in KTx recipients when compared to ESRD patients on the transplant waitlist [2,3]. However, due to immunosuppression, KTx patients are at a two-fold increased risk of developing malignancy in comparison to the general population [4,5,6]. Malignancies are among the top three leading causes of death in KTx recipients, following infection and cardiovascular complications [6].
Studies have demonstrated a higher incidence of renal cell carcinoma (RCC) among ESRD patients (0.3%) than its reported incidence in the general population (approximately 0.005%) [7,8]. Thus, the Clinical Practice Guidelines Committee of the American Society of Transplantation (AST) [9] has recommended RCC screening for high-risk candidates, such as ESRD patients on dialysis for more than 3 years [10]. Despite screening for RCC among KTx candidates, de novo RCC has been reported among KTx patients in both native kidneys [11,12,13,14,15,16,17,18], and transplanted kidneys [17,19,20]. However, the incidence and incidence trends of RCC among KTx patients remain unclear [11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42].
Thus, we performed a systematic review to (1) investigate the pooled incidence/incidence trends, and (2) assess the mortality/mortality trends in KTx patients with RCC.

2. Methods

2.1. Search Strategy and Literature Review

The protocol for this systematic review is registered with PROSPERO (International Prospective Register of Systematic Reviews; no. CRD42018108994). A systematic literature search of MEDLINE (1946 to October 2018), EMBASE (1988 to October 2018), and the Cochrane Database of Systematic Reviews (database inception to October 2018) was performed to assess (1) the pooled incidence/incidence trends, and (2) to assess the mortality/mortality trends in KTx patients with RCC. The systematic literature review was conducted independently by two investigators (C.T. and W.C) using a search strategy that consolidated the terms “kidney cancer” OR “renal cell carcinoma” AND “kidney transplantation” OR “renal transplantation” which is provided in the online Supplementary Data S1. The database searches were limited to English language articles only. A manual search for conceivably related studies using references of the included articles was also performed. This study was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) statement [43].

2.2. Selection Criteria

Eligible studies had to be clinical trials or observational studies (cohort, case-control, or cross-sectional studies) that reported the incidence or mortality of RCC among adult KTx recipients (age >/= 18 years old). Retrieved articles were individually reviewed for eligibility by two investigators (A.C. and C.T.). Discrepancies were addressed and solved by mutual consensus. Inclusion was not limited by the size of study.

2.3. Data Abstraction

A structured data collecting form was used to obtain the following information from each study: title, name of the first author, year of the study, publication year, country where the study was conducted, RCC definition, incidence of RCC, and mortality in KTx patients with RCC.

2.4. Statistical Analysis

Analyses were performed utilizing the Comprehensive Meta-Analysis 3.3 software (Biostat Inc, Englewood, NJ, USA). Adjusted point estimates from each study were consolidated by the generic inverse variance approach of DerSimonian and Laird, which designated the weight of each study based on its variance [44]. Given the possibility of between-study variance, we used a random-effect model rather than a fixed-effect model. Forest plots were constructed to visually evaluate the incidence and mortality of RCC among adult KTx recipients. Cochran’s Q test and I2 statistic were applied to determine the between-study heterogeneity. A value of I2 of 0–25% represents insignificant heterogeneity, 26–50% low heterogeneity, 51–75% moderate heterogeneity and 76–100% high heterogeneity [45]. The presence of publication bias was assessed using the Egger test [46]. Funnel plots were created to evaluate for the presence or absence of publication bias.

3. Results

A total of 7815 potentially eligible articles were identified using our search strategy. After the exclusion of 7629 articles based on their title and abstract for clearly not fulfilling the inclusion criteria on the basis of type of article, patient population, study design, or outcome of interest, and 81 due to being duplicates, 105 articles were left for full-length review. Fifty-nine of them were excluded from the full-length review as they did not report the outcome of interest. Twenty-one articles were case reports and three articles were not in English. Thus, 22 cohort studies [11,12,13,14,15,16,17,18,19,20,23,28,29,30,31,32,33,34,35,36,38,39] with a total of 320,190 KTx patients were enrolled. The literature retrieval, review, and selection process are demonstrated in Figure 1. The characteristics of the included studies are presented in Table 1.

3.1. Incidence of RCC after KTx

Eighteen studies provided data on the incidence of RCC after KTx [11,12,13,14,15,16,17,18,19,20,28,29,31,32,34,35]. Overall, the pooled estimated incidence of RCC after KTx was 0.7% (95% CI: 0.5–0.8%, I2 = 93%, Figure 2). While the pooled estimated incidence of de novo RCC in the native kidney was 0.7% (95% CI: 0.6–0.9%, I2 = 88%, Figure 3A), the pooled estimated incidence of RCC in the allograft kidney was 0.2% (95% CI: 0.1–0.4%, I2 = 64%, Figure 3B).
Meta-regression showed a significant negative correlation between year of study and incidence of de novo RCC post-KTx (slopes = −0.05, p = 0.01, Figure 4).

3.2. Mortality Rate in KTx Recipients with RCC

Eleven studies provided data the on mortality rate in KTx recipients with RCC [13,14,16,17,19,20,23,30,33,36,39]. Overall, the pooled estimated mortality rate in KTx recipients with RCC was 15.0% (95% CI: 7.4–28.1%, I2 = 80%, Figure 5) at a mean follow-up time of 42 months after RCC diagnosis. The data on the incidence and mortality of recurrent RCC among KTx recipients with a previous history of RCC prior to KTX were limited. A prior study demonstrated an incidence of recurrent RCC after KTX of 9.1% with an associated 5-year survival of 41.7% [23]. Sensitivity analysis, excluding the study of recurrent RCC among KTx recipients with a previous history of RCC prior to KTX (23), demonstrated a pooled estimated mortality rate of 11.5% in KTx recipients with RCC (95% CI: 6.4–19.8%, I2 = 67%).
Meta-regression showed no significant correlations between the year of study and mortality of patients with RCC (p = 0.50, Figure 6). When meta-regression was performed excluding the study of recurrent RCC among KTx recipients with a previous history of RCC prior to KTX [30], there were still no significant correlations between the year of study and mortality of patients with RCC (p = 0.56, Figure 7).

