Mammalian Target of Rapamycin as a Target in Hematological Malignancies he era of targeted therapy is here. Understanding the regulation of cellular protein synthesis is crucial in the development of novel agents that target certain pathways causing smarter cell kill. The mammalian target of rapamycin (mTOR) role in cellular protein synthesis affects different aspects of the cell growth, including differentiation, cell cycle progression, protein degradation, and apoptosis, as well as protein kinase C signaling and transcription.1-5 mTOR has other known names, including rapamycin-associated protein (FRAP), FK506-binding protein (FKBP12), rapamycin and FKBP12 target (RAFT1), rapamycin target (RAPT1), and sirolimus effector protein (SEP). It is a 290-kDa serine–threonine kinase orthologue of target of rapamycin 1 (TOR1) and target of rapamycin 2 (TOR2) in Saccharomyces cerevisia.6-10 The TOR1 and TOR2 genes encode two large, highly homologous proteins. The mTOR gene maps to chromosome 1p36.2 in humans. Human, mouse, and rat mTOR proteins share a 95% identity at the amino acid level,6,11,12 suggesting that these domains are critical for cellular function. Rapamycin (sirolimus) is a macrocyclic lactone that is naturally produced by Streptomyces hygroscopicus bacteria. It was initially identified as an antifungal agent.13-15 It was also used as an immunosuppressant agent for prevention of rejection after solid organ transplantation16,17 and in rheumatoid arthritis given its effect on lymphocyte proliferation and cytokine production, which affect the pathogenesis of rheumatoid arthritis significantly.18 It is widely used currently in drug-eluting stents to prevent restenosis given its antiproliferative effect.19 The demonstration of rapamycin’s antitumor properties was initially reported in the National Cancer Institute (NCI) screening program in the 1980s.20-22 This review will focus on the role of mTOR as a target in hematological malignancies.
T
Curr Probl Cancer 2008;32:161-177. 0147-0272/2008/$34.00 ⫹ 0 doi:10.1016/j.currproblcancer.2008.05.001
Curr Probl Cancer, July/August 2008
161
mTOR: Structure and Function mTOR is formed of up to 20 tandemly repeated motifs of HEAT (Huntington, EF3, A subunit of PP2A, and TOR) at the N terminus. The C terminus, on the other hand, contains: a FRAP–ataxia telangiectasiamutated, Transformation/transcription domain-associated protein (FAT) domain, a FKBP12–rapamycin binding (FRB) domain, a probable autoinhibitory or repressor domain, a catalytic kinase domain, and a FAT carboxy-terminal (FATC) domain.23,24 Given that the catalytic domain in the C terminus of mTOR is very similar to the lipid kinase domain of phosphatidylinositol 3=kinase (PI3K), mTOR is considered a member of the PI3K-related protein kinase family (PIKK).6,11 It is frequently referred to as the Pl3K/Akt/mTOR pathway, and it is critical in PIKKs regulating cellular responses to DNA damage, repair, and recombination.25,26 Rapamycin and its analogs bind to FKBP12, creating a complex which binds to the FRB domain of mTOR and inhibits its kinase activity.23 mTOR is functionally composed of two complexes: mTOR Complex 1 (mTORC1) and mTORC2.27 mTORC1 is composed of mTOR, a novel 150-kDa peptide regulatory associated protein of mTOR (raptor) and mammalian LST8/G-protein -subunit like protein (mLST8/GL).28-30 Raptor acts by presenting downstream target substrates to the mTOR kinase domain for phosphorylation, and it also binds to its targets p70S6K and 4E-BP1 via TOR signaling motif (TOS).31-34 The mLST8 protein also associates with mTOR, the yeast LST8 homologue, which negatively regulates RTG1/3 and GLN3 gene expression resulting in limiting ketoglutarate, glutamate, and glutamine synthesis, which is involved in the maintenance of cell wall integrity.24,30-32 GL binds to the mTOR kinase domain and stabilizes its interaction with mTOR.33 Raptor and mTOR share a strong interaction at the N terminus but a weak one at the C terminus. mTORC1 is activated in the presence of nutrients through significant weakness of the C terminus, leading to mTOR kinase activation. The C terminus is strengthened during starvation status, leading to deactivation of mTOR.29 On the other hand, mTORC2 is composed of mTOR, a companion of mTOR (rictor), GL, and mammalian stressactivated protein kinase interacting protein 1 (mSin1).35 The rictor is insensitive to rapamycin, leading to the fact that the mTOR sensitivity to rapamycin is exerted through mTORC1. mTORC2 stimulates Akt phosphorylation at threonine308 by the phosphoinositide-dependent kinase 1 (PDK1), leading to Akt activation.36 162
Curr Probl Cancer, July/August 2008
AKT/mTOR Pathway in Cancer The activation of mTOR has been implicated in many human malignancies,37-52 and it has also been described in the spread of cancer via lymphatics. Rapamycin has been shown in in vivo experimentation to decrease the number and lymphatic vessels in the primary tumors revealing anti-lymphangiogenic effect.53,54 An interesting concept is its role in minimal residual disease, which is surgical margins that are free from tumor but over-express eIF4E. This has been reported in head and neck cancer with rapamycin given as adjuvant therapy yielding survival advantage.55 Endothelial growth factor receptor (EGFR) regulates PI3K/ Akt pathway. Activation of Akt leads to the stimulation of antiapoptotic pathways, promoting cell survival. The activity of the EGFR inhibitor erlotinib in non-small-cell lung cancer is dependent on inhibition of the PI3K phosphoinositide-dependent kinase 1-Akt-mTOR pathway. Rapamycin inhibited significantly S6 in all cell lines derived for different tissue types (non-small-cell lung, pancreatic, colon, and breast), and this was accompanied by activation of Akt phosphorylation. Erlotinib