ABSTRACT

In kidney transplant recipients, cancer is one of the leading causes of death with a functioning graft beyond the first year of kidney transplantation, and malignancies account for 8–10% of all deaths in the USA (2.6 deaths/1000 patient-years) and exceed 30% of deaths in Australia (5/1000 patient-years) in kidney transplant recipients. Patient-, transplant- and medication-related factors contribute to the increased cancer risk following kidney transplantation. While it is well established that the overall immunosuppressive dose is associated with an increased risk for cancer following transplantation, the contributive effect of different immunosuppressive agents is not well established. In this review we will discuss the different risk factors for malignancies after kidney transplantation.

Introduction

Malignancy is one of the most common causes of death in kidney transplant recipients [1, 2]. In kidney transplant recipients, the incidence of cancer is generally increased 2- to 3-fold compared with the general population [3, 4]. This increased cancer risk is not spread evenly over all types of cancers; while some cancer incidences are not increased (breast, prostate, ovarian, brain and cervical cancer), others are increased substantially (lung, colon, liver, lymphoma, melanoma and non-melanoma skin cancer). Cancer-related mortality rates are also higher in kidney transplant recipients compared with the general population [5].

Patient-, transplant- and medication-related factors contribute to the increased cancer risk following kidney transplantation. Immunosuppression is considered the most important risk factor, as it decreases the immunologic control of oncogenic viral infection and cancer immunosurveillance [4, 6]. Although it is accepted that the overall immunosuppressive dose is associated with the increased cancer risk following transplantation, the contributive effect of different immunosuppressive agents is not well established at this time. Currently available immunosuppressive agents influence different anticancer pathways and mammalian target of rapamycin (mTOR) inhibitors have been reported to have a decreased cancer risk compared with alternative immunosuppressive therapies. However, recent studies have not been able to demonstrate improved survival in kidney transplant recipients taking mTOR inhibitors. T cell–depleting agents are very potent immunosuppressive agents used as induction therapy and to treat acute rejection (AR) in kidney transplant recipients. While some studies have suggested an association between antibody induction and cancer after transplantation [7–11], others have failed to demonstrate this association [12–15].

Epidemiology and clinical presentation

Analyses from different registry data estimate the general increase in cancer incidence in kidney transplant recipients to be two- to three-fold compared with the general population [3, 4, 16–25]. Estimates of cancer incidence obtained from different registries differ widely, suggesting that data quality is problematic. This was confirmed in a recent study by Yanik et al. [26], who compared cancer diagnoses collected in the Scientific Registry of Transplant Recipients (SRTR) database with 15 linked cancer registries for colorectal, liver, lung, breast, prostate and kidney cancers, melanoma and non-Hodgkin lymphoma (NHL). They concluded that SRTR cancer data were strikingly incomplete, as only 36.8% of cancers were both registered in the SRTR database and cancer registries, whereas 47.5% of cancers were only documented in cancer registries and 15.7% were only documented in the SRTR database [26]. The estimated sensitivity for identifying cancer was only 52.5% for the SRTR and 84.3% for cancer registries [26].

Data from the USA concerning 175 732 solid organ transplant recipients (58.4% kidney transplant recipients) during the period 1987–2008 showed that the standardized incidence ratio (SIR) for cancer overall was 2.1 (95% CI 2.06–2.14) higher compared with the general population, with an excess absolute risk of 719 cancer cases per 100 000 person-years [3]. The majority of the patients included in these studies were kidney transplant recipients [27]. It is important to note that this increase is not uniform for all cancer types; some cancers are not increased following kidney transplantation, e.g. breast, prostate, ovarian, cervical and brain cancers [3, 4, 20], and the incidence of breast cancer might even be reduced [3, 28]. In contrast, lymphoma, lung cancer, colon cancer, melanoma and non-melanoma skin cancer and liver cancer are increased 2- to 4-fold. In a study by Engels et al. [3], skin cancer was the most common malignancy in solid organ transplant recipients, with a SIR for Kaposi sarcoma and non-melanoma skin cancer of 61.46 and 13.85, respectively. In addition, the SIRs for non-Hodgkin and Hodgkin lymphoma, liver cancer, gastrointestinal cancer and melanoma were also increased [3]. In more recent reports from both the Australia and New Zealand Dialysis and Transplant Registry (ANZDATA) registry [29] and European and North American registries [23], excluding non-melanocytic skin cancers, genitourinary tract cancers are the most frequent malignancies in renal transplant recipients.

In an analysis of the Collaborative Transplant Study (CTS) database, the incidence and impact of malignant lymphoma after solid organ transplantation in 195 938 solid organ transplant recipients (145 104 cadaveric kidney transplant recipients) between 1985 and 2001 were studied [30]. Over the 10-year observation period, the risk for malignant lymphoma in renal transplant recipients was 11.8-fold higher compared with a matched non-transplanted population, and most lymphomas occurred in the first post-transplant year [30]. Recent data suggest that from 2005 to 2010, the 5-year incidence of post-transplant lymphoproliferative disease (PTLD) in adult kidney transplant recipients has remained stable [31]. There was, however, a substantial decline in PTLD rates for paediatric recipients reported in patients transplanted from 2002 to 2012 compared with those transplanted from 2000 to 2009 [31]. In all groups, PTLD risk was highest in Epstein–Barr virus (EBV)-seronegative recipients [31]. In kidney transplant recipients, there is a slight predilection for the lymphoma to occur in the transplanted kidney. In addition, central nervous system lymphomas were most common after renal transplantation in the CTS [30].

On average, the age at diagnosis of post-transplant cancer is 40 years and the time from transplantation is 3–5 years [12, 28, 32]. However, these numbers vary substantially according to the cancer subtype, with lymphoma and Kaposi sarcoma occurring early after transplantation [30, 33] and epithelial cancers later on [33, 34]. Although in other types of solid organ transplantation cancer tends to occur in the transplanted organ, in kidney transplant recipients, kidney cancers almost exclusively occur in the native kidneys [1] and there is a greater incidence of papillary type relative to the general population [35]. Acquired cystic kidney disease is common in patients with advanced renal failure and is associated with the development of kidney cancer [25, 36]. In dialysis patients, the risks for thyroid cancer, myeloma and urinary tract cancers are increased, and this is mirrored in kidney transplant recipients [25]. This parallel between dialysis patients and kidney transplant recipients does not hold true for all cancer types, as ovarian and prostate cancer were less frequent in kidney transplant recipients than in the dialysis cohorts [23].

Pathogenesis and transplant-specific risk factors

Several factors have been linked to the increased incidence of malignancies among transplant recipients [6], including age, sun exposure, previous cancer, concomitant viral infection, cumulative dose of immunosuppression, type of immunosuppression, AR and the duration of pre-transplant dialysis (Table 1 and Figure 1) [38, 39]. Risk factors for patient death from cancer include male gender, a history of prior cancer and immunosuppression and lymphocyte-depleting antibodies [5].

Table 1.

Risk factor for post-transplant malignancies

Patient-related risk factorsRecipient age
Previous cancer
Sun exposure
Viral infection
Duration of dialysis
Transplant-related risk factorsDonor transmission
Donor type
Rejection
Medication-related risk factorsNet immunosuppression
Induction therapy
Maintenance therapy
Patient-related risk factorsRecipient age
Previous cancer
Sun exposure
Viral infection
Duration of dialysis
Transplant-related risk factorsDonor transmission
Donor type
Rejection
Medication-related risk factorsNet immunosuppression
Induction therapy
Maintenance therapy
Table 1.

Risk factor for post-transplant malignancies

Patient-related risk factorsRecipient age
Previous cancer
Sun exposure
Viral infection
Duration of dialysis
Transplant-related risk factorsDonor transmission
Donor type
Rejection
Medication-related risk factorsNet immunosuppression
Induction therapy
Maintenance therapy
Patient-related risk factorsRecipient age
Previous cancer
Sun exposure
Viral infection
Duration of dialysis
Transplant-related risk factorsDonor transmission
Donor type
Rejection
Medication-related risk factorsNet immunosuppression
Induction therapy
Maintenance therapy

Cancerogenesis following kidney transplantation (adapted from Riella [37]).
Fig. 1.

Cancerogenesis following kidney transplantation (adapted from Riella [37]).

Donor transmission

A variety of donor-transmitted malignancies have been documented, including melanoma and cancers of the lung, breast, colon, rectum and kidney, Kaposi sarcoma and glioblastoma multiforme. Donor transmission as a cause of post-transplant malignancy is a rare but dreaded event, as it might result in metastatic disease in the transplant recipient [40–47]. Reported transmission rates are <0.03%, but these are likely under-reported and underdiagnosed [41, 48, 49]. The most common transmitted cancer types are renal cancer, lung cancer, melanoma and lymphoma [46, 50, 51]. The risk of donor transmission depends on the type and extent of the original donor cancer. A donor history of melanoma, lung carcinoma or choriocarcinoma seems to be associated with high transmission risk and death and organs from such donors should not be accepted for transplantation [46]. In contrast, organs from donors with renal cell cancer without capsular invasion and central nervous system tumours (except medulloblastoma) are acceptable, as the risk seems to be low, reflecting the limited metastatic potential of these tumours [46, 52]. Regarding outcome, early donor-transmitted cancer (diagnosed ≤6 weeks of transplantation) was associated with a better outcome compared with late donor-transmitted cancer [51]; 5-year survival was 83% for kidney recipients with donor-transmitted cancer compared with 93% for recipients without donor-transmitted cancer (P = 0.077) [50, 51]. Recipients with transmitted renal cancers had the best outcomes, with >70% 2-year survival post-transplantation [50], while patients with melanoma and lung cancers had <50% 2-year survival post-transplantation [50].

Donor type

Differences in the type of transplant (living versus deceased) have been associated with cancer risk. In a study by Ma et al. [53], the overall risks for cancer were 1080, 1444 and 2018 per 100 000 patient-years for recipients of living donor, standard and expanded criteria deceased donor kidney recipients, respectively. This increased risk with different donor types was independent of age, sex, and time on dialysis [53]. Recipients of living-donor kidneys had a lower risk of cancer, particularly for genitourinary cancer and PTLD [53].

Recipient age and time on dialysis

Both in paediatric and adult kidney transplant recipients, recipient age has been identified as an independent risk factor of post–kidney transplant malignancies [54, 55]. With increasing recipient age, this is an important factor in the overall increasing incidence of post-transplant cancer in kidney transplant recipients. Time on dialysis before transplantation has also been identified as a risk factor for developing post-transplant malignancy. In a study based on the ANZDATA database, Wong et al. [38] reported a linear relationship between the duration of dialysis and the risk of solid organ cancer after transplantation, irrespective of recipient age. In a very interesting article, Yanik et al. [56] evaluated the incidence of cancer types depending on non-renal function interval (time on dialysis either on wait list or after transplant failure) or kidney function interval (time with a functioning graft and thus on immunosuppression), applying a linkage between the SRTR and several US cancer registries. While the incidence of infection-related and immune-related cancer (Kaposi sarcoma, NHL, lip cancer and non-epithelial skin cancer) was higher during kidney function intervals, end-stage renal disease (ESRD)-related cancer incidence (kidney cancer and thyroid cancer) was lower during kidney function intervals. Every change of status (non-renal function interval/kidney function interval) was associated with a changing incidence for NHL, melanoma, lung, pancreatic and non-epithelial skin cancers (higher during function intervals) and kidney and thyroid cancers (higher during non-function intervals), suggesting potent short-term effects of kidney dysfunction and immunosuppression on cancer incidence [56].

Previous cancer

A history of cancer prior to kidney transplantation in the recipient increases the risk of death by 30% [57].These findings were also confirmed in another study showing that kidney transplant recipients with a pre-transplant cancer are 3.7 times more likely to die of cancer post-transplantation [5]. Acuna et al. [58] performed an interesting meta-analysis including 32 cohort studies on solid organ transplant recipients with a pre-transplant malignancy in remission. They demonstrated that pre-transplant malignancy is associated with an increased risk of all-cause mortality (pooled hazard ratio 1.51), cancer-specific mortality (pooled hazard ratio 3.13) and of developing de novo malignancies (pooled hazard ratio 1.92) after transplantation compared with solid organ transplant recipients without a pre-transplant malignancy [58]. These studies clearly identify kidney transplant recipients with pre-transplant cancer as a high-risk patient population requiring tailored screening and management strategies.

Organ predilection

The incidence of specific malignancies varies according to the transplanted organ [3]. While in some types of transplantation (lung and liver), post-transplant malignancies tend to occur in the transplanted organ, in kidney transplantation this does not appear to be the case (kidney cancer in kidney transplant recipients primarily affects the native kidney) [3, 4, 16–23]. In addition, other cancer types vary depending on the transplanted organ. For example, the risk of NHL in lung transplant recipients is doubled compared with kidney, heart or liver transplant [3].

