Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cancer Therapy: Clinical

Phase I/II Study of the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients with Relapsed or Refractory Hematologic Malignancies

Karen W.L. Yee, Zhihong Zeng, Marina Konopleva, Srdan Verstovsek, Farhad Ravandi, Alessandra Ferrajoli, Deborah Thomas, William Wierda, Efrosyni Apostolidou, Maher Albitar, Susan O'Brien, Michael Andreeff and Francis J. Giles
Karen W.L. Yee
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Zhihong Zeng
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Marina Konopleva
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Srdan Verstovsek
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Farhad Ravandi
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alessandra Ferrajoli
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Deborah Thomas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
William Wierda
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Efrosyni Apostolidou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maher Albitar
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Susan O'Brien
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Michael Andreeff
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francis J. Giles
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-06-0764 Published September 2006
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: Everolimus (RAD001, Novartis), an oral derivative of rapamycin, inhibits the mammalian target of rapamycin (mTOR), which regulates many aspects of cell growth and division. A phase I/II study was done to determine safety and efficacy of everolimus in patients with relapsed or refractory hematologic malignancies.

Experimental Design: Two dose levels (5 and 10 mg orally once daily continuously) were evaluated in the phase I portion of this study to determine the maximum tolerated dose of everolimus to be used in the phase II study.

Results: Twenty-seven patients (9 acute myelogenous leukemia, 5 myelodysplastic syndrome, 6 B-chronic lymphocytic leukemia, 4 mantle cell lymphoma, 1 myelofibrosis, 1 natural killer cell/T-cell leukemia, and 1 T-cell prolymphocytic leukemia) received everolimus. No dose-limiting toxicities were observed. Grade 3 potentially drug-related toxicities included hyperglycemia (22%), hypophosphatemia (7%), fatigue (7%), anorexia (4%), and diarrhea (4%). One patient developed a cutaneous leukocytoclastic vasculitis requiring a skin graft. One patient with refractory anemia with excess blasts achieved a major platelet response of over 3-month duration. A second patient with refractory anemia with excess blasts showed a minor platelet response of 25-day duration. Phosphorylation of downstream targets of mTOR, eukaryotic initiation factor 4E-binding protein 1, and/or, p70 S6 kinase, was inhibited in six of nine patient samples, including those from the patient with a major platelet response.

Conclusions: Everolimus is well tolerated at a daily dose of 10 mg daily and may have activity in patients with myelodysplastic syndrome. Studies of everolimus in combination with therapeutic agents directed against other components of the phosphatidylinositol 3-kinase/Akt/mTOR pathway are warranted.

  • mTOR inhibitor
  • myelodysplastic syndrome
  • chronic lymphocytic leukemia
  • 4E-BP1
  • p70S6K
  • hypoxia-inducible factor

The phosphatidylinositol 3-kinase/Akt signaling pathway is important for cell growth and survival (1, 2). Akt is activated downstream of phosphatidylinositol 3-kinase and can activate several effector proteins, including the serine/threonine kinase mammalian target of rapamycin (mTOR; Fig. 1 ). Activated Akt can phosphorylate tuberin (TSC2), causing disruption of the inhibitory hamartin (TSC1)/TSC2 complex, and activate the GTPase Rheb, which in turn promotes the formation of the mTOR-raptor complex (TORC1). TORC1 regulates cell cycle progression (i.e., G1 to S phase transition) in part by controlling the mammalian translation machinery via activation of the p70 S6 kinase (p70S6K) protein kinase and via inhibition of the elongation initiation factor 4E inhibitor 4E-binding protein 1 (4E-BP1 refs. 1, 2). Hence, TORC1 activation results in up-regulation of effectors required for protein translation and cell cycle progression and proliferation, such as hypoxia-inducible factor-1α and cyclin D1. Up-regulation of hypoxia-inducible factor-1α leads to increased expression of angiogenic factors, such as vascular endothelial growth factor, platelet-derived growth factor, and growth-stimulatory molecules [e.g., glucose transporter 1 (Glut1); ref. 3]. mTOR has been implicated in both transformation and therapeutic resistance (1, 2).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

The mTOR pathway. PI3K, phosphatidylinositol 3-kinase; Hif-1α, hypoxia-inducible factor-1α; eIF3, elongation initiation factor 3; eIF4E, elongation initiation factor 4E; eIF4G, elongation initiation factor 4G.

The phosphatidylinositol 3-kinase/Akt/mTOR pathway is heavily dysregulated in hematologic malignancies and is activated by several upstream proteins, such as ras, TCL1, and bcr-abl, and membrane receptor tyrosine kinases, including vascular endothelial growth factor receptor, platelet-derived growth factor receptor, c-kit, and Flt3 (1). Increased expression and constitutive activation of the catalytic subunit of phosphatidylinositol 3-kinase and Akt and/or decreased or absent PTEN protein expression have been reported in primary samples from patients with acute myelogenous leukemia (AML) and/or myelodysplastic syndrome (MDS; ref. 1). Furthermore, p70S6K and 4E-BP1 are constitutively phosphorylated in primary AML cells. Tumors, such as mantle cell lymphoma, may express high levels of mTOR-regulated mRNAs (e.g., cyclin D1; ref. 1).

Everolimus [RAD001; 40-O-[2-hydroxyethyl]-rapamycin], an orally available ester derivative of the macrolide antifungal antibiotic sirolimus (rapamycin), is currently approved in Europe as an immunosuppressive agent to prevent rejection in adult cardiac and renal transplant recipients (4, 5). Everolimus has better oral bioavailability compared with the parent compound sirolimus, with steady state levels achieved within 7 days (6). Maintenance immunosuppression with everolimus or sirolimus has been associated with a significantly reduced risk of developing de novo malignancies after renal transplant (7).

