TY - JOUR AU - Abdollahi, Amir AB - cancer, immunosuppressive drugs, transplantation Introduction Organ transplantation is a well-established method for the therapy of end-stage organ failure. The emergence of novel immunosuppressive regimens has reduced the risk of rejection and extended the life expectancy of organ recipients. The long-term outcome of these patients is now challenged by life-threatening complications such as cardiovascular disease, infections and post-transplant malignancies. Malignancy is a well-recognized complication of transplantation and can manifest as de novo cancer, as a recurrence of a pre-existing malignancy or from transmission of malignancy from the donor. Recent studies show that tumour incidence increases with time after organ transplantation and is related to the intensity of immunosuppression [ 1,2 ]. Overall, a 3- to 4-fold increased incidence of cancer has been observed in transplant patients compared with age-matched controls in the general population. During immunosuppressive therapy, there is a higher frequency of some relatively rare tumours that tend to be biologically more aggressive than those that occur in the general population [ 3 ]. The relative risk for developing skin cancer in allografted patients is increased up to 70% in regions with high sun exposure [ 4 ]. Post-transplantation lymphoproliferative disorder (PTLD) occurs in up to 11% of renal transplant recipients and holds the major cause for cancer-related mortality. There is a 400- to 500-fold increase in Kaposi sarcoma compared with controls of the same ethnic origin and a 100-fold increase in vulval and anal carcinomas in transplant recipients [ 5 ]. The aetiology of post-transplant malignancy is believed to be multifactorial in nature and probably involves impaired immunosurveillance of the host, direct carcinogenic effects of some immunosuppressive agents and depressed antiviral immune activity referring to a number of common viral-related post-transplant malignancies. To minimize the risk of malignancy after transplantation, it is important to evaluate the effects of an immunosuppressive agent on cancer incidence and tumour growth. However, the common use of combination therapies limits the interpretation of single-agent effects on the tumourigenesis process. There is now growing evidence that newer immunosuppressive drugs exert anti-neoplastic effects and could therefore simultaneously address the risk of allograft rejection and cancer in transplant recipients. The focus of this review is to illustrate known pro- and anti-neoplastic mechanisms attributed to some currently used immunosuppressive drugs. mTOR inhibitors The mammalian target of rapamycin (mTOR) is a critical modulator of protein translation. Emerging data indicate that mTOR plays a pivotal role in regulating a plethora of signalling pathways activated in response to growth factors and metabolic changes [ 6 ]. Inhibitors of mTOR (mTORis) exert their immunosuppressive activity by impeding the response to interleukin-2 (IL-2) and thereby blocking the activation of T- and B-cells. After binding of the mTOR inhibitor to the 12-kDa immunophilin FK506-binding protein (FKBP12), this complex interacts with and inhibits the serine/threonine kinase mTOR [ 6 ]. Currently used mTOR inhibitors in transplantation medicine include rapamycin (Rapamune®, Wyeth, Madison, NJ, USA), rapamycin-derivatives RAD001 (Everolimus®, Novartis, Basel, Switzerland) and CCI779 (Temsirolimus®, Wyeth), and the rapamycin analogue ap23573 (Deforolimus®, ARIAD Pharmaceuticals, Cambridge, MA, USA). Multiple effects of mTORis on cellular signalling have been described. Rapamycin induces inhibition of the phosphatidylinositol-3-kinase (PI3K) signalling pathway that plays a key role in regulation of proliferation, survival, mobility and angiogenesis [ 7 ]. Rapamycin further inhibits signal transducer and transcription activator 3 (STAT3) signalling [ 8 ]. STAT3 mediates the expression of a variety of genes in response to cell stimuli and is involved in many cellular processes such as cell growth and apoptosis. STAT3 is upregulated in many tumour types and elevated expression levels often correlate with a poor prognosis [ 9 ]; hence, mTOR inhibition may serve as a reasonable approach for targeting STAT3 in cancer therapy. A rare but serious complication in transplant recipients is the development of PTLD. In most cases, PTLD is associated with Epstein-Barr virus (EBV) infection of B-cells, either as a consequence of reactivation of the virus after transplantation or from primary EBV infection acquired from the donor. RAD001 was shown to inhibit the growth of Epstein-Barr virus-transformed B lymphocytes both in vitro and in vivo ; thus RAD001 therapy might prevent PTLD development [ 10 ]. In alignment with this experimental data, some patients with PTLD were treated successfully with rapamycin and the anti-CD20 antibody rituximab; however, controlled prospective randomized studies are pending [ 11 ]. Another virus-related disease, the Kaposi sarcoma, is caused by the human herpes virus 8 (HHV8). It occurs mostly in immunosuppressed patients with HIV or after solid organ transplantation. In renal transplant recipients, Kaposi sarcomas were dramatically regressed after the immunosuppressive therapy was switched from cyclosporine (CsA) to mTOR inhibitors [ 12–14 ]. One plausible molecular explanation for this phenomenon was recently provided by Sodhi et al . They showed that HHV8 encodes a G protein-coupled receptor (vGPCR) that activates the mTOR pathway via phosphorylation of tuberin (TSC2). The inhibition of mTOR with rapamycin prevented vGPCR-induced sarcomagenesis, thus supporting the proposed promoting role of the activated mTOR pathway in endothelial cell transformation [ 15 ]. Further, mTOR inhibitors were shown to exert potent antiangiogenic activity in vitro and in vivo in primary and metastatic tumours via inhibition of VEGF production [ 16 ]. This antiangiogenic activity of mTORis might at least in part contribute to the regression of the highly vascularized Kaposi sarcomas. Evidence for anti-neoplastic effects of mTOR inhibitors are further provided by recent clinical cancer trials. The most encouraging data were reported for mTOR administration in mantle cell lymphoma [ 17 ] and renal cell carcinoma [ 18,19 ]. Additional ongoing cancer trials include malignant melanoma, hepatocellular carcinoma, glioblastoma, lung cancer, breast cancer and other tumour entities [ 20 ]. In contrast to their status in clinical oncology, in the setting of organ transplantation, mTORis are anticipated to exert a dual role by providing adequate immunosuppression to prevent organ rejection and simultaneously impair tumour development. A multivariate analysis of renal transplant patients examined the incidence of post-transplant malignancies in patients receiving mTOR inhibitors alone (everolimus or sirolimus), calcineurin inhibitors alone (CsA or tacrolimus) or a combination of both types of agents. Maintenance of immunosuppression with the mTOR inhibitor drugs sirolimus and everolimus alone or even in combination with calcineurin inhibitors (CNI) showed a significantly reduced risk of developing any post-transplant de novo malignancy and non-skin solid malignancy compared to a therapy with CNI alone [ 21 ]. Analysis of five multicentre studies of patients after renal transplantation revealed that patients receiving a rapamycin-based therapy without CsA or a rapamycin maintenance therapy after early CsA withdrawal had a significantly lower rate of malignancy in the first 2 years after transplantation [ 22 ]. Recent clinical trials in renal transplant recipients have further confirmed the significantly reduced risk of developing any malignancy under treatment with mTOR inhibitors [ 23,24 ]. However, in the Symphony study that investigated the efficacy and relative toxicity of four different immunosuppressive regimens in 1645 renal transplant recipients, the highest cancer incidence was noted in the rapamycin group after 12 months of follow-up [ 25 ]. Azathioprine and mycophenolic acid Azathioprine (AZA) is a purine analogue that is incorporated into cellular DNA where it inhibits the purine nucleotide synthesis and interferes with RNA synthesis and metabolism. Its strong correlation with tumour incidence in transplant recipients is reflected by the fact that it is listed as a human carcinogen in the 11th Report on Carcinogens of the US Department of Health and Human Services [ 26 ]. Two large prospective epidemiological studies reported a high incidence of non-Hodgkin's lymphoma, squamous cell cancers of the skin, hepatobiliary carcinomas and mesenchymal tumours in renal transplant patients, who were treated with AZA and prednisone [ 26 ]. The high incidence of skin cancers in the course of treatment with AZA is proposed to be due to its synergism with UV radiation in carcinogenesis. It has been shown that AZA causes the accumulation of 6-thioguanine in patients’ DNA, and 6-thioguanine and UVA are synergistically mutagenic [ 27 ]. Furthermore, the intercalation of thioguanine into the DNA inhibits repair and induces codon misreads that could encounter for AZA-mediated cytotoxicity and carcinogenesis [ 28 ]. However, in the era of combination therapies the effects of lower doses of AZA on tumour development in a transplanted patient are difficult to determine. Compared with the more powerful immunosuppressant CsA, AZA has been associated with a lower cumulative incidence of tumours after transplantation [ 29 ]. Hence, a clear correlation of less intensive AZA use with cancer development in transplant recipients has not been established. Mycophenolic acid (MPA) reversibly inhibits inosine 5′-monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme in the de novo synthesis of guanine nucleotides. MPA displays high lymphocyte specificity and cytotoxicity due to the higher dependence of activated lymphocytes on both salvage and de novo synthesis of guanine nucleotides relative to other cell types. The morpholinoethyl ester of MPA, mycophenolate mofetil (MMF), has potent immunosuppressive properties and is used worldwide in transplantation medicine to prevent organ rejection. In addition to the well-known immunosuppressive effects, an increasing number of studies document the antiproliferative properties of MPA on a variety of cell lines [ 30 ]. IMPDH activity is increased significantly in cancer cells, and it was thus considered a sensitive target for chemotherapy [ 31 ]. In fact, MPA has been shown to inhibit the growth of tumour cells in vitro and in mouse xenografts [ 32 ]. This observation led to testing of MPA in small cohorts of patients with a variety of cancers in the 1970s. Considerable dose-limiting gastrointestinal toxicity limited the success of these trials due to the use of the diethanolamine salt of MPA rather than the currently prescribed MMF prodrug [ 33,34 ]. One molecular mechanism contributing to MPA's antitumour activity might be its ability to induce apoptosis as demonstrated in human neuroblastoma-, B-lymphoma- and multiple myeloma cells [ 35–37 ]. Furthermore, the antitumour activity of MPA is linked to its antiangiogenic properties. MPA was found to potently inhibit endothelial cell proliferation in vitro and block tumour-induced angiogenesis in vivo [ 38 ]. We have recently shown a preferential sensitivity of endothelial cells to MPA treatment compared to different tumour cell lines. A glioblastoma cell line (U87) displayed resistance to MPA treatment in vitro but MPA significantly inhibited U87 tumour growth in vivo suggesting an important role for the tumour microenvironment in MPA response [ 39 ]. In another study that used RNA interference, the knockdown of one of the two known isoforms of inosine monophosphate dehydrogenase (IMPDH-1) was sufficient to cause endothelial cell cycle arrest. In addition to that data, anti-endothelial activity of MPA was detected in two recent pharmacological screens among FDA-approved drugs that aimed to identify compounds with antiangiogenic properties [ 40,41 ]. The interaction of MMF with adhesion molecules seems to play an important role in preventing integrin-dependent tumour dissemination and metastasis. MMF was shown to induce alterations of the beta1 integrin profile and thereby block tumour cell adhesion to vascular endothelium [ 42 ]. Another mechanism discussed for the antitumour activity of MPA is the induction of differentiation in tumour cells. Addition of MPA to cultures of HL-60, neuroblastoma and melanoma cells resulted in a decreased cellular level of guanine nucleotides and the induction of cell differentiation [ 43–45 ]. In a phase I clinical trial, MPA was tested as an anticancer agent in patients with advanced multiple myeloma. Partial response to the drug adds evidence that inhibition of the IMPDH pathway may be a promising novel targeted therapy to overcome drug resistance in multiple myeloma [ 46 ]. Furthermore, synergy between imatinib and MPA in the treatment of chronic myleogenous leukaemia (CML) has been reported [ 47 ]. Evidence of potential anti-neoplastic effects of MPA in transplant recipients are provided by a number of clinical studies. In a large cohort study that examined the risk of malignancies with MMF in renal transplant recipients, a trend towards a lower risk of malignancy and a significant increase in time to malignancy in the MMF group were found. [ 48 ]. Data from the transplant registry of the International Society for Heart and Lung Transplantation (ISHLT) showed in a multivariate analysis that the use of MMF in standard immunosuppressive regimens is associated with a significantly lower risk of developing malignancy after orthotopic heart transplantation [ 49 ]. An independent report compared the cancer incidence in AZA- versus MMF-treated patients using data from the SRTR database [the SRTR database contains information collected by the Organ Procurement and Transplantation Network (OPTN)]. In 17 145 adult renal transplant recipients with pre-existing diabetes mellitus, the incidence of any post-transplant malignancy was higher in AZA-treated patients compared with MMF-treated patients (3.7% versus 2.2%, respectively). This report also indicated a significant difference in lymphoproliferative malignancies (0.6% versus 0.3%) and de novo solid tumours (2.5% versus 1.6%) [ 50 ]. However, the Tricontinental Multicenter study and the US Randomized study that compared the treatment of MMF with AZA in cadaveric renal recipients showed no difference in the incidence of any malignancy between the two drugs [ 51,52 ]. Since MMF is more effective than AZA in preventing allograft rejection [ 53 ] and might even lower the risk of malignancy, it offers a favourable profile in transplantation medicine. FTY720 The immunosuppressive agent FTY720 is a synthetic analogue of sphingosine and was isolated from culture filtrates of the ascomycete Isaria sinclairii . FTY720 inhibits the egression of lymphocytes from lymph nodes to efferent lymphatics and blood via modulation of sphingosine-1-phosphate (S1P) receptors on lymphocytes [ 54 ]. The efficacy and safety of FTY720 in de novo kidney transplant patients was proven in combination with CsA and corticoids in a phase II study [ 55 ]. However, the multifaceted mode of action of FTY720 opens up other possibilities for its use, e.g., as anti-cancer agent. FTY720 was shown to exert potent anti-tumour and anti-metastatic activities in different tumour types in vitro and in vivo. Pro-apoptotic activity of FTY720 was linked to its ability to inhibit Akt- or downregulate Bcl2 survival signalling [ 56 ]. In multiple myeloma cell lines, Yasui and colleagues showed that FTY720 induces mitochondria-mediated apoptosis followed by the release of mitochondria proteins, cleavage of PARP and activation of caspase-8, -3 and -9. FTY720 also inhibits other survival signals triggered by cytokines such as IL-6 via inhibition of STAT3, Akt and ERK phosphorylation in a dose-dependent manner [ 57 ]. The potent antiangiogenic activity of FTY720 is linked to its functional antagonism of vascular S1P receptors and transactivation of the CXCR4 chemokine receptor [ 58,59 ]. Recent studies showed that FTY720 decreases vascular permeability induced by the key angiogenic protein VEGF [ 60 ]. Azuma and colleagues showed that FTY720 caused cytoskeletal changes in cancer cells that resulted in mis-shaped cells with reduced filopodias. In addition, it reduced the expression of integrins on the surface of cancer cells and impeded their ability to adhere and migrate to extracellular matrix proteins [ 61 ]. Evidence that FTY720 could inhibit cancer metastasis process comes from a study of Zhou et al . in 2006. They showed that FTY720 markedly inhibited prostate cancer cell invasion by down-regulating GTP-bound RhoA, a protein that is associated with metastasis in many types of malignancies [ 62 ]. Recent published data support the investigation of FTY720 as a novel therapeutic approach for patients with blast crisis in chronic myelogenous leukaemia (CML) or adult acute lymphoblastic leukaemia that express the BCR-ABL oncogene. The prognosis in these patients is usually poor. FTY720 was shown to induce cell death and reduce clonogenicity in myeloid and lymphoid cell lines and patient-derived progenitor cells that were either sensitive or resistant to imatinib and/or dasatinib. Of note, no adverse effects of FTY720 on normal bone marrow progenitor cells were detected in this study [ 63 ]. Although Novartis discontinued the development of FTY720 for allograft recipients due to a lack of benefits compared with MMF in a large phase III clinical trial, the multifaceted activities of this compound described here warrant further clinical testing. Calcineurin inhibitors CsA and tacrolimus are among the most widely used immunosuppressive agents belonging to the family of CNI. Their major effect is to block the IL-2 expression and to increase the production of transforming growth factor beta (TGFβ), thereby impairing immune response [ 64,65 ]. CNIs are linked to a higher incidence and aggressive progression of neoplasm. These effects were mainly attributed to the impairment of the organ recipient's immune surveillance [ 66–68 ]. However, Hojo and colleagues were able to prove that CsA-treated cells underwent significant morphological alterations, including induction of pseudopodial protrusions, increased cell motility and invasive growth, thereby inducing cancer progression by a cell-autonomous mechanism probably mediated by TGFβ1 [ 69 ]. TGFβ is a pleiotropic cytokine that has a dual role in carcinogenesis. Initially, it acts as a tumour suppressor and causes growth arrest of epithelial cells and cells in the early stages of cancer [ 70 ]. In established tumours, TGFβ exerts an effect that is favourable for the survival, progression and metastasis of the tumour [ 71 ]. These data suggest that TGFβ-mediated effects of CsA might rather promote the growth of dormant tumours than inducing de novo cancer. Possible mechanisms for enhanced de novo cancer development induced by CsA include its ability to inhibit the DNA repair apparatus that facilitates the accumulation of DNA mutations [ 72,73 ]. Combined with depletion of activated T-cells, this could result in decreased clearance of altered cells. The results of clinical studies evaluating the risk of cancer development in patients treated with CNIs are conflicting. In a prospective, randomized study, patients receiving full-dose CsA had an increased incidence of post-transplant malignancy, as compared with those receiving low doses (32% versus 20% of patients, respectively). This trend was apparent for all types of cancers, including skin cancer, PTLD and other cancers [ 1 ]. In a series of 106 patients with hepatocellular carcinoma who underwent liver transplantation, a higher cumulative dosage of CsA showed a negative effect on the recurrence-free survival rate [ 74 ]. The comparison of cancer incidence in patients treated with CsA versus tacrolimus yielded conflicting results. Although tacrolimus is associated with a slightly higher immunosuppressive potency [ 75 ], there are studies that report a reduction in the incidence of de novo solid tumours if these patients were treated with tacrolimus versus CsA [ 76,77 ]. On the other hand, tacrolimus treatment was associated with a higher rate of PTLD that might be correlated with its greater immunosuppressive effect. The analysis of 41 686 renal allograft recipients included in the Scientific Registry of Transplant Recipients (SRTR database) demonstrated that, in patients who did not receive induction therapy, the cumulative incidence of PTLD was lower in CsA-treated patients than in tacrolimus-treated patients. This difference disappeared if the patient had received induction therapy, which itself poses the recipients to a 1.8-fold higher risk for PTLD development. A possible explanation could be the strong immunosuppressive effect of induction therapy that might override the differences among the maintenance drugs [ 76 ]. Biological agents and small molecules Lymphocyte-depleting antibodies are strongly associated with a higher risk of post-transplant malignancy, in particular with virally induced cancer. Furthermore, the use of induction regimens containing lymphocyte-depleting antibodies is one of the best-known risk factors for the development of PTLD. A striking rise in the frequency of PTLD was first described in 1990 in heart transplant recipients, who received the monoclonal antibody against CD3 (OKT3, muromonab) in addition to standard immunosuppressive therapy [ 78 ]. Subsequent studies investigating the effect of anti-thymocyte globulins and OKT3 on PTLD development supported this data [ 79 ]. In contrast, treatment with anti-IL-2 receptor antibodies such as basiliximab or daclizumab was not correlated with an increased risk for post-transplant malignancy [ 2 , 80 , 81 ]. These antibodies target specifically activated T-cells, macrophages and monocytes whereas the total number of lymphocytes remains unaffected. Using the Collaborative Transplant Study (CTS) database, a recent study has evaluated the influence of different induction therapies on graft survival and the incidence of non-Hodgkin lymphoma in 112 122 renal allograft recipients. The highest rates of lymphoma were found in patients treated with equine-/rabbit-anti-thymocyte globulin or OKT3, whereas IL-2 receptor antibodies offered the best risk-to-benefit ratio with regard to graft survival and lymphoma incidence [ 82 ]. The emergence of novel targeted biologicals and small molecular agents has fuelled hope to develop therapies that deliver immunosuppression without long-term toxicity. Currently, a number of interesting biologicals are investigated in preclinical and clinical studies including (i) belatacept, a fusion protein of the Fc-fragment of human IgG1 immunoglobulin with the extracellular domain of CTLA-4 that blocks the interaction between CD80/86 and CD28 costimulatory pathways; (ii) anti-interleukin-15; (iii) anti-CD40 and (iv) efalizumab, a humanized anti-LFA1 monoclonal antibody. Further, targeted inhibition of a series of pathways is tested using novel small molecular inhibitors of pyrimidine synthesis (FK778), Janus kinase 3 (JAK3, CP-690550) and protein kinase C (PKC, AEB-071). It remains to be elucidated whether these promising novel agents will reduce the incidence of post-transplant malignancies as well as infectious and cardiovascular-related complications [ 83 ]. Conclusion The advent of novel immunosuppressive regimens has steadily reduced the incidence of acute transplant rejection. With the increased life expectancy of allograft recipients, post-transplant malignancy has emerged as an important cause for mortality in these patients. The tremendous growth of knowledge in cancer biology and the molecular mechanisms underlying anti-neoplastic effects of immunosuppressive agents offers new possibilities to address this issue. Therefore, prospective clinical studies are necessary to evaluate the impact of selected immunosuppressive agents on cancer development and progression. Conflict of interest statement . None declared. References 1 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, vol.  351 (pg.  623- 628) Google Scholar CrossRef Search ADS PubMed  2 Wimmer CD,  Rentsch M,  Crispin A, et al.  The janus face of immunosuppression—de novo malignancy after renal transplantation: the experience of the Transplantation Center Munich,  Kidney Int ,  2007, vol.  71 (pg.  1271- 1278) Google Scholar CrossRef Search ADS PubMed  3 Penn I.  Occurrence of cancers in immunosuppressed organ transplant recipients,  Clin Transpl ,  1998(pg.  147- 158) 4 Euvrard S,  Kanitakis J,  Claudy A.  Skin cancers after organ transplantation,  N Engl J Med ,  2003, vol.  348 (pg.  1681- 1691) Google Scholar CrossRef Search ADS PubMed  5 Penn I.  Post-transplant malignancy: the role of immunosuppression,  Drug Saf ,  2000, vol.  23 (pg.  101- 113) Google Scholar CrossRef Search ADS PubMed  6 Bjornsti MA,  Houghton PJ.  The TOR pathway: a target for cancer therapy,  Nat Rev Cancer ,  2004, vol.  4 (pg.  335- 348) Google Scholar CrossRef Search ADS PubMed  7 Vivanco I,  Sawyers CL.  The phosphatidylinositol 3-kinase AKT pathway in human cancer,  Nat Rev Cancer ,  2002, vol.  2 (pg.  489- 501) Google Scholar CrossRef Search ADS PubMed  8 Yokogami K,  Wakisaka S,  Avruch J, et al.  Serine phosphorylation and maximal activation of STAT3 during CNTF signaling is mediated by the rapamycin target mTOR,  Curr Biol ,  2000, vol.  10 (pg.  47- 50) Google Scholar CrossRef Search ADS PubMed  9 Klampfer L.  Signal transducers and activators of transcription (STATs): novel targets of chemopreventive and chemotherapeutic drugs,  Curr Cancer Drug Targets ,  2006, vol.  6 (pg.  107- 121) Google Scholar CrossRef Search ADS PubMed  10 Majewski M,  Korecka M,  Kossev P, et al.  The immunosuppressive macrolide RAD inhibits growth of human Epstein-Barr virus-transformed B lymphocytes in vitro and in vivo: a potential approach to prevention and treatment of posttransplant lymphoproliferative disorders,  Proc Natl Acad Sci USA ,  2000, vol.  97 (pg.  4285- 4290) Google Scholar CrossRef Search ADS PubMed  11 Garcia VD,  Bonamigo Filho JL,  Neumann J, et al.  Rituximab in association with rapamycin for post-transplant lymphoproliferative disease treatment,  Transpl Int ,  2003, vol.  16 (pg.  202- 206) Google Scholar CrossRef Search ADS PubMed  12 Campistol JM,  Gutierrez-Dalmau A,  Torregrosa JV.  Conversion to sirolimus: a successful treatment for posttransplantation Kaposi's sarcoma,  Transplantation ,  2004, vol.  77 (pg.  760- 762) Google Scholar CrossRef Search ADS PubMed  13 Lebbe C,  Euvrard S,  Barrou B, et al.  Sirolimus conversion for patients with posttransplant Kaposi's sarcoma,  Am J Transplant ,  2006, vol.  6 (pg.  2164- 2168) Google Scholar CrossRef Search ADS PubMed  14 Stallone G,  Schena A,  Infante B, et al.  Sirolimus for Kaposi's sarcoma in renal-transplant recipients,  N Engl J Med ,  2005, vol.  352 (pg.  1317- 1323) Google Scholar CrossRef Search ADS PubMed  15 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, vol.  10 (pg.  133- 143) Google Scholar CrossRef Search ADS PubMed  16 Guba M,  von Breitenbuch P,  Steinbauer M, et al.  Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor,  Nat Med ,  2002, vol.  8 (pg.  128- 135) Google Scholar CrossRef Search ADS PubMed  17 Haritunians T,  Mori A,  O’Kelly J, et al.  Antiproliferative activity of RAD001 (everolimus) as a single agent and combined with other agents in mantle cell lymphoma,  Leukemia ,  2007, vol.  21 (pg.  333- 339) Google Scholar CrossRef Search ADS PubMed  18 Hudes G,  Carducci M,  Tomczak P, et al.  Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma,  N Engl J Med ,  2007, vol.  356 (pg.  2271- 2281) Google Scholar CrossRef Search ADS PubMed  19 Motzer RJ,  Escudier B,  Oudard S, et al.  Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial,  Lancet ,  2008, vol.  372 (pg.  449- 456) Google Scholar CrossRef Search ADS PubMed  20 Faivre S,  Kroemer G,  Raymond E.  Current development of mTOR inhibitors as anticancer agents,  Nat Rev Drug Discov ,  2006, vol.  5 (pg.  671- 688) Google Scholar CrossRef Search ADS PubMed  21 Kauffman HM,  Cherikh WS,  Cheng Y, et al.  Maintenance immunosuppression with target-of-rapamycin inhibitors is associated with a reduced incidence of de novo malignancies,  Transplantation ,  2005, vol.  80 (pg.  883- 889) Google Scholar CrossRef Search ADS PubMed  22 Mathew T,  Kreis H,  Friend P.  Two-year incidence of malignancy in sirolimus-treated renal transplant recipients: results from five multicenter studies,  Clin Transplant ,  2004, vol.  18 (pg.  446- 449) Google Scholar CrossRef Search ADS PubMed  23 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, vol.  17 (pg.  581- 589) Google Scholar CrossRef Search ADS PubMed  24 Kahan BD,  Yakupoglu YK,  Schoenberg L, et al.  Low incidence of malignancy among sirolimus/cyclosporine-treated renal transplant recipients,  Transplantation ,  2005, vol.  80 (pg.  749- 758) Google Scholar CrossRef Search ADS PubMed  25 Ekberg H,  Tedesco-Silva H,  Demirbas A, et al.  Reduced exposure to calcineurin inhibitors in renal transplantation,  N Engl J Med ,  2007, vol.  357 (pg.  2562- 2575) Google Scholar CrossRef Search ADS PubMed  26 U.S. Department of Health and Human Services, Public Health Service, National Toxicology Program  Report on Carcinogens, Eleventh Edition. Available at http://ntp.niehs.nih.gov/ntp/roc/toc11.html 27 O’Donovan P,  Perrett CM,  Zhang X, et al.  Azathioprine and UVA light generate mutagenic oxidative DNA damage,  Science ,  2005, vol.  309 (pg.  1871- 1874) Google Scholar CrossRef Search ADS PubMed  28 Swann PF,  Waters TR,  Moulton DC, et al.  Role of postreplicative DNA mismatch repair in the cytotoxic action of thioguanine,  Science ,  1996, vol.  273 (pg.  1109- 1111) Google Scholar CrossRef Search ADS PubMed  29 McGeown MG,  Douglas JF,  Middleton D.  One thousand renal transplants at Belfast City Hospital: post-graft neoplasia 1968–1999, comparing azathioprine only with cyclosporin-based regimes in a single centre,  Clin Transpl ,  2000(pg.  193- 202) 30 Morath C,  Zeier M.  Review of the antiproliferative properties of mycophenolate mofetil in non-immune cells,  Int J Clin Pharmacol Ther ,  2003, vol.  41 (pg.  465- 469) Google Scholar CrossRef Search ADS PubMed  31 Jackson RC,  Weber G,  Morris HP.  IMP dehydrogenase, an enzyme linked with proliferation and malignancy,  Nature ,  1975, vol.  256 (pg.  331- 333) Google Scholar CrossRef Search ADS PubMed  32 Carter SB,  Franklin TJ,  Jones DF, et al.  Mycophenolic acid: an anti-cancer compound with unusual properties,  Nature ,  1969, vol.  223 (pg.  848- 850) Google Scholar CrossRef Search ADS PubMed  33 Brewin TB,  Cole MP,  Jones CT, et al.  Mycophenolic acid (NSC-129185): preliminary clinical trials,  Cancer Chemother Rep ,  1972, vol.  56 (pg.  83- 87) Google Scholar PubMed  34 Knudtzon S,  Nissen NI.  Clinical trial with mycophenolic acid (NSC-129185), a new antitumor agent,  Cancer Chemother Rep ,  1972, vol.  56 (pg.  221- 227) Google Scholar PubMed  35 Messina E,  Gazzaniga P,  Micheli V, et al.  Guanine nucleotide depletion triggers cell cycle arrest and apoptosis in human neuroblastoma cell lines,  Int J Cancer ,  2004, vol.  108 (pg.  812- 817) Google Scholar CrossRef Search ADS PubMed  36 Vegso G,  Sebestyen A,  Paku S, et al.  Antiproliferative and apoptotic effects of mycophenolic acid in human B-cell non-Hodgkin lymphomas,  Leuk Res ,  2007, vol.  31 (pg.  1003- 1008) Google Scholar CrossRef Search ADS PubMed  37 Takebe N,  Cheng X,  Fandy TE, et al.  IMP dehydrogenase inhibitor mycophenolate mofetil induces caspase-dependent apoptosis and cell cycle inhibition in multiple myeloma cells,  Mol Cancer Ther ,  2006, vol.  5 (pg.  457- 466) Google Scholar CrossRef Search ADS PubMed  38 Koehl GE,  Wagner F,  Stoeltzing O, et al.  Mycophenolate mofetil inhibits tumor growth and angiogenesis in vitro but has variable antitumor effects in vivo, possibly related to bioavailability,  Transplantation ,  2007, vol.  83 (pg.  607- 614) Google Scholar CrossRef Search ADS PubMed  39 Domhan S,  Muschal S,  Schwager C, et al.  Molecular mechanisms of the antiangiogenic and antitumor effects of mycophenolic acid,  Mol Cancer Ther ,  2008, vol.  7 (pg.  1656- 1668) Google Scholar CrossRef Search ADS PubMed  40 Chong CR,  Qian DZ,  Pan F, et al.  Identification of type 1 inosine monophosphate dehydrogenase as an antiangiogenic drug target,  J Med Chem ,  2006, vol.  49 (pg.  2677- 2680) Google Scholar CrossRef Search ADS PubMed  41 Wu X,  Zhong H,  Song J, et al.  Mycophenolic acid is a potent inhibitor of angiogenesis,  Arterioscler Thromb Vasc Biol ,  2006, vol.  26 (pg.  2414- 2416) Google Scholar CrossRef Search ADS PubMed  42 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, vol.  5 pg.  4  Google Scholar CrossRef Search ADS PubMed  43 Collart FR,  Huberman E.  Expression of IMP dehydrogenase in differentiating HL-60 cells,  Blood ,  1990, vol.  75 (pg.  570- 576) Google Scholar PubMed  44 Kiguchi K,  Collart FR,  Henning-Chubb C, et al.  Induction of cell differentiation in melanoma cells by inhibitors of IMP dehydrogenase: altered patterns of IMP dehydrogenase expression and activity,  Cell Growth Differ ,  1990, vol.  1 (pg.  259- 270) Google Scholar PubMed  45 Messina E,  Gazzaniga P,  Micheli V, et al.  Low levels of mycophenolic acid induce differentiation of human neuroblastoma cell lines,  Int J Cancer ,  2004, vol.  112 (pg.  352- 354) Google Scholar CrossRef Search ADS PubMed  46 Takebe N,  Cheng X,  Wu S, et al.  Phase I clinical trial of the inosine monophosphate dehydrogenase inhibitor mycophenolate mofetil (Cellcept) in advanced multiple myeloma patients,  Clin Cancer Res ,  2004, vol.  10 (pg.  8301- 8308) Google Scholar CrossRef Search ADS PubMed  47 Gu JJ,  Santiago L,  Mitchell BS.  Synergy between imatinib and mycophenolic acid in inducing apoptosis in cell lines expressing Bcr-Abl,  Blood ,  2005, vol.  105 (pg.  3270- 3277) Google Scholar CrossRef Search ADS PubMed  48 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, vol.  5 (pg.  2954- 2960) Google Scholar CrossRef Search ADS PubMed  49 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, vol.  25 (pg.  1186- 1191) Google Scholar CrossRef Search ADS PubMed  50 David KM,  Morris JA,  Steffen BJ, et al.  Mycophenolate mofetil versus azathioprine is associated with decreased acute rejection, late acute rejection, and risk for cardiovascular death in renal transplant recipients with pre-transplant diabetes,  Clin Transplant ,  2005, vol.  19 (pg.  279- 285) Google Scholar CrossRef Search ADS PubMed  51 US Renal Transplant Mycophenolate Mofetil Study Group.  Mycophenolate mofetil in cadaveric renal transplantation,  Am J Kidney Dis ,  1999, vol.  34 (pg.  296- 303) CrossRef Search ADS PubMed  52 Mathew TH.  Tricontinental Mycophenolate Mofetil Renal Transplantation Study Group A blinded, long-term, randomized multicenter study of mycophenolate mofetil in cadaveric renal transplantation: results at three years,  Transplantation ,  1998, vol.  65 (pg.  1450- 1454) Google Scholar CrossRef Search ADS PubMed  53 Halloran P,  Mathew T,  Tomlanovich S, et al.  ,  The International Mycophenolate Mofetil Renal Transplant Study Groups.  Mycophenolate mofetil in renal allograft recipients: a pooled efficacy analysis of three randomized, double-blind, clinical studies in prevention of rejection.,  Transplantation ,  1997, vol.  63 (pg.  39- 47) Google Scholar CrossRef Search ADS PubMed  54 Yopp AC,  Ledgerwood LG,  Ochando JC, et al.  Sphingosine 1-phosphate receptor modulators: a new class of immunosuppressants,  Clin Transplant ,  2006, vol.  20 (pg.  788- 795) Google Scholar CrossRef Search ADS PubMed  55 Tedesco-Silva H,  Mourad G,  Kahan BD, et al.  FTY720, a novel immunomodulator: efficacy and safety results from the first phase 2A study in de novo renal transplantation,  Transplantation ,  2005, vol.  79 (pg.  1553- 1560) Google Scholar CrossRef Search ADS PubMed  56 Shen Y,  Cai M,  Xia W, et al.  FTY720, a synthetic compound from Isaria sinclairii, inhibits proliferation and induces apoptosis in pancreatic cancer cells,  Cancer Lett ,  2007, vol.  254 (pg.  288- 297) Google Scholar CrossRef Search ADS PubMed  57 Yasui H,  Hideshima T,  Raje N, et al.  FTY720 induces apoptosis in multiple myeloma cells and overcomes drug resistance,  Cancer Res ,  2005, vol.  65 (pg.  7478- 7484) Google Scholar CrossRef Search ADS PubMed  58 LaMontagne K,  Littlewood-Evans A,  Schnell C, et al.  Antagonism of sphingosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization,  Cancer Res ,  2006, vol.  66 (pg.  221- 231) Google Scholar CrossRef Search ADS PubMed  59 Schmid G,  Guba M,  Ischenko I, et al.  The immunosuppressant FTY720 inhibits tumor angiogenesis via the sphingosine 1-phosphate receptor 1,  J Cell Biochem ,  2007, vol.  101 (pg.  259- 270) Google Scholar CrossRef Search ADS PubMed  60 Sanchez T,  Estrada-Hernandez T,  Paik JH, et al.  Phosphorylation and action of the immunomodulator FTY720 inhibits vascular endothelial cell growth factor-induced vascular permeability,  J Biol Chem ,  2003, vol.  278 (pg.  47281- 47290) Google Scholar CrossRef Search ADS PubMed  61 Azuma H,  Takahara S,  Ichimaru N, et al.  Marked prevention of tumor growth and metastasis by a novel immunosuppressive agent, FTY720, in mouse breast cancer models,  Cancer Res ,  2002, vol.  62 (pg.  1410- 1419) Google Scholar PubMed  62 Zhou C,  Ling MT,  Kin-Wah LT, et al.  FTY720, a fungus metabolite, inhibits invasion ability of androgen-independent prostate cancer cells through inactivation of RhoA-GTPase,  Cancer Lett ,  2006, vol.  233 (pg.  36- 47) Google Scholar CrossRef Search ADS PubMed  63 Neviani P,  Santhanam R,  Oaks JJ, et al.  FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia,  J Clin Invest ,  2007, vol.  117 (pg.  2408- 2421) Google Scholar CrossRef Search ADS PubMed  64 Maluccio M,  Sharma V,  Lagman M, et al.  Tacrolimus enhances transforming growth factor-beta1 expression and promotes tumor progression,  Transplantation ,  2003, vol.  76 (pg.  597- 602) Google Scholar CrossRef Search ADS PubMed  65 Pawelec G,  Wernet P.  Cyclosporin A inhibits interleukin 2-dependent growth of alloactivated cloned human T-lymphocytes,  Int J Immunopharmacol ,  1983, vol.  5 (pg.  315- 321) Google Scholar CrossRef Search ADS PubMed  66 Masuhara M,  Ogasawara H,  Katyal SL, et al.  Cyclosporine stimulates hepatocyte proliferation and accelerates development of hepatocellular carcinomas in rats,  Carcinogenesis ,  1993, vol.  14 (pg.  1579- 1584) Google Scholar CrossRef Search ADS PubMed  67 Van d V,  Marquet RL,  Eggermont AM.  Cyclosporin A enhances locoregional metastasis of the CC531 rat colon tumour,  J Cancer Res Clin Oncol ,  1997, vol.  123 (pg.  21- 24) Google Scholar CrossRef Search ADS PubMed  68 Van Der EJ,  De Greve J,  Geerts F, et al.  Quantitative study of liver metastases from colon cancer in rats after treatment with cyclosporine A,  J Natl Cancer Inst ,  1986, vol.  77 (pg.  227- 232) Google Scholar PubMed  69 Hojo M,  Morimoto T,  Maluccio M, et al.  Cyclosporine induces cancer progression by a cell-autonomous mechanism,  Nature ,  1999, vol.  397 (pg.  530- 534) Google Scholar CrossRef Search ADS PubMed  70 Engle SJ,  Hoying JB,  Boivin GP, et al.  Transforming growth factor beta1 suppresses nonmetastatic colon cancer at an early stage of tumorigenesis,  Cancer Res ,  1999, vol.  59 (pg.  3379- 3386) Google Scholar PubMed  71 Siegel PM,  Shu W,  Cardiff RD, et al.  Transforming growth factor beta signaling impairs Neu-induced mammary tumorigenesis while promoting pulmonary metastasis,  Proc Natl Acad Sci USA ,  2003, vol.  100 (pg.  8430- 8435) Google Scholar CrossRef Search ADS PubMed  72 Andre N,  Roquelaure B,  Conrath J.  Molecular effects of cyclosporine and oncogenesis: a new model,  Med Hypotheses ,  2004, vol.  63 (pg.  647- 652) Google Scholar CrossRef Search ADS PubMed  73 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, vol.  137 (pg.  14- 20) Google Scholar CrossRef Search ADS PubMed  74 Vivarelli M,  Bellusci R,  Cucchetti A, et al.  Low recurrence rate of hepatocellular carcinoma after liver transplantation: better patient selection or lower immunosuppression?,  Transplantation ,  2002, vol.  74 (pg.  1746- 1751) Google Scholar CrossRef Search ADS PubMed  75 Henry ML.  Cyclosporine and tacrolimus (FK506): a comparison of efficacy and safety profiles,  Clin Transplant ,  1999, vol.  13 (pg.  209- 220) Google Scholar CrossRef Search ADS PubMed  76 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, vol.  4 (pg.  87- 93) Google Scholar CrossRef Search ADS PubMed  77 Kasiske BL,  Snyder JJ,  Gilbertson DT, et al.  Cancer after kidney transplantation in the United States,  Am J Transplant ,  2004, vol.  4 (pg.  905- 913) Google Scholar CrossRef Search ADS PubMed  78 Swinnen LJ,  Costanzo-Nordin MR,  Fisher SG, et al.  Increased incidence of lymphoproliferative disorder after immunosuppression with the monoclonal antibody OKT3 in cardiac-transplant recipients,  N Engl J Med ,  1990, vol.  323 (pg.  1723- 1728) Google Scholar CrossRef Search ADS PubMed  79 Gutierrez-Dalmau A,  Campistol JM.  Immunosuppressive therapy and malignancy in organ transplant recipients: a systematic review,  Drugs ,  2007, vol.  67 (pg.  1167- 1198) Google Scholar CrossRef Search ADS PubMed  80 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, vol.  76 (pg.  1289- 1293) Google Scholar CrossRef Search ADS PubMed  81 Opelz G,  Dohler B.  Lymphomas after solid organ transplantation: a collaborative transplant study report,  Am J Transplant ,  2004, vol.  4 (pg.  222- 230) Google Scholar CrossRef Search ADS PubMed  82 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, vol.  81 (pg.  1227- 1233) Google Scholar CrossRef Search ADS PubMed  83 Yabu JM,  Vincenti F.  Novel immunosuppression: small molecules and biologics,  Semin Nephrol ,  2007, vol.  27 (pg.  479- 486) Google Scholar CrossRef Search ADS PubMed  © The Author [2008]. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved. For Permissions, please e-mail: journals.permissions@oxfordjournals.org Oxford University Press TI - Immunosuppressive therapy and post-transplant malignancy JF - Nephrology Dialysis Transplantation DO - 10.1093/ndt/gfn605 DA - 2008-10-31 UR - https://www.deepdyve.com/lp/oxford-university-press/immunosuppressive-therapy-and-post-transplant-malignancy-3qOiExeGCE SP - 1097 EP - 1103 VL - 24 IS - 4 DP - DeepDyve ER -