It is well established that kidney cancer is greatly increased in dialysis patients and kidney transplant recipients [3, 4, 16–23]. Prolonged time on dialysis has been identified as a risk factor for the development of kidney cancer [59, 60] and the incidence of kidney cancer can be as high as 100 times the expected incidence [61, 62]. While kidney cancer in native kidneys is frequent, cancer in the transplanted kidney is rare. In a European retrospective study, 20 patients were identified with kidney cancer in the transplanted kidney: 85% were papillary renal cell carcinoma (RCC) and 15% were clear cell RCC [63]. The tumours were small at the time of diagnosis and all patients were managed with ablation therapy (cryoablation or radiofrequent ablation) without a reduction or change in their immunosuppressive therapy [63].

Sun exposure

In the development of skin cancer, sun exposure is an established risk factor [64–66]. The application of sun block and administration of nicotinamide have both been demonstrated to reduce the incidence of non-melanoma skin cancer [67–70].

Viral infection

At least four viruses are believed to be co-carcinogenic in transplanted patients: EBV (Hodgkin’s and NHL), human herpesvirus 8 (HHV8; Kaposi sarcoma) [71–73], human papillomavirus (HPV; cervix, vulva, vagina, anus and some oro-pharynx cancers) and Merkel cell polyomavirus (Merkel cell skin carcinoma). EBV has conclusively been implicated in the pathogenesis of PTLD following kidney transplantation [74, 75] and EBV status is one of the most important risk factors for PTLD. More than 50% of PTLD cases are EBV related, and EBV mismatch between donor and recipient (an EBV-negative receptor engrafted with an EBV-positive donor) is associated with a 20-fold increased risk for PTLD [76–78]. Moreover, primary EBV infection post-transplant is a major risk factor for EBV-positive PTLD in early onset PTLD [15]. Additionally, other viruses have been associated with the development of cancer, e.g. hepatitis B and C (HBV and HCV; liver cancer) and BK polyomavirus (urological cancers) [79–87]. The central role of the immune system in the control of oncogenic viruses was emphasized by the findings of Grulich et al. [4], where a similar increase of virus-associated cancers was observed in solid organ transplant patients and patients with HIV/AIDS. As far as cytomegalovirus (CMV) and post-transplant malignancy are concerned, conflicting results have been reported [88–92], so at this time it is not clear whether CMV infection is associated with an increased risk of post-transplant cancer. A recent study demonstrated that cancer risk after kidney transplantation during childhood is particularly increased for virus-related cancers [54].

Rejection and treatment

As the total dose of immunosuppression is related to the risk of post-transplant malignancy, it is no surprise that rejection episodes and anti-rejection therapy are associated with the risk of post-transplant malignancy, as doses of maintenance immunosuppression including calcineurin inhibitors, antimetabolite and/or corticosteroids are often increased during the treatment of rejection, thereby contributing to increased T cell dysfunction [93]. Besides T cell dysfunction, systemic inflammation and concomitant release of cytokines and chemokines may promote malignant transformation [94, 95]. In the CTS, anti-rejection therapy with OKT3 or anti-thymocyte globulin (ATG) increased the overall cancer risk [96]. In a recent analysis of the ANZDATA, Lim et al. [29] studied the risk of incident cancer among kidney transplant recipients who have experienced AR, stratified by the use of T cell–depleting antibodies. The study included 7153 kidney transplant recipients transplanted between 1997 and 2009, of which 6.5% developed cancers. The risk for cancer after first kidney transplantation was significantly higher in patients experiencing AR treated with T cell–depleting antibodies (adjusted hazard ratio 1.42) compared with kidney transplant recipients not experiencing AR and the excess cancer risk was mainly confined to genitourinary tract cancers [29]. Also, treatment of rejection with high-dose steroids can adversely affect the risk for PTLD [97].

Maintenance immunosuppression

Maintenance immunosuppression is essential after kidney transplantation to prevent allograft rejection. Although it is accepted that overall immunosuppression dose is associated with an increased cancer risk following transplantation, the contributive effect of different immunosuppressive agents is not established. The mechanisms linking immunosuppression dose to the increased incidence of cancer are numerous and include decreased immune surveillance of tumours, decreased antiviral responses resulting in a specific increase of virus-induced tumours and possibly the direct carcinogenic effect of immunosuppressive drugs such as cyclosporine and azathioprine (Table 2) [6].

Table 2.

Immunosuppressive drugs and oncogenesis

Immunosuppressant agentMethod of actionRole in carcinogenesis
Calcineurin inhibitorInhibition of IL-2 production through binding and inhibition of cyclophilin (cyclosporine) and FKBP-12 (tacrolimus), respectively
  • Production of TGF-β [98, 99]

  • Production of VEGF [98, 100]

  • Production of interleukin-6 (IL-6) (promotion of EBV-induced B-cell growth) [101]

  • Promotion of invasive behaviour of non-transformed cells [98]

  • Reduced ability to repair radiation-induced DNA damage

  • Enhanced apoptotic effects of taxol and IFN-γ on human gastric and bladder cancer cells [102, 103]

  • Increased rate of lymfoproliferative disorders in HSV-infected mice [104]

AzathiopurineInhibition of DNA and RNA synthesis through incorporation of thiopurine analogues
  • Intercalation at the DNA level, inhibiting repair splicing and eliciting codon misreads [105]

  • Increased development of microsatellite DNA instability [106]

Mycophenolate mofetilInhibition of inosine monophosphate dehydrogenase and de novo purine biosynthesis
  • Anti-proliferative effect on leukaemia and solid tumour

  • Inhibition of adhesion molecules [107, 108–114]

  • Suppressed glycosylation and expression of several adhesion molecules [109, 115]

  • Inhibition of adhesion of colon adenocarcinoma cells to endothelial cells [116]

mTOR inhibitorsInhibition of mTOR pathway
  • Direct antitumour effect by inhibition of mTOR pathway [117, 118]

  • Inhibition of angiogenesis

  • Inhibition of p70 S6K: decreasing cancer cell proliferation [119, 120

  • Inhibition of interleukin-10: decreasing tumour cell JAK/STATs activity [120]

  • Inhibition of cyclins: blocking cell-cycle activity [121]

  • Decreased VEGF-A and VEGF-C signalling: impaired tumour angiogenesis [101, 119, 122, 123]

  • Inhibition of growth signals in PTLD-associated EBV + B-cell lymphomas [124]

  • Inhibition of replication of EBV-positive B cells, T cells and NK cells [125, 126]

  • Inhibition of ultraviolet B–induced metalloproteinase activation [127]

Immunosuppressant agentMethod of actionRole in carcinogenesis
Calcineurin inhibitorInhibition of IL-2 production through binding and inhibition of cyclophilin (cyclosporine) and FKBP-12 (tacrolimus), respectively
  • Production of TGF-β [98, 99]

  • Production of VEGF [98, 100]

  • Production of interleukin-6 (IL-6) (promotion of EBV-induced B-cell growth) [101]

  • Promotion of invasive behaviour of non-transformed cells [98]

  • Reduced ability to repair radiation-induced DNA damage

  • Enhanced apoptotic effects of taxol and IFN-γ on human gastric and bladder cancer cells [102, 103]

  • Increased rate of lymfoproliferative disorders in HSV-infected mice [104]

AzathiopurineInhibition of DNA and RNA synthesis through incorporation of thiopurine analogues
  • Intercalation at the DNA level, inhibiting repair splicing and eliciting codon misreads [105]

  • Increased development of microsatellite DNA instability [106]

Mycophenolate mofetilInhibition of inosine monophosphate dehydrogenase and de novo purine biosynthesis
  • Anti-proliferative effect on leukaemia and solid tumour

  • Inhibition of adhesion molecules [107, 108–114]

  • Suppressed glycosylation and expression of several adhesion molecules [109, 115]

  • Inhibition of adhesion of colon adenocarcinoma cells to endothelial cells [116]

mTOR inhibitorsInhibition of mTOR pathway
  • Direct antitumour effect by inhibition of mTOR pathway [117, 118]

  • Inhibition of angiogenesis

  • Inhibition of p70 S6K: decreasing cancer cell proliferation [119, 120

  • Inhibition of interleukin-10: decreasing tumour cell JAK/STATs activity [120]

  • Inhibition of cyclins: blocking cell-cycle activity [121]

  • Decreased VEGF-A and VEGF-C signalling: impaired tumour angiogenesis [101, 119, 122, 123]

  • Inhibition of growth signals in PTLD-associated EBV + B-cell lymphomas [124]

  • Inhibition of replication of EBV-positive B cells, T cells and NK cells [125, 126]

  • Inhibition of ultraviolet B–induced metalloproteinase activation [127]

Table 2.

Immunosuppressive drugs and oncogenesis

Immunosuppressant agentMethod of actionRole in carcinogenesis
Calcineurin inhibitorInhibition of IL-2 production through binding and inhibition of cyclophilin (cyclosporine) and FKBP-12 (tacrolimus), respectively
  • Production of TGF-β [98, 99]

  • Production of VEGF [98, 100]

  • Production of interleukin-6 (IL-6) (promotion of EBV-induced B-cell growth) [101]

  • Promotion of invasive behaviour of non-transformed cells [98]

  • Reduced ability to repair radiation-induced DNA damage

  • Enhanced apoptotic effects of taxol and IFN-γ on human gastric and bladder cancer cells [102, 103]

  • Increased rate of lymfoproliferative disorders in HSV-infected mice [104]

AzathiopurineInhibition of DNA and RNA synthesis through incorporation of thiopurine analogues
  • Intercalation at the DNA level, inhibiting repair splicing and eliciting codon misreads [105]

  • Increased development of microsatellite DNA instability [106]

Mycophenolate mofetilInhibition of inosine monophosphate dehydrogenase and de novo purine biosynthesis
  • Anti-proliferative effect on leukaemia and solid tumour

  • Inhibition of adhesion molecules [107, 108–114]

  • Suppressed glycosylation and expression of several adhesion molecules [109, 115]

  • Inhibition of adhesion of colon adenocarcinoma cells to endothelial cells [116]

mTOR inhibitorsInhibition of mTOR pathway
  • Direct antitumour effect by inhibition of mTOR pathway [117, 118]

  • Inhibition of angiogenesis

  • Inhibition of p70 S6K: decreasing cancer cell proliferation [119, 120

  • Inhibition of interleukin-10: decreasing tumour cell JAK/STATs activity [120]

  • Inhibition of cyclins: blocking cell-cycle activity [121]

  • Decreased VEGF-A and VEGF-C signalling: impaired tumour angiogenesis [101, 119, 122, 123]

  • Inhibition of growth signals in PTLD-associated EBV + B-cell lymphomas [124]

  • Inhibition of replication of EBV-positive B cells, T cells and NK cells [125, 126]

  • Inhibition of ultraviolet B–induced metalloproteinase activation [127]

Immunosuppressant agentMethod of actionRole in carcinogenesis
Calcineurin inhibitorInhibition of IL-2 production through binding and inhibition of cyclophilin (cyclosporine) and FKBP-12 (tacrolimus), respectively
  • Production of TGF-β [98, 99]

  • Production of VEGF [98, 100]

  • Production of interleukin-6 (IL-6) (promotion of EBV-induced B-cell growth) [101]

  • Promotion of invasive behaviour of non-transformed cells [98]

  • Reduced ability to repair radiation-induced DNA damage

  • Enhanced apoptotic effects of taxol and IFN-γ on human gastric and bladder cancer cells [102, 103]

  • Increased rate of lymfoproliferative disorders in HSV-infected mice [104]

AzathiopurineInhibition of DNA and RNA synthesis through incorporation of thiopurine analogues
  • Intercalation at the DNA level, inhibiting repair splicing and eliciting codon misreads [105]

  • Increased development of microsatellite DNA instability [106]

Mycophenolate mofetilInhibition of inosine monophosphate dehydrogenase and de novo purine biosynthesis
  • Anti-proliferative effect on leukaemia and solid tumour

  • Inhibition of adhesion molecules [107, 108–114]

  • Suppressed glycosylation and expression of several adhesion molecules [109, 115]

  • Inhibition of adhesion of colon adenocarcinoma cells to endothelial cells [116]

mTOR inhibitorsInhibition of mTOR pathway
  • Direct antitumour effect by inhibition of mTOR pathway [117, 118]

  • Inhibition of angiogenesis

  • Inhibition of p70 S6K: decreasing cancer cell proliferation [119, 120

  • Inhibition of interleukin-10: decreasing tumour cell JAK/STATs activity [120]

  • Inhibition of cyclins: blocking cell-cycle activity [121]

  • Decreased VEGF-A and VEGF-C signalling: impaired tumour angiogenesis [101, 119, 122, 123]

  • Inhibition of growth signals in PTLD-associated EBV + B-cell lymphomas [124]

  • Inhibition of replication of EBV-positive B cells, T cells and NK cells [125, 126]

  • Inhibition of ultraviolet B–induced metalloproteinase activation [127]

The cumulative immunosuppressive dose (net immunosuppressive dose for the entire life) is associated with the risk for cancer post-transplant. For example, patients previously treated with immunosuppression for primary glomerular disease [128] or for AR [29] are at higher risk to develop cancer. Hibberd et al. [128] reported an association between pre-transplantation immunosuppression and increased risk for four cancer groups: anogenital cancer, NHL, breast cancer and urinary tract cancer (excluding kidney). Grulich et al. [4] analyzed seven studies of people with HIV/AIDS (n = 444 172) and five of transplant recipients (n = 31 977) for 20 of the 28 types of cancers. A significantly increased cancer incidence was found in both populations, and most cancers that occurred at increased rates involved oncogenic viruses (e.g. EBV, HHV8, HPV, HBV and HCV). The rates of most common epithelial cancers (breast or prostate cancer) were not increased [4]. The similarity of the pattern of increased risk of cancer in the two populations suggests that it is immune deficiency rather than other risk factors for cancer that is responsible for the increased risk. Of note, there were also some discrepancies noted, as some cancer types (thyroid, kidney, melanoma and bladder cancers) were increased in the transplant population but not in the HIV/AIDS cohorts.