Everolimus forms a complex with the immunophilin FKBP-12 (i.e., 12 kDa FK506 binding protein), which then binds to and disrupts TORC1, leading to mTOR inhibition and preventing the activation of p70S6K and inactivation of 4E-BP1. Everolimus causes G1 phase cell cycle arrest; the p21/cyclin D/cyclin-dependent kinases 2 and 4/proliferating cell nuclear antigen complexes are disrupted with down-regulation of p21, cyclin-dependent kinases 2 and 4, and subsequent inhibition of tumor growth (8–11). It may prevent nuclear factor-κB activation possibly by stabilization of its inhibitor IκBα (12). Everolimus may also exert its antitumor effect by inducing apoptosis (9, 10, 13) and suppressing angiogenesis (9).

In preclinical studies, everolimus inhibited proliferation and growth of a broad range of human tumor cell lines and xenograft models (8, 10, 13) and is being developed as an antitumor agent (14–16). The dose of everolimus influenced the duration of p70S6K inhibition in peripheral blood mononuclear cells in patients with solid tumors receiving escalating doses of single agent everolimus on a weekly schedule (16). At steady state, duration of p70S6K inhibition was 3 to 5 days in patients given doses of 5 to 10 mg and >7 days for those administered 20 to 30 mg (16). Pharmacokinetic and pharmacodynamic modeling has been done by combining this clinical data with preclinical data in rat pancreatic cancer xenograft models to predict intratumoral inhibition in patients (8, 16, 17). The pharmacokinetic and pharmacodynamic model predicted that the 20 mg weekly dose would be the minimal effective dose in patients and that sustained p70S6K inhibition in peripheral blood mononuclear cells should occur with daily administration as opposed to increasing the weekly dose >20 mg for the same total weekly drug administration (i.e., 10 mg daily compared with 70 mg weekly).

This was confirmed in a second phase I study of everolimus given to adults with solid tumors either weekly or daily (15). Inhibition of downstream effectors of mTOR in tumor biopsies correlated with findings in surrogate skin biopsies. Biomarker analyses showed dose-dependent and schedule-dependent inhibitions of the mTOR pathway: the daily schedule was associated with high inhibition of p70S6K and elongation initiation factor 4G at 5 mg daily, with complete inhibition occurring at 10 mg daily, whereas with the weekly schedule complete and sustained inhibition of p70S6K and elongation initiation factor 4G was observed at doses >20 mg weekly and >50 mg weekly, respectively, and nonsustained up-regulation of phosphorylated Akt was seen at doses of 50 mg. There was also a trend for decreased phosphorylation of 4E-BP1.

Although mTOR is clearly an attractive therapeutic target in leukemia, no clinical data on mTOR inhibition by rapamycin analogues have been published in this patient population. A particular concern is whether these agents, with established immunosuppressive activity, could increase infectious agent complications.

Therefore, a phase I/II study was conducted to determine the safety, tolerability, and activity of everolimus administered daily at one of two doses (5 or 10 mg daily) in patients with a variety of relapsed or refractory or advanced hematologic malignancies. Furthermore, in consenting patients, blood samples were collected to assess surrogate markers of mTOR inhibition at baseline and at various treatment time points.

Materials and Methods

Patients. Patients with histologically confirmed advanced, relapsed, or refractory AML or MDS, chronic lymphocytic leukemia (CLL), T-cell leukemia, myelofibrosis, or mantle cell lymphoma (MCL) were eligible for enrollment.

Treatment protocol. Patients were treated with 5 (first three patients) or 10 mg orally once daily continuously either in a fasted state or after a light fat-free meal. One treatment cycle consisted of 28 days of therapy. If emesis occurred after ingesting everolimus, the dose was not repeated. Novartis Pharmaceuticals (East Hanover, NJ) provided everolimus film-coated tablets at strengths of 5 mg. Treatment was stopped if any of the following events occurred: unacceptable toxicity, intercurrent illness or change in the patient's condition that rendered the patient unsuitable for further therapy in the judgment of the investigator, or patient withdrawal from the study.

Assessment of toxicity and response. All patients were evaluable for toxicity if they received at least one dose of everolimus. Adverse events were assessed at each visit and graded according to the National Cancer Institute Common Toxicity Criteria version 3.0. Hematologic dose-limiting toxicity (DLT) was defined as myelosuppression with bone marrow hypoplasia (cellularity <5%) without evidence of leukemia for >42 days. Nonhematologic DLT was defined as any grade 4 toxicity, except for the following: grade 3 nausea or vomiting, drug-related fever of any grade, alopecia, and grade 3 lipid or electrolyte abnormalities.

Sample collection. After obtaining written informed consent, whole-blood samples were collected into 10 mL sodium heparin tubes (Becton Dickinson, Sparks, MD) at baseline and 24 hours after administration of everolimus during the first cycle of therapy.

Western blot analysis. For the Western blot analyses, peripheral blood mononuclear cells were lysed in phosphoprotein lysis buffer (10 mmol/L NaF, 1 mmol/L Na3VO4, 150 mmol/L NaCl, 1 mmol/L MgCl2, 1 mmol/L CaCl2, 0.1% NaN3, 10 mmol/L iodoacetamide, 3 mmol/L phenylmethylsulfonyl fluoride, and 1% Triton X-100) supplemented with a protease inhibitor cocktail (Roche Diagnostics Corp., Indianapolis, IN). Equal amounts of proteins were then separated on a 12% polyacrylamide gel, transferred to Hybond-P membranes (Amersham Pharmacia Biotech, Little Chalfont, United Kingdom), immunoblotted with specific antibody, and further visualized by using enhanced chemiluminescence detection system (Amersham Pharmacia Biotech). Western blots were analyzed on a Storm 860 system by using ImageQuant software (Molecular Dynamics, Sunnyvale, CA). Integrated absorbance of each lane (band obtained with the phosphorylated-specific antibody) was recorded, and protein levels were determined after normalizing for levels of corresponding total protein or glyceraldehyde-3-phosphate dehydrogenase. The antibodies for phosphorylated p70S6K (Thr389) and total p70S6K, phosphorylated Akt (Ser473)/(Thr308) and total Akt, phosphorylated S6RK and S6RK, phosphorylated 4E-BP1 (Thr70)/(Thr37/Thr46) and 4E-BP1, and phosphorylated extracellular signal-regulated kinase (ERK; p44/42) were obtained from Cell Signaling Technology, Inc. (Danvers, MA) The antibody for ERK was from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA), and the antibody for glyceraldehyde-3-phosphate dehydrogenase was from Chemicon International (Temecula, CA).

