Transplantation Reviews 19 (2005) 20 – 31 www.elsevier.com/locate/trre
Rapamycin and tumor growth: mechanisms behind its anticancer activityB Gudrun E. Koehl, Hans J. Schlitt, Edward K. GeisslerT Department of Surgery, University of Regensburg, 93053 Regensburg, Germany
Abstract In the past few years, a heightened awareness has developed toward the problem of cancer occurrence and treatment in organ transplant recipients under immunosuppression. Treatment and prevention of cancer in transplant patients with an intentionally suppressed immune system are generally not considered an ideal situation because intact immunity is important for recognizing and destroying potentially neoplastic cells. Nonetheless, recent studies indicate that fighting tumors is not impossible under conditions of immune suppression, and in fact, immunosuppressive agents have been discovered that possess potent anticancer effects. In particular, one class of immunosuppressants, referred to as mammalian target of rapamycin (mTOR) inhibitors (mTORi), has experimentally shown an ability to suppress the immune system to protect allografts from rejection while simultaneously inhibiting tumor growth. To gain a better understanding of this dual effect, we will review the key intracellular signaling pathways controlled by mTOR. We will discuss how mTORi affects the growth and survival of a variety of nonimmune cells, with a special emphasis on cancer. A better understanding of mTOR-related pathways and the repertoire of normal and neoplastic cells affected by mTORi will likely improve our ability to treat transplant rejection, cancer, and other pathological conditions. D 2005 Elsevier Inc. All rights reserved.
1. Introduction Cancer in patients after organ transplantation is a wellknown life-threatening complication [1]. Malignancies develop in those immunosuppressed patients at a younger age and tend to be more aggressive. The incidence of cancer increases dramatically with time after transplantation [2- 4]. While the risk for cancer is generally increased, certain types of cancer including lymphomas, nonmelanoma skin tumors, and buncommonQ tumors, such as Kaposi sarcoma, are much more prevalent [3,5]. Depending on the type of organ transplantation, the risk for posttransplant non-Hodgkin lymphomas is 12- to 240-fold increased (for kidney and heart/lung transplant patients, respectively [6-8]). The most common malignancy is skin cancer [9]. Not surprisingly the skin cancer rate is exceptionally high in Australia and New Zealand [10]. Other types of cancer also tend to vary geographically [3-5]. For instance, in western countries Kaposi sarcoma has a high incidence in transplant patients,
B
This work has been supported by the Roche Organ Transplantation Research Foundation. T Corresponding author. Tel.: +49 941 6964; fax: +49 941 944 6886. E-mail address:
[email protected] (E.K. Geissler). 0955-470X/$ – see front matter D 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.trre.2005.01.001
whereas it is basically undetected in Japan. Japan in turn has a high incidence of Epstein-Barr virus (EBV) and T-cell lymphotrophic virus type I (HTLV-1) – related tumors in kidney transplant patients. Regardless of the prevalence and cause for cancer in transplant recipients, treatment of tumors that do occur is complicated by the need to prevent graft rejection with immunosuppressive drugs. Immunosuppression is thought to be the primary culprit in transplant-related cancer [11]. In particular, azathioprine and cyclosporine-based immunosuppression have been clinically linked to the development of malignancies [3]. Experimentally, cyclosporine has been shown to promote tumor growth by different research groups [12-14], including ours [15,16]. Meanwhile, the development of other agents with different modes of action, such as mammalian target of rapamycin inhibitors (mTORi) has opened the possibility that not all immunosuppressive drugs promote tumor growth and, in fact, may even be used to treat cancer. Indeed, rapamycin has shown potent antitumor effects in several experimental tumor models (Table 1). Moreover, case reports and the first results of clinical studies (Table 2) indicate that rapamycin may be effective against tumors in transplant and nontransplant patients. These studies are critical because they open the way to adjust immunosuppressive regimens in a way to fight
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
21
Table 1 Tumor-related nonimmune cells and experimental tumors responsive to the effect of mTOR inhibitors Cell type Nonneoplastic cells Endothelial cells
Cell line
Smooth muscle cells
Bovine aortic [17], HUVEC [15,18] HuAoSMC [19-21]
Fibroblasts Neurons
3T3 [17,19] embryonic neurons [22]
Tumor cells B-cell lymphoma
AB5, JB7, MF4 (PTLD-derived cell lines) [23], BKS-2, L1.2, NFS.1.1, WEHI-279 [24]
B-cell lymphoma B-lymphoblastoid cells
Bladder cell carcinoma Brain cancer Brain cancer (glioma) Breast cancer
Colon carcinoma
Colon carcinoma Colon carcinoma Hepatoma/hepatocellular carcinoma Mast cell –derived tumor Medulloblastoma Melanoma
BJAB [27], PTLD-like EBV infected cell lines (panel) [25] T2 [28] DAOY [29]
Rhabdomyosarcoma T-cell lymphoma
AB5 [23], PTLD-derived Human tumors [24], PTLD-like EBV- infected cell lines [25]
EBV infection
breast cancer cell line panels [32-35]
SF295 [30], U-251 [31] MDA-MB-468 MDA-MB-435 [32]
Human Human
CT26 [15]
CT26 [15,16,18]
Murine
CT38 [36,37] GC3 [30]
Murine Human
H4IIEC [38], HepG2 [39], SK-Hep1, HepB3, PCL/PRF/5 [40], H4IIE [41] V2D1, V3D6, V3D8 (autocrine tumor cell lines) [42]
HL60 [27] DU-145, PC-3 PPC-1, TSU, LNCap [30,53,54] 786-O [56] RH18, RH30 [30,57-59] HTLV-I–transformed cells [19], YAC-1 [60], EL-4 [61]
Immature and mature lymphomas, EBV infection
Murine
Murine
B16 [15]
Additional information
Regulation of translation
T24 [28]
Muscle BC3H1 [44] Neuroblastoma KP-N-RT [45] Non–small cell lung cancer H69, H345, H510 [46], KLN-205, A-549 [47,48] Ovarian cancer IGROV1, OVCAR3, A2780, SKOV3, OVCAR4, OVCAR5 [49,50] Pancreatic cancer L3.6pl [51]
Renal cell cancer
Tumor origin
bFGF and VEGF induction, wound-healing, angiogenesis bFGF and PDGF induction, angiogenesis, wound healing
genetically induced lymphoma models [26]
Multiple myeloma
Promyelotic cells Prostate cancer
Tumor
Sensitive and resistant cell lines/tumors, depending on oncogene and Akt-pathway status Angiogenesis, combination with cyclosporine and 5-fluorouracil, combination with transplantation
Autophagy
v-H-ras DAOY, TE-671 [29] B16 [15,16,37]
Human Murine
8226, OPM-2, U266 [43]
Human
Combination with cyclosporine, combination with transplantation Reduction of tumor cell size induction of differentiation
KLN-205 [46], A-549 [47] OVCAR5 [30]
L3.6pl [51], CA20948 [52] DU-145, PC-3 [30], PIN [55] spontaneous murine tumor [28], 786-O [56] RH18 [30]
HTLV-I, T-cell lymphotrophic virus type I; PDGF, platelet-derived growth factor.