Although results suggest that currently available immunosuppressive agents influence different anticancer pathways [101], it is not clear whether currently used medications such as cyclosporine, tacrolimus, azathioprine or mycophenolate are associated with different cancer risks [14, 129–131]. mTOR inhibitors have been reported to have less cancer risk compared with alternative immunosuppressive therapies [132]; however, in a recent systematic review, decreased cancer incidence in kidney transplant recipients treated with mTOR inhibitors did not result in improved overall survival [133]. As induction therapy is concerned, interleukin-2 (IL-2) receptor antagonist (IL-2Ra) induction does not appear to be associated with an increase in cancer [39], whereas some studies find a small increase in cancer and cancer death with lymphocyte-depleting antibodies [5, 134, 135]. Moreover, there appear to be differences in the different types of lymphocyte-depleting antibodies.

Induction therapy

Multiple agents have been used as induction therapy at the time of kidney transplantation [e.g. OKT3/muromonab, polyclonal lymphocyte-depleting antibodies, anti-IL-2 receptor (CD25) antibodies and alemtuzumab (anti-CD52). As both CD4+ and CD8+ T cells are crucial in adaptive antiviral immunity, depletion of both populations of T cells with T cell–depleting antibodies would increase the susceptibility of individuals to a higher risk of virus-associated diseases [136]. Direct anti-tumour effects have also been attributed to CD4+ T helper 1 cells, CD8+ cytotoxic T cells and natural killer (NK) cells [137]. Polyclonal T cell–depleting antibodies target a variety of T and NK cell-derived antigens, including CD2, CD3, CD4, CD8 and CD16, but also markers expressed by leucocytes, B cells and plasma cells, which may explain the predisposition to infections and cancer complications associated with the use of these agents [138–140].

The immunosuppressive potency of OKT3 is greater than that of polyclonal lymphocyte–depleting agents and the use of OKT3 has clearly been associated with an increase in lymphoma risk [14, 141–143]. OKT3 is no longer commercially available, but other forms of induction therapy are still currently in use and from the late 1990s onwards, rabbit ATG (rATG) became the most commonly used polyclonal agent in the USA [144, 145], and later worldwide [146, 107].

Polyclonal induction therapy: When evaluating the cancer-inducing effect of different types of polyclonal lymphocyte-depleting agents, the data are limited and hard to interpret. Available registry analyses have often combined all polyclonal lymphocyte-depleting agents into one category and often span multiple decades. Combining different types of induction agents (e.g. polyclonal induction agents or ATG) is problematic, as there are clear differences in the risk of PTLD associated with different preparations [141, 147]. Furthermore, over time the type of lymphocyte-depleting agent, the average dose of rATG and the type and dose of concomitant immunosuppressive agents have changed significantly. During the 1980s and early 1990s, OKT3 and non-rATG preparations were most widely used [144, 145], and this was associated with a marked increase in the incidence of PTLD [148, 149]. From the late 1990s onwards, rATG became the most commonly used polyclonal agent in the USA [144, 145], and later worldwide [146]. Finally, in the 1980s rATG dosing was markedly higher than it is now (e.g. total dose 14 mg/kg versus 6 mg/kg now) [150] and it has been demonstrated that higher rATG dosing is associated with a higher risk of PTLD [13].

Earlier studies have suggested an association of induction therapy with T cell–depleting antibodies with an increased risk of PTLD. In an analysis of the CTS, the SIR of lymphoma compared with a similar non-transplant population was higher with T cell–depleting antibody induction as compared with IL-2Ra or no induction therapy [143]. Also, a study of the SRTR and the United States Renal Data System databases reported similar results (70% increased risk of PTLD in renal transplant recipients receiving monoclonal and/or polyclonal T cell–depleting antibodies as induction therapy) [10, 14]. Also, an earlier analysis of the ANZDATA registry demonstrated that the use of T cell–depleting antibodies (as induction or as treatment for rejection) was associated with a more than two-fold increased risk of early onset NHL after transplantation [9]. Registry database studies reported results regarding rATG use and the occurrence of PTLD have been mixed. Only three studies looked at rATG specifically and two found an increased risk for PTLD while one did not [10, 11, 151]. Other registry studies of PTLD risk have grouped multiple lymphocyte-depleting induction agents together for the purpose of analysis, in some cases including OKT3 [14, 152–155]. Three prospective randomized trials followed patients up to 5 years after kidney transplantation [156–158]. The incidence of PTLD and the follow-up time were too limited to allow for meaningful conclusions [159–162]. Finally, a systematic review by Marks et al. [13] evaluated the rate of PTLD in recipients of kidney or heart allografts and pointed to the importance of antiviral prophylaxis, as in this study; the absence of antiviral prophylaxis was the greatest risk factor for the development of PTLD rather than the use of induction therapy.

IL-2R antagonist induction: In the CTS, induction therapy with polyclonal and IL-2Ra induction was not associated with significant increases in the risk of PTLD when compared with no induction therapy [14]. However, universal use of IL-2Ra induction is increasingly questioned, as it does not provide benefit in low-risk kidney transplant recipients compared with no induction therapy, while being inferior compared with ATG in high-risk kidney transplant recipients [161, 163–165]. In a recent observation study, rATG was associated with a decreased risk of adverse outcomes (including mortality) compared with alemtuzumab and basiliximab as induction therapy [166].

Alemtuzumab: In the Transplant Cancer Match study, the use of alemtuzumab as induction therapy was associated with a 79% increase in NHL, a 2.5-fold increase in colorectal cancer and 3-fold increase in thyroid cancer after transplantation [142]. Other studies have reported mixed results regarding the use of alemtuzumab and PTLD; although one study did not find an association [11], another using a more recent Organ Procurement and Transplantation Network cohort did [78]. A recent study in small bowel allograft recipients receiving alemtuzumab demonstrated earlier onset of lymphoplasmacytic hyperplasia, the most indolent form of B lymphocyte clonal expansion, compared with patients receiving the IL-2Ra induction agent daclizumab [167].

Maintenance therapy

Calcineurin inhibitors: In kidney transplant recipients, both cyclosporine and tacrolimus are associated with an increased risk of malignancy [101]. In a French prospective randomized study involving 231 renal allograft recipients, low-dose (75–125 ng/mL) cyclosporine was associated with a lower incidence of secondary cancers (particularly skin cancers) compared with normal-dose (150–250 ng/mL) cyclosporine at a median of 66 months follow-up [168]. Some evidence suggested a higher risk for PTLD under tacrolimus versus cyclosporine [169, 170]. However, subsequent analyses by the same group postulated that this was the result of a lack of experience with the agent and overaggressive dosing at the time of introduction of tacrolimus in clinical practice. Ultimately, reduced tacrolimus trough levels led to substantial declines in the risk of PTLD [171]. An analysis of the CTS demonstrated that cyclosporine did not confer added risk for the development of NHL compared with azathioprine/steroid treatment, whereas treatment with FK506 increased the risk approximately 2-fold [96].

Azathioprine: The use of azathioprine has long been recognized as an a etiologic factor in the development of neoplasia, especially in the development of late non-melanoma skin malignancies (particularly squamous cell cancer) [23, 101, 172, 173]. Furthermore, azathioprine is associated with the development of myelodysplastic syndrome [106].

Mycophenolate mofetil: Some patient studies have suggested that the risk of developing malignancies is decreased with the use of mycophenolate mofetil [107, 174, 175], while other studies could not demonstrate a reduction in cancer incidence with mycophenolate- versus non-mycophenolate-based therapy [174]. An SRTR analysis reported that the introduction of mycophenolate mofetil was associated with the greatest decrease in relative risk for the development of PTLD [14]. When patient outcomes during different eras of immunosuppression were compared, the use of MMF was also found to be associated with a reduction in the incidence of PTLD [9, 174, 176]. In patients, the principal anti-tumour mechanism associated with mycophenolate mofetil use may be due to the decreased incidence of AR.

mTOR inhibitors: While for most classes of immunosuppressive agents there is a dose-dependent relationship between the dosage of the immunosuppressive agent and secondary malignancies, this does not hold true for mTOR inhibitors such as sirolimus and everolimus. In humans, evidence suggests that sirolimus may confer a decreased risk of malignancy compared with other immunosuppressive medications [101, 133, 177–181]. Case reports and case series have reported that in renal transplant recipients with Kaposi sarcoma, switching from cyclosporine to sirolimus resulted in total resolution of the Kaposi sarcoma [179, 182, 183]. In the TUMORAPA study, where patients with a history of squamous cell carcinoma were studied, conversion to sirolimus significantly reduced the risk for relapse when compared with those who were maintained on calcineurin inhibitor–based therapy [184]. For non-melanoma skin cancer, Campbell et al. [185] reported that conversion to sirolimus after 1-year post-transplant resulted in a lower yearly incidence rate of non-melanoma skin cancer and also a lower incidence of new or recurrent non-melanoma skin cancer. Individual studies regarding the incidence of cancer associated with the use of sirolimus have been conflicting [177, 186–193]. In a study linking the US SRTR database with 15 population-based cancer registries and national pharmacy claims, cancer incidence in 32 604 sirolimus-exposed and sirolimus non-exposed kidney transplant recipients was studied. The incidence of prostate cancer was higher during sirolimus use (hazard ratio 1.86), while the incidence of other cancers was similar or lower, with a 26% decrease in overall cancer incidence excluding prostate carcinoma (hazard ratio 0.74). The authors postulate that the increase in prostate cancer diagnosis is due to sirolimus, effects on screen detection. In addition, two meta-analyses have been published demonstrating a lower overall cancer incidence with the use of sirolimus [133, 194]. In the meta-analysis of Knoll et al. [133], including 21 randomized controlled trials with patient-level data from 5876 patients, it was demonstrated that sirolimus was associated with a 40% reduction in malignancy risk and a 56% reduction in the risk of non-melanoma skin cancer (0.44, 0.30 to 0.63) compared with controls. This effect is restricted to patients converting to sirolimus from another immunosuppressive regimen, as analysis of de novo sirolimus trials revealed no difference in malignancy risk between sirolimus and controls [133]. Moreover, in a study analyzing Medicare claims data for transplant recipients, de novo use of sirolimus was associated with a 22% increased risk of PTLD [195]. Remarkably, in the meta-analysis of Knoll et al. [133], the decreased risk of cancer development was associated with an increased overall mortality risk due to cardiovascular and infection-related deaths. The authors speculate that increased sirolimus-induced cardiovascular risk factors (anaemia, proteinuria, hyperglycaemia and hyperlipidemia) and overimmunosuppression with sirolimus might have contributed to these findings [133]. Based on these studies, universal sirolimus use in kidney transplant recipients cannot be recommended at this time.

Belatacept: For the co-stimulation blocker belatacept, an inhibitor of T cell proliferation, PTLD risk appears similar to that seen under calcineurin inhibitor therapy [196], however, belatacept is contraindicated in EBV-seronegative recipients. Although initially a number of PTLDs of the central nervous system were reported in patients treated with belatacept [197, 198], follow-up data of both the BENEFIT study and, more recently, a Phase 2 study where low immunologic risk patients were switched from calcineurin inhibitor therapy to belatacept showed a mild albeit small increase in post-transplant malignancies [199–202].

Screening

Since cancer before transplantation increases the risk of post-transplant malignancy, guidelines have been developed outlining waiting times for different types and stages of cancer (Table 3). A systematic review by Batabyal et al. [207] concluded that none of the available recommendations are backed by strong evidence. We recommend seeking an expert oncologist's opinion regarding cancer-free survival, patient life expectancies and optimal cancer surveillance. Clinicians have to realize that even longer waiting times do not eliminate the risk for cancer recurrence and cancer-related death [57]. In a Swedish population-based cohort of solid organ transplant recipients, the increased rate of death was greatest for patients with waiting times of ≤5 years but persisted with waiting times of >10 years among recipients with prior aggressive cancer types (gastrointestinal, breast, kidney/urothelial and hematologic malignancies) [57].

Table 3.