only inhibited extracellular signal-regulated kinase, Akt, and S6 in most sensitive cell lines. The combination of rapamycin and erlotinib downregulated rapamycin-stimulated Akt activity, so adding rapamycin to erlotinib could improve its activity.56 A phase II study of a combination of temsirolimus or everolimus with letrozole demonstrated a better progression-free survival in the combination arm than in the letrozole alone arm.57 Myelodysplastic syndrome (MDS) and acute myelogenous leukemia (AML) are believed to be two spectrums of the same disease process. The activation of mTOR pathway has been shown in both diseases.58-60 This pathway has also been described in diffuse large B-cell lymphoma61 and anaplastic large cell lymphoma.62 Mantle cell lymphoma is characterized by an over-expression of cyclin D1 resulting from chromosomal translocation (t11,14). The drug suberoylanilide hydroxamic acid (SAHA; vorinostat) was shown to decrease the level of cyclin D1 by 90% through the inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR/ eIF4E-BP pathway.63
Side Effects of mTOR Inhibitors A meta-analysis to study the effect of TOR-1 inhibitors (sirolimus and everolimus) on male gonads showed that these agents have a negative effect on spermatogenesis as evidenced by the decrease in testosterone level and the increase in LH and FSH.64 Oligospermia has been Curr Probl Cancer, July/August 2008
163
implicated in sirolimus-associated infertility.65 Although preclinical studies on animal models showed sirolimus to be a potentially non-nephrotoxic drug, clinical data reported nephrotoxicity to be a concern.66 Sirolimus has been reported to be associated with proteinuria and acute renal failure, especially in patients with pre-existing renal damage. Sirolimus-induced nephrotoxicity is mostly mulifactorial but may involve glomerular injury and suppression of compensatory renal cell proliferation and repair processes. It is advised to monitor patients for occurrence of proteinuria and renal dysfunction. Like in diabetic nephropathy, use of angiotensin-converting enzyme inhibitors or angiotensin II receptor blockers may alleviate proteinuria and kidney damage.67 Temsirolimus and its metabolite, sirolimus, are cytochrome P450 (CYP) 3A4/5 substrates resulting in potential interaction with drugs that induce CYP3A activity.68 A retrospective study evaluated pulmonary side effects of temsirolimus. Medical records of 22 patients who were treated with weekly doses of temsirolimus 25 mg were reviewed. Eight patients (36%) developed drug-induced pneumonitis. Two different patterns were described on imaging: ground glass opacities and lung parenchymal consolidation. The interesting finding of this study was that half of these patients had symptoms, with dyspnea and dry cough being the most common, but the other half were asymptomatic, raising the possibility of the role of screening imaging in patients receiving temsirolimus.69 There seems to be a difference in the side effects on lipid profile between the mTOR inhibitors. Sirolimus was found to increase serum triglyceride and total cholesterol concentrations in contrast to everolimus, which increased HDL concentration in heart transplant recipients with renal insufficiency. This resulted in increased frequency of statin use in the sirolimus group. Both drugs resulted in reliable immunosuppression in this setting.70
Resistance to mTOR Inhibitors With the growing role of mTOR inhibitors in cancer treatment, resistance is a concern. Several mechanisms have been reported, although their interaction is complex and still a lot remains unknown. Mutations in FKBP12 prevent rapamycin from binding to mTOR, leading to rapamycin resistance.71 Defects in mTOR-regulated proteins, like 4E-BP1, PP2Arelated phosphatases, and p27(Kip1), cause rapamycin insensitivity, hence resistance.72,73 Cancer cell lines with intrinsic rapamycin resistance were found to have low 4E-BP:eIF4E ratios.74 Point mutations in S6K1 affect rapamycin sensitivity in vitro.75,76 P53 status has been influential in 164
Curr Probl Cancer, July/August 2008
rapamycin resistance as well.77 Mutated ataxia telangiectasia cells are rapamycin-resistant. Defects in signaling could lead to resistance, but additional mutations could also play a role.78,79 Other factors have been implicated as well such as 14-3-3 proteins80 and PTEN/Akt status.81 Mutation in the FRB domain of mTOR has been implicated in high-level resistance.82 PIM-2 is a key component in hematopoietic cells growth and survival, and its overexpression has been implicated in developing resistance to rapamycin.83
mTOR in Specific Hematolgical Malignancies Malignancies with PTEN mutations are resistant to apoptosis but could be especially sensitive to mTOR inhibitors. A study by Shi et al.84 demonstrated that myeloma cells that are deficient in PTEN were not apoptosis-resistant. But these particular cells were significantly more sensitive to CCI-779 (sirilomus), which was the mTOR inhibitor the authors used in their experiment.84
Mantle Cell Lymphoma Mantle cell lymphoma (MCL) is characterized by t(11;14) and cyclin D1 overexpression. Through immunohistochemical methods and tissue microarray, it has been proved that mTOR signaling pathway is activated in this disease and it contributes to tumor cell survival.85 MCL has especially poor prognosis, and it is an attractive area for testing new targeted therapy. Several agents are currently being tested in this disease.86 Rapamycin has been shown in vivo to inhibit proliferation in three MCL cell lines, resulting in reduction of cyclin D3 expression with no effect on cyclin D1 levels. This finding was duplicated in cells from a MCL patient.87 This led to granting an FDA approval for the use of sirolimus in this disease.