Murine, human No apoptosis, reduction in tumor size and metastasis Human Some resistant cell lines, regulation of apoptosis pathways, no significant effect on tumor growth Human, rat Thrombosis in the tumor, combination with anti-VEGF antibody, 5-fluorouracil Human
Induction of Akt, HIF1a neoplasms induced by Akt overexpression Murine, human Combination with cyclosporine, reduced number of tumors in metastasis model Human Dependency on IGF-1/IGF receptor Cell lines with acquired resistance
22
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
Table 2 Clinical studies, case reports, and analysis of patient material regarding mTORi Type
Tumors
Drug
Additional information
Patient samples Case report
Head and neck cancer [62] Kaposi sarcoma [63]
No drug Sirolimus
Phase I
Hepatocellular carcinoma, metastatic renal cell carcinoma, metastatic sarcoma [64] Renal cell carcinoma, breast adenocarcinoma [65] Advanced solid tumors (including gastrointestinal tumors) [66] Fibrosarcoma, non–small cell lung carcinoma [67] Advanced gastrointestinal tumors [68] Refractory renal cell carcinoma [69,70] Renal transplant patients [71]
AP23573
Akt pathway, no rapamycin Conversion to rapamycin leads to tumor regression Dose escalation, preliminary meeting reports
Phase I Phase I Phase I Phase I/II Phase II Results from 5 multicenter studies (phases II and III) Retrospective single-center analyses
PTLD, hepatoblastoma (pediatric liver transplant patients) [72]
cancer while simultaneously maintaining a safe level of immunosuppression to protect allografts —understandably, this concept can be viewed with skepticism. Nonetheless, a better knowledge of the biology of tumors highly prevalent in the transplant population will likely lead to the development of rationally balanced tumor-transplant drug treatment options. Therefore, a special focus should be placed on understanding how immunosuppressive drugs affect vital signaling pathways that promote tumor transformation, proliferation, angiogenesis, and metastasis. Recently, we have been interested in the potential for mTORi to play the dual role of immunosuppressive and antitumor agent in transplant recipients. In this review, we discuss the mode of action of mTORi with a main focus on their effects on intracellular signaling pathways. These effects will be discussed in view of mutational and biochemical changes that are typical for transformed cells leading to tumor genesis and growth. A special emphasis will be placed on tumor types with a high prevalence in transplant patients. 2. Rapamycin: discovery and characterization Today, the class of mTORi consists of rapamycin (Rapamune/sirolimus, Wyeth) and its derivatives (CCI779, Wyeth, RAD001/everolimus, Novartis) [73] and the analogue ap23573 (ARIAD Pharmaceuticals). Rapamycin was originally found in a discovery program for antimicrobial agents from natural resources [19]. A strain of Streptomyces hygroscopicus with antifungal properties was isolated from a soil sample collected at Easter Island (Rapa Nui). The active substance, named rapamycin, is a lipophilic macrocyclic lactone with strong antimicrobial and, as later found,
CCI779 CCI779 RAD001 RAD001 CCI779 Sirolimus
Sirolimus
Safety, tolerability, and pharmacokinetics Combination with leucovorin and 5-fluorouracil, discontinued because of toxicity Dose escalation, toxicity, pharmacokinetics, pharmacodynamics Combination with imatinib in imatinib refractory tumors Efficacy, safety, and pharmacokinetics of multiple doses Rate of malignancy after transplantation, 2-y results, sirolimus in addition to cyclosporine Only sirolimus-treated patients analyzed
immunosuppressive activity. Since first characterized in 1975, rapamycin’s structure, biosynthesis, and binding partner(s) have been reported [19]. Rapamycin binds to intracellular FK binding protein (FKBP) 12, forming a drug/ immunophilin complex that modulates the activity of intracellular targets. In a similar fashion, cyclosporine binds to the immunophilin cyclophilin A, causing inactivation of calcineurin and consequently blocking nuclear factor of activation of T cells (NFAT) translocation into the nucleus. This action basically defines the class of calcineurin inhibitors within the immunosuppressants [19,74]. Interestingly, FK506 (tacrolimus) also binds to FKBP12. Whereas the tacrolimus/FKBP12 complex forms a ternary complex with calcineurin, thereby exhibiting the same immunosuppressive mode of action as cyclosporine [74], rapamycin has a different target and consequently a different mode of action. The rapamycin/FKBP12 complex binds to mTOR (mammalian target of rapamycin) [19,75,76]. This 289-kDa protein (a member of the PI-kinase family) is a pivotal regulator of cell growth and proliferation for many cell types. Although mTOR inhibition interferes with several intracellular mechanisms [77], one of its best known effects is G1 cell cycle arrest [78]. Moreover, very diverse cell types ranging from cells of the immune system [19,79], to smooth muscle cells [36,80], to endothelial cells [15,80], and to tumor cells (Table 1) respond poorly to cytokines and growth factors in the presence of rapamycin. The effects on different cell types not only have led to a variety of potential therapeutic uses but also indicate a potential for undesired effects. Indeed, rapamycin’s diverse effects have made for a curious trail of therapeutic applications. Although originally discovered for its antifungal action [19], enthusiasm for rapamycin’s therapeutic usefulness against infections was
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
trolled by mTOR. One key pathway controlled by mTOR, which is particularly important for cancer cells, is the phosphatidylinositol-3 (PI3) kinase/Akt signaling pathway [19,76,77,81,85-87] (Fig. 1). Basically, PI3 kinase is activated by growth factor/cytokine-triggered receptors, leading to phosphorylation of Akt and subsequent downstream effectors including 40S ribosomal p70S6 kinase and 4E-BP1 (eukaryotic translation initiation factor 4E binding protein 1). In more detail, growth factors and cytokines [19], including interleukin 2, vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and insulin-like growth factor (IGF), lead into the mTOR pathway by activating PI3 kinase via receptor tyrosine kinases and G-protein–coupled receptors [88]. Activation of PI3 kinase leads to production of the second messenger phosphatidylinositol (3,4,5)-P3 (PIP3). PIP3 activates Akt, leading to its translocation to the cytoplasmic membrane. PTEN (phosphatase and tensin homolog deleted from chromosome 10) counteracts Akt activation by eliminating PIP3 and can thereby bswitch offQ the PI3 kinase/Akt pathway [89]. Loss of PTEN function occurs in many cancers and is consequently correlated with increased Akt activity, making cancer cells resistant to apoptosis [90], but potentially highly sensitive to mTORi [26,91]. Akt in turn phosphorylates tuberoses sclerosis complex II (TSCII) leading to inactivation of the TSCI/TSCII complex, which negatively regulates mTOR. TSCI and TSCII are associated with dominant genetic or spontaneous mutations causing a disorder called tuberous sclerosis [81]. Loss-of-function mutations in TSCI or II cause enlarged cells and organs in Drosophila, whereas overexpression of both proteins dramatically slows cell growth. Deficiency in TSCII leads to constitutive phosphorylation of p70S6 kinase, but cells remain sensitive to rapamycin. Therefore, both Akt and TSCI/II
diminished because of its immunosuppressive effect. Sehgal [79] insightfully pursued the drug’s immunosuppressive potential to actually use rapamycin against the immune system, and it has since been fully developed in transplantation medicine for this purpose. Another property recognized early in rapamycin’s development was its anticancer activity [37]. Less than desirable potency at cytotoxic doses and its known immunosuppressive activity undoubtedly reduced early enthusiasm for its use in oncology. However, in recent years, the antitumor activities of mTORi have come back into focus with the development of rapamycin analogues CCI779 and RAD001, which are now being tested as pure oncological agents in phases I and II trials (Table 2). Phases I and II trials with ap23573 are also under way. In addition, the transplant community has become increasingly aware of the high cancer incidence in transplant recipients and is seeking a means of immunosuppression that could decrease this problem —mTORi could provide new hope for this purpose. 3. Mammalian target of rapamycin action and the phosphatidylinositol-3 kinase/Akt pathway Mammalian target of rapamycin is a central factor for integrating adaptive responses to external and internal signals [81] (eg, growth factors, hypoxia [80,82], and amino acid/ nutrient levels [83]) with regulation of transcription [77] and translation [84]. These responses in many cases alter critical processes such as cell cycle progression and cell proliferation. Evolutionary conservation of mTOR, as seen by wellcharacterized homologues in yeast (Saccharomyces cerevisiae) and Drosophila, underscores its important role in the regulation of cell functions in response to nutrient status. In mammals, nutrient status has been integrated with highly developed regulatory mechanisms, including those conGF
23
IL-2, VEGF, PDGF, IGF, EGF, FGF
ErbB2 receptor
P
PIP2
PI3-kinase
PTEN Akt
br a
integrins
em
P
lm
PIP3
ce l
ras
ne
nutrients TSCI TSCII P
upstream
Rheb
rapamycin FKBP12 mTOR raptor Fig. 1. PI3-kinase Akt pathway upstream of mTOR.