Recommendations for waiting times after cancer [203–206]

Decisions regarding waiting time should be individualized and it should be expalined to all patients with a history of cancer that they are at increased risk of de novo malignancy post-transplantation. Recommended waiting periods before transplantation do not guarantee no recurrence of malignancy after transplantation
Absolute contraindication for transplantation
  • Uncontrolled or untreated malignancies

  • Multiple myeloma

  • Advanced breast cancer (Stage III or IV)

  • Colorectal cancer (Stage D)

  • Advanced prostate cancer (Grade 4 or 5, T3c, T4, N+, M+)

No waiting time
  • Superficial bladder cancer

  • Non-metastatic, basal cell carcinoma

  • Prostatic cancer microscopic (focal, microscopic low-grade (Gleason’s grade ≤3), low risk) (T1a, T1c) disease

  • Incidentally discovered T1 renal cell carcinoma (with no suspicious histological features)

  • Monoclonal gammopathy of undetermined significance

2-year waiting time
  • Invasive bladder cancer

  • In situ breast cancer

  • Localized cervical cancer

  • Duke’s Stage A and B1 colorectal cancer

  • Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, post-transplant lymphoproliferative disorder, leukaemia

  • In situ melanoma

  • Lung cancer

  • Prostatic cancer

  • Testicular cancer

  • Thyroid cancer

  • Wilm’s tumour (a 1-year waiting period might be acceptable)

5-year waiting time
  • Stage II breast cancer

  • Extensive cervical cancer and non-in situ cancer of the uterus

  • Colorectal cancer Stage C

  • Melanoma

  • Large or invasive or symptomatic renal cell carcinoma

Decisions regarding waiting time should be individualized and it should be expalined to all patients with a history of cancer that they are at increased risk of de novo malignancy post-transplantation. Recommended waiting periods before transplantation do not guarantee no recurrence of malignancy after transplantation
Absolute contraindication for transplantation
  • Uncontrolled or untreated malignancies

  • Multiple myeloma

  • Advanced breast cancer (Stage III or IV)

  • Colorectal cancer (Stage D)

  • Advanced prostate cancer (Grade 4 or 5, T3c, T4, N+, M+)

No waiting time
  • Superficial bladder cancer

  • Non-metastatic, basal cell carcinoma

  • Prostatic cancer microscopic (focal, microscopic low-grade (Gleason’s grade ≤3), low risk) (T1a, T1c) disease

  • Incidentally discovered T1 renal cell carcinoma (with no suspicious histological features)

  • Monoclonal gammopathy of undetermined significance

2-year waiting time
  • Invasive bladder cancer

  • In situ breast cancer

  • Localized cervical cancer

  • Duke’s Stage A and B1 colorectal cancer

  • Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, post-transplant lymphoproliferative disorder, leukaemia

  • In situ melanoma

  • Lung cancer

  • Prostatic cancer

  • Testicular cancer

  • Thyroid cancer

  • Wilm’s tumour (a 1-year waiting period might be acceptable)

5-year waiting time
  • Stage II breast cancer

  • Extensive cervical cancer and non-in situ cancer of the uterus

  • Colorectal cancer Stage C

  • Melanoma

  • Large or invasive or symptomatic renal cell carcinoma

Table 3.

Recommendations for waiting times after cancer [203–206]

Decisions regarding waiting time should be individualized and it should be expalined to all patients with a history of cancer that they are at increased risk of de novo malignancy post-transplantation. Recommended waiting periods before transplantation do not guarantee no recurrence of malignancy after transplantation
Absolute contraindication for transplantation
  • Uncontrolled or untreated malignancies

  • Multiple myeloma

  • Advanced breast cancer (Stage III or IV)

  • Colorectal cancer (Stage D)

  • Advanced prostate cancer (Grade 4 or 5, T3c, T4, N+, M+)

No waiting time
  • Superficial bladder cancer

  • Non-metastatic, basal cell carcinoma

  • Prostatic cancer microscopic (focal, microscopic low-grade (Gleason’s grade ≤3), low risk) (T1a, T1c) disease

  • Incidentally discovered T1 renal cell carcinoma (with no suspicious histological features)

  • Monoclonal gammopathy of undetermined significance

2-year waiting time
  • Invasive bladder cancer

  • In situ breast cancer

  • Localized cervical cancer

  • Duke’s Stage A and B1 colorectal cancer

  • Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, post-transplant lymphoproliferative disorder, leukaemia

  • In situ melanoma

  • Lung cancer

  • Prostatic cancer

  • Testicular cancer

  • Thyroid cancer

  • Wilm’s tumour (a 1-year waiting period might be acceptable)

5-year waiting time
  • Stage II breast cancer

  • Extensive cervical cancer and non-in situ cancer of the uterus

  • Colorectal cancer Stage C

  • Melanoma

  • Large or invasive or symptomatic renal cell carcinoma

Decisions regarding waiting time should be individualized and it should be expalined to all patients with a history of cancer that they are at increased risk of de novo malignancy post-transplantation. Recommended waiting periods before transplantation do not guarantee no recurrence of malignancy after transplantation
Absolute contraindication for transplantation
  • Uncontrolled or untreated malignancies

  • Multiple myeloma

  • Advanced breast cancer (Stage III or IV)

  • Colorectal cancer (Stage D)

  • Advanced prostate cancer (Grade 4 or 5, T3c, T4, N+, M+)

No waiting time
  • Superficial bladder cancer

  • Non-metastatic, basal cell carcinoma

  • Prostatic cancer microscopic (focal, microscopic low-grade (Gleason’s grade ≤3), low risk) (T1a, T1c) disease

  • Incidentally discovered T1 renal cell carcinoma (with no suspicious histological features)

  • Monoclonal gammopathy of undetermined significance

2-year waiting time
  • Invasive bladder cancer

  • In situ breast cancer

  • Localized cervical cancer

  • Duke’s Stage A and B1 colorectal cancer

  • Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, post-transplant lymphoproliferative disorder, leukaemia

  • In situ melanoma

  • Lung cancer

  • Prostatic cancer

  • Testicular cancer

  • Thyroid cancer

  • Wilm’s tumour (a 1-year waiting period might be acceptable)

5-year waiting time
  • Stage II breast cancer

  • Extensive cervical cancer and non-in situ cancer of the uterus

  • Colorectal cancer Stage C

  • Melanoma

  • Large or invasive or symptomatic renal cell carcinoma

The optimal cancer screening strategy to detect post-transplant cancers in an early stage is not defined (Table 4). In general, many experts recommend using general practice guidelines in kidney transplant recipients [208–215]. Several centres routinely screen for native kidney cancer, as the risk for kidney cancer is greatly increased in both the dialysis and kidney transplant populations [3, 4, 16–23, 216, 217]. In a medical decision analysis, screening for kidney cancer in all transplant recipients would have a small benefit at relatively high cost [218]. However, directed screening using ultrasound in those with documented acquired cystic kidney disease or those with a previous cancer in a contralateral kidney might be cost effective. Modelling studies by Wong et al. [219, 220] suggest that screening for colon and cervical cancer would be cost effective in the kidney transplant population. In a population-based cohort of Ontario between 1997 and 2010, 77.5, 69.8 and 91.4% of eligible solid organ transplant recipients were not up to date with colorectal, cervical and breast cancer screening, respectively [221]. Solid organ transplant recipients with fewer co-morbidities, assessment by a primary care provider and continuity of care by a transplant specialist at a transplant centre were associated with higher rates of becoming screen up to date in this study [221].

Table 4.

Recommendations for post-transplant cancer screening

GuidelineSkin cancerCervical cancerBreast cancerColorectal cancerProstate cancerLiverLymphomaOthers
AST kidney 2000 [208]Self-screening AnnuallyAnnually including PAPEvery 1–2 yearsAs in general populationAnnually PSA and DREEvery 6–12 months AFP and USClinically every 3 months in first yearAnnually prostate, lung and bladder
EBPG 2002 [209]Annually including PAPRecommendedRecommendedAnnually PSA and DREAnnually prostate and kidney
KDIGO 2009 [210]Self-screening AnnuallyAs in general populationAs in general populationAs in general populationAnnual AFP and US
NKF 2009 [211]Annually
CST and CSN 2010 [212]Self-screening AnnuallyAnnually PAPAs in general populationAs in general populationAnnual AFP and US
RA 2011 [213]Self-screening AnnuallyAs in general populationAs in general populationAs in general population
KHA-CARI 2012 [214]Self-screening AnnuallyAnnually
GuidelineSkin cancerCervical cancerBreast cancerColorectal cancerProstate cancerLiverLymphomaOthers
AST kidney 2000 [208]Self-screening AnnuallyAnnually including PAPEvery 1–2 yearsAs in general populationAnnually PSA and DREEvery 6–12 months AFP and USClinically every 3 months in first yearAnnually prostate, lung and bladder
EBPG 2002 [209]Annually including PAPRecommendedRecommendedAnnually PSA and DREAnnually prostate and kidney
KDIGO 2009 [210]Self-screening AnnuallyAs in general populationAs in general populationAs in general populationAnnual AFP and US
NKF 2009 [211]Annually
CST and CSN 2010 [212]Self-screening AnnuallyAnnually PAPAs in general populationAs in general populationAnnual AFP and US
RA 2011 [213]Self-screening AnnuallyAs in general populationAs in general populationAs in general population
KHA-CARI 2012 [214]Self-screening AnnuallyAnnually

DRE, digital rectal examination; AFP, Alpha fetoprotein; US, Ultra sound examination; PAP, PAP-mean

Table 4.

Recommendations for post-transplant cancer screening

GuidelineSkin cancerCervical cancerBreast cancerColorectal cancerProstate cancerLiverLymphomaOthers
AST kidney 2000 [208]Self-screening AnnuallyAnnually including PAPEvery 1–2 yearsAs in general populationAnnually PSA and DREEvery 6–12 months AFP and USClinically every 3 months in first yearAnnually prostate, lung and bladder
EBPG 2002 [209]Annually including PAPRecommendedRecommendedAnnually PSA and DREAnnually prostate and kidney
KDIGO 2009 [210]Self-screening AnnuallyAs in general populationAs in general populationAs in general populationAnnual AFP and US
NKF 2009 [211]Annually
CST and CSN 2010 [212]Self-screening AnnuallyAnnually PAPAs in general populationAs in general populationAnnual AFP and US
RA 2011 [213]Self-screening AnnuallyAs in general populationAs in general populationAs in general population
KHA-CARI 2012 [214]Self-screening AnnuallyAnnually
GuidelineSkin cancerCervical cancerBreast cancerColorectal cancerProstate cancerLiverLymphomaOthers
AST kidney 2000 [208]Self-screening AnnuallyAnnually including PAPEvery 1–2 yearsAs in general populationAnnually PSA and DREEvery 6–12 months AFP and USClinically every 3 months in first yearAnnually prostate, lung and bladder
EBPG 2002 [209]Annually including PAPRecommendedRecommendedAnnually PSA and DREAnnually prostate and kidney
KDIGO 2009 [210]Self-screening AnnuallyAs in general populationAs in general populationAs in general populationAnnual AFP and US
NKF 2009 [211]Annually
CST and CSN 2010 [212]Self-screening AnnuallyAnnually PAPAs in general populationAs in general populationAnnual AFP and US
RA 2011 [213]Self-screening AnnuallyAs in general populationAs in general populationAs in general population
KHA-CARI 2012 [214]Self-screening AnnuallyAnnually

DRE, digital rectal examination; AFP, Alpha fetoprotein; US, Ultra sound examination; PAP, PAP-mean

Treatment of post-transplant cancer

In general, a reduction in immunosuppression is recommended or kidney transplant recipients upon cancer diagnosis. In the above-mentioned study of Yanik et al. [56], it was noted that the incidence of infection-related cancers was higher and the incidence of ESRD-related cancers was lower during kidney function intervals (time on immunosuppression), suggesting that a reduction of immunosuppression affects different cancer types differently. Similar findings were reported by van Leeuwen et al. [222], who performed a population-based retrospective cohort study and compared the cancer incidence in kidney transplant recipients during periods of transplant function (and immunosuppression) and after transplant failure (when immunosuppression is ceased or reduced) [222]. The SIRs for NHL, lip cancer and melanoma were significantly elevated during periods of transplant function. For leukaemia and lung carcinoma, SIRs remained elevated after transplant failure, while the SIRs for kidney/urinary tract and thyroid cancers significantly increased after transplant failure. These data suggest that while the effect of immunosuppression on cancer risk is rapidly reversible for some cancers (mainly infectious-related cancers), this does not hold true for other cancer types (ESRD-related cancers) [222]. Some centres convert patients with non-melanoma skin cancer to mTOR therapy, as randomized clinical trials have shown fewer skin cancers in mTOR-treated patients [184, 185]. Also, for other solid and hematologic cancers, mTOR inhibitors have had marginal success [223, 224]. However, routine conversion to mTOR inhibitors to improve outcomes in all cancers or to prevent long-term cancer development in all solid organ transplant recipients is not widely practiced at this time, as data are lacking to support this practice.