Multiple Myeloma Activation of adenosine monophosphate activated protein kinase inhibits mTOR and P70S6 kinase (P70S6K) as well as AKT phosphorylation in multiple myeloma (MM) cell lines. It induced S-phase cell cycle arrest. This inhibition persisted in the face of adding IL-6, IGF-1, and HS-5 stromal cell-conditioned medium.88 Rapamycin also induces G0/G1 arrest in MM cell lines, leading to increase in the cyclin-dependent kinase inhibitor p27 and a decrease in cyclins D2 and D3. Rapamycin induced apoptosis and it sensitized MM cells and cell lines to dexamethasoneinduced apoptosis despite adding the antiapoptotic agents insulin growth factor-I (IGF-I) and interleukin 6 (IL-6). This effect was associated with Curr Probl Cancer, July/August 2008
165
a decreased expression of cyclin D2 and surviving.89 Immunoblot assays had shown that rapamycin inhibited expressions of XIAP, CIAP1, HSP-27, and BAG-3, which may explain what has been noticed about rapamycin sensitizing multiple myeloma cells to dexamethasone-induced apoptosis.90 This suggests that the combination of rapamycin and dexamethasone is active in vivo in MM. Inhibitors of HSP90 protein are active in vitro and in vivo in MM. The combination of these agents with mTOR inhibitors have been shown to have synergistic activity in MM cell lines through induction of apoptosis and cell cycle arrest. Both groups of inhibitors inhibit osteoclast cells affecting the microenvironment of the bone marrow, which is especially attractive in the treatment of MM.91
Acute Leukemia PI3k/Akt pathway is involved in the pathophysiology of childhood acute lymphoblastic leukemia (ALL) and it has a role in blast survival. Avellino et al.92 showed rapamycin to induce apoptosis of blasts in 56% of samples of the bone marrow. Rapamycin also increased doxorubicininduced apoptosis, even in non-responder samples. This is probably explained by their finding that FKBP51, an immunophilin inhibited by rapamycin, is essential for drug-induced nuclear factor B (NF-B) activation in leukemia. Anthracyclines activate NF-B, and rapamycin inhibited doxorubicin-induced NF-B. Interestingly, rapamycin did not increase doxorubicin-induced apoptosis when NF-B was overexpressed. NF-B pathway through FKBP51 is a newly discovered pathway that rapamycin can target along with the previously described PI3k/Akt pathway leading to therapeutic benefit in ALL.92 Rapamycin is an emerging treatment in ALL,93 and its activity as a single agent in B-precursor ALL cell lines has been documented. It caused apoptotic cell death which was reversible by adding IL-7. This is possibly due to p70S6 kinase being regulated by both rapamycin and IL-7. E mu-ret transgenic mice with advanced disease treated with daily rapamycin as a single agent more than doubled their survival as compared with symptomatic controls.94 All-trans-retinoic acid (RA) is an active agent in acute promyelocytic leukemia (APL). RA treatment of primary APL blasts and RA-sensitive NB-4 cells resulted in phosphorylation/activation of mTOR and downstream induction of p70S6 kinase activity. Interestingly, this was not the case in NB-4 variant cell line (NB-4.007/6) that is resistant to the biologic effects of RA. RA also resulted in phosphorylation of the 4E-BP1 repressor of mRNA translation through mTOR pathway, causing its 166
Curr Probl Cancer, July/August 2008
deactivation and dissociation from the eukaryotic initiation factor-4E (eIF-4E) complex.95 PI3-kinase-Akt-mTOR pathway has been shown to be activated in up to 60% of AML patient cells.96 In a series of 23 AML cases, rapamycin inhibited the growth of the most immature AML cell lines through G0/G1 cell cycle arrest. Rapamycin spared normal hematopoietic progenitors. It induced significant clinical responses in 4 of 9 patients with AML (either refractory/relapsed de novo or secondary), making it an attractive agent in this disease.97,98 It has been hypothesized that inhibition of both the mTOR pathway and glycolysis can work synergistically to affect the energy status in cancer cells. Combining rapamycin and glycolytic inhibitor produced synergistic apoptosis and cell growth inhibition in human lymphoma cells and leukemia cells. This combination suppressed glucose uptake and critically depleted cellular ATP pools, leading to cell death. No cytotoxicity was observed in vitro when rapamycin was combined with cytarabine.99
Chronic Myelogenous Leukemia Ribosomal protein S6 and 4E-BP1 are phosphorylated in chronic myelogenous leukemia (CML) cells. This phosphorylation is downstream of Bcr-Abl kinase and mTOR, meaning that mTOR is activated in CML. Rapamycin was found to potentiate arsenic trioxide-mediated suppression of primitive leukemic progenitors taken from the bone marrow of patients with CML.100 Rapamycin was also shown to enhance imatinib-mediated killing of CML cell lines in vitro, and it overcame imatinib resistance.101 Resistance to tyrosine kinases is unfortunate given their marked activity in this disease. BCR/ABL-kinase mutations are mostly to blame. Burchert et al.102 showed that Imatinib treatment activated the PI3K/Akt/mTOR pathway in BCR/ABL-positive LAMA-cells and primary leukemia cells in vitro, as well as in a chronic phase CML patient in vivo. In the early phase of imatinib resistance, PI3K/Akt-activation mediated survival before the kinase mutation manifested. Patients who were imatinibresistant or refractory had patterns of Akt-pathway activation independent of the kinase mutation. mTOR-inhibition overcame imatinib resistance only if Akt was strongly activated. This gives rationale to test combinations of imatinib and mTOR inhibitors in patients with positive BCR/ ABL.102 Rapamycin inhibited growth of CML cells of both imatinibresponsive or imatinib-resistant nature. This effect was also shown in an imatinib-resistant positive BCR-ABL leukemic patient. This was associated with G1 cell cycle arrest, induction of apoptosis, and downregulation of expression of vascular endothelial growth factor (VEGF). Curr Probl Cancer, July/August 2008
167
Exogenously added VEGF did not correct the rapamycin-induced decrease in proliferation.103 Like imatinib, interferon has also been shown to activate mTOR in BCR-ABL positive cells. It resulted in downstream activation of p70S6 kinase. Imatinib, on the other hand, suppressed p70S6 kinase activity, consistent with inhibition of BCR-ABL-mediated activation of the mTOR/p70 S6 kinase pathway. Rapamycin had a synergistic effect with imatinib on primary leukemic granulocyte macrophage– colony-forming unit (CFU-GM) progenitors from patients with CML.104