Quantitative real-time reverse transcription-PCR. Total RNAs were prepared using Trizol reagent as described by the manufacturer (Life Technologies, Inc., Gaithersburg, MD). Total RNA (1 μg) was reverse transcribed by avian myeloblastosis virus reverse transcriptase (Roche Diagnostics) under standard conditions. Duplicate samples of 1 μL of each cDNA were amplified by PCR in the ABI Prism 7700 Sequence Detection System (PE Applied Biosystems, Foster City, CA). The amplification reaction mixture (25 μL) contained cDNAs, forward primers, reverse primers, probes, and Taqman Universal PCR Master Mix (PE Applied Biosystems). β2-microglobulin was coamplified as an internal control to normalize for variable amounts of cDNA in each sample. The thermocycler variables were as follows: 50°C for 2 minutes, 95°C for 10 minutes, 40 cycles of 95°C for 15 seconds, and 60°C for 1 minute. Taqman gene expression assay for cyclin D1 (assay Hs00277039_m1), cyclin D2 (assay Hs00277041_m1), and Glut1 (assay Hs00197884_m1) was purchased from PE Applied Biosystems and used as directed by the manufacturer. Results were collected and analyzed to determine the PCR cycle number that generated the first fluorescence signal above a threshold (threshold cycle, Ct; 10 SD above the mean fluorescence generated during the baseline cycles). The abundance of each transcript of interest relative to that of β2-microglobulin was calculated as follows: relative expression = 100 × 2−ΔCt, where ΔCt is the mean Ct of the transcript of interest less the mean Ct of the transcript for β2-microglobulin.

Statistical analysis. At least three patients had to be observed for at least 4 weeks during their first cycle of treatment with 5 mg daily without a DLT. With each DLT, three additional assessable patients had to be accrued, and further escalation could occur only if no more DLTs were observed. In the absence of such toxicity, the highest dose level (i.e., 10 mg daily) was to be used for phase II testing.

For the phase II part of the study, the primary objective was to estimate the overall response rate (responses were defined as per standard criteria; refs. 18–21). As everolimus has a noncytotoxic mechanism of action, a response rate of 10% in a very poor prognosis group of patients was considered of sufficient interest to warrant additional investigation. A maximum total of 25 evaluable patients were to be entered in each group of patients with advanced or refractory disease: (a) AML or MDS, (b) myelofibrosis, and (c) CLL, T-cell leukemia, or MCL. This sample size would yield an 82% posterior credibility interval for probability of response of width ∼0.16. Interim analyses were to be done in each group after 14 patients had been evaluated for response. Thus, in each patient cohort, the trial would be terminated after the first 14 patients if no responses were observed. As a further safeguard to prevent exposure of patients to an agent with minimal activity, the overall response rate was reviewed periodically. If overall response rate was <1 in 26 patients, 2 in 42 patients, or 3 in 55 patients, this would indicate that the probability of an overall response rate of <10% among this patient population was >90%. The study was closed to accrual after 26 evaluable patients were enrolled.

Results

Study group. A total of 27 patients with AML, MDS, CLL, MCL, myelofibrosis, natural killer cell/T-cell cell leukemia, and T-cell prolymphocytic leukemia was entered onto the study between April 2004 and May 2005. Patient characteristics are listed in Table 1 . Fourteen patients had received three or more prior treatment regimens for their disease, including 2 patients who had received an allogeneic stem cell transplant. Two patients had not received prior therapy for their disease: a 75-year-old patient who developed therapy-related AML 2 years after being treated for Burkitt-like non–Hodgkin's lymphoma and a 59-year-old man who developed MDS 9 years after being treated with uracil and tegafur plus leucovorin for metastatic colon cancer. A third patient had been diagnosed with essential thrombocythemia in 1992, which evolved to myelofibrosis in 1999; the patient progressed to refractory anemia with excess blasts (RAEB) in transformation while receiving therapy with imatinib for myelofibrosis. The patients with myelofibrosis, natural killer cell/T-cell leukemia, and T-cell prolymphocytic leukemia had received two or more prior treatment regimens. Three of four patients with MCL were in leukemic phase with circulating lymphoma cells. All six patients with CLL had received prior fludarabine-based therapy.

View this table:
  • View inline
  • View popup
Table 1.

Patient characteristics (N = 27)

Side effects. All 27 patients were evaluable for toxicity. No DLT occurred in the first three patients treated with 5 mg daily; all subsequent patients received 10 mg daily. The frequency and grading of potentially treatment-related adverse effects are summarized in Table 2 . These included grade 1 or 2 hyperlipidemia (44%), elevation of transaminases and/or alkaline phosphatase (41%), anorexia (37%), oral aphthous ulcers (37%), diarrhea (29%), hyperglycemia (26%), and hypomagnesemia (22%). Uncomplicated grade 3 hyperglycemia occurred in six patients requiring insulin therapy; five had a history of diabetes mellitus with grade 2 hyperglycemia noted at baseline in four patients. The hyperglycemia was resolved to baseline or better with cessation of everolimus. Other grade 3 toxicities included hypophosphatemia and fatigue in two patients each and anorexia and diarrhea in one patient.

View this table:
  • View inline
  • View popup
Table 2.