24
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
brain) [81]. Small G-proteins, are only active in the GTP (guanosine triphosphate) – bound state, generally using a specific GTPase-activating protein as a bhelper Q to convert it to the inactive GDF (guanosine diphosphate) – bound state. The GTPase-activating protein for Rheb is TSCII, thus linking Akt and mTOR. Finally, to phosphorylate and therefore activate the downstream effectors of this pathway (p70S6 kinase and 4E-BP1), mTOR must be in a complex with raptor (regulatory associated protein of mTOR) [77]. Raptor serves as a docking site for p70S6 kinase and 4E-BP1 and brings the target proteins in position for phosphorylation by mTOR. Rapamycin causes disruption of the raptor/ mTOR complex, leading to mTOR inactivation (Fig. 1).
rapamycin FKBP12 mTOR raptor
P
P
downstream
4E-BP1
p70S6K
eIF4E
eIF4B
cyclins translation
CDKs p53
P
pRb
ribosomal biosynthesis
apoptosis
4. What can be expected of mTORi use?
cell proliferation
tumor immune system Fig. 2. Downstream targets of mTOR I: cell cycle and apoptosis.
are upstream of mTOR and rapamycin inhibition. More recently, it has been shown that the interaction of the TSCI/II complex with mTOR is indirectly mediated by the small G-protein, Rheb (Ras homologue enriched in
Inhibition of mTOR leads to a 15% to 20% decrease in overall protein synthesis in many cell types [91], with its main effect being, as mentioned above, cell cycle arrest in G1. In yeast, mTOR signaling modulates transcription of genes involved in nutrient use, respiration, protein degradation, and amino acid biosynthesis [76,77]. Other effects include apoptosis (depending on retinoblastoma protein [pRB] and p53 status) [91], inhibition of T-cell activation [92], inhibition of cell migration [93] and invasion [93], changes in the (actin-)cytoskeleton [78], membrane trafficking [78], autophagy [94], and decreased expression of specific proteins such as VEGF [95]. The two major downstream effectors of mTOR are p70S6 kinase and eukaryotic translation initiation factor 4E (eIF4E). Both of these proteins and their targets are
rapamycin FKBP12 mTOR raptor
P
c-Myc
STAT3 P
downstream
transcription
p70S6K
HIF1α
HIF1α
glucose use genes
VEGF
P
translation
VHL
ribosomal biosynthesis cell proliferation
degradation
signalling to other cells
tumor angiogenesis
immune system
Fig. 3. Downstream targets of mTOR II: physiology and adaptive responses.
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
needed for assembly of translation machinery and for the recruitment of ribosomes to mRNA (Figs. 2 and 3). The effect of mTOR on eIF4E is indirectly mediated via 4E-BP1 [77,84,91], which is a suppressor of eIF4E. Phosphorylation of 4E-BP1 by mTOR leads to release of suppression of eIF4E and thereby to efficient initiation of CAP-dependent translation of mRNA. Rapamycin treatment keeps 4E-BP1 in its suppressor state and blocks not only efficient translation of key proteins for the translation machinery, but also for the synthesis of proteins necessary for cell cycle progression (eg, cyclin D1 [90]) and survival (eg, c-Myc [96]). Thus, rapamycin use leads to reduced cell growth and cell cycle arrest in G1. Interestingly, phosphorylation of 4E-BP1 by protein kinase Cd can be blocked by rapamycin, although it is Akt-independent [97]. Indeed, signaling through MAP-kinase pathways [98] (eg, the Erk and p38 families) also leads to eIF4E activation. Therefore, other important signal transduction pathways have the potential to interact with mTOR signaling or possibly provide a bypass around the effects of rapamycin [77,91]. Activated p70S6 kinase increases translation of the so-called 5VTOP (terminal oligopyrimidine) tract mRNAs [84], which encode ribosomal proteins, elongation factors, and other proteins of the translational machinery. Notably, several proteins involved in recruiting ribosomes including eIF4B, eIF4G, and indirectly by its repressor, eIF4E, are sensitive to rapamycin [77]. Therefore, mTOR has a profound effect on cell growth and cell cycle progression by regulating ribosomal protein synthesis and recruitment. In terms of sufficient use of resources and energy, it is only logical that mTOR can regulate the upstream processes of translation and transcription. In effect, mTOR can stimulate the transcription of rRNA genes, ribosomal proteins, tRNA genes, and some transcription factors [77]. With specific regard to the cell cycle (Fig. 2), mTOR is needed for efficient translation of cyclin D1 [90]. Cyclin D1 in complex with cell cycle –dependent kinase (CDK) 4 is needed for phosphorylation of pRB [91]. pRB is involved in the regulation of Pol I/II/III –dependent transcription and provides a link to the apoptosis pathways. In addition, rapamycin stabilizes p27, which inhibits the activity of the cyclin/CDK complexes [91]. Moreover, it has been shown that rapamycin inhibits phosphorylation of STAT3 (signal transducer and activator of transcription 3) via mTOR [99]. The transcription factor STAT3 mediates stabilization of cyclin D1 and up-regulates c-Myc. Other cyclins and CDKs are involved in mTOR-regulated events, and once again, mTOR/rapamycin acts as an important regulator in several different ways. For example, by inhibiting CDK1 rapamycin induces G1 arrest in T lymphocytes [91], thereby preventing T-cell proliferation, which contributes to an immunosuppressive effect exploited in transplant patients. From a different perspective, inhibition of the cell cycle in tumor and endothelial cells provides 1 potential explanation for rapamycin’s antitumor and antiangiogenic properties. Interestingly, the cell cycle also plays a role in rapamycin’s
25
ability to promote cell apoptosis. Of special interest with regard to tumors is the fact that the p53 status of a cell can determine its fate with regard to rapamycin-induced apoptosis. For instance, cells with wild-type p53 arrest in G1 and remain viable, whereas p53 mutant rhabdomyosarcoma cells undergo apoptosis [100]. This underscores the importance of the G1/S –phase checkpoint control by p53, which is lost in many tumor cells and may be a mechanism for rapamycin to more or less selectively target tumor cells by sending them into death by unsuccessful cell division. Therefore, although rapamycin is generally considered an antiproliferative agent, there are circumstances where it also promotes cell death. Integration of these intracellular pathways with the environment surrounding individual cells is controlled in a large part by growth factors. Growth factors not only activate the PI3 kinase/Akt pathway; expression of these molecules is controlled by the same signaling pathway. For example, inhibition of mTOR leads to less production and release of VEGF [53], thus having the potential to systemically reduce growth factor–dependent responses. VEGF is regulated at the translational and transcriptional level by rapamycin via 4EB-P1 [101] and hypoxia-induced factor 1a (HIF1a) [53], respectively. The transcription factor HIF1a is activated and translocated to the nucleus during hypoxic conditions. Mammalian target of rapamycin induces a protein stabilization function that leads to accumulation of activated HIF1a [102]. Consequently, rapamycin can potentially reduce accumulation of HIF1a in the nucleus independent of the cell reaction to hypoxia (Fig. 3). This could be important for tumor growth, where hypoxia leads to increased VEGF production, which in turn promotes tumor angiogenesis. In addition, in cancers where HIF1a is constitutively boverexpressed,Q rapamycin can have an antitumor effect independent of hypoxia by reducing HIF1a-induced transcription of growth factors, glycolytic enzymes, and glucose transporters [103]. For instance, this type of HIF1a overactivation occurs in cells where mutations in the von HippelLindau (VHL) gene prevent efficient HIF1a degradation. Interestingly, patients with renal cell carcinoma (often with VHL mutations) have recently been shown to respond to the rapamycin analogue CCI-779 [69]. In summary, rapamycin inhibits pivotal downstream regulators of cell growth, division, and survival and therefore can be expected to affect many normal, as well as neoplastic, cell types. The response of each cell type to rapamycin treatment likely depends substantially on the physiological and genetic state of the cell. Most affected will be not only cells depending on growth factor– or cytokine-induced cell growth and division, such as T cells and tumor cells, but also cells needed for proliferative processes such as angiogenesis and wound healing. Different genetic mutations may make tumor cells susceptible to direct rapamycin effects, particularly apoptosis. Screening for such tumors and tumors highly dependent on angiogenesis will make best use of rapamycin’s antitumor efficacy.