Lymphoma Everolimus has antiproliferative activity in vitro and in NOD/SCID mice in vivo against Hodgkin lymphoma (HL) and anaplastic large cell lymphoma (ALCL) cells. It down-regulated the truncated isoform of the transcription factor CCAAT enhancer binding protein  (C/EBP), leading to a disruption in the terminal differentiation with a transformed phenotype. Everolimus also inhibited NF-B activity, which is a critical survival factor of HL cells.105 Rapamycin is also effective in B-Chronic lymphocytic leukemia (B-CLL).106 mTOR is activated in follicular lymphoma (FL) through Syk. Inhibition of Syk by siRNA plasmids or piceatannol results in mTOR inhibition in FL cells. This effect was also seen in other lymphomas like mantle-cell and diffuse-large B-cell. Targeting Syk-mTOR pathway is a possible new target in the treatment of these disorders.107
Novel mTOR Inhibitors Sirolimus (CCI-779) Sirolimis is the most extensively studied mTOR inhibitor and the most widely used one in clinical practice. A phase I trial established the safety of sirolimus (CCI-779) given weekly over 30 minutes at doses of 25, 75, and 250 mg/m2. The most common toxicities were maculopapular rash, mucositis, and stomatitis, and they were reversible. Thrombocytopenia was the main dose-limiting toxicity but was also reversible. The maximum tolerated dose was not reached. No immunosuppression was observed even at the highest dose.108 Another phase I study reported the maximum tolerated dose to be 15 mg/m2/day for patients with extensive prior treatment and 19 mg/m2/day for patients who were minimally pretreated. Terminal half-life in that study was 13 to 25 hours.109 In phase II testing, the side effect profile was similar with maculopapular rash (76%), mucositis (70%), asthenia (50%), and nausea (43%). The most frequently occurring grade 3 or 4 adverse 168
Curr Probl Cancer, July/August 2008
events were hyperglycemia (17%), hypophosphatemia (13%), anemia (9%), and hypertriglyceridemia (6%).110 It is known that CCI-779 inhibits the kinase activity of mTOR, resulting in inhibition of the mTOR-regulated translational controllers p70(s6) kinase and 4E-BP1. This led to postulate that measuring p70(s6) kinase activity may mirror the effect of sirolimus on cells. It was shown that the activity of inhibition of p70(s6) kinase is identical between peripheral blood mononuclear cells and cells from the tumor tissue. This makes the ease of measurement of the assay of p70(s6) kinase from peripheral blood an attractive tool in judging biochemical response to sirolimus.111 Witzig et al.112 treated 35 patients with relapsed or refractory mantle cell lymphoma with temsirolimus 250 mg intravenously every week as a single agent. The median age was 70 years and 91% of patients had stage 4. The overall response rate was an impressive 38% with the response lasting for 6.9 months. The most common toxicities were hematological with thrombocytopenia being the most dose-limiting side effect, although it was reversible.112 It is FDA approved in mantle cell lymphoma.
Everolimus (RAD001) Everolimus is an immunosuppressive macrolide with a stable 2-hydroxyethyl chain substitution at position 40 on the rapamycin structure. It was developed to be orally bioavailable more than sirolimus. It is currently the only mTOR inhibitor in oral form. It acts by blocking growth-driven transduction signals in the T-cell response to alloantigen so its site of action is at a later stage than tacrolimus. It has a high affinity for the intracellular receptor protein FKBP-12 through which it affects interleukin and growth-factor-dependent proliferation of cells. In vitro experiments with human umbilical endothelial cells (HUVECS) and animal models revealed a possible antiangiogenic effect of everolimus, possibly through inhibition of proliferating endothelial cells.113 Everolimus was developed as an immunosuppressive agent and is approved in Europe for that purpose in the setting of solid organ transplantation.114-118 Oral everolimus is readily bioavailable, and it reaches peak concentration after 1.3–1.8 hours. It is metabolized by the cytochrome P450 (CYP) 3A4, 3A5, and 2C8. Dosage should be reduced by half in patients with hepatic impairment.119 It was studied in a phase I/II study in relapsed or refractory hematologic malignancies (9 acute myelogenous leukemia, 5 myelodysplastic syndrome, 6 B-chronic lymphocytic leukemia, 4 mantle cell lymphoma, 1 of each: myelofibrosis, natural killer cell/T-cell leukemia, and T-cell prolymphocytic leukemia). The phase I part established the maximum Curr Probl Cancer, July/August 2008
169
tolerated dose to be 10 mg orally once daily. Grade 3 toxicities included hyperglycemia (22%), hypophosphatemia (7%), fatigue (7%), diarrhea (4%), and anorexia (4%). Analysis of the phosphorylation of downstream targets of mTOR, eukaryotic initiation factor 4E-binding protein 1, and/or p70 S6 kinase showed inhibition in 66% of patients (6 of 9).44 Measuring S6K activity in peripheral blood mononuclear cells has been proposed as a surrogate of measuring the clinical effect of everolimus, given that prolonged inactivation of S6K1 has been noticed after intermittent treatment doses.120 When given as a single agent in mantle cell lymphoma cell lines, it induced G1 cell-cycle arrest and reduced phosphorylation of the mTOR downstream target, 4E-BP1. It also showed synergistic cytotoxicity with other agents that are commonly used in this disease, including doxorubicin, vincristine, or rituximab as well as other agents like bortezomib, paclitaxel, and vorinostat.121
Deforolimus (AP23573) A phase 2 clinical trial tested AP23573 in 12 patients with relapsed or refractory hematologic malignancies in which it was administered at a dose of 12.5 mg intravenously daily for 5 days every 2 weeks. The median age was 76 years. Three patients had minor responses in terms of hematologic improvement, and another 3 patients had stable disease. Serious drug-related side events were hypertriglyceridemia, mucositis, and neutropenic sepsis.122 A later report from the same study included 51 patients with a median age of 61 years. Serious side effects additionally reported were diarrhea, dyspnea, syncope, pleural effusion, and pneumonia. Nineteen of the 46 evaluable patients (41%) showed at least stable disease.123
Summary Despite significant advances in the treatment of hematological malignancies over the previous decades, morbidity and mortality from theses disorders still remain high. New discoveries in the pathways involved in the pathophysiology led to new thinking in drug discovery. mTOR inhibitors are new and viable options for patients with these malignancies. We hope that newer clinical data will bring about new options in this field.
REFERENCES 1. Meijer AJ, Dubbelhuis PF. Amino acid signalling and the integration of metabolism. Biochem Biophys Res Commun 2004;313(2):397-403. 170
Curr Probl Cancer, July/August 2008
2. Long X, Muller F, Avruch J. TOR action in mammalian cells and in Caenorhabditis elegans. Curr Top Microbiol Immunol 2004;279:115-38. 3. Gingras AC, Raught B, Sonenberg N. mTOR signaling to translation. Curr Top Microbiol Immunol 2004;279:169-97. 4. Fingar DC, Richardson CJ, Tee AR, et al. mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol Cell Biol 2004;24(1):200-16. 5. Schmelzle T, Hall MN. TOR, a central controller of cell growth [see comment]. Cell 2000;103(2):253-62. 6. Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/mTOR. Genes Dev 2001;15(7):807-26. 7. Sabers CJ, Martin MM, Brunn GJ, et al. Isolation of a protein target of the FKBP12-rapamycin complex in mammalian cells. J Biol Chem 1995;270(2): 815-22. 8. Sabatini DM, Erdjument-Bromage H, Lui M, et al. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994;78(1):35-43. 9. Chiu MI, Katz H, Berlin V. RAPT1, a mammalian homolog of yeast Tor, interacts with the FKBP12/rapamycin complex. Proc Natl Acad Sci U S A 1994; 91(26):12574-8. 10. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 1994;369(6483):756-8. 11. Jacinto E, Hall MN. Tor signalling in bugs, brain and brawn. Nat Rev Mol Cell Biol 2003;4(2):117-26. [Published erratum appears in Nat Rev Mol Cell Biol 2003;4(3):249.] 12. Abraham RT. Identification of TOR signaling complexes: more TORC for the cell growth engine. Cell 2002;111(1):9-12. 13. 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(5):335-40. [Reprint in Clin Biochem 2006;39(5):484-9, PMID: 16783879.] 14. Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot 1975;28(10):721-6. 15. Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22989), a new antifungal antibiotic. II. Fermentation, isolation and characterization. J Antibiot 1975; 28(10):727-32. 16. Calne RY, Collier DS, Lim S, et al. Rapamycin for immunosuppression in organ allografting [see comment]. Lancet 1989;2(8656):227. 17. Augustine JJ, Bodziak KA, Hricik DE. Use of sirolimus in solid organ transplantation. Drugs 2007;67(3):369-91. 18. Temsirolimus: CCI 779, CCI-779, cell cycle inhibitor-779. Drugs R D 2004; 5(6):363-7. 19. Steffel J, Latini RA, Akhmedor A, et al. Rapamycin, but not FK-506, increases endothelial tissue factor expression: implications for drug-eluting stent design. Circulation 2005;112(13):2002-11. 20. Eng CP, Sehgal SN, Vezina C. Activity of rapamycin (AY-22989) against transplanted tumors. J Antibiot 1984;37(10):1231-7. Curr Probl Cancer, July/August 2008
171
21. 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(6):799-805. 22. Douros J, Suffness M. New antitumor substances of natural origin. Cancer Treat Rev 1981;8(1):63-87. 23. Huang S, Houghton PJ. Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol 2003;3(4):371-7. 24. Dennis PB, Thomas G. Quick guide: target of rapamycin. Curr Biol 2002; 12(8):R269. 25. Sarkaria JN, Busby EC, Tibbetts RS, et al. Inhibition of phosphoinositide 3-kinase related kinases by the radiosensitizing agent wortmannin. Cancer Res 1998; 58(19):4375-82. 26. Yuille MA, Coignet LJ. The ataxia telangiectasia gene in familial and sporadic cancer. Recent Results Cancer Res 1998;154:156-73. 27. Kim DH, Sarbassor DD, Ali SM, et al. GbetaL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR. Mol Cell 2003;11(4):895-904. 28. Hara K, Maruki Y, Long X, et al. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 2002;110(2):177-89. 29. Kim DH, Sarbassor DD, Ali SM, et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002;110(2):163-75. 30. Yonezawa K, Tokunaga C, Oshiro N, et al. Raptor, a binding partner of target of rapamycin. Biochem Biophys Res Commun 2004;313(2):437-41. 31. Harris TE, Lawrence JC Jr. TOR signaling. Science’s Stke [Electronic Resource]: Signal Transduction Knowledge Environment, 2003. 2003(212):re15. 32. Nojima H, Tokunaga C, Eguchi S, et al. The mammalian target of rapamycin (mTOR) partner, raptor, binds the mTOR substrates p70 S6 kinase and 4E-BP1 through their TOR signaling (TOS) motif. J Biol Chem 2003;278(18):15461-4. 33. Proud CG. mTOR-mediated regulation of translation factors by amino acids. Biochem Biophys Res Commun 2004;313(2):429-36. 34. Yonezawa K, Yoshino KI, Tokunaga C, et al. Kinase activities associated with mTOR. Curr Top Microbiol Immunol 2004;279:271-82. 35. Sarbassov DD, Ali SM, Kim DH, et al. Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 2004;14(14):1296-302. 36. Sarbassov DD, Guertin DA, Ali SM, et al. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307(5712):1098-101. 37. Motzer RJ, Hudes GR, Curti BD, et al. Phase I/II trial of temsirolimus combined with interferon alfa for advanced renal cell carcinoma. J Clin Oncol 2007; 25(25):3958-64. 38. Mabuchi S, Altomare DA, Connolly DC, et al. RAD001 (Everolimus) delays tumor onset and progression in a transgenic mouse model of ovarian cancer. Cancer Res 2007;67(6):2408-13. 39. Karbowniczek M, Spittle CS, Morrison T, Wu H, Henske EP. mTOR is activated in the majority of malignant melanomas. J Invest Dermatol 2008;128(4):980-7. 40. Johnson BE, Jackman D, Janne PA. Rationale for a phase I trial of erlotinib and the mammalian target of rapamycin inhibitor everolimus (RAD001) for patients with relapsed non small cell lung cancer. Clin Cancer Res 2007;13(15):s4628-31. 172
Curr Probl Cancer, July/August 2008
41. Fouladi M, Laningham F, Wu J, et al. Phase I study of everolimus in pediatric patients with refractory solid tumors. J Clin Oncol 2007;25(30):4806-12. 42. Costa LJ. Aspects of mTOR biology and the use of mTOR inhibitors in non-Hodgkin’s lymphoma. Cancer Treat Rev 2007;33(1):78-84. 43. Cho D, Signoretti S, Regan M, et al. The role of mammalian target of rapamycin inhibitors in the treatment of advanced renal cancer. Clin Cancer Res 2007;13(2):758s-63s. 44. Yee KW, Zeng Z, Konoplera M, et al. Phase I/II study of the mammalian target of rapamycin inhibitor everolimus (RAD001) in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res 2006;12(17):5165-73. 45. Hynes NE, Boulay A. The mTOR pathway in breast cancer. J Mammary Gland Biol Neoplasia 2006;11(1):53-61. 46. Panwalkar A, Verstovsek S, Giles FJ. Mammalian target of rapamycin inhibition as therapy for hematologic malignancies. Cancer 2004;100(4):657-66. 47. Dancey JE. mTOR inhibitors in hematologic malignancies. Clin Adv Hematol Oncol 2003;1(7):419-23. 48. Herberger B, Puhalla H, Lehnert M, et al. Activated mammalian target of rapamycin is an adverse prognostic factor in patients with biliary tract adenocarcinoma. Clin Cancer Res 2007;13(16):4795-9. 49. Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell 2007;12(1):9-22. 50. Hutson TE, Figlin RA. Evolving role of novel targeted agents in renal cell carcinoma. Oncology 2007;21(10):1175-80. 51. Brown RE, Zhang PL, Lun M, et al. Morphoproteomic and pharmacoproteomic rationale for mTOR effectors as therapeutic targets in head and neck squamous cell carcinoma. Ann Clin Lab Sci 2006;36(3):273-82. 52. Clark PE. Recent advances in targeted therapy for renal cell carcinoma. Curr Opin Urol 2007;17(5):331-6. 53. Kobayashi S, Kishimoto T, Kamata S, et al. Rapamycin, a specific inhibitor of the mammalian target of rapamycin, suppresses lymphangiogenesis and lymphatic metastasis. Cancer Sci 2007;98(5):726-33. 54. Huber S, Bruns CJ, Schmid G, et al. Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis. Kidney Int 2007;71(8):771-7. 55. Nathan CO, Amirghahari N, Rong X, et al. Mammalian target of rapamycin inhibitors as possible adjuvant therapy for microscopic residual disease in head and neck squamous cell cancer. Cancer Res 2007;67(5):2160-8. 56. Buck E, Eyzaguirre A, Brown E, et al. Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther 2006;5(11):2676-84. 57. Chollet P, Abrial C, Tacca O, et al. Mammalian target of rapamycin inhibitors in combination with letrozole in breast cancer. Clin Breast Cancer 2006;7(4):336-8. 58. Martelli AM, Tazzari PL, Evangelisti C, et al. Targeting the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin module for acute myelogenous leukemia therapy: from bench to bedside. Curr Med Chem 2007;14(19):2009-23. 59. Follo MY, Mongiorgi S, Bosi C, et al. The Akt/mammalian target of rapamycin signal transduction pathway is activated in high-risk myelodysplastic syndromes and influences cell survival and proliferation. Cancer Res 2007;67(9):4287-94. Curr Probl Cancer, July/August 2008
173
60. Xu Q, Thompson JE, Carroll M. mTOR regulates cell survival after etoposide treatment in primary AML cells. Blood 2005;106(13):4261-8. 61. Wanner K, Hipp S, Oelsner M, et al. Mammalian target of rapamycin inhibition induces cell cycle arrest in diffuse large B cell lymphoma (DLBCL) cells and sensitises DLBCL cells to rituximab. Br J Haematol 2006;134(5):475-84. 62. Vega F, Medeiros LJ, Leventaki V, et al. Activation of mammalian target of rapamycin signaling pathway contributes to tumor cell survival in anaplastic lymphoma kinase-positive anaplastic large cell lymphoma. Cancer Res 2006; 66(13):6589-97. 63. Kawamata N, Chen J, Koeffler HP. Suberoylanilide hydroxamic acid (SAHA; vorinostat) suppresses translation of cyclin D1 in mantle cell lymphoma cells. Blood 2007;110(7):2667-73. 64. Huyghe E, Zairi A, Nohra J, et al. Gonadal impact of target of rapamycin inhibitors (sirolimus and everolimus) in male patients: an overview. Transpl Int 2007;20(4):305-11. 65. Deutsch MA, Kaczmarek I, Huber S, et al. Sirolimus-associated infertility: case report and literature review of possible mechanisms. Am J Transplant 2007; 7(10):2414-21. 66. Pallet N, Thervet E, Legendre C, et al. [Nephrotoxicity of sirolimus: experimental and clinical data]. Nephrol Ther 2006;2(4):183-90. 67. Rangan GK. Sirolimus-associated proteinuria and renal dysfunction. Drug Saf 2006;29(12):1153-61. 68. Boni J, Leister C, Burns J, et al. Pharmacokinetic profile of temsirolimus with concomitant administration of cytochrome P450-inducing medications. J Clin Pharmacol 2007;47(11):1430-9. 69. Duran I, Siu LL, Oza AM, et al. Characterisation of the lung toxicity of the cell cycle inhibitor temsirolimus. Eur J Cancer 2006;42(12):1875-80. 70. Tenderich G, Fuchs U, Zittermann A, et al. Comparison of sirolimus and everolimus in their effects on blood lipid profiles and haematological parameters in heart transplant recipients. Clin Transplant 2007;21(4):536-43. 71. Fruman DA, Wood MA, Gjertson CK, et al. FK506 binding protein 12 mediates sensitivity to both FK506 and rapamycin in murine mast cells. Eur J Immunol 1995;25(2):563-71. 72. Luo Y, Marx SO, Kiyokawa H, et al. Rapamycin resistance tied to defective regulation of p27Kip1. Mol Cell Biol 1996;16(12):6744-51. 73. Shima H, Pende M, Chen Y, et al. Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse phenotype and a new functional S6 kinase. EMBO J 1998; 17(22):6649-59. 74. Dilling MB, Germain GS, Dudkin L, et al. 4E-binding proteins, the suppressors of eukaryotic initiation factor 4E, are down-regulated in cells with acquired or intrinsic resistance to rapamycin. J Biol Chem 2002;277(16):13907-17. 75. Dennis PB, Pullen N, Kozma SC, et al. The principal rapamycin-sensitive p70(s6k) phosphorylation sites, T-229 and T-389, are differentially regulated by rapamycininsensitive kinase kinases. Mol Cell Biol 1996;16(11):6242-51. 76. Sugiyama H, Papst P, Gelfand EW, et al. p70 S6 kinase sensitivity to rapamycin is eliminated by amino acid substitution of Thr229. J Immunol 1996;157(2):656-60. 77. Huang S, Shu L, Dilling MB, et al. Sustained activation of the JNK cascade and 174
Curr Probl Cancer, July/August 2008
78. 79. 80. 81. 82.
83.
84. 85.
86. 87.
88.
89.
90.
91.
92. 93. 94.
95.
rapamycin-induced apoptosis are suppressed by p53/p21(Cip1). Mol Cell 2003;11(6):1491-501. Kurmasheva RT, Huang S, Houghton PJ. Predicted mechanisms of resistance to mTOR inhibitors. Br J Cancer 2006;95(8):955-60. Beamish H, Williams R, Chen P, et al. Rapamycin resistance in ataxia-telangiectasia. Oncogene 1996;13(5):963-70. Bertram PG, Zeng C, Thorson J, et al. The 14-3-3 proteins positively regulate rapamycin-sensitive signaling. Curr Biol 1998;8(23):1259-67. O’Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res 2006;66(3):1500-8. Dumont FJ, Staruch MJ, Grammer T, et al. Dominant mutations confer resistance to the immunosuppressant, rapamycin, in variants of a T cell lymphoma. Cell Immunol 1995;163(1):70-9. Hammerman PS, Fox CJ, Birnbaum MJ, et al. Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival. Blood 2005; 105(11):4477-83. Shi Y, Gera J, Hu L, et al. Enhanced sensitivity of multiple myeloma cells containing PTEN mutations to CCI-779. Cancer Res 2002;62(17):5027-34. Peponi E, Drakos E, Reyes G, et al. Activation of mammalian target of rapamycin signaling promotes cell cycle progression and protects cells from apoptosis in mantle cell lymphoma. Am J Pathol 2006;169(6):2171-80. Goy A, Feldman T. Expanding therapeutic options in mantle cell lymphoma. Clin Lymphoma Myeloma 2007;7:S184-91 (suppl 5). Hipp S, Ringshausen I, Oelsner M, et al. Inhibition of the mammalian target of rapamycin and the induction of cell cycle arrest in mantle cell lymphoma cells. Haematologica 2005;90(10):1433-4. Baumann P, Mandl-Weber S, Emmerich B, et al. Activation of adenosine monophosphate activated protein kinase inhibits growth of multiple myeloma cells. Exp Cell Res 2007;313(16):3592-603. Strömberg T, Dimberg A, Hammarberg A, et al. Rapamycin sensitizes multiple myeloma cells to apoptosis induced by dexamethasone. Blood 2004;103(8): 3138-47. Yan H, Frost P, Shi Y, et al. Mechanism by which mammalian target of rapamycin inhibitors sensitize multiple myeloma cells to dexamethasone-induced apoptosis. Cancer Res 2006;66(4):2305-13. Francis LK, Alsayed Y, Leleu X, et al. Combination mammalian target of rapamycin inhibitor rapamycin and HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin has synergistic activity in multiple myeloma. Clin Cancer Res 2006;12(22):6826-35. Avellino R, Romano S, Parasole R, et al. Rapamycin stimulates apoptosis of childhood acute lymphoblastic leukemia cells. Blood 2005;106(4):1400-6. Thomas X. Emerging drugs for adult acute lymphoblastic leukaemia. Expert Opin Emerg Drugs 2005;10(3):591-617. Brown VI, Fang J, Alcom K, et al. Rapamycin is active against B-precursor leukemia in vitro and in vivo, an effect that is modulated by IL-7-mediated signaling. Proc Natl Acad Sci U S A 2003;100(25):15113-8. Lal L, Li Y, Smith J, et al. Activation of the p70 S6 kinase by all-trans-retinoic acid in acute promyelocytic leukemia cells. Blood 2005;105(4):1669-77.
Curr Probl Cancer, July/August 2008
175
96. Dos Santos C, Récher C, Demur C, et al. [The PI3K/Akt/mTOR pathway: a new therapeutic target in the treatment of acute myeloid leukemia]. Bull Cancer 2006;93(5):445-7. 97. Récher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 2005;105(6):2527-34. 98. Récher C, Dos Santos C, Demur C, et al. mTOR, a new therapeutic target in acute myeloid leukemia. Cell Cycle 2005;4(11):1540-9. 99. Xu RH, Pelicano H, Zhang H, et al. Synergistic effect of targeting mTOR by rapamycin and depleting ATP by inhibition of glycolysis in lymphoma and leukemia cells. Leukemia 2005;19(12):2153-8. 100. Yoon P, Giafis N, Smith J, et al. Activation of mammalian target of rapamycin and the p70 S6 kinase by arsenic trioxide in BCR-ABL-expressing cells. Mol Cancer Ther 2006;5(11):2815-23. 101. Ly C, Arechiga AF, Melo JV, et al. Bcr-Abl kinase modulates the translation regulators ribosomal protein S6 and 4E-BP1 in chronic myelogenous leukemia cells via the mammalian target of rapamycin. Cancer Res 2003;63(18):5716-22. 102. Burchert A, Wang Y, Cai D, et al. Compensatory PI3-kinase/Akt/mTor activation regulates imatinib resistance development. Leukemia 2005;19(10):1774-82. 103. Mayerhofer M, Aichberger KJ, Florian S, et al. Identification of mTOR as a novel bifunctional target in chronic myeloid leukemia: dissection of growth-inhibitory and VEGF-suppressive effects of rapamycin in leukemic cells. FASEB J 2005;19(8):960-2. 104. Parmar S, Smith J, Sassano A, et al. Differential regulation of the p70 S6 kinase pathway by interferon alpha (IFNalpha) and imatinib mesylate (STI571) in chronic myelogenous leukemia cells. Blood 2005;106(7):2436-43. 105. Jundt F, Raetzel N, Müller C, et al. A rapamycin derivative (everolimus) controls proliferation through down-regulation of truncated CCAAT enhancer binding protein {beta} and NF-{kappa}B activity in Hodgkin and anaplastic large cell lymphomas. Blood 2005;106(5):1801-7. 106. Ringshausen I, Peschel C, Decker T. Mammalian target of rapamycin (mTOR) inhibition in chronic lymphocytic B-cell leukemia: a new therapeutic option. Leuk Lymphoma 2005;46(1):11-9. 107. Leseux L, Hamdi SM, Al Saati T, et al. Syk-dependent mTOR activation in follicular lymphoma cells. Blood 2006;108(13):4156-62. 108. Raymond E, Alexandre J, Faivre S, et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004;22(12):2336-47. 109. Hidalgo M, Buckner JC, Erlichman C, et al. A phase I and pharmacokinetic study of temsirolimus (CCI-779) administered intravenously daily for 5 days every 2 weeks to patients with advanced cancer. Clin Cancer Res 2006;12(19):5755-63. 110. 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(5):909-18. 111. Peralba JM, DeGraffenreid L, Friedrichs W, et al. Pharmacodynamic evaluation of CCI-779, an inhibitor of mTOR, in cancer patients. Clin Cancer Res 2003; 9(8):2887-92. 176
Curr Probl Cancer, July/August 2008
112. Witzig TE, Geyer SM, Ghobrial I, et al. Phase II trial of single-agent temsirolimus (CCI-779) for relapsed mantle cell lymphoma. J Clin Oncol 2005;23(23):5347-56. 113. Grube E, Gerckens U, Buellesfeld L. Drug-eluting stents: clinical experiences and perspectives. Minerva Cardioangiol 2002;50(5):469-73. 114. 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(8):4285-90. 115. Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients. N Engl J Med 2003;349(9):847-58. 116. Majewski M, Korecka M, Joergensen J, et al. Immunosuppressive TOR kinase inhibitor everolimus (RAD) suppresses growth of cells derived from posttransplant lymphoproliferative disorder at allograft-protecting doses. Transplantation 2003;75(10):1710-7. 117. Azzola A, Havry KA, Chhajed P, et al. Everolimus and mycophenolate mofetil are potent inhibitors of fibroblast proliferation after lung transplantation. Transplantation 2004;77(2):275-80. 118. Pascual J. Everolimus in clinical practice: renal transplantation. Nephrol Dial Transplant 2006;21:iii18-23 (suppl 3). 119. Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet 2004;43(2):83-95. 120. 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(1):252-61. 121. 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;21(2):333-9. 122. Feldman E, Giles F, Roboz G, et al. A phase 2 clinical trial of AP23573, an mTOR inhibitor, in patients with relapsed or refractory hematologic malignancies. J Clin Oncol 2005 ASCO Annual Meeting Proceedings. Vol 23, No. 16S, Part I of II (June 1 Supplement), Abstract 6631, 2005. 123. Rizzieri DA, Feldman E, Moore JO, et al. A phase 2 clinical trial of AP23573, an mTOR inhibitor, in patients with relapsed or refractory hematologic malignancies. Blood 2005;106(11):Abstract 2980.
Curr Probl Cancer, July/August 2008
177