Potentially everolimus-related adverse effects

One patient with refractory anemia with ringed sideroblasts developed a biopsy-proven grade 3 cutaneous leukocytoclastic vasculitis (LCV) after 120 days of therapy, which was believed to be related to everolimus. Everolimus was discontinued. She did not have any systemic manifestations of vasculitis nor malignancy. Systemic immunosuppressive therapy, including corticosteroids, was not used, and no new lesions have appeared. She required surgical debridement and skin grafting.

Thirty-eight infectious episodes were noted in 21 (78%) patients at some time during the therapy. These included fever of unknown origin in 6 (22%) patients and documented infections in 20 (74%) patients: 2 patients with sinusitis, 2 patients with bronchitis, 2 patients with bacteremia involving Enterococcus faecalis and Staphylococcus aureus, four urinary tract infections in 4 patients (one of whom developed a vaginal yeast infection and another Clostridium difficile–positive diarrhea while on antibiotics), 14 pneumonias in 12 patients (one of whom developed oral thrush while on antibiotics), and 1 patient each with cellulitis, folliculitis, paronychia, and presumed herpetic stomatitis/mucositis. Fourteen (37%) of the 38 infectious episodes that occurred in 9 patients required either hospitalization or prolonged hospitalization.

One patient with RAEB-1 developed a prolonged Escherichia coli urinary tract infection during the eleventh cycle of therapy, which was complicated by recurrent hematuria, requiring platelet transfusions, antibiotics, and hospitalization. Therefore, everolimus was held during the twelfth cycle of therapy. Cytoscopy with bladder biopsy revealed a marked chronic inflammatory infiltrate, vascular congestion, and hemorrhage with no leukemic cells present. The hematuria was resolved 37 days after discontinuation of everolimus. This has been observed with prolonged symptomatic urinary tract infections and was not believed to be associated with administration of everolimus.

Of the 12 patients with pneumonia, only 2 had positive cultures (Aspergillus species and coagulase-negative Staphylococcus and Aspergillus niger, Klebsiella species, and methicillin-resistant Staphylococcus aureus). Two episodes of pneumonia occurred in one patient with myelofibrosis who was subsequently diagnosed with biopsy-proven bronchiolitis obliterans with organizing pneumonia, which was treated successfully with steroids. Of the remaining 11 patients with pneumonia, 1 patient had a chronic esophageal stricture, requiring dilations every 2 months, due to prior radiotherapy for Burkitt-like lymphoma and was believed to have an aspiration pneumonia involving the right lower lobe, 4 had grade 3 or 4 neutropenia, and 2 had CLL with hypogammaglobulinemia. Two patients died while on study: one patient with natural killer cell/T-cell leukemia from neutropenic sepsis and one with CLL from uncontrolled gastrointestinal bleeding secondary to arteriovenous malformations.

Response to treatment. The median number of cycles administered was 1 (range, 0-11) with a median of 50 doses given (range, 4-318). Nine patients missed a median of two doses of everolimus (range, 1-6), two patients missed three doses, and one patient missed six doses at some time during the therapy. No patients required dose reduction; only one patient had dose escalation from 5 to 10 mg daily. One patient with AML received hydroxyurea for 4 days during the first cycle of therapy to control blast counts, was subsequently taken off study without completing one cycle of therapy because of intercurrent illness, and was replaced. Therefore, 26 patients were evaluable for response.

One objective response (hematologic improvement) was observed. A patient with RAEB-1 (patient 5) who was pancytopenic and red cell transfusion dependent had a major hematologic improvement in platelet count from baseline values of 22 to 26 × 109/L to a maximum of 102 × 109/L. Time to response was 50 days. Relapse occurred 104 days after achievement of major hematologic improvement in platelet count. Duration of minor and major platelet responses was 210 and 177 days, respectively. Fifty days after starting therapy with everolimus, this patient also had an improvement in the absolute neutrophil count by >0.5 × 109/L from baseline values of 1.3 to 1.4 × 109/L to a maximum of 3.0 × 109/L.

A second patient with therapy-related RAEB-1 (patient 3) had an improvement in platelet count from baseline values of 11 to 14 × 109/L to a maximum of 34 × 109/L; the improvement was noted 113 days after the dose of everolimus was increased from 5 mg daily to 10 mg daily. However, duration of response was only 25 days and, therefore, did not fulfill the criteria for minor hematologic improvement in platelet count. Response was lost during the episodes of recurrent hematuria. While off treatment and after resolution of the hematuria, this patient has had a sustained increase in platelet counts ranging from 29 to 42 × 109/L of >2-month duration. The patient with refractory anemia with ringed sideroblasts (patient 7), who developed a cutaneous LCV, had decreased red cell transfusion requirements, with only three transfusions administered over a 9-month period since discontinuing everolimus. The maximum interval between transfusions was 116 days compared with a baseline of every 21 to 26 days before treatment with everolimus.

Although no patients with CLL patients achieved a complete or partial response, three patients had a 27% to 34% reduction in adenopathy, documented radiographically, after two to three cycles of therapy. A fourth patient had a 65% reduction in peripheral lymphadenopathy, documented clinically, after two cycles of therapy; this patient subsequently died from uncontrolled gastrointestinal bleeding due to arteriovenous malformations.

Fifteen patients subsequently received other therapies for their underlying disease (median, 1; range, 1-4), including hydroxyurea (n = 2), induction chemotherapy with cytarabine and an anthracycline or purine analogue (n = 3), fludarabine-based regimen (n = 2), azacytidine (n = 1), vincristine and decadron (n = 1), hyperCVAD and alemtuzumab (n = 1), allogeneic stem cell transplant (n = 1), and other investigational agents (n = 8). After a median follow-up of 18 weeks (range, 3 to 68+) from the time of study entry, 16 patients have died, including the 2 patients who died while on study.