3.3. Evaluation for Publication Bias

Funnel plots (Supplementary Figures S1 and S2) and Egger’s regression asymmetry tests were performed to evaluate publication bias in the analysis evaluating the incidence and mortality of KTx recipients with RCC. There was no significant publication bias, with p-values of 0.58 and 0.54, respectively.

4. Discussion

In this systematic review, we found that RCC after KTx occurs with an incidence of 0.7%. RCC can occur in the native kidney with an incidence of 0.7% or in the allograft kidney with an incidence of 0.2%. Our findings also showed a statistically significant negative correlation between the incidence of RCC after KTx and study year, representing a potential decrease in the RCC incidence among KTx patients. However, mortality in KTx patients with RCC has not decreased over time.
Post-KTx malignancy is a common cause of death [5,6,47,48,49,50,51] and RCC is the most common solid-organ malignancy in this population [52,53]. Due to the increased risk of RCC among ESRD patients [7,8], the Clinical Practice Guidelines Committee of the AST has suggested RCC screening in ESRD patients on dialysis for longer than 3 years [9,10]. In addition, it is suggested that most KTx candidates with a history of RCC should wait at least 2 years from successful cancer treatment to KTx (unless candidates have only small localized incidental tumors, which may not require any waiting period) [54,55]. Candidates with large, invasive or symptomatic RCC may require a longer waiting period of 5 years [54,55]. Despite RCC screening prior to KTx, the findings from our study suggest that RCC can still occur post-KTx at a higher incidence (0.7%) than its reported incidence among ESRD patients (0.3%) [8]. In addition, studies have demonstrated that KTx recipients have a relative increased risk of five- to ten-fold for RCC compared with an age-matched general population, and that the majority of these tumors arise in the setting of acquired kidney cystic disease (AKCD) which develops with chronic renal failure [5,8,35,56,57,58,59,60,61,62,63,64]. Although RCC occurrence is more frequent in the native kidneys of KTx recipients, RCC can also occur in the renal allograft (incidence of 0.2%) [17,19,20].
While the exact etiology of the increased risk of RCC in KTx remains unclear, it is likely linked to the immunosuppressed state [4]. Reported risk factors for post-KTx RCC include older age, male sex, African descent, excess body weight, smoking, hypertension, history of acquired cystic kidney disease (ACKD), previous RCC prior to KTx, and longer pre-transplant dialysis duration [3,6,18,23,29,31,34,35,65,66,67]. Studies have shown that causes of ESRD before KTx may also affect the incidence of post-KTx RCC [14,32,35]. While KTx recipients with ESRD due to glomerulonephritis, hypertensive nephrosclerosis, and vascular diseases have been shown to have a higher incidence of post-KTx RCC, recipients with ESRD due to diabetic nephropathy carry a lower risk of post-KTx RCC [14,32,35,68]. KTx recipients are usually under intensified medical surveillance and the higher incidence of RCC among KTx recipients compared to general populations and ESRD patients might be due to detection bias. On the other hand, the lack of consensual RCC screening among KTx recipients may also have underestimated the exact incidence among the KTx patient population. Currently, there are no universal recommendations for RCC screening among KTx patients [3,22,69,70,71,72]. While the European Renal Best Practice (ERBP) guidelines recommend native kidney ultrasound as RCC screening in kidney transplant recipients, and the European Association of Urology (EAU) recommends an annual ultrasound of native kidneys and allografts for anyone with ACKD, previous RCC, or von Hippel–Lindau disease [3,71,72], the Kidney Disease Improving Global Outcomes (KDIGO) and AST guidelines for post-KTx care currently do not suggest universal screening for RCC among KTx recipients [22,69,70]. Thus, there are various RCC screening approaches for KTx recipients at different transplant centers. Many cases of RCC have been discovered during investigations for post-transplant erythrocytosis, elevated serum creatinine, hematuria, urinary infection, or incidentally from imaging for other indications [33,73,74,75]. The majority of studies with available data on surveillance programs performed screening for RCC post-KTx annually by ultrasonography of native and allograft kidneys. Among KTx recipients with ACKD, acquired multicystic dysplasia, or a prior history of RCC required more frequent screenings, every 6 months [16,17,19,20,28,36,76]. Given that the risk is greatest in the first year post-KTx and the majority of RCCs occur in the first 5 years after KTx [15,29,31,65,77], previous reports suggest that KTx recipients should routinely undergo ultrasonography to screen RCC on the native kidney during the first 30 days post-KTx and every 5 years afterwards in the absence of renal cysts, or every 2 years in the presence of renal cysts [65,77,78,79]. Our study’s findings suggest the need for future studies to identify a cost-effective surveillance strategy for RCC among KTx recipients. This strategy would need to take into consideration both native and allograft kidneys, and differentiate KTx recipients with non-simple renal cysts [3,80].
Several limitations of our systematic review are worth mentioning. First, there are statistical heterogeneities in our meta-analysis. Potential sources for heterogeneities were the variations in the renal transplant recipient screening methods, patient characteristics, and differences in the immunosuppressive regimens used at various transplant centers, which may have affected the incidence of RCC and mortality rate in this population. Second, there is a lack of data from included studies on immunosuppressive regimens [81,82,83,84,85]. Mammalian target of rapamycin (mTOR) inhibitors have shown antineoplastic activities [86]. Although the effects of mTOR among KTx recipients have been shown mostly for non-melanoma skin cancer [87,88,89], future studies evaluating the effects of different immunosuppressive regimens on mortality in KTx patients with RCC are needed. Lastly, this is a meta-analysis of cohort studies and the data from population-based studies were limited. Thus, large population-based studies evaluating the incidence of RCC in KTx patients are required in the future.
In summary, the overall estimated incidence of RCC after KTX was 0.6%, with an associated high mortality rate in KTx recipients of 13.9%. Despite potential improvements in the post-KTx RCC incidence, the mortality in KTx patients with RCC has remained unchanged over time.

Supplementary Materials

The following are available online at https://www.mdpi.com/2077-0383/8/4/530/s1, Data S1: Search terms for systematic review; Figure S1: Funnel plot evaluating for publication bias evaluating incidence of KTx recipients with RCC; Figure S2: Funnel plot evaluating for publication bias evaluating mortality of KTx recipients with RCC.