26
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
5. Which nonimmune cells respond to rapamycin? mTOR’s central role in controlling cell proliferation and cell cycle progression in living organisms from yeast and Drosophila to mammals suggests that rapamycin affects several cell types. Besides effects on T cells, rapamycin influences at least 4 other types of cells: (1) tumor cells, (2) vascular cells, (3) virally infected cells, and (4) developmentally or regeneratively active cells. Effects on tumor cells were reported shortly after rapamycin’s discovery [37]. Subsequent screening of the National Cancer Institute human tumor cell line panel [73,85] has shown significant growth inhibition in breast cancer, prostate cancer, renal cell cancer, pancreatic cancer, melanoma, leukemia, and glioblastoma. At present, different antiproliferative effects and changes in signal transduction, as well as changes in translation and transcription, have been elucidated in many cancer cell types (Table 1). Studies with murine and human tumors in mice have shown an antitumor effect of rapamycin as a single agent or in combination with other chemotherapeutics (Tables 1 and 2). In general, good targets for rapamycin treatment would theoretically be tumors with mutations in proteins with a close connection to the PI3 kinase/Akt pathway. These include gain-offunction receptor mutations, such as ErbB2 overexpression in breast cancer. PTEN mutations occurs in up to 50% of human solid tumors [104] and have been extensively studied in melanoma [89], breast cancer [105], glioblastoma [89], and prostate carcinoma [106]. Both ErbB2 and PTEN mutations lead to constitutive activation of PI3 kinase and Akt, therefore making those tumors potential targets for treatment by mTORi [90,107,108]. Other, somewhat unexpected, activators of PI3 kinase include the oncogenic small G-protein ras [90] and integrins [109] (Fig. 1). Because ras generally transmits signals to the MAP kinase pathway, and the integrins provide a link to cell movements, two other aspects of cell function and tumor biology are linked to mTOR. As discussed earlier, mutations in the tumor suppressor p53 may promote rapamycin-induced apoptosis of different tumor cells, thereby potentially enhancing its cytotoxic properties (Fig. 2). Another oncogene that is often associated with renal cell cancer [78] and could be a key factor for mTORi sensitivity is VHL. Despite being blocatedQ downstream of mTOR, HIF1a activation by VHL mutations [107] may be counterbalanced by rapamycin (Fig. 3). Of particular importance to transplant recipients, skin cancer has a high correlation to sun exposure and ultraviolet-induced damage. The combination of general immunosuppression and promotion of aberrant repair mechanisms by drugs such as cyclosporine [110] is thought to promote transformation of cells. Interestingly, studies indicate that ultravioletinduced damage may activate mTOR-dependent p70S6 kinase, providing a potential target for mTORi [111]. Even cancers highly refractory to treatment, including pancreatic cancer, may be responsive to rapamycin therapy. In vitro studies and xenogenic (human to mouse) models have
demonstrated effective rapamycin monotherapy against pancreatic cancer that synergizes with standard chemotherapy (gemcitabine [51], 5-fluorouracil [52]) — curiously, rapamycin also works well in combination with anti-VEGF antibody [112]. In addition, we have shown that one mechanism of the effect of rapamycin on pancreatic cancer relates to the promotion of thrombosis specifically in tumor vessels, thus reducing the tumor blood supply and inhibiting tumor growth; no effects on normal blood vessels were observed in these experiments [51]. Whether this activity has any relationship to rare cases of thrombosis in rapamycin-coated vascular stents or to hepatic artery thrombosis after liver transplantation will require additional research. Recently, we have correlated mTORi anticancer effects with inhibition of angiogenesis in animal models [15]. In fact, vascular-associated tissue contains a third group of cells targeted by mTORi. The mechanisms of this antiangiogenic effect are not completely understood, but as discussed above, HIF1a-dependent signaling can be inhibited by rapamycin, thereby hindering angiogenic signaling at its origin. In addition, vascular endothelial and smooth muscle cells are affected by mTOR inhibition. Rapamycin not only inhibits the proliferation and migration of these vascular cells in response to VEGF, but it can also promote endothelial cell death under proliferative conditions [51]. These antiangiogenic effects have been correlated with markedly reduced tumor growth in different mouse models [15,18,28,51]. The effects of rapamycin on bnormalQ vascular cells embedded within a neoplasm suggest that although cancer cells may not necessarily be directly affected by rapamycin, indirect effects on angiogenesis could help to control tumor growth [15,51,112]. In a different respect, angiogenesis is essential for tissue regeneration and for processes such as wound healing [113,114]. Therefore, caution should be taken when considering mTORi use in patients with wounds or transplant recipients demanding tissue regeneration (eg, period immediately after living-related liver transplantation). With regard to virally infected cells, it is of special interest that transplant patients infected with EBV tend to develop posttransplant lymphoproliferative disorders (PTLDs). Interestingly, it has been shown that mTOR inhibition blocks proliferation of human EBV-transformed B cells in immunodeficient mice and induces their apoptosis in culture [23]. A key effect of rapamycin on EBV-infected B cell proliferation described in this study is through an inhibitory action on interleukin 10 secretion. Therefore, mTORi may prove to be valuable in treating PTLD. Viral infections may also be important for other types of cancer in transplant patients, and the effects of immunosuppression could be critical. For instance, hepatitis C virus (HCV) is thought to be important for development and recurrence of hepatocellular carcinoma (after liver transplantation). Interestingly, it has recently been reported that the mTOR pathway controls the phosphorylation/activation of a critical HCV-replication protein (nonstructural protein 5A, NS5A) [115]. Moreover, rapamycin was shown to inhibit NS5A activity, potentially impacting
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