Effects of everolimus on mTOR signaling pathways. The ability of everolimus to modulate mTOR signaling was examined in samples from nine patients by comparing profiles of phosphorylated mTOR target proteins before and 24 hours after everolimus administration. To investigate everolimus-specific effects on mTOR signaling in vivo, phosphorylation of the 70-kDa 40S ribosomal protein kinase (p70S6K) and 4E-BP1 was examined. Both phosphorylated p70S6K and 4E-BP1 in Thr70 were simultaneously inhibited after 24 hours of everolimus administration in samples from four patients (patients 2, 3, 7, and 9; Table 3 ). 4E-BP1 exhibited a decrease in Thr70 phosphorylation in two additional samples (patients 5 and 6). Interestingly, the phosphorylation of another rapamycin-sensitive residue (Thr37/Thr46) was affected only in the sample from patient 3, indicating that mTOR-insensitive phosphorylation of this site can occur as previously suggested (22). No significant changes in mTOR signaling were observed in samples from four other patients. Notably, patients 3 (CLL), 5 (RAEB-1), 6 (RAEB-1), and 7 (refractory anemia with ringed sideroblasts) displayed some evidence of clinical responses to everolimus as detailed above, and patient 5 had objective response (hematologic improvement, see above). Examples of Western blot analyses for samples 5 and 3 are represented in Fig. 2 .

View this table:
  • View inline
  • View popup
Table 3.

Effects of everolimus on intracellular signaling pathways and transcription of target genes

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Peripheral blood mononuclear cells from the responding patient with RAEB-1 (sample #5) and from a patient with CLL (sample #3) were subjected to immunoblot analyses of phosphorylated 4E-BP1 (p4E-BP1), phosphorylated p70S6K (pp70S6K), total 4E-BP1, phosphorylated Akt (pAkt), total Akt, phosphorylated ERK (pERK), total ERK, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and quantitiative real-time PCR analysis of Glut1 and cyclin D1 transcripts. The abundance of transcripts of either cyclin D1 or Glut1 relative to that of β2-microglobulin (B2M) was assessed by real-time PCR as described in Materials and Methods. Error bars, half the difference between the maximum and minimum values that arose on substituting ΔCt − SD or ΔCt + SD, respectively, for ΔCt in the following formula: relative expression = 100 × 2−ΔCt.

Akt and mTOR are linked to each other via positive and negative regulatory circuits, and in model systems, tumors exhibiting activation of Akt are hypersensitive to mTOR inhibitors (23). We therefore analyzed effects of everolimus in the context of Akt signaling. At baseline, Akt was phosphorylated on Thr308 in all seven of seven samples tested and on Ser473 in eight of nine samples. Unexpectedly, we found inhibition of phosphorylation of Akt at Thr308 in four of six (patients 2, 3, 5, and 7) and at Ser473 in five of eight (patients 2, 3, 5, 6, and 9) patient samples (see examples on Fig. 2). Interestingly, Akt was down-regulated only in samples where inhibition of mTOR signaling was observed, suggesting that mTOR inhibition in vivo may inhibit Akt signaling. Activation (phosphorylation) of ERK was documented in samples from eight of nine patients, and unanticipated decrease in phosphorylated ERK levels was observed in samples from two patients (patients 3 and 8).

mTOR signaling has been reported to affect the expression of the target genes by different mechanisms (transcription, translation, or protein degradation; refs. 24–26). We examined the effects of mTOR inhibition on the transcription of the glucose transporter Glut1 and D-type cyclins that control the G1-S transition in the cell cycle. In samples from six patients, in which administration of everolimus inhibited mTOR signaling, down-regulation of Glut1 mRNA (>40%) was observed in three samples (patients 2, 3, and 6) and down-regulation of either cyclin D1 or cyclin D2 in three and three samples, respectively (Table 3). In samples 3 (CLL) and 5 (RAEB-1), concomitant decreases in mRNA expression of both cyclin D1 and D2 were observed.

Discussion

Because of accumulating evidence documenting the importance of the phosphatidylinositol 3-kinase/Akt/mTOR pathway in the pathogenesis of a variety of hematologic malignancies, several therapeutic strategies are being developed to modulate this signaling pathway. The importance of mTOR is underscored by its key regulatory role in protein translation (1, 2). The current phase I/II study is the first to evaluate everolimus in patients with advanced hematologic malignancies. In addition, the effect of everolimus on multiple mTOR-regulated proteins was evaluated.

Therapy with everolimus at a dose of 10 mg orally once daily was relatively well tolerated. Toxicities observed with everolimus were consistent with those reported in previous studies using mTOR inhibitors and consisted of hyperglycemia, hyperlipidemia, anorexia, elevated liver enzymes, oral aphthous ulcers, diarrhea, hypophosphatemia, fatigue, and hypomagnesemia (27–30). The frequency of infectious episodes did not seem to be increased in the study patients, an important consideration for future studies in the hematologic malignancies. There were no documented episodes of cytomegalovirus infections or varizella zoster infections.

One patient with advanced myelofibrosis had recurrent pulmonary infiltrates while receiving therapy with everolimus. The first episode occurred 57 days after initiating therapy and resolved with antibiotics and ongoing therapy with everolimus. This was subsequently biopsy proven to be bronchiolitis obliterans with organizing pneumonia and treated successfully with steroids. Everolimus was discontinued due to the intercurrent illness. She was not rechallenged. Bronchiolitis obliterans with organizing pneumonia is usually idiopathic, although it has been associated with hematologic malignancies and cytotoxic drugs (31, 32) Nonspecific pneumonitis has been described in patients treated with temsirolimus and sirolimus (27, 33). Six of 42 patients were diagnosed with bronchiolitis obliterans with organizing pneumonia, all of whom were recipients of solid organ transplants. Approximately one third of patients experienced improvement of the infiltrates after discontinuation of the drug, and two of four patients with renal cell cancer experienced recurrent pneumonitis on rechallenge (27). Preclinical studies indicated that everolimus could prevent the development of bronchiolitis obliterans in a porcine bronchial model (34). Similarly, the combination of mycophenolate and sirolimus could attenuate the progression of bronchiolitis obliterans syndrome in lung and heart-lung transplant recipients (35). Therefore, the relationship of bronchiolitis obliterans with organizing pneumonia to everolimus is unclear.