Author Contributions

Conceptualization, A.C., C.T., W.K., M.A.M., N.J.K. and W.C.; Data curation, A.C. and C.T.; Formal analysis, C.T. and W.C.; Investigation, A.C., C.T. and W.C.; Methodology, A.C., T.B., B.B., W.K., P.L., N.L. and W.C.; Project administration, K.W. and K.S.; Resources, K.W. and K.S.; Software, K.W.; Supervision, T.B., N.R.A., B.B., W.K., A.T.-O., N.L., M.A., N.J. and W.C.; Validation, A.C., C.T., P.L. and W.C.; Visualization, T.B. and K.S.; Writing—original draft, A.C. and N.R.; Writing—review and editing, C.T., T.B., N.R.A., B.B., W.K., K.W., P.L., K.S., A.T.-O., N.L., M.A.M., N.J.K. and W.C.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kaballo, M.A.; Canney, M.; O’Kelly, P.; Williams, Y.; O’Seaghdha, C.M.; Conlon, P.J. A comparative analysis of survival of patients on dialysis and after kidney transplantation. Clin. Kidney J. 2018, 11, 389–393. [Google Scholar] [CrossRef] [PubMed]
  2. Wolfe, R.A.; Ashby, V.B.; Milford, E.L.; Ojo, A.O.; Ettenger, R.E.; Agodoa, L.Y.; Held, P.J.; Port, F.K. Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant. N. Engl. J. Med. 1999, 341, 1725–1730. [Google Scholar] [CrossRef] [PubMed]
  3. Hickman, L.A.; Sawinski, D.; Guzzo, T.; Locke, J.E. Urologic malignancies in kidney transplantation. Am. J. Transpl. 2018, 18, 13–22. [Google Scholar] [CrossRef] [PubMed]
  4. Engels, E.A.; Pfeiffer, R.M.; Fraumeni, J.F.; Jr Kasiske, B.L.; Israni, A.K.; Snyder, J.J.; Wolfe, R.A.; Goodrich, N.P.; Bayakly, A.R.; Clarke, C.A.; et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 2011, 306, 1891–1901. [Google Scholar] [CrossRef] [PubMed]
  5. Bennett, W.M.; Simonich, E.L.; Garre, A.M.; McEvoy, K.M.; Farinola, M.A.; Batiuk, T.D. Renal Cell Carcinoma in Renal Transplantation: The Case for Surveillance. Transpl. Proc. 2017, 49, 1779–1782. [Google Scholar] [CrossRef]
  6. Briggs, J.D. Causes of death after renal transplantation. Nephrol Dial. Transpl. 2001, 16, 1545–1549. [Google Scholar] [CrossRef]
  7. Dy, G.W.; Gore, J.L.; Forouzanfar, M.H.; Naghavi, M.; Fitzmaurice, C. Global Burden of Urologic Cancers, 1990. Eur. Urol. 2017, 71, 437–446. [Google Scholar] [CrossRef] [PubMed]
  8. Kompotiatis, P.; Thongprayoon, C.; Manohar, S.; Cheungpasitporn, W.; Gonzalez Suarez, M.L.; Craici, I.M.; Mao, M.A.; Herrmann, S.M. Association between urologic malignancies and end-stage renal disease: A meta-analysis. Nephrology 2019, 24, 65–73. [Google Scholar] [CrossRef] [PubMed]
  9. Kasiske, B.L.; Cangro, C.B.; Hariharan, S.; Hricik, D.E.; Kerman, R.H.; Roth, D.; Rush, D.N.; Vazquez, M.A.; Weir, M.R. The evaluation of renal transplantation candidates: Clinical practice guidelines. Am. J. Transplant. 2001, 1 (Suppl. 2), 3–95. [Google Scholar]
  10. Holley, J.L. Screening, diagnosis, and treatment of cancer in long-term dialysis patients. Clin. J. Am. Soc. Nephrol. 2007, 2, 604–610. [Google Scholar] [CrossRef] [PubMed]
  11. Hoshida, Y.; Tsukuma, H.; Yasunaga, Y.; Xu, N.; Fujita, M.Q.; Satoh, T.; Ichikawa, Y.; Kurihara, K.; Imanishi, M.; Matsuno, T.; et al. Cancer risk after renal transplantation in Japan. Int. J. Cancer 1997, 71, 517–520. [Google Scholar] [CrossRef]
  12. Agraharkar, M.L.; Cinclair, R.D.; Kuo, Y.F.; Daller, J.A.; Shahinian, V.B. Risk of malignancy with long-term immunosuppression in renal transplant recipients. Kidney Int. 2004, 66, 383–389. [Google Scholar] [CrossRef] [PubMed]
  13. Neuzillet, Y.; Lay, F.; Luccioni, A.; Daniel, L.; Berland, Y.; Coulange, C.; Lechevallier, E. De novo renal cell carcinoma of native kidney in renal transplant recipients. Cancer 2005, 103, 251–257. [Google Scholar] [CrossRef] [PubMed]
  14. Moudouni, S.M.; Lakmichi, A.; Tligui, M.; Rafii, A.; Tchala, K.; Haab, F.; Gattegno, B.; Thibault, P.; Doublet, J.D. Renal cell carcinoma of native kidney in renal transplant recipients. BJU Int. 2006, 98, 298–302. [Google Scholar] [CrossRef]
  15. Tsai, M.K.; Yang, C.Y.; Lee, C.Y.; Yeh, C.C.; Hu, R.H.; Lee, P.H. De novo malignancy is associated with renal transplant tourism. Kidney Int. 2011, 79, 908–913. [Google Scholar] [CrossRef]
  16. Filocamo, M.T.; Zanazzi, M.; Li Marzi, V.; Guidoni, L.; Villari, D.; Dattolo, E.; Nicita, G. Renal cell carcinoma of native kidney after renal transplantation: Clinical relevance of early detection. Transpl. Proc. 2009, 41, 4197–4201. [Google Scholar] [CrossRef] [PubMed]
  17. Leveridge, M.; Musquera, M.; Evans, A.; Cardella, C.; Pei, Y.; Jewett, M.; Robinette, M.; Finelli, A. Renal cell carcinoma in the native and allograft kidneys of renal transplant recipients. J. Urol. 2011, 186, 219–223. [Google Scholar] [CrossRef]
  18. Hwang, J.K.; Moon, I.S.; Kim, J.I. Malignancies after kidney transplantation: A 40-year single-center experience in Korea. Transpl. Int. 2011, 24, 716–721. [Google Scholar] [CrossRef] [PubMed]
  19. Tillou, X.; Doerfler, A.; Collon, S.; Kleinclauss, F.; Patard, J.J.; Badet, L.; Barrou, B.; Audet, M.; Bensadoun, H.; Berthoux, E.; et al. De novo kidney graft tumors: Results from a multicentric retrospective national study. Am. J. Transpl. 2012, 12, 3308–3315. [Google Scholar] [CrossRef]
  20. Ploussard, G.; Chambade, D.; Meria, P.; Gaudez, F.; Tariel, E.; Verine, J.; De Bazelaire, C.; Peraldi, M.N.; Glotz, D.; Desgrandchamps, F.; et al. Biopsy-confirmed de novo renal cell carcinoma (RCC) in renal grafts: A single-centre management experience in a 2396 recipient cohort. BJU Int. 2012, 109, 195–199. [Google Scholar] [CrossRef]
  21. Scandling, J.D. Acquired cystic kidney disease and renal cell cancer after transplantation: Time to rethink screening? Clin. J. Am. Soc. Nephrol. 2007, 2, 621–622. [Google Scholar] [CrossRef]