HCV replication, and therefore cancer recurrence in hepatocellular carcinoma patients receiving a liver transplant. A fourth group of cells depending on mTOR is developmental [94] and regenerative cells. For example, a mutation in mTOR can cause severe brain defects in mice and is homozygous embryonic lethal; the same phenotype is induced by rapamycin injections during pregnancy. Although the mechanism by which rapamycin causes such severe developmental problems is not clear, it has been hypothesized that the drug induces autophagy through mTOR dysregulation. Indeed, nutrient-scavenging autophagy plays an important role in developmental processes requiring controlled cell death (eg, insect morphogenesis and Xenopus spinal cord development). Nonetheless, mTOR has also been implicated in bnormalQ neuronal function because neurotransmitters, such as serotonin and dopamine, and stimulation of the l-opioid receptors (mediating morphine effects) operate via the PI3 kinase/p70S6 kinase/4E-BP1 pathway. There are also a number of examples of how mTORi affects cell regeneration. For instance, bone marrow suppression is a well-known side effect of mTORi use in transplant recipients [116]. Cell regeneration could be critical as well for liver regeneration in patients who receive either a living-related or split liver transplant. In fact, in an experimental rat model where a partial hepatectomy is performed, it has been reported that liver regeneration is impaired by rapamycin treatment [117]. In summary, mTORi has diverse effects on normal cells that are important for tissue-organ development and regeneration. These potential effects need to be taken into consideration when using mTORi during periods where these processes are critical. 6. Will mTORi synergize with standard chemotherapy against cancer? A practical aspect of mTORi as anticancer agents is that they will likely be best used in combination with additional chemotherapy. Examples of this strategy are emerging with time. For instance, we have recently reported experimentally that rapamycin works best against pancreatic cancer when combined with gemcitabine chemotherapy [51]. This work has led to the initiation of a clinical pilot study at our institution using this combination treatment strategy for pancreatic cancer. For colon cancer, we have shown that rapamycin synergizes well with 5-fluorouracil chemotherapy in mice [18]. In this case, we could show mechanistically that rapamycin blocks a proangiogenic escape pathway that may allow tumors to compensate for 5-fluorouracil treatment. Other studies have also shown that mTORi treatment can be effective to break resistance that tumor cells may develop against other chemotherapeutics. One example is that chronic myelogenous leukemia cells tend to become resistant to the effects of the tyrosine kinase inhibitor imatinib (STI571, Gleevec, Novartis). However, resistant chronic myelogenous leukemia cells are sensitive to the effects of imatinib when combined with rapamycin because the cells use mTOR-
27
dependent pathways as an escape mechanism [118]. In ovarian cancer models, inhibition of the PI3 kinase/Akt pathway increases the sensitivity of tumors to paclitaxel [119] and cisplatin [49]. In yet another example, chemoresistance to tamoxifen because of overactivation of Akt can be reversed in breast cancer xenografts by mTORi (CCI779) [120]. Therefore, the activity of molecules in the PI3 kinase/ Akt/mTOR pathway may not only be involved in tumor chemoresistance, but their activation status may be indicative of tumor cell sensitivity to mTORi and potential synergism with standard chemotherapy. 7. Will mTORi be effective against cancer in transplant recipients? When considering the treatment of cancer in transplant recipients, a complicated situation emerges. The goal is to attack a tumor in the face of immunosuppression used to protect the allograft. Because of the known antitumor effects of the immune system, this seems an uphill battle. However, the emergence of immunosuppressants with both immunosuppressive and antitumor effects (at least experimentally) could provide one solution to the problem. To better understand the potential for mTORi to function simultaneously as an immunosuppressant and anticancer treatment, we have developed mouse models mimicking the scenario in humans [16]. Indeed, we have recently reported that a mouse allograft can be protected by rapamycin against immunologic rejection with simultaneous antitumor effects. Furthermore, we show that the antitumor effect remains fully intact when rapamycin is used in combination with cyclosporine, which otherwise promotes tumor growth in these models. Our results demonstrate in principle that mTORi can exhibit a dual function against allograft rejection and tumors in an organ transplant situation. At present, controlled prospective clinical data are not yet available to confirm these experimental results. However, some hints toward a potential effect have been recently published. For example, Campistol et al [63] have reported two renal transplant recipients who had developed Kaposi sarcoma under cyclosporine treatment and showed complete tumor regression when the patients were switched to a sirolimus/mTORi immunosuppressive regimen. Although it is not yet clear whether the tumor regression can be attributed to sirolimus in these cases (because this was not a controlled study), this report points to the fact that tumor regression can occur simultaneously with sirolimus immunosuppressive treatment. Interestingly, Kaposi sarcoma is a highly vascular tumor where mTORi might be expected to exhibit antiangiogenic effects. There are also clues in the literature that sirolimus might be useful in liver transplant patients with a pretransplant hepatocellular carcinoma. In a pilot study from Kneteman et al [121] using sirolimus-based immunosuppression, two groups of patients were examined; one group of patients was within the Milan criteria (n = 19), whereas a second group of patients presented with extended hepatocel-
28
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
lular carcinoma (n = 21). Tumor recurrence rate in the Milan group at a median follow-up of 17 months was 5.3%, which compares favorably to published results of patient cohorts adhering to the Milan criteria under conventional immunosuppression. The tumor recurrence rate in the extended criteria group was 19%, which again compares favorably to data from patient cohorts with extended criteria under standard immunosuppression [122]. Although this study only reports on a small number of patients and is not controlled, it suggests a potential for sirolimus in a tumortransplant setting. Finally, regarding the prevention of de novo cancer in transplant recipients, only early data are available. Kauffman et al [123] from United Network for Organ Sharing have examined more than 33 000 renal transplant patients over a 2-year period and found that the relative risk for developing de novo cancer was decreased by 59% in patients on an mTORi-based (used together with a calcineurin inhibitor) versus calcineurin inhibitor–based immunosuppressive therapy. Whether this early trend will continue to diverge in favor of mTORi, narrow, or even reverse itself, as these patients enter the middle to late posttransplant period (when most cancers occur), is simply unknown. Skin cancer, in particular, will be interesting to observe over time because this most common type of transplant-related cancer tends to occur only after several years of immunosuppression. 8. Conclusions mTOR is emerging as a central molecule for integrating several important internal and external signals with essential cell functions such as biosynthesis of ribosomes, translation, and even transcription. Therefore, mTOR plays an important role for survival and differentiation at the single-cell level and for complex multicellular entities such as the immune system and tumors. Because operational mTOR pathways are needed by the immune system, mTORi has been incorporated successfully into immunosuppressive regimens to prevent organ allograft rejection. The effects of mTORi on endothelial cells and smooth muscle cells led also to the development of rapamycin-coated vascular stints for vascular stenosis. These devises have been used with great success clinically in preventing restenosis of blood vessels. A major focus of this review has been the potential for mTORi to be used against cancer. Clearly, mTORi not only blocks normal tumor cell proliferation, but they can also mitigate the effects oncogenic mutations have on intracellular pathways connected to cell proliferation and apoptosis. Furthermore, the normal process of angiogenesis, essential for tumor survival, is inhibited by drugs such as rapamycin, thus providing yet another avenue of anticancer activity. Because of this broad spectrum of effects, mTORi must be used with caution in clinical situations where processes such as tissue regeneration or wound healing are necessary. To minimize these side effects and to achieve its most favorable therapeutic effects in patients, we must continue to better
understand the complex network of intracellular pathways that mTOR influences. References [1] Penn I. Malignancies associated with immunosuppressive or cytotoxic therapy. Surgery 1978;83:492 - 502. [2] Penn I. Incidence and treatment of neoplasia after transplantation. J Heart Lung Transplant 1993;12:S328-36. [3] Haberal M, Karakayali H, Emiroglu R, et al. Malignant tumors after renal transplantation. Artif Organs 2002;26:778 - 81. [4] Hoshida Y, Aozasa K. Malignancies in organ transplant recipients. Pathol Int 2004;54:649 - 58. [5] Penn I. Tumors after renal and cardiac transplantation. Hematol Oncol Clin North Am 1993;7:431 - 45. [6] Opelz G, Dohler B. Lymphomas after solid organ transplantation: a collaborative transplant study report. Am J Transplant 2004;4: 222 - 30. [7] Scadden DT. The malignant side of successful transplantation. Am J Transplant 2004;4:153 - 4. [8] Lutz J, Heemann U. Tumours after kidney transplantation. Curr Opin Urol 2003;13:105 - 9. [9] Euvrard S, Ulrich C, Lefrancois N. Immunosuppressants and skin cancer in transplant patients: focus on rapamycin. Dermatol Surg 2004;30:628 - 33. [10] Ramsay HM, Fryer AA, Hawley CM, et al. Non-melanoma skin cancer risk in the Queensland renal transplant population. Br J Dermatol 2002;147:950 - 6. [11] Geissler EK, Schlitt HJ. The relation between immunosuppressive agents and malignancy. Curr Opin Organ Transpl 2004;9:394 - 9. [12] Freise CE, Ferrell L, Liu T, et al. Effect of systemic cyclosporine on tumor recurrence after liver transplantation in a model of hepatocellular carcinoma. Transplantation 1999;67:510 - 3. [13] Hojo M, Morimoto T, Maluccio M, et al. Cyclosporine induces cancer progression by a cell-autonomous mechanism. Nature 1999; 397:530 - 4. [14] Masuhara M, Ogasawara H, Katyal SL, et al. Cyclosporine stimulates hepatocyte proliferation and accelerates development of hepatocellular carcinomas in rats. Carcinogenesis 1993;14:1579 - 84. [15] 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;8: 128 - 35. [16] Koehl GE, Andrassy J, Guba M, et al. Rapamycin protects allografts from rejection while simultaneously attacking tumors in immunosuppressed mice. Transplantation 2004;77:1319 - 26. [17] Akselband Y, Harding MW, Nelson PA. Rapamycin inhibits spontaneous and fibroblast growth factor beta–stimulated proliferation of endothelial cells and fibroblasts. Transplant Proc 1991;23: 2833 - 6. [18] Seeliger H, Guba M, Koehl GE, et al. Blockage of 2-deoxy-d-ribose– induced angiogenesis with rapamycin counteracts a thymidine phosphorylase–based escape mechanism available for colon cancer under 5-fluorouracil therapy. Clin Cancer Res 2004;10:1843 - 52. [19] Sehgal SN. Sirolimus: its discovery, biological properties, and mechanism of action. Transplant Proc 2003;35:7S - 14S. [20] Marx SO, Jayaraman T, Go LO, et al. Rapamycin-FKBP inhibits cell cycle regulators of proliferation in vascular smooth muscle cells. Circ Res 1995;76:412 - 7. [21] Cao W, Mohacsi P, Shorthouse R, et al. Effects of rapamycin on growth factor–stimulated vascular smooth muscle cell DNA synthesis. Inhibition of basic fibroblast growth factor and platelet-derived growth factor action and antagonism of rapamycin by FK506. Transplantation 1995;59:390 - 5. [22] Schratt GM, Nigh EA, Chen WG, et al. BDNF regulates the translation of a select group of mRNAs by a mammalian target of
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
[23]
[24]
[25]
[26]
[27] [28]
[29]
[30]
[31]
[32]
[33]
[34] [35]
[36]
[37]
[38] [39]
[40]
[41]
[42]
rapamycin-phosphatidylinositol 3-kinase–dependent pathway during neuronal development. J Neurosci 2004;24:9366 - 77. 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 - 89. Muthukkumar S, Ramesh TM, Bondada S. Rapamycin, a potent immunosuppressive drug, causes programmed cell death in B lymphoma cells. Transplantation 1995;60:264 - 70. 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 U S A 2000;97:4285 - 90. Wendel HG, De Stanchina E, Fridman JS, et al. Survival signaling by Akt and eIF4E in oncogenesis and cancer therapy. Nature 2004;428: 332 - 7. Kay JE, Smith MC, Frost V, et al. Hypersensitivity to rapamycin of BJAB B lymphoblastoid cells. Immunology 1996;87:390 - 5. Luan FL, Hojo M, Maluccio M, et al. Rapamycin blocks tumor progression: unlinking immunosuppression from antitumor efficacy. Transplantation 2002;73:1565 - 72. Geoerger B, Kerr K, Tang CB, et al. Antitumor activity of the rapamycin analog CCI-779 in human primitive neuroectodermal tumor/medulloblastoma models as single agent and in combination chemotherapy. Cancer Res 2001;61:1527 - 32. Dudkin L, Dilling MB, Cheshire PJ, et al. Biochemical correlates of mTOR inhibition by the rapamycin ester CCI-779 and tumor growth inhibition. Clin Cancer Res 2001;7:1758 - 64. Houchens DP, Ovejera AA, Riblet SM, et al. Human brain tumor xenografts in nude mice as a chemotherapy model. Eur J Cancer Clin Oncol 1983;19:799 - 805. Yu K, Toral-Barza L, Discafani C, et al. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer 2001; 8:249 - 58. Pang H, Faber LE. Estrogen and rapamycin effects on cell cycle progression in T47D breast cancer cells. Breast Cancer Res Treat 2001;70:21 - 6. Chen Y, Zheng Y, Foster DA. Phospholipase D confers rapamycin resistance in human breast cancer cells. Oncogene 2003;22:3937 - 42. Noh WC, Mondesire WH, Peng J, et al. Determinants of rapamycin sensitivity in breast cancer cells. Clin Cancer Res 2004; 10:1013 - 23. Gregory CR, Pratt RE, Huie P, et al. Effects of treatment with cyclosporine, FK 506, rapamycin, mycophenolic acid, or deoxyspergualin on vascular muscle proliferation in vitro and in vivo. Transplant Proc 1993;25:770 - 1. Eng CP, Sehgal SN, Vezina C. Activity of rapamycin (AY22,989) against transplanted tumors. J Antibiot (Tokyo) 1984;37: 1231 - 7. Price DJ, Grove JR, Calvo V, et al. Rapamycin-induced inhibition of the 70-kilodalton S6 protein kinase. Science 1992;257:973 - 7. Leung-Pineda V, Pan Y, Chen H, et al. Induction of p21 and p27 expression by amino acid deprivation of HepG2 human hepatoma cells involves mRNA stabilization. Biochem J 2004;379:79 - 88. Schumacher G, Oidtmann M, Rosewicz S, et al. Sirolimus inhibits growth of human hepatoma cells in contrast to tacrolimus which promotes cell growth. Transplant Proc 2002;34:1392 - 3. Shigemitsu K, Tsujishita Y, Hara K, et al. Regulation of translational effectors by amino acid and mammalian target of rapamycin signaling pathways. Possible involvement of autophagy in cultured hepatoma cells. J Biol Chem 1999;274:1058 - 65. Banholzer R, Nair AP, Hirsch HH, et al. Rapamycin destabilizes interleukin-3 mRNA in autocrine tumor cells by a mechanism requiring an intact 3V untranslated region. Mol Cell Biol 1997; 17:3254 - 60.
29
[43] Frost P, Moatomed F, Hoang B, et al. In vivo anti-tumor effects of the mTOR inhibitor, CCI-779, against human multiple myeloma cells in a xenograft model. Blood 2004;104:4181 - 7 [Prepublished online August 10, 2004]. [44] Jayaraman T, Marks AR. Rapamycin-FKBP12 blocks proliferation, induces differentiation, and inhibits cdc2 kinase activity in a myogenic cell line. J Biol Chem 1993;268:25385 - 8. [45] Misawa A, Hosoi H, Tsuchiya K, et al. Rapamycin inhibits proliferation of human neuroblastoma cells without suppression of MycN. Int J Cancer 2003;104:233 - 7. [46] Boffa DJ, Luan F, Thomas D, et al. Rapamycin inhibits the growth and metastatic progression of non–small cell lung cancer. Clin Cancer Res 2004;10:293 - 300. [47] Seufferlein T, Rozengurt E. Rapamycin inhibits constitutive p70s6k phosphorylation, cell proliferation, and colony formation in small cell lung cancer cells. Cancer Res 1996;56:3895 - 7. [48] Pardo OE, Arcaro A, Salerno G, et al. Novel cross talk between MEK and S6K2 in FGF-2 induced proliferation of SCLC cells. Oncogene 2001;20:7658 - 67. [49] Altomare DA, Wang HQ, Skele KL, et al. Akt and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene 2004;23:5853 - 7. [50] Aguirre D, Boya P, Bellet D, et al. Bcl-2 and CCND1/CDK4 expression levels predict the cellular effects of mTOR inhibitors in human ovarian carcinoma. Apoptosis 2004;9:797 - 805. [51] Bruns CJ, Koehl GE, Guba M, et al. Rapamycin-induced endothelial cell death and tumor vessel thrombosis potentiate cytotoxic therapy against pancreatic cancer. Clin Cancer Res 2004;10: 2109 - 19. [52] Boulay A, Zumstein-Mecker S, Stephan C, et al. Antitumor efficacy of intermittent treatment schedules with the rapamycin derivative RAD001 correlates with prolonged inactivation of ribosomal protein S6 kinase 1 in peripheral blood mononuclear cells. Cancer Res 2004;64:252 - 61. [53] Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxiainducible factor 1alpha expression by the epidermal growth factor/ phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res 2000;60:1541 - 5. [54] Van der Poel HG. Mammalian target of rapamycin and 3phosphatidylinositol 3-kinase pathway inhibition enhances growth inhibition of transforming growth factor–beta1 in prostate cancer cells. J Urol 2004;172:1333 - 7. [55] Majumder PK, Febbo PG, Bikoff R, et al. mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1–dependent pathways. Nat Med 2004;10:594 - 601. [56] Luan FL, Ding R, Sharma VK, et al. Rapamycin is an effective inhibitor of human renal cancer metastasis. Kidney Int 2003;63: 917 - 26. [57] Dilling MB, Dias P, Shapiro DN, et al. Rapamycin selectively inhibits the growth of childhood rhabdomyosarcoma cells through inhibition of signaling via the type I insulin-like growth factor receptor. Cancer Res 1994;54:903 - 7. [58] Hosoi H, Dilling MB, Shikata T, et al. Rapamycin causes poorly reversible inhibition of mTOR and induces p53-independent apoptosis in human rhabdomyosarcoma cells. Cancer Res 1999; 59:886 - 94. [59] Gallicchio MA, van Sinderen M, Bach LA. Insulin-like growth factor binding protein-6 and CCI-779, an ester analogue of rapamycin, additively inhibit rhabdomyosarcoma growth. Horm Metab Res 2003;35:822 - 7. [60] Brunn GJ, Williams J, Sabers C, et al. Direct inhibition of the signaling functions of the mammalian target of rapamycin by the phosphoinositide 3-kinase inhibitors, wortmannin and LY294002. EMBO J 1996;15:5256 - 67.
30
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31
[61] Dumont FJ, Fischer P, Sirotina A. Increased LFA-1–mediated homotypic cell adhesion is associated with the G1 growth arrest induced by rapamycin in a T cell lymphoma. Exp Cell Res 1995;219: 146 - 58. [62] Nathan CA, Amirghahari N, Abreo F, et al. Overexpressed eIF4E is functionally active in surgical margins of head and neck cancer patients via activation of the Akt/mammalian target of rapamycin pathway. Clin Cancer Res 2004;10:5820 - 7. [63] Campistol JM, Gutierrez-Dalmau A, Torregrosa JV. Conversion to sirolimus: a successful treatment for posttransplantation Kaposi’s sarcoma. Transplantation 2004;77:760 - 2. [64] Mita M, Rowinsky E, Mita A, et al. AP23573, an mTOR inhibitor, administered IV daily x 5 every other week in patients with refractory or advanced malignancies—a phase I, pharmacokinetic (PK), and pharmacodynamic (PD) study. Eur J Cancer Suppl 2004; 2(8):122. [65] Raymond E, Alexandre J, Faivre S, et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004;22:2336 - 47. [66] Punt CJ, Boni J, Bruntsch U, et al. Phase I and pharmacokinetic study of CCI-779, a novel cytostatic cell-cycle inhibitor, in combination with 5-fluorouracil and leucovorin in patients with advanced solid tumors. Ann Oncol 2003;14:931 - 7. [67] O’Donnell A, Faivre S, Judson I, et al. A phase I study of the oral mTOR inhibitor RAD001 as monotherapy to identify the optimal biologically effective dose using toxicity, pharmacokinetic (PK) and pharmacodynamic (PD) endpoints in patients with solid tumours. Proc Am Soc Clin Oncol 2003;22:200. [68] Van Oosterom AT, Dumez H, Desai J, et al. Combination signal transduction inhibition: a phase I/II trial of the oral mTOR-inhibitor everolimus (E, RAD001) and imatinib mesylate (IM) in patients (pts) with gastrointestinal stromal tumor (GIST) refractory to IM. 2004 ASCO Annual Meeting Proceedings (Post-Meeting Edition). J Clin Oncol 2004;22(14S):3002 [15-7-2004]. [69] Atkins MB, Hidalgo M, Stadler WM, et al. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 2004;22:909 - 18. [70] Trump DL. Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma [Adapted from Atkins MB, Hidalgo M, Stadler WM, Logan TF, Dutcher JP, Hudes GR, Park Y, Liou SH, Marshall B, Boni JP, Dukart G, Sherman ML, Department of Medicine, Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Boston, MA. J Clin Oncol 2004;22:909-18]. Urol Oncol 2004;22:431 - 2. [71] 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;18:446 - 9. [72] Jimenez-Rivera C, Avitzur Y, Fecteau AH, et al. Sirolimus for pediatric liver transplant recipients with post-transplant lymphoproliferative disease and hepatoblastoma. Pediatr Transplant 2004; 8:243 - 8. [73] Huang S, Houghton PJ. Mechanisms of resistance to rapamycins. Drug Resist Updat 2001;4:378 - 91. [74] Ke H, Huai Q. Structures of calcineurin and its complexes with immunophilins-immunosuppressants. Biochem Biophys Res Commun 2003;311:1095 - 102. [75] Chan S. Targeting the mammalian target of rapamycin (mTOR): a new approach to treating cancer. Br J Cancer 2004;91:1420 - 4. [76] Asnaghi L, Bruno P, Priulla M, et al. mTOR: a protein kinase switching between life and death. Pharmacol Res 2004;50:545 - 9. [77] Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev 2004;18:1926 - 45. [78] Dutcher JP. Mammalian target of rapamycin inhibition. Clin Cancer Res 2004;10:6382S - 7S.
[79] Sehgal SN. Rapamune (RAPA, rapamycin, sirolimus): mechanism of action immunosuppressive effect results from blockade of signal transduction and inhibition of cell cycle progression. Clin Biochem 1998;31:335 - 40. [80] Humar R, Kiefer FN, Berns H, et al. Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)dependent signaling. FASEB J 2002;16:771 - 80. [81] Fingar DC, Blenis J. Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 2004;23:3151 - 71. [82] Brugarolas J, Lei K, Hurley RL, et al. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev 2004;18:2893 - 904. [83] Proud CG. Role of mTOR signaling in the control of translation initiation and elongation by nutrients. Curr Top Microbiol Immunol 2004;279:215 - 44. [84] Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/mTOR. Genes Dev 2001;15:807 - 26. [85] Carraway H, Hidalgo M. New targets for therapy in breast cancer: mammalian target of rapamycin (mTOR) antagonists. Breast Cancer Res 2004;6:219 - 24. [86] Thompson JE, Thompson CB. Putting the rap on Akt. J Clin Oncol 2004;22:4217 - 26. [87] Zhang H, Cicchetti G, Onda H, et al. Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through downregulation of PDGFR. J Clin Invest 2003;112:1223 - 33. [88] Roymans D, Slegers H. Phosphatidylinositol 3-kinases in tumor progression. Eur J Biochem 2001;268:487 - 98. [89] Sansal I, Sellers WR. The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol 2004;22:2954 - 63. [90] Panwalkar A, Verstovsek S, Giles FJ. Mammalian target of rapamycin inhibition as therapy for hematologic malignancies. Cancer 2004;100:657 - 66. [91] Huang S, Houghton PJ. Resistance to rapamycin: a novel anticancer drug. Cancer Metastasis Rev 2001;20:69 - 78. [92] Dumont FJ, Su Q. Mechanism of action of the immunosuppressant rapamycin. Life Sci 1996;58:373 - 95. [93] Gomez-Cambronero J. Rapamycin inhibits GM-CSF–induced neutrophil migration. FEBS Lett 2003;550:94 - 100. [94] Raught B, Gingras AC, Sonenberg N. The target of rapamycin (TOR) proteins. Proc Natl Acad Sci U S A 2001;98:7037 - 44. [95] Brugarolas JB, Vazquez F, Reddy A, et al. TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell 2003;4:147 - 58. [96] Zimmermann T, Holtappels M, Buhr J, et al. The efficacy of adjuvant cytostatic therapy after organ transplantation for malignancy: an experimental study with a combined transplantation/tumor model. Transpl Int 2000;13:136 - 41. [97] Kumar V, Pandey P, Sabatini D, et al. Functional interaction between RAFT1/FRAP/mTOR and protein kinase cdelta in the regulation of cap-dependent initiation of translation. EMBO J 2000; 19:1087 - 97. [98] Pyronnet S, Imataka H, Gingras AC, et al. Human eukaryotic translation initiation factor 4G (eIF4G) recruits mnk1 to phosphorylate eIF4E. EMBO J 1999;18:270 - 9. [99] 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;10:47 - 50. [100] Huang S, Liu LN, Hosoi H, et al. p53/p21(CIP1) cooperate in enforcing rapamycin-induced G(1) arrest and determine the cellular response to rapamycin. Cancer Res 2001;61:3373 - 81. [101] Feliers D, Duraisamy Sl, Barnes JL, et al. Translational regulation of vascular endothelial growth factor expression in renal epithelial cells by angiotensin II. Am J Physiol Renal Physiol 2004;271. [102] Hudson CC, Liu M, Chiang GG, et al. Regulation of hypoxiainducible factor 1 alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol 2002;22:7004 - 14.
G.E. Koehl et al. / Transplantation Reviews 19 (2005) 20 –31 [103] Zundel W, Schindler C, Haas-Kogan D, et al. Loss of PTEN facilitates HIF-1–mediated gene expression. Genes Dev 2000; 14:391. [104] Simpson L, Li J, Liaw D, et al. PTEN expression causes feedback upregulation of insulin receptor substrate 2. Mol Cell Biol 2001; 21:3947 - 58. [105] DeGraffenried LA, Fulcher L, Friedrichs WE, et al. Reduced PTEN expression in breast cancer cells confers susceptibility to inhibitors of the PI3 kinase/Akt pathway. Ann Oncol 2004;15: 1510 - 6. [106] Tolcher AW. Novel therapeutic molecular targets for prostate cancer: the mTOR signaling pathway and epidermal growth factor receptor. J Urol 2004;171:S41-3. [107] Laughner E, Taghavi P, Chiles K, et al. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1–mediated vascular endothelial growth factor expression. Mol Cell Biol 2001;21:3995 - 4004. [108] Manning BD, Tee AR, Logsdon MN, et al. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/Akt pathway. Mol Cell 2002;10:151 - 62. [109] Guo W, Giancotti FG. Integrin signaling during tumour progression. Nat Rev Mol Cell Biol 2004;5:816 - 26. [110] 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. [111] Huang C, Li J, Ke Q, et al. Ultraviolet-induced phosphorylation of p70(S6K) at Thr(389) and Thr(421)/Ser(424) involves hydrogen peroxide and mammalian target of rapamycin but not Akt and atypical protein kinase C. Cancer Res 2002;62:5689 - 97. [112] Stephan S, Datta K, Wang E, et al. Effect of rapamycin alone and in combination with antiangiogenesis therapy in an orthotopic model of human pancreatic cancer. Clin Cancer Res 2004;10: 6993 - 7000. [113] Oberbauer R, Kreis H, Johnson RW, et al. Long-term improvement in renal function with sirolimus after early cyclosporine withdrawal in
[114]
[115]
[116]
[117]
[118]
[119]
[120]
[121]
[122]
[123]
31
renal transplant recipients: 2-year results of the Rapamune Maintenance Regimen Study. Transplantation 2003;76:364 - 70. MacDonald AS. Rapamycin in combination with cyclosporine or tacrolimus in liver, pancreas, and kidney transplantation. Transplant Proc 2003;35:201S - 8S. Coito C, Diamond DL, Neddermann P, et al. High-throughput screening of the yeast kinome: identification of human serine/ threonine protein kinases that phosphorylate the hepatitis c virus NS5A protein. J Virol 2004;78:3502 - 13. Kahan BD. The limitations of calcineurin and mTOR inhibitors: new directions for immunosuppressive strategies. Transplant Proc 2002; 34:130 - 3. Dahmen U, Gu Y, Shen K, et al. Onset of liver regeneration after subtotal resection is inhibited by the use of new immunosuppressive drugs. Transplant Proc 2002;34:2312 - 3. Mohi MG, Boulton C, Gu TL, et al. Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci U S A 2004;101:3130 - 5. Hu L, Hofmann J, Lu Y, et al. Inhibition of phosphatidylinositol 3V- kinase increases efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Cancer Res 2002;62:1087 - 92. DeGraffenried LA, Friedrichs WE, Russell DH, et al. Inhibition of mTOR activity restores tamoxifen response in breast cancer cells with aberrant Akt activity. Clin Cancer Res 2004;10:8059 - 67. Kneteman NM, Oberholzer J, Al Saghier M, et al. Sirolimus-based immunosuppression for liver transplantation in the presence of extended criteria for hepatocellular carcinoma. Liver Transpl 2004; 10:1301 - 11. Yao FY, Ferrell L, Bass NM, et al. Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology 2001;33:1394 - 403. Kauffman HM, Cherikh WS, Cheng Y, et al. Maintenance immunosuppression with TOR inhibitors is associated with a reduced incidence of de novo malignancies. American Transplant Congress 2004 [abstract 505;2004].