A patient with MDS developed cutaneous LCV while receiving therapy with everolimus on this study. Causes of LCV include malignancy, including MDS, and drugs (36). LCV has been reported with sirolimus in four patients. This occurred 0.75 to 4 months after initiation of sirolimus to prevent solid organ rejection. The involved sites were skin in three cases and gastrointestinal in the fourth. Symptoms were resolved within 0.2 to 4 months after discontinuation of sirolimus. Rechallenge with sirolimus resulted in recurrence of the LCV in one case.

Only one patient with MDS achieved an objective response (i.e., major hematologic improvement in platelet count), although one other patient achieved a minor response in platelet count of 25-day duration. Although only five patients with MDS were treated with everolimus, the response rate (20%) was comparable with what has been observed with sirolimus in this patient population (16%; ref. 28). None of the nine patients with AML treated with everolimus had an objective response, in contrast to an overall response rate of 44% (44% partial response) reported with sirolimus in patients with AML (37). It is unclear whether this is due to differences in patient characteristics, duration of therapy (only 28 days with sirolimus), or significant differences in the agents (1). We observed no responses in 10 patients with AML treated with temsirolimus (38). On interim analysis, we observed that only 2 of 26 (8%) of patients with AML or MDS treated with AP23573 had a hematologic improvement (39). Response rates of 38% (3% complete response; 35% partial response) were observed in patients with relapsed or refractory MCL receiving single agent temsirolimus; median time to response was 1 month (range, 1-8 months; ref. 29). All four patients with MCL treated with everolimus had progressive disease; three of whom did not complete a course of therapy. Furthermore, three patients had circulating lymphoma cells at baseline and all four patients in the current study had refractory disease compared with 54% of those given temsirolimus.

Variability in everolimus concentrations has been observed in different ethnic groups, with lower bioavailability in African-Americans compared with Caucasians or non-African-Americans (40). In the current study, 3 of the 26 evaluable patients were African-Americans (1 CLL, 1 natural killer cell/T-cell leukemia, and 1 AML); none of whom achieved an objective response, although the patient with CLL did have a 33% reduction in lymphadenopathy after three cycles of therapy with everolimus.

Akt was activated as evidenced by phosphorylation of Ser473 and Thr308 in all but one sample tested, confirming the high frequency of Akt activation in primary leukemia cells. (1) Analysis of phosphorylation of 4E-BP1 and p70S6K, established biomarkers of mTOR blockade by rapamycin analogues, showed inhibition of phosphorylation of both proteins in four of nine samples and inhibition of phosphorylated Thr70 in 4E-BP1 in two additional samples (i.e., inhibition in ∼66% of patients). Notably, evidence of inhibition of mTOR signaling by everolimus was documented in samples from all patients with clinical responses, with the exception of samples from three patients with CLL, where the patients did not consent to provide samples. mTOR inhibition was associated with transcriptional down-regulation of D-type cyclins (either cyclin D1 or cyclin D2) and a decrease in Glut1 mRNA levels in a subset of patients, suggesting that, in leukemic cells, everolimus attenuates transcription of these genes. However, inhibition of mTOR downstream targets did not translate into clinical responses in AML patients 2 and 9, indicating that the ability to inhibit the mTOR pathway is not always sufficient to elicit clinical responses.

Modest antitumor activity of mTOR inhibitors may also be associated with activation of alternative prosurvival signaling pathways, such as mitogen-activated protein kinase/ERK. Furthermore, ERK1/2 has been shown to phosphorylate and inactivate TSC2 and 4E-BP1 and activate p70S6K in vitro (41–43), and preclinical data indicate that inhibition of both mTOR and ERK signaling pathways is required to prevent de novo protein translation. We observed high levels of phosphorylation of ERK in the majority of samples studied, confirming our previously reported data in AML that showed phosphorylated ERK in >80% of AML patients and providing a rationale for future combination studies with Raf or mitogen-activated protein kinase/ERK kinase inhibitors (44, 45).

Recent work has shown that selective inhibition of mTOR downstream signals paradoxically increases phosphorylated Akt (46, 47), likely attributable to lack of feedback inhibition by p70S6K on the insulin-like growth factor-I receptor. We unexpectedly observed inhibition of Akt phosphorylation in five of six samples, in which everolimus modulated mTOR signaling. mTOR partitions between two scaffold proteins, the rapamycin-sensitive raptor and the rapamycin-insensitive rictor (23). Recent data show that the rictor-mTOR complex (TORC2) directly phosphorylates Akt on Ser473 and facilitates Thr308 phosphorylation by PDK1 (48). Although rapamycin does not bind to a preformed rictor-mTOR complex (49), it has been proposed that, during prolonged rapamycin treatment, the drug will sequester the newly synthesized mTOR molecules, interfere with reassembly of rictor-mTOR complex, and thereby inhibit rictor-mTOR activity. In fact, prolonged, but not acute, treatment of some human cells with rapamycin has been reported to partially inhibit Akt phosphorylation (50).

The data reported here provide the first in vivo evidence that rapamycin analogues are capable of suppressing Akt activity in hematopoietic cells, perhaps in contrast to solid tumor cell types. Although the antitumor activity of single agent mTOR inhibitors is modest in patients with hematologic malignancies, combination therapy of these agents with modulators of upstream stimulants of the pathway (e.g., vascular endothelial growth factor, Flt3, bcr-abl) is warranted. Based on data from the current study, concerns about the immunosuppressive effects of mTOR inhibitors in patients with advanced hematologic malignancies should not pose a barrier to the conduct of further studies, although this issue will require careful monitoring.

Footnotes

  • Grant support: Cancer Care Ontario Fellowship (K.W.L. Yee), American Cancer Society grant 1 RSG-06-054-01-LIB (M. Konopleva), and NIH grant P01 CA55164 (M. Andreeff).