  22. Kasiske, B.L.; Vazquez, M.A.; Harmon, W.E.; Brown, R.S.; Danovitch, G.M.; Gaston, R.S.; Roth, D.; Scandling, J.D.; Singer, G.G. Recommendations for the outpatient surveillance of renal transplant recipients. American Society of Transplantation. J. Am. Soc. Nephrol 2000, 11 (Suppl. 15), S1–S86. [Google Scholar] [PubMed]
  23. Cognard, N.; Anglicheau, D.; Gatault, P.; Girerd, S.; Essig, M.; Hurault de Ligny, B.; Le Meur, Y.; Le Roy, F.; Garrouste, C.; Thierry, A.; et al. Recurrence of Renal Cell Cancer After Renal Transplantation in a Multicenter French Cohort. Transplantation 2018, 102, 860–867. [Google Scholar] [CrossRef] [PubMed]
  24. EBPG Expert Group on Renal Transplantation. European best practice guidelines for renal transplantation. Section IV: Long-term management of the transplant recipient. IV.6. Cancer risk after renal transplantation. Solid organ cancers: Prevention and treatment. Nephrol Dial. Transplant. 2002, 17 (Suppl. 4), 34–36. [Google Scholar]
  25. Wiesel, M.; Carl, S.; Drehmer, I.; Hofmann, W.J.; Zeier, M.; Staehler, G. [The clinical significance of renal cell carcinoma in dialysis dependent patients in comparison with kidney transplant recipients]. Urol. A 1997, 36, 126–129. [Google Scholar] [CrossRef]
  26. Gulanikar, A.C.; Daily, P.P.; Kilambi, N.K.; Hamrick-Turner, J.E.; Butkus, D.E. Prospective pretransplant ultrasound screening in 206 patients for acquired renal cysts and renal cell carcinoma. Transplantation 1998, 66, 1669–1672. [Google Scholar] [CrossRef]
  27. Brunner, F.P.; Landais, P.; Selwood, N.H. Malignancies after renal transplantation: The EDTA-ERA registry experience. European Dialysis and Transplantation Association-European Renal Association. Nephrol. Dial. Transplant. 1995, 10 (Suppl. 1), 74–80. [Google Scholar] [CrossRef] [PubMed]
  28. Lee, H.H.; Choi, K.H.; Yang, S.C.; Han, W.K. Renal cell carcinoma in kidney transplant recipients and dialysis patients. Korean J. Urol. 2012, 53, 229–233. [Google Scholar] [CrossRef] [PubMed]
  29. Einollahi, B.; Rostami, Z.; Nourbala, M.H.; Lessan-Pezeshki, M.; Simforoosh, N.; Nemati, E.; Pourfarziani, V.; Beiraghdar, F.; Nafar, M.; Pour-Reza-Gholi, F.; et al. Incidence of malignancy after living kidney transplantation: A multicenter study from iran. J. Cancer 2012, 3, 246–256. [Google Scholar] [CrossRef] [PubMed]
  30. Gigante, M.; Neuzillet, Y.; Patard, J.J.; Tillou, X.; Thuret, R.; Branchereau, J.; Timsit, M.O.; Terrier, N.; Boutin, J.M.; Sallusto, F.; et al. Renal cell carcinoma (RCC) arising in native kidneys of dialyzed and transplant patients: Are they different entities? BJU Int. 2012, 110, E570–E573. [Google Scholar] [CrossRef] [PubMed]
  31. Cheung, C.Y.; Lam, M.F.; Chu, K.H.; Chow, K.M.; Tsang, K.Y.; Yuen, S.K.; Wong, P.N.; Chan, S.K.; Leung, K.T.; Chan, C.K.; et al. Malignancies after kidney transplantation: Hong Kong renal registry. Am. J. Transpl. 2012, 12, 3039–3046. [Google Scholar] [CrossRef]
  32. Piselli, P.; Busnach, G.; Citterio, F.; Richiardi, L.; Cimaglia, C.; Angeletti, C.; Doringhet, P.V.; Pozzetto, U.; Perrino, M.L.; Serraino, D. [Kidney transplant and cancer risk: An epidemiological study in Northern and Central Italy]. Epidemiol. Prev. 2008, 32, 205–211. [Google Scholar]
  33. Ryosaka, M.; Ishida, H.; Takagi, T.; Shimizu, T.; Tanabe, K.; Kondo, T. Solid-type RCC originating from native kidneys in renal transplant recipients should be monitored cautiously. Transpl. Int. 2015, 28, 813–819. [Google Scholar] [CrossRef]
  34. Kalil, R.S.; Lynch, C.F.; Engels, E.A. Risk of cancer in retransplants compared to primary kidney transplants in the United States. Clin. Transpl. 2015, 29, 944–950. [Google Scholar] [CrossRef]
  35. Karami, S.; Yanik, E.L.; Moore, L.E.; Pfeiffer, R.M.; Copeland, G.; Gonsalves, L.; Hernandez, B.Y.; Lynch, C.F.; Pawlish, K.; Engels, E.A. Risk of Renal Cell Carcinoma Among Kidney Transplant Recipients in the United States. Am. J. Transpl. 2016, 16, 3479–3489. [Google Scholar] [CrossRef]
  36. Takagi, T.; Kondo, T.; Okumi, M.; Ishida, H.; Tanabe, K. Differences in Clinical and Pathological Features of Renal Cell Carcinoma Between Japanese Patients After Kidney Transplantation and Those on Hemodialysis. Ther. Apher. Dial. 2017, 21, 133–138. [Google Scholar] [CrossRef]
  37. Olsson, C.A. Renal cell carcinoma (RCC) arising in native kidneys of dialyzed and transplant patients: Are they different entities? BJU Int. 2012, 110, E574. [Google Scholar] [CrossRef]
  38. Schwarz, A.; Vatandaslar, S.; Merkel, S.; Haller, H. Renal cell carcinoma in transplant recipients with acquired cystic kidney disease. Clin. J. Am. Soc. Nephrol. 2007, 2, 750–756. [Google Scholar] [CrossRef] [PubMed]
  39. Ianhez, L.E.; Lucon, M.; Nahas, W.C.; Sabbaga, E.; Saldanha, L.B.; Lucon, A.M.; Srougi, M. Renal cell carcinoma in renal transplant patients. Urology 2007, 69, 462–464. [Google Scholar] [CrossRef] [PubMed]
  40. Levine, E. Renal cell carcinoma in uremic acquired renal cystic disease: Incidence, detection, and management. Urol Radiol 1992, 13, 203–210. [Google Scholar] [CrossRef] [PubMed]
  41. Doublet, J.D.; Peraldi, M.N.; Gattegno, B.; Thibault, P.; Sraer, J.D. Renal cell carcinoma of native kidneys: Prospective study of 129 renal transplant patients. J. Urol. 1997, 158, 42–44. [Google Scholar] [CrossRef]
  42. Nie, H.; Wang, W.; Zhao, Y.; Zhang, X.; Xiao, Y.; Zeng, Q.; Zhang, C.; Zhang, L. New-Onset Diabetes After Renal Transplantation (NODAT): Is It a Risk Factor for Renal Cell Carcinoma or Renal Failure? Ann. Transpl. 2019, 24, 62–69. [Google Scholar] [CrossRef]
  43. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef]
  44. DerSimonian, R.; Laird, N. Meta-analysis in clinical trials. Controll. Clin. Trials 1986, 7, 177–188. [Google Scholar] [CrossRef]
  45. Higgins, J.P.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef]
  46. Easterbrook, P.J.; Berlin, J.A.; Gopalan, R.; Matthews, D.R. Publication bias in clinical research. Lancet 1991, 337, 867–872. [Google Scholar] [CrossRef]
  47. Penn, I. Primary kidney tumors before and after renal transplantation. Transplantation 1995, 59, 480–485. [Google Scholar] [CrossRef]
  48. Kasiske, B.L.; Snyder, J.J.; Gilbertson, D.T.; Wang, C. Cancer after kidney transplantation in the United States. Am. J. Transplant. 2004, 4, 905–913. [Google Scholar] [CrossRef]
  49. Pedotti, P.; Cardillo, M.; Rossini, G.; Arcuri, V.; Boschiero, L.; Caldara, R.; Cannella, G.; Dissegna, D.; Gotti, E.; Marchini, F.; et al. Incidence of cancer after kidney transplant: Results from the North Italy transplant program. Transplantation 2003, 76, 1448–1451. [Google Scholar] [CrossRef]
  50. Bellini, M.I.; Gopal, J.P.; Hill, P.; Nicol, D.; Gibbons, N. Urothelial Carcinoma arising from the transplanted kidney: A single centre experience and literature review. Clin. Transplant. 2019. [Google Scholar] [CrossRef]
  51. Benoni, H.; Eloranta, S.; Ekbom, A.; Wilczek, H.; Smedby, K.E. Survival among solid organ transplant recipients diagnosed with cancer compared to nontransplanted cancer patients-A nationwide study. Int. J. Cancer 2019. [Google Scholar] [CrossRef] [PubMed]
  52. Vajdic, C.M.; McDonald, S.P.; McCredie, M.R.; van Leeuwen, M.T.; Stewart, J.H.; Law, M.; Chapman, J.R.; Webster, A.C.; Kaldor, J.M.; Grulich, A.E. Cancer incidence before and after kidney transplantation. JAMA 2006, 296, 2823–2831. [Google Scholar] [CrossRef] [PubMed]
  53. Birkeland, S.A.; Lokkegaard, H.; Storm, H.H. Cancer risk in patients on dialysis and after renal transplantation. Lancet 2000, 355, 1886–1887. [Google Scholar] [CrossRef]
  54. Knoll, G.; Cockfield, S.; Blydt-Hansen, T.; Baran, D.; Kiberd, B.; Landsberg, D.; Rush, D.; Cole, E. Canadian Society of Transplantation: Consensus guidelines on eligibility for kidney transplantation. CMAJ 2005, 173, S1–S25. [Google Scholar] [CrossRef] [PubMed]
  55. Knoll, G.; Cockfield, S.; Blydt-Hansen, T.; Baran, D.; Kiberd, B.; Landsberg, D.; Rush, D.; Cole, E. Canadian Society of Transplantation consensus guidelines on eligibility for kidney transplantation. CMAJ 2005, 173, 1181–1184. [Google Scholar] [CrossRef] [PubMed]
  56. Butler, A.M.; Olshan, A.F.; Kshirsagar, A.V.; Edwards, J.K.; Nielsen, M.E.; Wheeler, S.B.; Brookhart, M.A. Cancer incidence among US Medicare ESRD patients receiving hemodialysis, 1996–2009. Am. J. Kidney Dis. 2015, 65, 763–772. [Google Scholar] [CrossRef]
  57. Zorbas, K.A.; Karhadkar, S.S.; Lau, K.N.; Di Carlo, A. Renal Cell Carcinoma in Kidney Transplant Candidates. Transpl. Proc. 2017, 49, 1312–1317. [Google Scholar] [CrossRef]
  58. Tillou, X.; Guleryuz, K.; Doerfler, A.; Bensadoun, H.; Chambade, D.; Codas, R.; Devonec, M.; Dugardin, F.; Erauso, A.; Hubert, J.; et al. Nephron sparing surgery for De Novo kidney graft tumor: Results from a multicenter national study. Am. J. Transpl. 2014, 14, 2120–2125. [Google Scholar] [CrossRef]
  59. Krisl, J.C.; Doan, V.P. Chemotherapy and Transplantation: The Role of Immunosuppression in Malignancy and a Review of Antineoplastic Agents in Solid Organ Transplant Recipients. Am. J. Transpl. 2017, 17, 1974–1991. [Google Scholar] [CrossRef] [PubMed]
  60. Purdue, M.P.; Moore, L.E.; Merino, M.J.; Boffetta, P.; Colt, J.S.; Schwartz, K.L.; Bencko, V.; Davis, F.G.; Graubard, B.I.; Janout, V.; et al. An investigation of risk factors for renal cell carcinoma by histologic subtype in two case-control studies. Int. J. Cancer 2013, 132, 2640–2647. [Google Scholar] [CrossRef]
  61. Ishikawa, I. Uremic acquired renal cystic disease. Natural history and complications. Nephron 1991, 58, 257–267. [Google Scholar] [CrossRef] [PubMed]
  62. Ishikawa, I. Uremic acquired cystic disease of kidney. Urology 1985, 26, 101–108. [Google Scholar] [CrossRef]
  63. Levine, E.; Slusher, S.L.; Grantham, J.J.; Wetzel, L.H. Natural history of acquired renal cystic disease in dialysis patients: A prospective longitudinal CT study. Am. J. Roentgenol. 1991, 156, 501–506. [Google Scholar] [CrossRef] [PubMed]
  64. Matson, M.A.; Cohen, E.P. Acquired cystic kidney disease: Occurrence, prevalence, and renal cancers. Medicine 1990, 69, 217–226. [Google Scholar] [CrossRef] [PubMed]
  65. Goh, A.; Vathsala, A. Native renal cysts and dialysis duration are risk factors for renal cell carcinoma in renal transplant recipients. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2011, 11, 86–92. [Google Scholar] [CrossRef] [PubMed]
  66. Moris, D.; Kakavia, K.; Argyrou, C.; Garmpis, N.; Bokos, J.; Vernadakis, S.; Diles, K.; Sotirchos, G.; Boletis, J.; Zavos, G. De Novo Renal Cell Carcinoma of Native Kidneys in Renal Transplant Recipients: A Single-center Experience. Anticancer Res. 2017, 37, 773–779. [Google Scholar] [CrossRef]
  67. Chow, W.H.; Devesa, S.S. Contemporary epidemiology of renal cell cancer. Cancer J. 2008, 14, 288–301. [Google Scholar] [CrossRef]
  68. Dhakal, P.; Giri, S.; Siwakoti, K.; Rayamajhi, S.; Bhatt, V.R. Renal Cancer in Recipients of Kidney Transplant. Rare Tumors 2017, 9, 6550. [Google Scholar] [CrossRef] [PubMed]
  69. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am. J. Transplant. 2009, 9 (Suppl. 3), S1–S155. [Google Scholar] [CrossRef]
  70. Bia, M.; Adey, D.B.; Bloom, R.D.; Chan, L.; Kulkarni, S.; Tomlanovich, S. KDOQI US commentary on the 2009 KDIGO clinical practice guideline for the care of kidney transplant recipients. Am. J. Kidney Dis. 2010, 56, 189–218. [Google Scholar] [CrossRef]
  71. European Renal Best Practice Transplantation Guideline Development Group. ERBP Guideline on the Management and Evaluation of the Kidney Donor and Recipient. Nephrol. Dial. Transplant. 2013, 28 (Suppl. 2), ii1–ii71. [Google Scholar]
  72. Kalble, T.; Lucan, M.; Nicita, G.; Sells, R.; Burgos Revilla, F.J.; Wiesel, M. EAU guidelines on renal transplantation. Eur. Urol. 2005, 47, 156–166. [Google Scholar] [CrossRef]
  73. Fuiano, G.; Zoccali, C.; Bertoni, E.; Bartolomeo, F.; Cambareri, F.; Salvadori, M.; Altieri, P.; Ponticelli, C.; Sandrini, S.; Schena, F.P.; et al. [Guidelines for ambulatory monitoring of kidney transplant patients. Adaptation of the Guidelines of the American Society of Transplantation (J Am Soc Nephrol 2000; 11 (S1): 86)]. Giornale italiano di nefrologia Organo ufficiale della Societa italiana di nefrologia 2004, 21 (Suppl. 28), S11–S50. [Google Scholar]
  74. Marinella, M.A. Hematologic abnormalities following renal transplantation. Int. Urol. Nephrol. 2010, 42, 151–164. [Google Scholar] [CrossRef] [PubMed]
  75. Park, P.; Kim, W.Y.; Lee, J.B.; Choi, S.B.; Kim, W.B.; Choi, S.Y. Incidental renal cell carcinoma originating from a native kidney after en-bloc resection for adrenal carcinoma in a kidney transplant recipient. Transpl. Proc. 2014, 46, 637–639. [Google Scholar] [CrossRef] [PubMed]
  76. Brennan, J.F.; Stilmant, M.M.; Babayan, R.K.; Siroky, M.B. Acquired renal cystic disease: Implications for the urologist. Br. J. Urol. 1991, 67, 342–348. [Google Scholar] [CrossRef]
  77. Klatte, T.; Seitz, C.; Waldert, M.; de Martino, M.; Kikic, Z.; Bohmig, G.A.; Haitel, A.; Schmidbauer, J.; Marberger, M.; Remzi, M. Features and outcomes of renal cell carcinoma of native kidneys in renal transplant recipients. BJU Int. 2010, 105, 1260–1265. [Google Scholar] [CrossRef] [PubMed]
  78. Muruve, N.A.; Shoskes, D.A. Genitourinary malignancies in solid organ transplant recipients. Transplantation 2005, 80, 709–716. [Google Scholar] [CrossRef]
  79. Ljungberg, B.; Cowan, N.C.; Hanbury, D.C.; Hora, M.; Kuczyk, M.A.; Merseburger, A.S.; Patard, J.J.; Mulders, P.F.; Sinescu, I.C. EAU guidelines on renal cell carcinoma: The 2010 update. Eur. Urol. 2010, 58, 398–406. [Google Scholar] [CrossRef] [PubMed]
  80. Israel, G.M.; Bosniak, M.A. An update of the Bosniak renal cyst classification system. Urology 2005, 66, 484–488. [Google Scholar] [CrossRef]
  81. Kauffman, H.M.; Cherikh, W.S.; McBride, M.A.; Cheng, Y.; Hanto, D.W. Post-transplant de novo malignancies in renal transplant recipients: The past and present. Transpl. Int. Off. J. Eur. Soc. Organ. Transplant. 2006, 19, 607–620. [Google Scholar] [CrossRef] [PubMed]
  82. Taylor, A.L.; Marcus, R.; Bradley, J.A. Post-transplant lymphoproliferative disorders (PTLD) after solid organ transplantation. Crit. Rev. Oncol. Hematol. 2005, 56, 155–167. [Google Scholar] [CrossRef]
  83. Barrett, W.L.; First, M.R.; Aron, B.S.; Penn, I. Clinical course of malignancies in renal transplant recipients. Cancer 1993, 72, 2186–2189. [Google Scholar] [CrossRef]
  84. Kliem, V.; Kolditz, M.; Behrend, M.; Ehlerding, G.; Pichlmayr, R.; Koch, K.M.; Brunkhorst, R. Risk of renal cell carcinoma after kidney transplantation. Clin. Transplant. 1997, 11, 255–258. [Google Scholar]
  85. Sandock, D.S.; Seftel, A.D.; Resnick, M.I. A new protocol for the followup of renal cell carcinoma based on pathological stage. J. Urol. 1995, 154, 28–31. [Google Scholar] [CrossRef]
  86. Kapoor, A. Malignancy in kidney transplant recipients. Drugs 2008, 68 (Suppl. 1), 11–19. [Google Scholar] [CrossRef]
  87. Opelz, G.; Unterrainer, C.; Susal, C.; Dohler, B. Immunosuppression with mammalian target of rapamycin inhibitor and incidence of post-transplant cancer in kidney transplant recipients. Nephrol. Dial. Transpl. 2016, 31, 1360–1367. [Google Scholar] [CrossRef] [PubMed]
  88. Kao, C.C.; Liu, J.S.; Lin, M.H.; Hsu, C.Y.; Chang, F.C.; Lin, Y.C.; Chen, H.H.; Chen, T.W.; Hsu, C.C.; Wu, M.S. Impact of mTOR Inhibitors on Cancer Development in Kidney Transplantation Recipients: A Population-Based Study. Transpl. Proc. 2016, 48, 900–904. [Google Scholar] [CrossRef]
  89. Karpe, K.M.; Talaulikar, G.S.; Walters, G.D. Calcineurin inhibitor withdrawal or tapering for kidney transplant recipients. Cochrane Database Syst. Rev. 2017, 7, CD006750. [Google Scholar] [CrossRef]
Figure 1. Outline of our search methodology.
Figure 1. Outline of our search methodology.
Jcm 08 00530 g001
Figure 2. Forest plots of the included studies [11,12,13,14,15,16,17,18,19,20,28,29,31,32,34,35,38,39] assessing incidence rates of RCC after KTx. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Figure 2. Forest plots of the included studies [11,12,13,14,15,16,17,18,19,20,28,29,31,32,34,35,38,39] assessing incidence rates of RCC after KTx. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Jcm 08 00530 g002
Figure 3. Forest plots of the included studies [11,12,13,14,15,16,17,18,28,29,31,32,34,35,38,39] assessing incidence rates of (A) de novo RCC in the native kidney and (B) RCC in the allograft kidney [17,19,20,38,39]. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Figure 3. Forest plots of the included studies [11,12,13,14,15,16,17,18,28,29,31,32,34,35,38,39] assessing incidence rates of (A) de novo RCC in the native kidney and (B) RCC in the allograft kidney [17,19,20,38,39]. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Jcm 08 00530 g003
Figure 4. Meta-regression analyses showed a significant negative correlation between the year of study and incidence of de novo RCC post-KTx (slopes = −0.05, p = 0.01). The solid line represents the weighted regression line based on variance-weighted least squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate the log event rates in each study.
Figure 4. Meta-regression analyses showed a significant negative correlation between the year of study and incidence of de novo RCC post-KTx (slopes = −0.05, p = 0.01). The solid line represents the weighted regression line based on variance-weighted least squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate the log event rates in each study.
Jcm 08 00530 g004
Figure 5. Forest plots of the included studies [13,14,16,17,19,20,23,30,33,36,39] assessing mortality rate in KTx recipients with RCC. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Figure 5. Forest plots of the included studies [13,14,16,17,19,20,23,30,33,36,39] assessing mortality rate in KTx recipients with RCC. A diamond data marker represents the overall rate from each included study (square data marker) and 95% confidence interval.
Jcm 08 00530 g005
Figure 6. Meta-regression analyses showed no significant correlations between the year of study and mortality of patients with RCC (p = 0.50). The solid line represents the weighted regression line based on variance-weighted least-squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate the log event rates in each study.
Figure 6. Meta-regression analyses showed no significant correlations between the year of study and mortality of patients with RCC (p = 0.50). The solid line represents the weighted regression line based on variance-weighted least-squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate the log event rates in each study.
Jcm 08 00530 g006
Figure 7. Meta-regression analyses, excluding the study of recurrent RCC among KTx recipients with a previous history of RCC prior to KTX, showed no significant correlations between the year of study and mortality of patients with RCC (p = 0.56). The solid line represents the weighted regression line based on variance-weighted least-squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate log event rates in each study.
Figure 7. Meta-regression analyses, excluding the study of recurrent RCC among KTx recipients with a previous history of RCC prior to KTX, showed no significant correlations between the year of study and mortality of patients with RCC (p = 0.56). The solid line represents the weighted regression line based on variance-weighted least-squares. The inner and outer lines show the 95% confidence interval and prediction interval around the regression line. The circles indicate log event rates in each study.
Jcm 08 00530 g007
Table 1. Main characteristics of studies included in the meta-analysis of incidence and mortality of renal cell carcinoma (RCC) after kidney transplantation (KTx) [11,12,13,14,15,16,17,18,19,20,23,28,29,30,31,32,33,34,35,36,38,39].
Table 1. Main characteristics of studies included in the meta-analysis of incidence and mortality of renal cell carcinoma (RCC) after kidney transplantation (KTx) [11,12,13,14,15,16,17,18,19,20,23,28,29,30,31,32,33,34,35,36,38,39].
StudyYearType of StudyNumber of PatientsIncidence of RCCFollow-Up Time after TransplantTime from Transplant to Cancer DiagnosisMortality of RCCQuality Assessment
Hoshida et al. [11]1997Cohort174415/1744N/AN/AN/AS4 C0 O3
Agraharkar et al. [12]2004Cohort17396/1739Mean 6.1 yearsN/AN/AS4 C1 O3
Neuzillet et al. [13]2004Cohort93311/933N/AMean 70.9 ± 49.4 (range 8–156) months2/11 (1 died due to cancer)S4 C0 O2
Moudouni et al. [14]2006Cohort37310/373N/AMean 12.8 years
Median 127 in patients treated with cyclosporine A and 114 months in patients not treated with cyclosporine A
1/10 (1 died due to cancer)S4 C0 O3
Ianhez et al. [39]2007Cohort137510/1375
9 in native kidney
1 in allograft kidney
N/AN/A3/10 (2 died due to myocardial infarction and one due to penile cancer)S4 C0 O2
Schwarz et al. [38]2007Cohort5618/561
7 de novo in native kidney
1 de novo in allograft kidney
N/A105.2 ± 62.39 monthsN/AS4C2O3
Tsai et al. [15]2008Cohort3259Touring group
15/215 kidney cancer
Domestic group
4/321 kidney cancer
Touring group
Mean 76.2 ± 48.1 months
Domestic group
Mean 81.5 ± 53.4 months
N/AN/AS4 C1 O3
Filocamo et al. [16]2009cohort694Native de novo
10/694
N/A61.8 months (12–156 months)3/10 (3 died due to cancer other than RCC)S4 C1 O3
Leveridge et al. [17]2010cohort356839/3568 native kidney
8/3568 allograft kidney
6.6 yearsNative 10.6 years
allograft 12.1 years
5 native died (not RCC cause), 1 allograft died due to cardiac causeS4 C0 O3
Hwang et al. [18]2011Cohort16957/16959.1 ± 6.9 yearsMean 11.8 ± 6.0 yearsN/AS4 C0 O3
Lee et al. [28]2011Cohort275721/2757N/AMean 119 (range 0–264) monthsN/AS4 C1 O2
Ploussard et al. [20]2012Cohort2396Allograft kidney
12/2396
N/AMean 13 (range 4–20) years0/12S4 C0 O3
Einollahi et al. [29]2012Cohort12,5256/12,525N/AMedian 16 monthsN/AS4 C0 O3
Gigante et al. [30]2012Cohort213N/AN/AMean 91 ± 82 months6/213 due to RCCS4 C0 O2
Tillou et al. [19]2012Cohort41,806Allograft kidney
79/41,806
N/AMean 131.7 (0.9–244) months4/79S4 C0 O3
Cheung et al. [31]2012Cohort489526/4895N/AMedian 4 (0.2–16.5) yearsN/AS4 C1 O3
Piselli et al. [32]2013Cohort721731/7217Median 5.2 years (2.9–7.8)N/AN/AS4 C1 O3
Ryosaka et al. [33]2015Cohort202N/AN/AN/ASolid-type renal cell carcinoma
2/17
Cystic-type renal cell carcinoma
2/27
S4 C0 O3
Kalil et al. [34]2015Cohort115,845Primary kidney transplant
514/109,224
Retransplant
43/6621
Mean
1st–4.6 years
2nd–3.7 years
3rd–2.9 years
4th–3.4 years
N/AN/AS4 C2 O3
Karami et al. [35]2016Cohort116,208683/116,208Median 4.2 years (range 0.003–23.1)N/AN/AS4 C0 O2
Takagi et al. [36]2017Cohort42N/AN/AMean 86 ± 69 months9/42 (5 died due to cancer)S4 C0 O3
Cognard et al. [23]2018Cohort143 with history of pre-transplant kidney cancer13/143Mean 5.6 ± 3.2 yearsMean 3 ± 2.3 years (range 45 days–7 years)10/13 (9 died due to cancer)S4 C0 O3
KTx, kidney transplantation; N/A, not available; RCC, renal cell carcinoma; S, C, O, selection, comparability, and outcome.

Share and Cite

MDPI and ACS Style

Chewcharat, A.; Thongprayoon, C.; Bathini, T.; Aeddula, N.R.; Boonpheng, B.; Kaewput, W.; Watthanasuntorn, K.; Lertjitbanjong, P.; Sharma, K.; Torres-Ortiz, A.; et al. Incidence and Mortality of Renal Cell Carcinoma after Kidney Transplantation: A Meta-Analysis. J. Clin. Med. 2019, 8, 530. https://doi.org/10.3390/jcm8040530

AMA Style

Chewcharat A, Thongprayoon C, Bathini T, Aeddula NR, Boonpheng B, Kaewput W, Watthanasuntorn K, Lertjitbanjong P, Sharma K, Torres-Ortiz A, et al. Incidence and Mortality of Renal Cell Carcinoma after Kidney Transplantation: A Meta-Analysis. Journal of Clinical Medicine. 2019; 8(4):530. https://doi.org/10.3390/jcm8040530

Chicago/Turabian Style

Chewcharat, Api, Charat Thongprayoon, Tarun Bathini, Narothama Reddy Aeddula, Boonphiphop Boonpheng, Wisit Kaewput, Kanramon Watthanasuntorn, Ploypin Lertjitbanjong, Konika Sharma, Aldo Torres-Ortiz, and et al. 2019. "Incidence and Mortality of Renal Cell Carcinoma after Kidney Transplantation: A Meta-Analysis" Journal of Clinical Medicine 8, no. 4: 530. https://doi.org/10.3390/jcm8040530

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

Article Metrics

Back to TopTop