Outcome

Data suggest that cancer as a cause of death is on the rise. For example, in Australia and New Zealand, cardiovascular deaths are decreasing while cancer mortality is increasing [1]. Malignancy accounts for 8–10% of all deaths in the USA (2.6 deaths/1000 patient-years) and >30% of deaths in Australia (5/1000 patient-years) [1, 2]. The data regarding standardized mortality rates (SMRs) have been conflicting. While some studies have suggested that the cancer-related SMR has increased with the same magnitude as the SIR in transplant recipients [224], other studies have shown a more nuanced picture [5]. In a study from Hong Kong, the cancer SIR and SMR in kidney transplant recipients were very similar (2.9 and 2.3, respectively) [225]; high SMRs were associated with lymphoma, leukaemia, kidney, colon, lung, bladder, melanoma and stomach cancers, while lymphoma, liver, colorectal and lung were associated with excess absolute risk of >25 deaths/100 000 patient-years [225]. In a US registry analysis by Kiberd et al. [5], no overall excess mortality was observed in kidney transplant recipients. Cancer SMRs varied substantially with age group; cancer SMRs were 23-fold and 4.4-fold higher in patients <20 years and 20–39 years of age, respectively, while cancer SMRs were lower in patients >60 years of age [5]. The cancer death rates were >500/100 000 patient-years for patients >60 years of age compared with 13/100 000 patient-years for patients 20–39 years of age [5]. So in older patients who are at the highest risk to die from cancer, there is no increased risk to die from cancer in kidney transplant recipients. More specific data are available concerning post-transplant lymphoma. The 1-year survival in cadaveric kidney transplant recipients developing lymphoma was 60% and showed little improvement over the study period, while the 5-year survival was ∼40% [30]. Interestingly, in this analysis the time of lymphoma development after transplantation did not influence survival: 5-year survival in kidney transplantation with lymphoma development <90 days post-transplant and >365 days post-transplant was 41.4% and 37.0%, respectively [30]. Post-transplant lymphoma with lymph node involvement had a good prognosis while disseminated disease had a poor prognosis [30].

Conclusion

Malignancy is one of the most common causes of death in kidney transplant recipients. In general, the cancer incidence in solid organ transplant recipients is increased 2- to 3-fold compared with the general population [3, 4]. Moreover, cancer-related mortality rates are also higher in solid organ transplant recipients compared with the general population [5]. Several risk factors for post-transplantation cancer development have been identified and immunosuppression is considered the most important risk factor, as it decreases the immunologic control of oncogenic viral infection and immunosurveillance. Currently available immunosuppressive agents influence different anticancer pathways and mTOR inhibitors seem to have a favourable profile in this respect. However, the increased mortality associated with mTOR inhibitor use in a recent meta-analysis argues against their universal use in renal allograft recipients or switching to mTOR inhibition in all patients with post-transplant malignancies. Intense collaboration between nephrologists and oncologists is needed in this field to design safer immunosuppressive regimens and define optimal screening and treatment strategies in kidney transplant recipients.

Funding

No funding was involved in the preparation of this Manuscript.

References

1

Pilmore
H
,
Dent
H
,
Chang
S
et al. .
Reduction in cardiovascular death after kidney transplantation
.
Transplantation
2010
;
89
:
851
857

2

Collins
AJ
,
Foley
RN
,
Chavers
B
et al. .
United States Renal Data System 2011 annual data report: atlas of chronic kidney disease & end-stage renal disease in the United States
.
Am J Kidney Dis
2012
;
59
:
e1
e420

3

Engels
EA
,
Pfeiffer
RM
,
Fraumeni
JF
Jr
et al. .
Spectrum of cancer risk among US solid organ transplant recipients
.
JAMA
2011
;
306
:
1891
1901

4

Grulich
AE
,
van Leeuwen
MT
,
Falster
MO
et al. .
Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis
.
Lancet
2007
;
370
:
59
67

5

Kiberd
BA
,
Rose
C
,
Gill
JS.
Cancer mortality in kidney transplantation
.
Am J Transplant
2009
;
9
:
1868
1875

6

Stallone
G
,
Infante
B
,
Grandaliano
G.
Management and prevention of post-transplant malignancies in kidney transplant recipients
.
Clin Kidney J
2015
;
8
:
637
644

7

Caillard
S
,
Dharnidharka
V
,
Agodoa
L
et al. .
Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression
.
Transplantation
2005
;
80
:
1233
1243

8

Hibberd
AD
,
Trevillian
PR
,
Wlodarzcyk
JH
et al. .
Cancer risk associated with ATG/OKT3 in renal transplantation
.
Transplant Proc
1999
;
31
:
1271
1272

9

van Leeuwen
MT
,
Grulich
AE
,
Webster
AC
et al. .
Immunosuppression and other risk factors for early and late non-Hodgkin lymphoma after kidney transplantation
.
Blood
2009
;
114
:
630
637

10

Bustami
RT
,
Ojo
AO
,
Wolfe
RA
et al. .
Immunosuppression and the risk of post-transplant malignancy among cadaveric first kidney transplant recipients
.
Am J Transplant
2004
;
4
:
87
93

11

Kirk
AD
,
Cherikh
WS
,
Ring
M
et al. .
Dissociation of depletional induction and posttransplant lymphoproliferative disease in kidney recipients treated with alemtuzumab
.
Am J Transplant
2007
;
7
:
2619
2625

12

Pedotti
P
,
Cardillo
M
,
Rossini
G
et al. .
Incidence of cancer after kidney transplant: results from the North Italy transplant program
.
Transplantation
2003
;
76
:
1448
1451

13

Marks
WH
,
Ilsley
JN
,
Dharnidharka
VR.
Posttransplantation lymphoproliferative disorder in kidney and heart transplant recipients receiving thymoglobulin: a systematic review
.
Transplant Proc
2011
;
43
:
1395
1404

14

Cherikh
WS
,
Kauffman
HM
,
McBride
MA
et al. .
Association of the type of induction immunosuppression with posttransplant lymphoproliferative disorder, graft survival, and patient survival after primary kidney transplantation
.
Transplantation
2003
;
76
:
1289
1293

15

Quinlan
SC
,
Pfeiffer
RM
,
Morton
LM
et al. .
Risk factors for early-onset and late-onset post-transplant lymphoproliferative disorder in kidney recipients in the United States
.
Am J Hematol
2011
;
86
:
206
209

16

Collett
D
,
Mumford
L
,
Banner
NR
et al. .
Comparison of the incidence of malignancy in recipients of different types of organ: a UK Registry audit
.
Am J Transplant
2010
;
10
:
1889
1896

17

Villeneuve
PJ
,
Schaubel
DE
,
Fenton
SS
et al. .
Cancer incidence among Canadian kidney transplant recipients
.
Am J Transplant
2007
;
7
:
941
948

18

Webster
AC
,
Craig
JC
,
Simpson
JM
et al. .
Identifying high risk groups and quantifying absolute risk of cancer after kidney transplantation: a cohort study of 15,183 recipients
.
Am J Transplant
2007
;
7
:
2140
2151

19

Adami
J
,
Gabel
H
,
Lindelof
B
et al. .
Cancer risk following organ transplantation: a nationwide cohort study in Sweden
.
Br J Cancer
2003
;
89
:
1221
1227

20

Maisonneuve
P
,
Agodoa
L
,
Gellert
R
et al. .
Cancer in patients on dialysis for end-stage renal disease: an international collaborative study
.
Lancet
1999
;
354
:
93
99

21

Kyllonen
L
,
Salmela
K
,
Pukkala
E.
Cancer incidence in a kidney-transplanted population
.
Transpl Int
2000
;
13
:
S394
S398

22

Li
WH
,
Chen
YJ
,
Tseng
WC
et al. .
Malignancies after renal transplantation in Taiwan: a nationwide population-based study
.
Nephrol Dial Transplant
2012
;
27
:
833
839

23

Kasiske
BL
,
Snyder
JJ
,
Gilbertson
DT
et al. .
Cancer after kidney transplantation in the United States
.
Am J Transplant
2004
;
4
:
905
913

24

Vajdic
CM
,
van Leeuwen
MT
,
McDonald
SP
et al. .
Increased incidence of squamous cell carcinoma of eye after kidney transplantation
.
J Natl Cancer Inst
2007
;
99
:
1340
1342

25

Stewart
JH
,
Vajdic
CM
,
van Leeuwen
MT
et al. .
The pattern of excess cancer in dialysis and transplantation
.
Nephrol Dial Transplant
2009
;
24
:
3225
3231

26

Yanik
EL
,
Nogueira
LM
,
Koch
L
et al. .
Comparison of cancer diagnoses between the US solid organ transplant registry and linked central cancer registries
.
Am J Transplant
2016
;
16
:
2986
2993

27

Lanza
LL
,
Wang
L
,
Simon
TA
et al. .
Epidemiologic critique of literature on post-transplant neoplasms in solid organ transplantation
.
Clin Transplant
2009
;
23
:
582
588

28

Stewart
T
,
Tsai
SC
,
Grayson
H
et al. .
Incidence of de-novo breast cancer in women chronically immunosuppressed after organ transplantation
.
Lancet
1995
;
346
:
796
798

29

Lim
WH
,
Turner
RM
,
Chapman
JR
,
Ma
MK
et al. .
Acute rejection, T-cell-depleting antibodies, and cancer after transplantation
.
Transplantation
2014
;
97
:
817
825

30

Opelz
G
,
Dohler
B.
Lymphomas after solid organ transplantation: a collaborative transplant study report
.
Am J Transplant
2004
;
4
:
222
230

31

Kotton
CN
,
Huprikar
S
,
Kumar
D.
Transplant infectious diseases: a review of the scientific registry of transplant recipients published data
.
Am J Transplant
2017
;
17
:
1439
1446

32

Saeian
K
,
Franco
J
,
Komorowski
RA
et al. .
Hepatocellular carcinoma after renal transplantation in the absence of cirrhosis or viral hepatitis: a case series
.
Liver Transpl Surg
1999
;
5
:
46
49

33

Euvrard
S
,
Kanitakis
J
,
Claudy
A.
Skin cancers after organ transplantation
.
N Engl J Med
2003
;
348
:
1681
1691

34

Penn
I.
De novo malignances in pediatric organ transplant recipients
.
Pediatr Transplant
1998
;
2
:
56
63

35

Karami
S
,
Yanik
EL
,
Moore
LE
et al. .
Risk of renal cell carcinoma among kidney transplant recipients in the United States
.
Am J Transplant
2016
;
16
:
3479
3489

36

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
.
J Am Soc Nephrol
2003
;
14
:
197
207

37

Riella
LV.
Malignancy after kidney transplantation
.
Kidney Transplant eBook
2015
, version 1.
3
, pp.
172
178

38

Wong
G
,
Turner
RM
,
Chapman
JR
et al. .
Time on dialysis and cancer risk after kidney transplantation
.
Transplantation
2013
;
95
:
114
121

39

Webster
AC,
,
Ruster
LP,
,
McGee
R
et al. .
Interleukin 2 receptor antagonists for kidney transplant recipients
.
Cochrane Database Syst Rev
2010
;
1
:
1
146

40

Penn
I.
Malignant melanoma in organ allograft recipients
.
Transplantation
1996
;
61
:
274
278

41

Myron
KH
,
McBride
MA
,
Cherikh
WS
et al. .
Transplant tumour registry: donor related malignancies
.
Transplantation
2002
;
74
:
358
362

42

Birkeland
SA
,
Storm
HH.
Risk for tumour and other disease transmission by transplantation: a population-based study of unrecognized malignancies and other diseases in organ donors
.
Transplantation
2002
;
74
:
1409
1413

43

Pedotti
P
,
Poli
F
,
Longhi
E
et al. .
Epidemiologic study on the origin of cancer after kidney transplantation
.
Transplantation
2004
;
77
:
426
428

44

Armanios
MY
,
Grossman
SA
,
Yang
SC
et al. .
Transmission of glioblastoma multiforme following bilateral lung transplantation from an affected donor: case study and review of the literature
.
Neuro-Oncology
2004
;
6
:
259
263

45

Penn
I.
Donor transmitted disease: cancer
.
Transplant Proc
1991
;
23
:
2629
2631

46

Buell
JF
,
Trofe
J
,
Hanaway
MJ
et al. .
Transmission of donor cancer into cardiothoracic transplant recipients
.
Surgery
2001
;
130
:
660
666

47

Nalesnik
MA
,
Woodle
ES
,
Dimaio
JM
et al. .
Donor-transmitted malignancies in organ transplantation: assessment of clinical risk
.
Am J Transplant
2011
;
11
:
1140
1147

48

Feng
S
,
Buell
JF
,
Cherikh
WS
et al. .
Organ donors with positive viral serology or malignancy: risk of disease transmission by transplantation
.
Transplantation
2002
;
74
:
1657
1663

49

Kauffman
HM.
The United Network for Organ Sharing position on using donors with primary central nervous system malignancies
.
Transplantation
2005
;
79
:
622
623

50

Xiao
D
,
Craig
JC
,
Chapman
JR
et al. .
Donor cancer transmission in kidney transplantation: a systematic review
.
Am J Transplant
2013
;
13
:
2645
2652

51

Desai
R
,
Collett
D
,
Watson
CJ
et al. .
Cancer transmission from organ donors-unavoidable but low risk
.
Transplantation
2012
;
94
:
1200
1207

52

Kauffman
HM
,
McBride
MA
,
Delmonico
FL.
First report of the United Network for Organ Sharing Transplant Tumour Registry: donors with a history of cancer
.
Transplantation
2000
;
70
:
1747
1751

53

Ma
MK
,
Lim
WH
,
Turner
RM
et al. .
The risk of cancer in recipients of living-donor, standard and expanded criteria deceased donor kidney transplants: a registry analysis
.
Transplantation
2014
;
98
:
1286
1293

54

Francis
A,
,
Johnson
DW,
,
Craig
JC
et al. .
Incidence and predictors of cancer following kidney transplantation in childhood
.
Am J Transplant
2017
;
17
:
2650
2658

55

Farrugia
D
,
Mahboob
S
,
Cheshire
J
et al. .
Malignancy-related mortality following kidney transplantation is common
.
Kidney Int
2014
;
85
:
1395
1403

56

Yanik
EL
,
Clarke
CA
,
Snyder
JJ.
Variation in cancer incidence among patients with ESRD during kidney function and nonfunction intervals
.
J Am Soc Nephrol
2016
;
27
:
1495
1504

57

Brattstrom
C
,
Granath
F
,
Edgren
G
et al. .
Overall and cause-specific mortality in transplant recipients with a pretransplantation cancer history
.
Transplantation
2013
;
96
:
297
305

58

Acuna
SA
,
Huang
JW
,
Daly
C
et al. .
Outcomes of solid organ transplant recipients with preexisting malignancies in remission: a systematic review and meta-analysis
.
Transplantation
2017
;
101
:
471
481

59

Hiesse
C
,
Rieu
P
,
Kriaa
F
et al. .
. Malignancy after renal transplantation: analysis of incidence and risk factors in 1700 patients followed during a 25-year period
.
Transplant Proc
1997
;
29
:
831
833

60

Muruve
NA
,
Shoskes
DA.
Genitourinary malignancies in solid organ transplant recipients
.
Transplantation
2005
;
80
:
709
716

61

Doublet
JD
,
Peraldi
MN
,
Gattegno
B
et al. .
Renal cell carcinoma of native kidneys: prospective study of 129 renal transplant patients
.
J Urol
1997
;
158
:
42
44

62

Denton
MD
,
Magee
CC
,
Ovuworie
C
et al. .
Prevalence of renal cell carcinoma in patients with ESRD pre-transplantation: a pathologic analysis
.
Kidney Int
2002
;
61
:
2201
2209

63

Cornelis
F
,
Buy
X
,
Andre
M
et al. .
De novo renal tumours arising in kidney transplants: midterm outcome after percutaneous thermal ablation
.
Radiology
2011
;
260
:
900
907

64

Yarosh
DB.
DNA repair, immunosuppression, and skin cancer
.
Cutis
2004
;
74
:
10
13

65

Kricker
A
,
Armstrong
BK
,
English
DR.
Sun exposure and non-melanocytic skin cancer
.
Cancer Causes Control
1994
;
5
:
367
392

66

Armstrong
BK
,
Kricker
A.
The epidemiology of UV-induced skin cancer
.
J Photochem Photobiol B
2001
;
63
:
8
18

67

Thompson
SC
,
Jolley
D
,
Marks
R.
Reduction of solar keratoses by regular sunscreen use
.
N Engl J Med
1993
;
329
:
1147
1151

68

Green
A
,
Williams
G
,
Neale
R
et al. .
Daily sunscreen application and betacarotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the skin: a randomised controlled trial
.
Lancet
1999
;
354
:
723
729

69

Green
AC
,
Williams
GM
,
Logan
V
et al. .
Reduced melanoma after regular sunscreen use: randomized trial follow-up
.
J Clin Oncol
2011
;
29
:
257
263

70

Chen
AC
,
Martin
AJ
,
Choy
B
et al. .
A phase 3 randomized trial of nicotinamide for skin-cancer chemoprevention
.
N Engl J Med
2015
;
373
:
1618
1626

71

Sodhi
A
,
Chaisuparat
R
,
Hu
J
et al. .
The TSC2/mTOR pathway drives endothelial cell transformation induced by the Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor
.
Cancer Cell
2006
;
10
:
133
143

72

Montaner
S
,
Sodhi
A
,
Ramsdell
AK
et al. .
The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor as a therapeutic target for the treatment of Kaposi's sarcoma
.
Cancer Res
2006
;
66
:
168
174

73

Hosseini-Moghaddam
SM
,
Soleimanirahbar
A
,
Mazzulli
T
et al. .
Post renal transplantation Kaposi's sarcoma: a review of its epidemiology, pathogenesis, diagnosis, clinical aspects, and therapy
.
Transpl Infect Dis
2012
;
14
:
338
345

74

Young
LS
,
Rickinson
AB.
Epstein-Barr virus: 40 years on
.
Nat Rev Cancer
2004
;
4
:
757
768

75

Grywalska
E
,
Rolinski
J.
Epstein-Barr virus-associated lymphomas
.
Semin Oncol
2015
;
42
:
291
303

76

Walker
RC
,
Paya
CV
,
Marshall
WF
et al. .
Pretransplantation seronegative Epstein-Barr virus status is the primary risk factor for posttransplantation lymphoproliferative disorder in adult heart, lung, and other solid organ transplantations
.
J Heart Lung Transplant
1995
;
14
:
214
221

77

Sampaio
MS
,
Cho
YW
,
Shah
T
et al. .
Impact of Epstein-Barr virus donor and recipient serostatus on the incidence of post-transplant lymphoproliferative disorder in kidney transplant recipients
.
Nephrol Dial Transplant
2012
;
27
:
2971
2979

78

Dharnidharka
VR
,
Lamb
KE
,
Gregg
JA
et al. .
Associations between EBV serostatus and organ transplant type in PTLD risk: an analysis of the SRTR National Registry Data in the United States
.
Am J Transplant
2012
;
12
:
976
983

79

Kenan
DJ
,
Mieczkowski
PA
,
Burger-Calderon
R
et al. .
The oncogenic potential of BK-polyomavirus is linked to viral integration into the human genome
.
J Pathol
2015
;
237
:
379
389

80

Kenan
DJ
,
Mieczkowski
PA,
,
Latulippe
E.
Polyomavirus genomic integration and large T antigen expression: evolving paradigms in human oncogenesis
.
Am J Transplant
2017
;
17
:
1674
1680

81

Liu
S
,
Chaudhry
MR
,
Berrebi
AA
et al. .
Polyomavirus replication and smoking are independent risk factors for bladder cancer after renal transplantation
.
Transplantation
2017
;
101
:
1488
1494

82

Papadimitriou
JC
,
Randhawa
P
,
Rinaldo
CH
et al. .
BK polyomavirus infection and renourinary tumorigenesis
.
Am J Transplant
2016
;
16
:
398
406

83

Oikawa
M
,
Hatakeyama
S
,
Fujita
T
et al. .
BK virus-associated urothelial carcinoma of a ureter graft in a renal transplant recipient: a case report
.
Transplant Proc
2014
;
46
:
616
619

84

Yan
L
,
Salama
ME
,
Lanciault
C
et al. .
Polyomavirus large T antigen is prevalent in urothelial carcinoma post-kidney transplant
.
Hum Pathol
2016
;
48
:
122
131

85

Nickeleit
V
,
Singh
HK
,
Goldsmith
CS
et al. .
BK virus-associated urinary bladder carcinoma in transplant recipients: productive or nonproductive polyomavirus infections in tumor cells?
Hum Pathol
2013
;
44
:
2870
2871

86

Kausman
JY
,
Somers
GR
,
Francis
DM
et al. .
Association of renal adenocarcinoma and BK virus nephropathy post transplantation
.
Pediatr Nephrol
2004
;
19
:
459
462

87

Emerson
LL
,
Carney
HM
,
Layfield
LJ
et al. .
Collecting duct carcinoma arising in association with BK nephropathy post-transplantation in a pediatric patient. A case report with immunohistochemical and in situ hybridization study
.
Pediatr Transplant
2008
;
12
:
600
605

88

Couzi
L
,
Levaillant
Y
,
Jamai
A
et al. .
Cytomegalovirus-induced γδ T cells associate with reduced cancer risk after kidney transplantation
.
J Am Soc Nephrol
2010
;
21
:
181
188

89

Courivaud
C
,
Bamoulid
J
,
Gaugler
B
et al. .
Cytomegalovirus exposure, immune exhaustion and cancer occurrence in renal transplant recipients
.
Transpl Int
2012
;
25
:
948
955

90

Desai
R
,
Collett
D
,
Watson
CJ
et al. .
Impact of cytomegalovirus on long-term mortality and cancer risk after organ transplantation
.
Transplantation
2015
;
99
:
1989
1994

91

Wong
G
,
Chakera
A
,
Chapman
JR
et al. .
Cytomegalovirus and cancer after kidney transplantation: Role of the human leukocyte antigen system?
Transpl Infect Dis
2017
;
19
:
10

92

Verghese
PS
,
Schmeling
DO
,
Knight
JA.
Valganciclovir administration to kidney donors to reduce the burden of cytomegalovirus and Epstein-Barr virus transmission during transplantation
.
Transplantation
2015
;
99
:
1186
1191

93

Halloran
PF.
Immunosuppressive drugs for kidney transplantation
.
N Engl J Med
2004
;
351
:
2715
2729

94

Borsig
L
,
Wolf
MJ
,
Roblek
M
et al. .
Inflammatory chemokines and metastasis–tracing the accessory
.
Oncogene
2014
;
33
:
3217
3224

95

Eiro
N
,
Vizoso
FJ.
Inflammation and cancer
.
World J Gastrointest Surg
2012
;
4
:
62
72

96

Opelz
G
,
Henderson
R.
Incidence of non-Hodgkin lymphoma in kidney and heart transplant recipients
.
Lancet
1993
;
342
:
1514
1516

97

Kremers
WK
,
Devarbhavi
HC
,
Wiesner
RH
et al. .
Post-transplant lymphoproliferative disorders following liver transplantation: incidence, risk factors and survival
.
Am J Transplant
2006
;
6
:
1017
1024

98

Hojo
M
,
Morimoto
T
,
Maluccio
M
et al. .
Cyclosporine induces cancer progression by a cell-autonomous mechanism
.
Nature
1999
;
397
:
530
534

99

Herman
M
,
Weinstein
T
,
Korzets
A
et al. .
Effect of cyclosporin A on DNA repair and cancer incidence in kidney transplant recipients
.
J Lab Clin Med
2001
;
137
:
14
20

100

Shihab
FS
,
Bennett
WM
,
Isaac
J.
Nitric oxide modulates vascular endothelial growth factor and receptors in chronic cyclosporine nephrotoxicity
.
Kidney Int
2003
;
63
:
522
533

101

Guba
M
,
Graeb
C
,
Jauch
KW.
Pro- and anti-cancer effects of immunosuppressive agents used in organ transplantation
.
Transplantation
2004
;
77
:
1777
1782

102

Morisaki
T
,
Matsunaga
H
,
Beppu
K
et al. .
A combination of cyclosporin-A (CsA) and interferon-gamma (INF-gamma) induces apoptosis in human gastric carcinoma cells
.
Anticancer Res
2000
;
20
:
3363
3373

103

Nomura
T
,
Yamamoto
H
,
Mimata
H
et al. .
Enhancement by cyclosporin A of taxol-induced apoptosis of human urinary bladder cancer cells
.
Urol Res
2002
;
30
:
102
111

104

Mistrikova
J
,
Mrmusova
M
,
Durmanova
V
et al. .
Increased neoplasm development due to immunosuppressive treatment with FK-506 in BALB/C mice persistently infected with the mouse herpesvirus (MHV-72)
.
Viral Immunol
1999
;
12
:
237
247

105

Swann
PF
,
Waters
TR
,
Moulton
DC
et al. .
Role of postreplicative DNA mismatch repair in the cytotoxic action of thioguanine
.
Science
1996
;
273
:
1109
1111

106

Offman
J
,
Opelz
G
,
Doehler
B
et al. .
Defective DNA mismatch repair in acute myeloid leukemia/myelodysplastic syndrome after organ transplantation
.
Blood
2004
;
104
:
822
828

107

Buell
JF
,
Gross
TG
,
Woodle
ES.
Malignancy after transplantation
.
Transplantation
2005
;
80
:
S254
S264

108

Nagai
M
,
Natsumeda
Y
,
Konno
Y
et al. .
Selective up-regulation of type II inosine 5′-monophosphate dehydrogenase messenger RNA expression in human leukemias
.
Cancer Res
1991
;
51
:
3886
3890

109

Engl
T
,
Makarevic
J
,
Relja
B
et al. .
Mycophenolate mofetil modulates adhesion receptors of the beta1 integrin family on tumor cells: impact on tumor recurrence and malignancy
.
BMC Cancer
2005
;
5
:
4

110

Weber
G
,
Hager
JC
,
Lui
MS
et al. .
Biochemical programs of slowly and rapidly growing human colon carcinoma xenografts
.
Cancer Res
1981
;
41
:
854
859

111

Jackson
RC
,
Weber
G
,
Morris
HP.
IMP dehydrogenase, an enzyme linked with proliferation and malignancy
.
Nature
1975
;
256
:
331
333

112

Yu
J
,
Lemas
V
,
Page
T
et al. .
Induction of erythroid differentiation in K562 cells by inhibitors of inosine monophosphate dehydrogenase
.
Cancer Res
1989
;
49
:
5555
5560

113

Ohsugi
Y
,
Suzuki
S
,
Takagaki
Y.
Antitumor and immunosuppressive effects of mycophenolic acid derivatives
.
Cancer Res
1976
;
36
:
2923
2927

114

Carter
SB
,
Franklin
TJ
,
Jones
DF
et al. .
Mycophenolic acid: an anti-cancer compound with unusual properties
.
Nature
1969
;
223
:
848
850

115

Heemann
U
,
Azuma
H
,
Hamar
P
et al. .
Mycophenolate mofetil inhibits lymphocyte binding and the upregulation of adhesion molecules in acute rejection of rat kidney allografts
.
Transpl Immunol
1996
;
4
:
64
67

116

Leckel
K
,
Beecken
WD
,
Jonas
D
et al. .
The immunosuppressive drug mycophenolate mofetil impairs the adhesion capacity of gastrointestinal tumour cells
.
Clin Exp Immunol
2003
;
134
:
238
245

117

Vignot
S
,
Faivre
S
,
Aguirre
D
et al. .
mTOR-targeted therapy of cancer with rapamycin derivatives
.
Ann Oncol
2005
;
16
:
525
537

118

Muthukkumar
S
,
Ramesh
TM
,
Bondada
S.
Rapamycin, a potent immunosuppressive drug, causes programmed cell death in B lymphoma cells
.
Transplantation
1995
;
60
:
264
270

119

Luan
FL
,
Ding
R
,
Sharma
VK
et al. .
Rapamycin is an effective inhibitor of human renal cancer metastasis
.
Kidney Int
2003
;
63
:
917
926

120

Nepomuceno
RR
,
Balatoni
CE
,
Natkunam
Y
et al. .
Rapamycin inhibits the interleukin 10 signal transduction pathway and the growth of Epstein Barr virus B-cell lymphomas
.
Cancer Res
2003
;
63
:
4472
4480

121

Garcia-Morales
P
,
Hernando
E
,
Carrasco-Garcia
E
et al. .
Cyclin D3 is down-regulated by rapamycin in HER-2-overexpressing breast cancer cells
.
Mol Cancer Ther
2006
;
5
:
2172
2181

122

Guba
M
,
von
BP
,
Steinbauer
M
et al. .
Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor
.
Nat Med
2002
;
8
:
128
135

123

Huber
S
,
Bruns
CJ
,
Schmid
G
et al. .
Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis
.
Kidney Int
2007
;
71
:
771
777

124

Krams
SM
,
Martinez
OM.
Epstein-Barr virus, rapamycin, and host immune responses
.
Curr Opin Organ Transplant
2008
;
13
:
563
568

125

Kawada
J
,
Ito
Y
,
Iwata
S
et al. .
mTOR inhibitors induce cell-cycle arrest and inhibit tumor growth in Epstein-Barr virus-associated T and natural killer cell lymphoma cells
.
Clin Cancer Res
2014
;
20
:
5412
5422

126

Adamson
AL
,
Le
BT
,
Siedenburg
BD.
Inhibition of mTORC1 inhibits lytic replication of Epstein-Barr virus in a cell-type specific manner
.
Virol J
2014
;
11
:
110
111

127

Brenneisen
P
,
Sies
H
,
Scharffetter-Kochanek
K.
Ultraviolet-B irradiation and matrix metalloproteinases: from induction via signaling to initial events
.
Ann N Y Acad Sci
2002
;
973
:
31
43

128

Hibberd
AD
,
Trevillian
PR
,
Wlodarczyk
JH
et al. .
Effect of immunosuppression for primary renal disease on the risk of cancer in subsequent renal transplantation: a population-based retrospective cohort study
.
Transplantation
2013
;
95
:
122
127

129

Marcen
R.
Immunosuppressive drugs in kidney transplantation: impact on patient survival, and incidence of cardiovascular disease, malignancy and infection
.
Drugs
2009
;
69
:
2227
2243

130

Gallagher
MP
,
Kelly
PJ
,
Jardine
M
et al. .
Long-term cancer risk of immunosuppressive regimens after kidney transplantation
.
J Am Soc Nephrol
2010
;
21
:
852
858

131

Knight
SR
,
Russell
NK
,
Barcena
L
et al. .
Mycophenolate mofetil decreases acute rejection and may improve graft survival in renal transplant recipients when compared with azathioprine: a systematic review
.
Transplantation
2009
;
87
:
785
794

132

Kauffman
HM
,
Cherikh
WS
,
Cheng
Y.
Maintenance immunosuppression with target-of-rapamycin inhibitors is associated with a reduced incidence of de novo malignancies
.
Transplantation
2005
;
80
:
883
889

133

Knoll
GA
,
Kokolo
MB
,
Mallick
R
et al. .
Effect of sirolimus on malignancy and survival after kidney transplantation: systematic review and meta-analysis of individual patient data
.
BMJ
2014
;
349
:
g6679

134

Kauffman
HM
,
Cherikh
WS
,
McBride
MA.
Post-transplant de novo malignancies in renal transplant recipients: the past and present
.
Transpl Int
2006
;
19
:
607
620

135

Sampaio
MS
,
Cho
YW
,
Qazi
Y
et al. .
Posttransplant malignancies in solid organ adult recipients: an analysis of the U.S. National Transplant Database
.
Transplantation
2012
;
94
:
990
998

136

Sant
AJ
,
McMichael
A.
Revealing the role of CD4+ T cells in viral immunity
.
J Exp Med
2012
;
209
:
1391
1395

137

Lakshmi
NB
,
Eshvendar
RK
,
Shantikumar
S
et al. .
Immune system: a double-edged sword in cancer
.
Inflamm Res
2013
;
62
:
823
834

138

Zand
MS.
B-cell activity of polyclonal antithymocyte globulins
.
Transplantation
2006
;
82
:
1387
1395

139

Hardinger
KL.
Rabbit antithymocyte globulin induction therapy in adult renal transplantation
.
Pharmacotherapy
2006
;
26
:
1771
1783

140

Midtvedt
K
,
Fauchald
P
,
Lien
B
et al. .
Individualized T cell monitored administration of ATG versus OKT3 in steroid-resistant kidney graft rejection
.
Clin Transplant
2003
;
17
:
69
74

141

Opelz
G
,
Naujokat
C
,
Daniel
V
et al. .
Disassociation between risk of graft loss and risk of non-Hodgkin lymphoma with induction agents in renal transplant recipients
.
Transplantation
2006
;
81
:
1227
1233

142

Hall
EC
,
Engels
EA
,
Pfeiffer
RM
et al. .
Association of antibody induction immunosuppression with cancer after kidney transplantation
.
Transplantation
2015
;
99
:
1051
1057

143

Swinnen
LJ
,
Fisher
RI.
OKT3 monoclonal antibodies induce interleukin-6 and interleukin-10: a possible cause of lymphoproliferative disorders associated with transplantation
.
Curr Opin Nephrol Hypertens
1993
;
2
:
670
678

144

Shapiro
R
,
Young
JB
,
Milford
EL
et al. .
Immunosuppression: evolution in practice and trends, 1993-2003
.
Am J Transplant
2005
;
5
:
874
886

145

Meier-Kriesche
HU
,
Li
S
et al. .
Immunosuppression: evolution in practice and trends, 1994-2004
.
Am J Transplant
2006
;
6
:
1111
1131

146

Markmann
JF
,
Fishman
JA.
Alemtuzumab in kidney-transplant recipients
.
N Engl J Med
2011
;
364
:
1968
1969

147

Dharnidharka
VR.
Post-transplant lymphoproliferative disease: association with induction therapy?
Drugs
2006
;
66
:
429
438

148

Dharnidharka
VR
,
Tejani
AH
,
Ho
PL
et al. .
Post-transplant lymphoproliferative disorder in the United States: young Caucasian males are at highest risk
.
Am J Transplant
2002
;
2
:
993
998

149

Dharnidharka
VR
,
Sullivan
EK
,
Stablein
DM
et al. .
Risk factors for posttransplant lymphoproliferative disorder (PTLD) in pediatric kidney transplantation: a report of the North American Pediatric Renal Transplant Cooperative Study (NAPRTCS)
.
Transplantation
2001
;
71
:
1065
1068

150

Mohty
M
,
Bacigalupo
A
,
Saliba
F
et al. .
New directions for rabbit antithymocyte globulin (Thymoglobulin®) in solid organ transplants, stem cell transplants and autoimmunity
.
Drugs
2014
;
74
:
1605
1634

151

Dharnidharka
VR
,
Stevens
G.
Risk for post-transplant lymphoproliferative disorder after polyclonal antibody induction in kidney transplantation
.
Pediatr Transplant
2005
;
9
:
622
626

152

Kasiske
BL
,
Kukla
A
,
Thomas
D
et al. .
Lymphoproliferative disorders after adult kidney transplant: epidemiology and comparison of registry report with claims-based diagnoses
.
Am J Kidney Dis
2011
;
58
:
971
980

153

Faull
RJ
,
Hollett
P
,
McDonald
SP.
Lymphoproliferative disease after renal transplantation in Australia and New Zealand
.
Transplantation
2005
;
80
:
193
197

154

Gajarski
RJ
,
Blume
ED
,
Urschel
S
et al. .
Infection and malignancy after pediatric heart transplantation: the role of induction therapy
.
J Heart Lung Transplant
2011
;
30
:
299
308

155

Caillard
S
,
Lamy
FX
,
Quelen
C
et al. .
Epidemiology of posttransplant lymphoproliferative disorders in adult kidney and kidney pancreas recipients: report of the French registry and analysis of subgroups of lymphomas
.
Am J Transplant
2012
;
12
:
682
693

156

Brennan
DC
,
Daller
JA
,
Lake
KD
et al. .
Rabbit antithymocyte globulin versus basiliximab in renal transplantation
.
N Engl J Med
2006
;
355
:
1967
1977

157

Brennan
DC
,
Flavin
K
,
Lowell
JA
et al. .
A randomized, double-blinded comparison of Thymoglobulin versus ATGAM for induction immunosuppressive therapy in adult renal transplant recipients
.
Transplantation
1999
;
67
:
1011
1018

158

Mourad
G
,
Rostaing
L
,
Legendre
C
et al. .
Sequential protocols using basiliximab versus antithymocyte globulins in renal-transplant patients receiving mycophenolate mofetil and steroids
.
Transplantation
2004
;
78
:
584
590

159

Hardinger
KL
,
Schnitzler
MA
,
Miller
B
et al. .
Five-year follow up of thymoglobulin versus ATGAM induction in adult renal transplantation
.
Transplantation
2004
;
78
:
136
141

160

Noel
C
,
Abramowicz
D
,
Durand
D
et al. .
Daclizumab versus antithymocyte globulin in high-immunological-risk renal transplant recipients
.
J Am Soc Nephrol
2009
;
20
:
1385
1392

161

Hellemans
R
,
Hazzan
M
,
Durand
D
et al. .
Daclizumab versus rabbit antithymocyte globulin in high-risk renal transplants: five-year follow-up of a randomized study
.
Am J Transplant
2015
;
15
:
1923
1932

162

Brennan
DC
,
Schnitzler
MA.
Long-term results of rabbit antithymocyte globulin and basiliximab induction
.
N Engl J Med
2008
;
359
:
1736
1738

163

Hellemans
R
,
Bosmans
JL
,
Abramowicz
D.
Induction therapy for kidney transplant recipients: do we still need anti-il2 receptor monoclonal antibodies?
Am J Transplant
2017
;
17
:
22
27

164

Tanriover
B
,
Zhang
S
,
MacConmara
M
et al. .
Induction therapies in live donor kidney transplantation on tacrolimus and mycophenolate with or without steroid maintenance
.
Clin J Am Soc Nephrol
2015
;
10
:
1041
1049

165

Tanriover
B
,
Jaikaransingh
V
,
MacConmara
MP
et al. .
Acute rejection rates and graft outcomes according to induction regimen among recipients of kidneys from deceased donors treated with tacrolimus and mycophenolate
.
Clin J Am Soc Nephrol
2016
;
11
:
1650
1661

166

Koyawala
N
,
Silber
JH
,
Rosenbaum
PR
et al. .
Comparing outcomes between antibody induction therapies in kidney transplantation
.
J Am Soc Nephrol
2017
;
28
:
2188
2200

167

Ruiz
P
,
Soares
MF
,
Garcia
M
et al. .
Lymphoplasmacytic hyperplasia (possibly pre-PTLD) has varied expression and appearance in intestinal transplant recipients receiving Campath immunosuppression
.
Transplant Proc
2004
;
36
:
386
387