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • Note: Presented in part at the 46th Annual Meeting of the American Society of Hematology, November 16, 2004, San Diego, California.

    • Accepted June 20, 2006.
    • Received March 28, 2006.
    • Revision received May 12, 2006.

References

  1. ↵
    Panwalkar A, Verstovsek S, Giles FJ. Mammalian target of rapamycin inhibition as therapy for hematologic malignancies. Cancer 2004;100:657–66.
    OpenUrlCrossRefPubMed
  2. ↵
    Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy. Nat Rev Cancer 2004;4:335–48.
    OpenUrlCrossRefPubMed
  3. ↵
    Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer 2003;3:721–32.
    OpenUrlCrossRefPubMed
  4. ↵
    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;349:847–58.
    OpenUrlCrossRefPubMed
  5. ↵
    Lorber MI, Mulgaonkar S, Butt KM, et al. Everolimus versus mycophenolate mofetil in the prevention of rejection in de novo renal transplant recipients: a 3-year randomized, multicenter, phase III study. Transplantation 2005;80:244–52.
    OpenUrlCrossRefPubMed
  6. ↵
    Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet 2004;43:83–95.
    OpenUrlCrossRefPubMed
  7. ↵
    Kauffman HM, Cherikh WS, Cheng Y, Hanto DW, Kahan BD. Maintenance immunosuppression with target-of-rapamycin inhibitors is associated with a reduced incidence of de novo malignancies. Transplantation 2005;80:883–9.
    OpenUrlCrossRefPubMed
  8. ↵
    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.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    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.
    OpenUrlCrossRefPubMed
  10. ↵
    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.
    OpenUrlCrossRefPubMed
  11. ↵
    Law M, Forrester E, Chytil A, et al. Rapamycin disrupts cyclin/cyclin-dependent kinase/p21/proliferating cell nuclear antigen complexes and cyclin D1 reverses rapamycin action by stabilizing these complexes. Cancer Res 2006;66:1070–80.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Jundt F, Raetzel N, Muller C, et al. A rapamycin derivative (everolimus) controls proliferation through down-regulation of truncated CCAAT enhancer binding protein β and NF-κB activity in Hodgkin and anaplastic large cell lymphomas. Blood 2005;106:1801–7.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Boehm A, Aichberger KJ, Mayerhofer Mea. Targeting of mTOR in AML is associated with decreased growth or leukemic cells and downregulation of VEGF [abstract 2544]. Blood 2004;104:697a.
    OpenUrl
  14. ↵
    Lerut E, Roskams T, Goossens E, et al. Molecular pharmacodynamic evaluation of dose and schedule of RAD001 (everolimus) in patients with operable prostate carcinoma [abstract 3071]. Proc Am Soc Clin Oncol Annu Meet 2005;23:209s.
    OpenUrl
  15. ↵
    Tabernero J, Rojo F, Burris H, et al. A phase I study with tumor molecular pharmacodynamic evaluation of dose and schedule of the oral mTOR-inhibitor Everolimus (RAD001) in patients with advanced solid tumors [abstract 3007]. Proc Am Soc Clin Oncol Annu Meet 2005;23:193s.
    OpenUrl
  16. ↵
    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, and pharmacodynamic endpoints in patients with solid tumours [abstract 803]. Proc Am Soc Clin Oncol Annu Meet 2003;22:200.
  17. ↵
    Lane H, Tanaka C, Kovarik J, et al. Preclinical and clinical pharmacokinetic/pharmacodynamic modeling to help define an optimal biological dose for the oral mTOR inhibitor, RAD001, in oncology [abstract 951]. Proc Am Soc Clin Oncol Annu Meet 2003;22:237.
    OpenUrl
  18. ↵
    Tefferi A, Barosi G, Mesa RA, et al. International Working Group (IWG) consensus criteria for treatment response in myelofibrosis with myeloid metaplasia: on behalf of the IWG for myelofibrosis research and treatment (IWG-MRT). Blood 2006.
  19. Cheson BD, Bennett JM, Grever M, et al. National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 1996;87:4990–7.
    OpenUrlFREE Full Text
  20. Cheson BD, Bennett JM, Kantarjian H, et al. Report of an international working group to standardize response criteria for myelodysplastic syndromes. Blood 2000;96:3671–4.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Cheson BD, Cassileth PA, Head DR, et al. Report of the National Cancer Institute-sponsored workshop on definitions of diagnosis and response in acute myeloid leukemia. J Clin Oncol 1990;8:813–9.
    OpenUrlAbstract
  22. ↵
    Jiang YP, Ballou LM, Lin RZ. Rapamycin-insensitive regulation of 4e-BP1 in regenerating rat liver. J Biol Chem 2001;276:10943–51.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Hay N. The Akt-mTOR tango and its relevance to cancer. Cancer Cell 2005;8:179–83.
    OpenUrlCrossRefPubMed
  24. ↵
    Raval RR, Lau KW, Tran MG, et al. Contrasting properties of hypoxia-inducible factor 1 (HIF-1) and HIF-2 in von Hippel-Lindau-associated renal cell carcinoma. Mol Cell Biol 2005;25:5675–86.
    OpenUrlAbstract/FREE Full Text
  25. Gera JF, Mellinghoff IK, Shi Y, et al. AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem 2004;279:2737–46.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Hashemolhosseini S, Nagamine Y, Morley SJ, Desrivieres S, Mercep L, Ferrari S. Rapamycin inhibition of the G1 to S transition is mediated by effects on cyclin D1 mRNA and protein stability. J Biol Chem 1998;273:14424–9.
    OpenUrlAbstract/FREE Full Text
  27. ↵
    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.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Platzbecker U, Haase M, Herbst R, et al. Activity of sirolimus in patients with myelodysplastic syndrome—results of a pilot study. Br J Haematol 2005;128:625–30.
    OpenUrlCrossRefPubMed
  29. ↵