168

Dantal
J
,
Hourmant
M
,
Cantarovich
D
et al. .
Effect of long-term immunosuppression in kidney-graft recipients on cancer incidence: randomised comparison of two cyclosporin regimens
.
Lancet
1998
;
351
:
623
628

169

Sampaio
MS
,
Cho
YW
,
Shah
T
et al. .
Association of immunosuppressive maintenance regimens with posttransplant lymphoproliferative disorder in kidney transplant recipients
.
Transplantation
2012
;
93
:
73
81

170

Dayton
JD
,
Richmond
ME
,
Weintraub
RG
et al. .
Role of immunosuppression regimen in post-transplant lymphoproliferative disorder in pediatric heart transplant patients
.
J Heart Lung Transplant
2011
;
30
:
420
425

171

Dharnidharka
VR
,
Ho
PL
,
Stablein
DM
et al. .
Mycophenolate, tacrolimus and post-transplant lymphoproliferative disorder: a report of the North American Pediatric Renal Transplant Cooperative Study
.
Pediatr Transplant
2002
;
6
:
396
399

172

Taylor
L
,
Hughes
RA
,
McPherson
K.
The risk of cancer from azathioprine as a treatment for multiple sclerosis
.
Eur J Neurol
2004
;
11
:
141

173

Beauparlant
P
,
Papp
K
,
Haraoui
B.
The incidence of cancer associated with the treatment of rheumatoid arthritis
.
Semin Arthritis Rheum
1999
;
29
:
148
158

174

Robson
R
,
Cecka
JM
,
Opelz
G
et al. .
Prospective registry-based observational cohort study of the long-term risk of malignancies in renal transplant patients treated with mycophenolate mofetil
.
Am J Transplant
2005
;
5
:
2954
2960

175

O'Neill
JO
,
Edwards
LB
,
Taylor
DO.
Mycophenolate mofetil and risk of developing malignancy after orthotopic heart transplantation: analysis of the transplant registry of the International Society for Heart and Lung Transplantation
.
J Heart Lung Transplant
2006
;
25
:
1186
1191

176

Birkeland
SA
,
Hamilton-Dutoit
S.
Is posttransplant lymphoproliferative disorder (PTLD) caused by any specific immunosuppressive drug or by the transplantation per se?
Transplantation
2003
;
76
:
984
988

177

Campistol
JM
,
Eris
J
,
Oberbauer
R
et al. .
Sirolimus therapy after early cyclosporine withdrawal reduces the risk for cancer in adult renal transplantation
.
J Am Soc Nephrol
2006
;
17
:
581
589

178

Luan
FL
,
Hojo
M
,
Maluccio
M
et al. .
Rapamycin blocks tumour progression: unlinking immunosuppression from antitumor efficacy
.
Transplantation
2002
;
73
:
1565
1572

179

Campistol
JM
,
Gutierrez-Dalmau
A
,
Torregrosa
JV.
Conversion to sirolimus: a successful treatment for posttransplantation Kaposi's sarcoma
.
Transplantation
2004
;
77
:
760
762

180

Euvrard
S
,
Ulrich
C
,
Lefrancois
N.
Immunosuppressants and skin cancer in transplant patients: focus on rapamycin
.
Dermatol Surg
2004
;
30
:
628
633

181

Kahan
BD
,
Yakupoglu
YK
,
Schoenberg
L
et al. .
Low incidence of malignancy among sirolimus/cyclosporine-treated renal transplant recipients
.
Transplantation
2005
;
80
:
749
758

182

Stallone
G
,
Schena
A
,
Infante
B
et al. .
Sirolimus for Kaposi's sarcoma in renal-transplant recipients
.
N Engl J Med
2005
;
352
:
1317
1323

183

Lebbe
C
,
Euvrard
S
,
Barrou
B
et al. .
Sirolimus conversion for patients with posttransplant Kaposi's sarcoma
.
Am J Transplant
2006
;
6
:
2164
2168

184

Euvrard
S
,
Morelon
E
,
Rostaing
L
et al. .
Sirolimus and secondary skin-cancer prevention in kidney transplantation
.
N Engl J Med
2012
;
367
:
329
339

185

Campbell
SB
,
Walker
R
,
Tai
SS
et al. .
Randomized controlled trial of sirolimus for renal transplant recipients at high risk for nonmelanoma skin cancer
.
Am J Transplant
2012
;
12
:
1146
1156

186

Kreis
H
,
Oberbauer
R
,
Campistol
JM
et al. .
Long-term benefits with sirolimus-based therapy after early cyclosporine withdrawal
.
J Am Soc Nephrol
2004
;
15
:
809
817

187

Alberu
J
,
Pascoe
MD
,
Campistol
JM
et al. .
Lower malignancy rates in renal allograft recipients converted to sirolimus-based, calcineurin inhibitor-free immunotherapy: 24-month results from the CONVERT trial
.
Transplantation
2011
;
92
:
303
310

188

Ekberg
H
,
Bernasconi
C
,
Noldeke
J
et al. .
Cyclosporine, tacrolimus and sirolimus retain their distinct toxicity profiles despite low doses in the Symphony study
.
Nephrol Dial Transplant
2010
;
25
:
2004
2010

189

Flechner
SM
,
Glyda
M
,
Cockfield
S
et al. .
The ORION study: comparison of two sirolimus-based regimens versus tacrolimus and mycophenolate mofetil in renal allograft recipients
.
Am J Transplant
2011
;
11
:
1633
1644

190

Budde
K
,
Becker
T
,
Arns
W
et al. .
Everolimus-based, calcineurin-inhibitor-free regimen in recipients of de-novo kidney transplants: an open-label, randomised, controlled trial
.
Lancet
2011
;
377
:
837
847

191

Budde
K
,
Lehner
F
,
Sommerer
C
et al. .
Conversion from cyclosporine to everolimus at 4.5 months posttransplant: 3-year results from the randomized ZEUS study
.
Am J Transplant
2012
;
12
:
1528
1540

192

Budde
K
,
Lehner
F
,
Sommerer
C
et al. .
Five-year outcomes in kidney transplant patients converted from cyclosporine to everolimus: the randomized ZEUS study
.
Am J Transplant
2015
;
15
:
119
128

193

Yanik
EL
,
Gustafson
SK
,
Kasiske
BL
et al. .
Sirolimus use and cancer incidence among US kidney transplant recipients
.
Am J Transplant
2015
;
15
:
129
136

194

Yanik
EL
,
Siddiqui
K
,
Engels
EA.
Sirolimus effects on cancer incidence after kidney transplantation: a meta-analysis
.
Cancer Med
2015
;
4
:
1448
1459

195

Nee
R
,
Hurst
FP
,
Dharnidharka
VR
et al. .
Racial variation in the development of posttransplant lymphoproliferative disorders after renal transplantation
.
Transplantation
2011
;
92
:
190
195

196

Masson
P
,
Henderson
L
,
Chapman
JR
et al. .
Belatacept for kidney transplant recipients
.
Cochrane Database Syst Rev
2014
;
11
:
1
65

197

Vincenti
F
,
Charpentier
B
,
Vanrenterghem
Y
et al. .
A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study)
.
Am J Transplant
2010
;
10
:
535
546

198

Durrbach
A
,
Pestana
JM
,
Pearson
T
et al. .
A phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors (BENEFIT-EXT study)
.
Am J Transplant
2010
;
10
:
547
557

199

Vincenti
F
,
Larsen
CP
,
Alberu
J
et al. .
Three-year outcomes from BENEFIT, a randomized, active-controlled, parallel-group study in adult kidney transplant recipients
.
Am J Transplant
2012
;
12
:
210
217

200

Rostaing
L
,
Vincenti
F
,
Grinyo
J
et al. .
Long-term belatacept exposure maintains efficacy and safety at 5 years: results from the long-term extension of the BENEFIT study
.
Am J Transplant
2013
;
13
:
2875
2883

201

Vincenti
F
,
Rostaing
L
,
Grinyo
J
et al. .
Belatacept and long-term outcomes in kidney transplantation
.
N Engl J Med
2016
;
374
:
333
343

202

Grinyo
JM
,
Del Carmen
RM
,
Alberu
J
et al. .
Safety and efficacy outcomes 3 years after switching to belatacept from a calcineurin inhibitor in kidney transplant recipients: results from a phase 2 randomized trial
.
Am J Kidney Dis
2017
;
69
:
587
594

203

Penn
I.
The effect of immunosuppression on pre-existing cancers
.
Transplantation
1993
;
55
:
742
747

204

Kasiske
BL
,
Cangro
CB
,
Hariharan
S
et al. .
The evaluation of renal transplantation candidates: clinical practice guidelines
.
Am J Transplant
2001
;
1
:
3
95

205

Knoll
G
,
Cockfield
S
,
Blydt-Hansen
T
et al. .
Canadian Society of Transplantation: consensus guidelines on eligibility for kidney transplantation
.
Can Med Assoc J
2005
;
173
:
S1
25

206

Campbell
S
,
Pilmore
H
,
Gracey
D
et al. .
KHA-CARI guideline: recipient assessment for transplantation
.
Nephrology
2013
;
18
:
455
462

207

Batabyal
P
,
Chapman
JR
,
Wong
G
et al. .
Clinical practice guidelines on wait-listing for kidney transplantation: consistent and equitable?
Transplantation
2012
;
94
:
703
713

208

Kasiske
BL,
,
Vazquez
MA,
,
Harmon
WE
et al. .
Recommendations for the outpatient surveillance of renal transplant recipients. American Society of Transplantation
.
J Am Soc Nephrol
2000
;
11
:
S1
S86

209

European best practice guidelines for renal transplantation
.
Section IV: long-term management of the transplant recipient
.
Nephrol Dial Transplant
2002
;
17
:
1
67

210

KDIGO clinical practice guideline for the care of kidney transplant recipients
.
Am J Transplant
2009
;
9(Suppl 3)
:
S1
S155

211

Bia
M
,
Adey
DB
,
Bloom
RD
et al. .
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

212

Knoll
GA
,
Blydt-Hansen
TD
,
Campbell
P
et al. .
Canadian Society of Transplantation and Canadian Society of Nephrology commentary on the 2009 KDIGO clinical practice guideline for the care of kidney transplant recipients
.
Am J Kidney Dis
2010
;
56
:
219
246

213

Baker
R
,
Jardine
A
,
Andrews
P.
Renal Association clinical practice guideline on post-operative care of the kidney transplant recipient
.
Nephron Clin Pract
2011
;
118
:
c311
c347

214

Chadban
SJ
,
Barraclough
KA
,
Campbell
SB
et al. .
KHA-CARI guideline: KHA-CARI adaptation of the KDIGO clinical practice guideline for the care of kidney transplant recipients
.
Nephrology
2012
;
17
:
204
214

215

Acuna
SA
,
Huang
JW
,
Scott
AL
et al. .
Cancer screening recommendations for solid organ transplant recipients: a systematic review of clinical practice guidelines
.
Am J Transplant
2017
;
17
:
103
114

216

Schwarz
A
,
Vatandaslar
S
,
Merkel
S
et al. .
Renal cell carcinoma in transplant recipients with acquired cystic kidney disease
.
Clin J Am Soc Nephrol
2007
;
2
:
750
756

217

Vajdic
CM
,
McDonald
SP
,
McCredie
MR
et al. .
Cancer incidence before and after kidney transplantation
.
JAMA
2006
;
296
:
2823
2831

218

Wong
G
,
Howard
K
,
Webster
AC
et al. .
Screening for renal cancer in recipients of kidney transplants
.
Nephrol Dial Transplant
2011
;
26
:
1729
1739

219

Wong
G
,
Howard
K
,
Craig
JC
et al. .
Cost-effectiveness of colorectal cancer screening in renal transplant recipients
.
Transplantation
2008
;
85
:
532
541

220

Wong
G
,
Howard
K
,
Webster
A
et al. .
The health and economic impact of cervical cancer screening and human papillomavirus vaccination in kidney transplant recipients
.
Transplantation
2009
;
87
:
1078
1091

221

Acuna
SA
,
Sutradhar
R
,
Camacho
X
et al. .
Uptake of cancer screening tests among recipients of solid organ transplantation
.
Am J Transplant
2017
;
17
:
2434
2443

222

van Leeuwen
MT
,
Webster
AC
,
McCredie
MR
et al. .
Effect of reduced immunosuppression after kidney transplant failure on risk of cancer: population based retrospective cohort study
.
BMJ
2010
;
340
:
c570

223

Khokhar
NZ
,
Altman
JK
,
Platanias
LC.
Emerging roles for mammalian target of rapamycin inhibitors in the treatment of solid tumors and hematological malignancies
.
Curr Opin Oncol
2011
;
23
:
578
586

224

Baldo
P
,
Cecco
S
,
Giacomin
E
et al. .
mTOR pathway and mTOR inhibitors as agents for cancer therapy
.
Curr Cancer Drug Targets
2008
;
8
:
647
665

225

Cheung
CY
,
Lam
MF
,
Chu
KH
et al. .
Malignancies after kidney transplantation: Hong Kong renal registry
.
Am J Transplant
2012
;
12
:
3039
3046

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected]

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.