    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;23:5347–56.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    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;22:2336–47.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Oymak FS, Demirbas HM, Mavili E, et al. Bronchiolitis obliterans organizing pneumonia. Clinical and roentgenological features in 26 cases. Respiration 2005;72:254–62.
    OpenUrlCrossRefPubMed
  32. ↵
    Mokhtari M, Bach PB, Tietjen PA, Stover DE. Bronchiolitis obliterans organizing pneumonia in cancer: a case series. Respir Med 2002;96:280–6.
    OpenUrlCrossRefPubMed
  33. ↵
    Morelon E, Stern M, Israel-Biet D, et al. Characteristics of sirolimus-associated interstitial pneumonitis in renal transplant patients. Transplantation 2001;72:787–90.
    OpenUrlCrossRefPubMed
  34. ↵
    Maasilta PK, Salminen US, Lautenschlager IT, Taskinen EI, Harjula AL. Immune cells and immunosuppression in a porcine bronchial model of obliterative bronchiolitis. Transplantation 2001;72:998–1005.
    OpenUrlPubMed
  35. ↵
    Groetzner J, Wittwer T, Kaczmarek I, et al. Conversion to sirolimus and mycophenolate can attenuate the progression of bronchiolitis obliterans syndrome and improves renal function after lung transplantation. Transplantation 2006;81:355–60.
    OpenUrlCrossRefPubMed
  36. ↵
    Yee KW, Hymes SR, Heller L, Prieto VG, Welch MA, Giles FJ. Cutaneous leukocytoclastic vasculitis in a patient with myelodysplatic syndrome after therapy with the rapamycin analogue everolimus: case report and review of the literature. Leuk Lymphoma 2006;47:926–9.
    OpenUrlCrossRefPubMed
  37. ↵
    Recher C, Beyne-Rauzy O, Demur C, et al. Antileukemic activity of rapamycin in acute myeloid leukemia. Blood 2005;105:2527–34.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Yee KWL, Garcia-Manero G, Thomas D, et al. A phase II study of temsirolimus (CCI-779) in patients with advanced leukemias [abstract 4523]. Blood 2004;104:214b.
    OpenUrl
  39. ↵
    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 [abstract 2980]. Blood 2005;106:835a.
    OpenUrl
  40. ↵
    Dirks NL, Huth B, Yates CR, Meibohm B. Pharmacokinetics of immunosuppressants: a perspective on ethnic differences. Int J Clin Pharmacol Ther 2004;42:701–18.
    OpenUrlPubMed
  41. ↵
    Naegele S, Morley SJ. Molecular cross-talk between MEK1/2 and mTOR signaling during recovery of 293 cells from hypertonic stress. J Biol Chem 2004;279:46023–34.
    OpenUrlAbstract/FREE Full Text
  42. Xu G, Zhang W, Bertram P, Zheng XF, McLeod H. Pharmacogenomic profiling of the PI3K/PTEN-AKT-mTOR pathway in common human tumors. Int J Oncol 2004;24:893–900.
    OpenUrlPubMed
  43. ↵
    Hammerman PS, Fox CJ, Birnbaum MJ, Thompson CB. Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival. Blood 2005;105:4477–83.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Ricciardi MR, McQueen T, Chism D, et al. Quantitative single cell determination of ERK phosphorylation and regulation in relapsed and refractory primary acute myeloid leukemia. Leukemia 2005;19:1543–9.
    OpenUrlCrossRefPubMed
  45. ↵
    Milella M, Precupanu CM, Gregorj C, et al. Beyond single pathway inhibition: MEK inhibitors as a platform for the development of pharmacological combinations with synergistic anti-leukemic effects. Curr Pharm Des 2005;11:2779–95.
    OpenUrlCrossRefPubMed
  46. ↵
    O'Reilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res 2006;66:1500–8.
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Sun SY, Rosenberg LM, Wang X, et al. Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res 2005;65:7052–8.
    OpenUrlAbstract/FREE Full Text
  48. ↵
    Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307:1098–101.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    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;14:1296–302.
    OpenUrlCrossRefPubMed
  50. ↵
    Edinger AL, Linardic CM, Chiang GG, Thompson CB, Abraham RT. Differential effects of rapamycin on mammalian target of rapamycin signaling functions in mammalian cells. Cancer Res 2003;63:8451–60.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Clinical Cancer Research: 12 (17)
September 2006
Volume 12, Issue 17
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Phase I/II Study of the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients with Relapsed or Refractory Hematologic Malignancies
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Phase I/II Study of the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients with Relapsed or Refractory Hematologic Malignancies
Karen W.L. Yee, Zhihong Zeng, Marina Konopleva, Srdan Verstovsek, Farhad Ravandi, Alessandra Ferrajoli, Deborah Thomas, William Wierda, Efrosyni Apostolidou, Maher Albitar, Susan O'Brien, Michael Andreeff and Francis J. Giles
Clin Cancer Res September 1 2006 (12) (17) 5165-5173; DOI: 10.1158/1078-0432.CCR-06-0764

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Phase I/II Study of the Mammalian Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients with Relapsed or Refractory Hematologic Malignancies
Karen W.L. Yee, Zhihong Zeng, Marina Konopleva, Srdan Verstovsek, Farhad Ravandi, Alessandra Ferrajoli, Deborah Thomas, William Wierda, Efrosyni Apostolidou, Maher Albitar, Susan O'Brien, Michael Andreeff and Francis J. Giles
Clin Cancer Res September 1 2006 (12) (17) 5165-5173; DOI: 10.1158/1078-0432.CCR-06-0764
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Biomarker Analysis from the BERIL-1 Study
  • Radiation and TGFβ Blockade in Metastatic Breast Cancer
  • Novel Intermediate Endpoint in Immunotherapy Studies
Show more Cancer Therapy: Clinical
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement