throbber
2257
`
`Prognostic Relevance of the mTOR Pathway
`in Renal Cell Carcinoma
`ImplicationsforMolecularPatientSelectionforTargetedTherapy
`
`1
`Allan J. Pantuck, MD
`2
`David B. Seligson, MD
`1
`Tobias Klatte, MD
`2
`Hong Yu, MD, PhD
`1
`John T. Leppert, MD
`Laurence Moore, MD, PhD
`3
`Timothy O’Toole, PhD
`4
`Jay Gibbons, PhD
`Arie S. Belldegrun, MD
`1,5
`Robert A. Figlin, MD
`
`1
`
`3
`
`1 Department of Urology, David Geffen School of
`Medicine, University of California, Los Angeles,
`California.
`
`2 Department of Pathology and Laboratory Medi-
`cine, David Geffen School of Medicine, University
`of California, Los Angeles, California.
`
`3 Wyeth Research, Cambridge, Massachusetts.
`
`4 Wyeth Research, Pearl River, New York.
`
`5 Department of Medicine, David Geffen School
`of Medicine, University of California, Los Angeles,
`California.
`
`BACKGROUND. The mammalian target of rapamycin (mTOR) pathway is up-regu-
`lated in many human cancers, and agents targeting the mTOR pathway are in
`
`various stages of clinical development. The goal of the study was to evaluate the
`
`potential and limitations of targeting the mTOR pathway in renal cell carcinoma
`
`(RCC).
`METHODS. Immunohistochemical analysis using antibodies against pAkt, PTEN,
`p27, and pS6 was performed on a tissue microarray constructed from paraffin-
`
`embedded specimens from 375 patients treated by nephrectomy for RCC. The
`
`expression was associated with pathological parameters and survival.
`RESULTS. The mTOR pathway was more significantly altered in clear-cell RCC,
`high-grade tumors, and tumors with poor prognostic features. PS6 and PTEN
`
`showed the strongest associations with pathological parameters. Survival tree
`
`analysis regarding expression of cytoplasmic pAkt, nuclear pAkt, PTEN, cytoplas-
`
`mic p27, and pS6 identified staining percentages of 40%, 10%, 75%, 7%, and 70%,
`
`respectively, as ideal cutoff values for stratification, with corresponding P-values
`of .03, .001, .02, .005, and <.0001, respectively. Interestingly, high nuclear pAkt
`expression was associated with a favorable prognosis, whereas high cytoplasmic
`
`pAkt expression was associated with a poor prognosis. In multivariate Cox regres-
`
`sion analysis, ECOG PS, T classification, N classification, M classification, cyto-
`
`plasmic Akt, nuclear pAkt, PTEN, and pS6 were independent prognostic factors
`
`of DSS.
`CONCLUSIONS. Components of the mTOR pathway are significantly associated
`with pathological features and survival. Not all RCC tumor types seem to be
`
`equally amenable to mTOR targeted therapy. PTEN, pAkt, p27, and pS6 may
`
`serve as surrogate parameters for patient selection and predicting prognosis.
`
`Patients with a highly activated mTOR pathway should benefit most from this
`recommended. Cancer
`
`results
`our
`validation of
`therapy. External
`2007;109:2257–67. Ó 2007 American Cancer Society.
`
`is
`
`Supported in part by Wyeth Research and The
`Richard
`and Nancy
`Bloch
`Kidney
`Cancer
`Research Fund.
`
`The first and second authors contributed equally
`to this article.
`
`Address for reprints: Robert A. Figlin, MD, Divi-
`sion of Medical Oncology and Experimental Ther-
`apeutics, City of Hope National Medical Center
`and Beckman Research Institute, 1500 East
`Duarte Rd., Duarte, CA 91010; Fax:
`(310) 267-
`1491; E-mail: rfiglin@coh.org
`
`revision received
`Received January 9, 2007;
`February 5, 2007; accepted February 8, 2007.
`
`KEYWORDS: mTOR, S6, Akt, PTEN, p27, survival.
`
`W ith over 38,000 new cases, representing an annual increase of
`
`2% to 3%, and over 12,000 cancer-related deaths in 2006 in the
`US,1,2 renal cell carcinoma (RCC) represents a major therapeutic
`challenge. Only a small number of patients with metastatic RCC can
`be cured by existing therapies. Approximately 20% to 30% of patients
`present with metastatic disease and an additional 20% to 40% de-
`velop recurrence after undergoing curative surgery for localized
`RCC.3,4 Advances in biological and immune-based therapies have
`produced response rates for patients with metastatic RCC of approxi-
`mately 15% to 30%, with some long-term durable remissions. Recent
`
`ª 2007 American Cancer Society
`DOI 10.1002/cncr.22677
`Published online 17 April 2007 in Wiley InterScience (www.interscience.wiley.com).
`
`

`

`2258
`
`CANCER June 1, 2007 / Volume 109 / Number 11
`
`advances in understanding the changes associated
`with von Hippel-Lindau (VHL) gene inactivation have
`led to several angiogenesis inhibitors (sunitinib, sora-
`fenib, bevacizumab) demonstrating enhanced re-
`sponse,
`improvement
`in progression-free survival,
`and trends toward improvement in overall survival for
`some of these agents administered in both the first-
`and second-line setting.5–11 A comparison of sunitinb
`to interferon-alpha (IFN) in a recent phase 3 trial
`demonstrating improvement in progression-free sur-
`vival has changed the paradigm for treatment of this
`disease.11 Despite these advances, most responses are
`partial in nature, with the majority of patients ulti-
`mately succumbing to their disease.
`Significant achievements in the basic sciences
`have led to a greater knowledge of the underlying sig-
`naling pathways in RCC,12 including the mammalian
`target of rapamycin (mTOR) pathway (phosphoinosi-
`tide 3-kinase/Akt pathway) (Fig. 1). The mTOR path-
`way has a central role in the regulation of cell growth
`and increasing evidence suggests its dysregulation in
`cancer.13 Receiving input
`from multiple signals,
`including growth factors, hormones, nutrients, and
`other stimulants or mitogens, the pathway stimulates
`protein synthesis by phosphorylating key translation
`regulators such as ribosomal S6 kinase. The mTOR
`pathway also contributes to many other critical cellu-
`lar
`functions,
`including protein degradation and
`angiogenesis. Hence, use of inhibitors of the pathway
`represents a new strategy for the targeted treatment of
`RCC.
`Temsirolimus (CCI-779) is an inhibitor of mTOR
`and, in a phase 2 trial, showed antitumor activity in
`heavily pretreated patients with advanced RCC.14 In a
`recent randomized phase 3 trial of patients with poor
`prognosis, previously untreated, metastatic RCC, tem-
`sirolimus demonstrated a statistically significant 49%
`improvement in overall survival when compared with
`IFN.15
`With mTOR inhibitors, it is unclear what clinical
`parameters and/or molecular pathways will predict
`which patients will derive the greatest benefit. These
`agents might have clinical activity only in selected
`patient cohorts in whose diseases this pathway drives
`their biology. An enhanced ability to predict patient
`survival would allow patients most likely to benefit
`from mTOR targeting therapies to be selected. For
`patient selection, a wide spectrum of molecular bio-
`markers is currently available including upstream and
`downstream targets of mTOR. Hence, the goals of our
`study were 1) to evaluate the prognostic relevance of
`the mTOR pathway in RCC in a large patient cohort,
`and 2) to identify patients whose tumor biology would
`most likely benefit from mTOR targeting therapy. For
`
`FIGURE 1. PI3K-Akt-mTOR pathway. Activation of the pathway leads to
`phosphorylation of S6 kinase and 4E-BP, activating the former and inactivat-
`ing the latter. S6 kinase and 4E-BP are critical components of the general
`translation machinery. Cell cycle regulator p27 is also regulated through
`mTOR. The pathway is negatively regulated by PTEN and rapamycin analogs
`like temsirolimus.
`
`these goals, we carried out an immunohistochemical
`study of the mTOR upstream and downstream targets
`phosphorylated Akt (pAkt), phosphorylated S6 ribo-
`somal protein (pS6), and p27, as well as the tumor
`suppressor PTEN (phosphatase and tensin homolog
`deleted on chromosome 10), and correlated our find-
`ings with pathological parameters and survival.
`
`MATERIALS AND METHODS
`Patients
`Our study cohort consisted of 375 patients who
`underwent radical or partial nephrectomy for spo-
`radic RCC at the University of California, Los Angeles
`(UCLA) between 1989 and 2000. After approval by
`the UCLA Institutional Review Board, a retrospective
`study was performed with outcome assessment
`based on chart review of clinical and pathological
`data. Clinical data included age, gender, and Eastern
`Cooperative Oncology Group performance status
`(ECOG PS). Pathological data included TNM staging
`and histologic subtyping, which was performed
`according to the 1997 Union Internationale Contre le
`Cancer (UICC) and American Joint Committee on
`Cancer (AJCC) classification, as well as Fuhrman
`grade. Localized RCC was defined as N0M0 RCC,
`whereas metastatic RCC was defined if
`regional
`
`

`

`lymph node metastasis and/or distant metastasis
`were present.
`
`TABLE 1
`Patient and Tumor Characteristics
`
`mTOR Pathway in RCC/Pantuck et al.
`
`2259
`
`Tissue Microarray Construction
`Formalin-fixed, paraffin-embedded tumor specimens
`from our patient cohort were obtained from the
`Department of Pathology. Three core tissue biopsies,
`0.6 mm in diameter, were taken from selected mor-
`phologically representative regions of each paraffin-
`embedded RCC and precisely arrayed using a custom-
`built instrument as described previously.16 Additional
`core tissue biopsies were taken from morphologically
`benign-appearing surrounding renal parenchyma tis-
`sue for each tumor. Sections of the resulting tumor tis-
`sue microarray block, 4 lm thick, were transferred to
`glass slides using the paraffin sectioning aid system
`(adhesive coated slides PSA-CS4x, adhesive tape, UV
`lamp, Instrumedics, Hackensack, NJ) to support the
`cohesion of 0.6 mm array elements.
`
`Immunohistochemical Staining and Evaluation
`Immunohistochemical staining was performed with a
`Dako Envision (Dako, Carpinteria, Calif) or Vectastain
`Elite ABC (Vector, Burlingame, Calif) staining system,
`as described previously.17,18 Rabbit monoclonal anti-
`body phospho-Akt Ser473 (Cell Signaling, Danvers,
`Mass) at a concentration of 1.5 lg/mL was used to
`stain for pAkt. Immunostaining for PTEN was per-
`formed using rabbit polyclonal antibody PN37 (Zymed,
`San Francisco, Calif) at 2 lg/mL. Mouse monoclonal
`antibody SX53G8 (Dako) was used at a concentration
`of 8 lg/mL to stain for p27. Staining for pS6 was per-
`formed with polyclonal rabbit antibody phospho-S6
`ribosomal protein Ser 235/236 (Cell Signaling) at a
`concentration of 0.125 lg/mL.
`The expression was evaluated by an anatomical
`pathologist (D.B.S.) in a blinded fashion to validate
`the diagnostic morphology of each array spot. The
`evaluation of expression involved site (subcellular lo-
`calization) and degree of reactivity (staining intensity:
`0 ¼ negative, 1 ¼ weak, 2 ¼ moderate, 3 ¼ strong,
`and staining frequency: percentage of positive cells).
`The overall score used for subsequent statistical
`analysis was the pooled mean from the 3 spots of
`the same tumor.
`
`Statistical Analysis
`The associations between protein expression and T
`classification, Fuhrman grade, metastatic status, and
`histologic subtype were evaluated using the nonpara-
`metric Mann-Whitney U-test (when 2 independent
`groups were compared) or the Kruskal-Wallis test
`(when more than 2 independent groups were com-
`pared). Correlations were determined using the Pear-
`
`ECOG PS
`0
`1
`2
`X
`Tumor size
`Median
`Range
`T classification
`T1
`T2
`T3
`T4
`N classification
`N0
`N1
`N2
`M classification
`M0
`M1
`Fuhrman grade
`G1
`G2
`G3
`G4
`Histological subtype
`Clear cell
`Papillary
`Chromophobe
`Collecting duct
`
`No.
`
`149
`207
`16
`3
`
`6.5
`1–18
`
`140
`49
`165
`21
`
`323
`23
`29
`
`216
`159
`
`49
`187
`126
`13
`
`323
`40
`8
`4
`
`%
`
`39.7
`55.2
`4.3
`0.8
`
`—
`—
`
`37.3
`13.1
`44.0
`5.6
`
`86.1
`6.1
`7.7
`
`57.6
`42.4
`
`13.1
`49.9
`33.6
`3.5
`
`86.1
`10.7
`2.1
`1.1
`
`ECOG PS indicates Eastern Cooperative Oncology Group performance status.
`
`son coefficient. The primary endpoint was disease-
`specific survival
`time (DSS). The Kaplan-Meier
`method was used to generate the survival functions.
`To find appropriate cutoffs for classifying patients
`according to the amount of expression, we used the
`recursive partitioning function in the R software
`(www.r-project.org).19 Subsequently,
`the dichoto-
`mized variable was used in univariate and multivari-
`ate survival analysis. Univariate survival analysis was
`performed by using the log-rank test and univariate
`Cox regression analysis. Independent prognostic vari-
`ables of survival were identified with a multivariate
`Cox regression analysis. A significance level of .05
`was used for all statistical tests. The statistical pack-
`age SPSS (Chicago, Ill) was used for the analysis.
`
`RESULTS
`Our study cohort consisted of 375 patients, 252 men
`and 123 women with a median age of 61 years
`(range, 27–88 years). Characteristics of the patients
`are summarized in Table 1.
`
`

`

`2260
`
`CANCER June 1, 2007 / Volume 109 / Number 11
`
`FIGURE 2. Immunohistochemical pattern of pAkt, PTEN, p27, and pS6 in (A) normal kidney tissue, (B) low-grade clear-cell renal cell carcinoma (RCC), and
`(C) high-grade clear-cell RCC.
`
`Expression and Association With Pathological
`Variables (Figs. 2, 3)
`Because staining intensity and frequency showed
`high intercorrelation (cytoplasmic pAkt: R ¼ 0.88,
`nuclear pAkt: R ¼ 0.90, cytoplasmic p27: R ¼ 0.84,
`nuclear p27: R ¼ 0.91, PTEN: R ¼ 0.61, pS6: R ¼ 0.94,
`each P < .0001), we restricted subsequent analyses to
`staining frequency.
`
`pAkt
`Anti-pAkt staining was seen in both cytoplasmic and
`nuclear cellular staining compartments. Cytoplasmic
`staining was detected in 93% of the RCCs. Higher
`cytoplasmic expression was noted in clear-cell than
`in nonclear-cell RCC, whereas no significant associa-
`tions were found with other variables. Nuclear stain-
`ing was positive in 61% of
`the tumors. Higher
`nuclear expression was observed in patients with
`localized disease. A significantly inverse correlation
`was found with tumor size (R ¼ 0.13, P ¼ .01).
`
`PTEN
`The anti-PTEN antibody stained the tissues of the
`array in the cytoplasmic cellular compartment in
`96% of the tumors. Tumors showed a lower expres-
`sion than normal renal tissues. PTEN expression was
`higher in tumors with lower T classification (T1/2),
`nonclear-cell subtypes, and in localized RCC.
`
`p27
`Cellular staining with anti-p27 antibody occurred in
`both nuclear and cytoplasmic compartments in 78%
`and 46% of the tumors, respectively. Nuclear expres-
`sion was higher
`in clear-cell RCC and inversely
`correlated with tumor size (R ¼ 0.12, P ¼ .02). Cyto-
`plasmic expression was higher in metastatic RCC.
`
`pS6
`Anti-pS6 staining was only seen in the cytoplasmic
`cellular compartment. Staining in tumors was noted
`in 85% of the RCCs, where it was generally increased
`
`

`

`mTOR Pathway in RCC/Pantuck et al.
`
`2261
`
`FIGURE 3. Relation of T classification, Fuhrman grade, localized/metastatic disease, and histologic type and mean expression frequency. The Kruskal-Wallis
`test was used to compare expression among T classification and Fuhrman grade. Mann-Whitney U-test was applied to compare expression between localized
`and metastatic renal cell carcinoma (RCC), clear-cell vs nonclear-cell RCC, and T1/2 vs T3/4 (PTEN).
`
`compared with matched normal tissue. Significantly
`higher expressions were observed in tumors with
`higher T classification, higher Fuhrman grades,
`in
`metastatic disease, and clear-cell subtype.
`
`Correlation Between Biomarkers
`The mean expression of nuclear and cytoplasmic
`pAkt (R ¼ 0.25, P < .001) and nuclear and cytoplas-
`mic p27 (R ¼ 0.14, P ¼ .01) were significantly corre-
`lated with each other. In addition, cytoplasmic pAkt
`expression was significantly correlated with nuclear
`p27 (R ¼ 0.32, P < .001), and pS6 (R ¼ 0.18,
`P ¼ .001). Nuclear pAkt expression was significantly
`correlated with nuclear p27 (R ¼ 0.38, P < .001),
`PTEN (R ¼ 0.13, P ¼ .02),
`(P ¼ .18,
`and pS6
`P ¼ .001). PTEN expression was further correlated
`with nuclear p27 expression (R ¼ 0.12, P ¼ .04).
`
`Survival Analysis
`The median follow-up time was 56.9 months (range,
`0.2–141.8) for the censored patients and 14.1 months
`(range, 0.4–115.0) for patients who died from RCC.
`Performing survival tree analysis with regard to
`expression of cytoplasmic pAkt, nuclear pAkt, PTEN,
`cytoplasmic p27, and pS6 for all patients, we identified
`staining percentages of 40%, 10%, 75%, 7%, and 70%,
`
`respectively, as ideal cutoff values for further patient
`stratification. The corresponding P-values
`for
`the
`dichotomized patient cohort, calculated with the log-
`.005, and <.0001,
`rank test, were .034,
`.001,
`.021,
`respectively (Fig. 4). Notably, nuclear p27 expression
`was not associated with DSS. High nuclear pAkt
`expression was associated with favorable prognosis,
`whereas high cytoplasmic pAkt
`expression was
`associated with poor prognosis.
`In univariate Cox
`regression analysis, ECOG PS, T classification, N classi-
`fication, M classification, Fuhrman grade, expression
`of cytoplasmic and nuclear pAkt, PTEN, cytoplasmic
`p27, and pS6 were all predictors of DSS. In multivari-
`ate Cox regression analysis, ECOG PS, T classification,
`N classification, M classification, Fuhrman grade, cyto-
`plasmic and nuclear pAkt, PTEN, and pS6 were inde-
`pendent prognostic factors (Table 2).
`We carried out
`further
`subanalysis dividing
`patients into localized and metastatic RCC at initial
`presentation. For patients with localized RCC
`(N0M0), pS6, nuclear p27, and nuclear pAkt expres-
`sion provided additional prognostic information to
`ECOG PS, T classification, and Fuhrman grade
`(Fig. 5A). Using a cutoff value of 65%, lower staining
`of pS6 predicted a better prognosis than a higher
`staining (P ¼ .0001). Higher nuclear p27 expression
`
`

`

`2262
`
`CANCER June 1, 2007 / Volume 109 / Number 11
`
`FIGURE 4. Kaplan-Meier survival estimates according to protein expression for all patients.
`
`(cutoff 35%) also predicted a more favorable out-
`come (P ¼ .039). Furthermore, higher nuclear pAkt
`expression (cutoff 5%) was associated with longer
`survival (P ¼ .027). Expression of PTEN (cutoff 70%)
`also seemed to be associated with outcome, but this
`difference did not
`reach statistical
`significance
`(P ¼ .079). Univariate Cox regression analysis showed
`prognostic significance for ECOG PS, T classification,
`Fuhrman grade, and expression of nuclear p27, nu-
`clear pAkt, and pS6. In multivariate Cox regression
`analysis, ECOG PS, T classification, and expression
`of pS6 were independent prognostic factors of DSS
`(Table 3A).
`In metastatic patients, expression of cytoplasmic
`pAkt, PTEN, and pS6 provided additional prognostic
`information (Fig. 5B). For pS6 (cutoff 70%),
`lower
`staining predicted a better survival than higher stain-
`ing, median survival 20.2 months (3.3, SE) vs 13.6
`months (1.8, SE), respectively (P ¼ .002). In addi-
`tion, higher staining of PTEN (cutoff 35%) predicted
`a median survival of 16.0 months ( 3.3, SE) vs 8.0
`(1.0, SE)
`months
`for
`lower PTEN expression
`
`(P ¼ .041). Lower cytoplasmic pAkt expression (cutoff
`35%) also predicted a more favorable prognosis, with
`a median survival of 25.5 months (4.6, SE) vs 15.1
`months (1.5, SE) for higher expression (P ¼ .045). In
`contrast, DSS of the metastatic patients was not sig-
`nificantly influenced by expression of p27 and nu-
`clear pAkt. Univariate Cox
`regression analysis
`showed prognostic significance for ECOG PS, T clas-
`sification, N classification, Fuhrman grade, expres-
`sion of cytoplasmic pAkt, PTEN, and pS6.
`In
`multivariate Cox regression analysis, T classification,
`N classification, and pS6 were independent prognos-
`tic factors of DSS (Table 3B).
`
`DISCUSSION
`Herein, we report a tissue-microarray based immu-
`nohistochemical analysis of the mTOR pathway in
`RCC. We evaluated pAkt, PTEN, p27, and pS6 and
`associated the expression of each molecular marker
`with relevant pathological parameters and survival.
`
`

`

`TABLE 2
`Multivariate Cox Regression Analysis for All Patients
`
`Factor
`
`ECOG PS
`
`T classification
`
`N classification
`
`M classification
`
`Fuhrman grade
`
`p27 cytoplasmic
`
`pAkt cytoplasmic
`
`pAkt nuclear
`
`PTEN
`
`pS6
`
`Category
`
`Hazard ratio [95% CI]
`
`0
`1
`T1
`T2
`T3
`T4
`N0
`N 1
`M0
`M1
`G1
`G2
`G3
`G4
`<7%
`7%
`<40%
`40%
`<10%
`10%
`<75%
`75%
`<70%
`70%
`
`1
`2.14 [1.31, 3.49]
`1
`0.99 [0.49, 1.99]
`2.18 [1.28, 3.71]
`3.86 [1.70, 8.78]
`1
`2.14 [1.33, 3.42]
`1
`2.60 [1.67, 4.04]
`1
`3.65 [1.12, 11.94]
`4.08 [1.23, 13.55]
`2.84 [0.65, 12.45]
`1
`1.00 [0.69, 1.45]
`1
`1.69 [1.12, 2.54]
`1
`0.60 [0.41, 0.89]
`1
`0.67 [0.47, 0.95]
`1
`1.61 [1.11, 2.32]
`
`P
`
`.002
`
`.972
`.004
`.001
`
`.002
`
`<.001
`
`.032
`.022
`.167
`
`.997
`
`.012
`
`.011
`
`.024
`
`.011
`
`ECOG PS indicates Eastern Cooperative Oncology Group performance status; 95% CI, 95% confi-
`dence interval.
`
`The ribosomal protein S6 reflects S6 kinase activ-
`ity,20 and the phosphorylated S6 protein effects its
`downstream targets, altering mRNA translation.21 In
`our study, pS6 expression significantly increased with
`T classification and Fuhrman grade, and was signifi-
`cantly higher in metastatic patients and in clear-cell
`RCC. Further, pS6 was the strongest predictor of sur-
`vival both in localized and metastatic RCC. This is of
`particular
`importance, because predicting which
`patients with localized disease will experience recur-
`rence is difficult.22 Our recent studies indicate that
`p21 and p53 are able to identify groups who are at
`greater risk of recurrence.23,24 In the current study,
`we identified pS6 and T classification as the strong-
`est independent prognostic factors of DSS in loca-
`lized RCC. Further, pS6 might be an important tool
`to stratify metastatic patients into different
`risk
`groups and to improve patient selection for mTOR
`targeted therapy.
`Taking the findings on pS6 together, we hypothe-
`size that patients with metastatic clear-cell RCC
`would benefit most
`from mTOR-targeted therapy
`because the pathway seems to be strongly activated
`in these patients. This hypothesis is further sup-
`ported by a study of Cho et al.25 In that study, tissue
`
`mTOR Pathway in RCC/Pantuck et al.
`
`2263
`
`specimens were obtained from 24 patients with met-
`astatic RCC participating in a study with the mTOR
`inhibitor temsirolimus. Using a staining index that
`combines the frequency and intensity of staining,
`they demonstrated that high expression of S6 kinase
`was associated with response to temsirolimus. Fur-
`ther shown in a recent study on patients with re-
`current glioblastoma multiforme, higher levels of
`phosphorylated p70S6 kinase in baseline tumor sam-
`ples appeared to predict a patient population more
`likely to benefit from treatment with temsirolimus.26
`However, objective response rates are not always pre-
`dictive for patient survival.27 Additionally, pS6 can
`also be phosphorylated in a mitogen-activated pro-
`tein kinase (MAPK)-dependent manner, presumably
`through p90 Rsk.28 This suggests a pathway for S6
`phosphorylation that is independent of mTOR activa-
`tion. In RCC with high activation of pS6 independent
`of the mTOR pathway, pS6 may then be less reliable
`as a surrogate marker for mTOR activation and
`response to mTOR inhibitors; however, the prognos-
`tic information provided by pS6 would be the same.
`PTEN is a tumor suppressor protein that
`is
`encoded by the tumor suppressor gene PTEN. It has
`been shown that PTEN loss occurs during carcino-
`genesis and is associated with adverse prognosis in
`RCC.29,30 In addition, Neshat et al.20 were able to
`demonstrate that PTEN-deficient
`tumors have an
`increased sensitivity to temsirolimus. Correlating to
`pathological parameters, we observed a significantly
`higher expression in RCC with lower T classification
`and localized disease. Furthermore, PTEN expression
`was significantly lower
`in clear-cell RCC. Taking
`these facts together, mTOR inhibitors would most
`likely benefit patients with metastatic clear-cell RCC
`and low PTEN expression.
`The subcellular localization of p27 and pAkt is of
`importance for pathobiology. Many studies have
`shown that low nuclear staining of p27 is associated
`with poor prognosis.31–34 However, little knowledge
`exists regarding cytoplasmic mislocalization of p27.
`We were able to show that cytoplasmic mislocaliza-
`tion and consequently higher cytoplasmic expression
`is also a poor prognostic finding. This has been
`shown in other cancers,35,36 but, to our knowledge,
`never before in RCC. The underlying mechanisms
`are poorly understood. It is speculated that pAkt
`induced phosphorylation mainly contributes to the
`retention.37,38 Because
`the
`growth-
`cytoplasmic
`restraining activity of p27 depends on its nuclear
`localization, the aberrant localization may impair its
`tumor suppressor functions.39,40 Thus,
`the mTOR
`pathway is increasingly activated in tumors with
`cytoplasmic mislocalized p27. In our study, cytoplasmic
`
`

`

`2264
`
`CANCER June 1, 2007 / Volume 109 / Number 11
`
`FIGURE 5. Kaplan-Meier survival estimates according to protein expression for patients (A) with localized renal cell carcinoma (RCC) and (B) with metastatic RCC.
`
`p27 expression was associated with metastatic RCC
`but not with histologic type. Therefore, we speculate
`that the cytoplasmic mislocalization of p27 occurs in
`metastatic patients irrespective of
`their histologic
`type.
`Further, we could show that the subcellular loca-
`lization of pAkt has a significant impact on pathology
`and survival. Akt, also called protein kinase B, regu-
`lates both growth and survival mechanisms by phos-
`phorylating a wide spectrum of substrates in the
`cytoplasm and the nucleus. Mainly, Akt is stimulated
`by a second messenger generated from phosphoino-
`sitide 3-kinase (PI3K), phosphatidylinosital (3,4,5)-
`trisphosphate.41 Akt can mediate phosphorylation of
`mTOR,42–44 a process that occurs through inactiva-
`tion of the tuberous sclerosis complex (TSC). The
`TSC complex is a heterodimer consisting of unphos-
`phorylated TSC2 (tuberin) and TSC1 (hamartin). Akt,
`by phosphorylating TSC2, disrupts the TSC complex,
`allowing Rheb (Ras homolog enriched in brain) to
`bind ATP and convert from the inactive GDP state to
`the active GTP state. GTP-bound Rheb in turn acti-
`vates mTOR.45,46 Our study showed a higher pAkt
`
`expression in the cytoplasm than in the nucleus. Fur-
`ther, nuclear pAkt expression was significantly higher
`in localized than in metastatic RCC. Consequently,
`patients with localized RCC might have less activa-
`tion of the mTOR pathway and therefore less aggres-
`sive tumors and better survival. Possibly, the recently
`described anti-oncogenic role of pAkt41 is more de-
`pendent on nuclear localization. In our study, cyto-
`plasmic as well as nuclear pAkt expressions were
`independent prognostic factors of DSS. Further,
`when performing univariate analysis classifying the
`patients in localized and metastatic RCC, higher nu-
`clear pAkt expression was associated with favorable
`prognosis in localized patients, whereas higher cyto-
`plasmic pAkt expression was a poor prognostic find-
`ing in metastatic patients.
`In conclusion,
`the
`localization of pAkt seems to be highly relevant in
`both localized and metastatic RCC.
`At present, there are at least 3 mTOR inhibitors
`that have been widely characterized in preclinical
`models and are in clinical development as anticancer
`agents: temsirolimus, AP23573, and RAD001 (Fig. 1),
`esters of rapamycin with improved bioavailability
`
`

`

`TABLE 3
`Multivariate Cox Regression Analysis for Patients (A) With Localized
`RCC and (B) With Metastatic RCC
`
`Category
`
`Hazard ratio [95% CI]
`
`P
`
`(A) With localized RCC factor
`ECOG PS
`
`T classification
`
`Fuhrman grade
`
`p27 nuclear
`
`pAkt nuclear
`
`pS6
`
`(B) With Metastatic RCC Factor
`ECOG PS
`
`T classification
`
`N classification
`
`Fuhrman grade
`
`pAkt cytoplasmic
`
`PTEN
`
`pS6
`
`0
`1
`T1-2
`T3-4
`G1-G2
`G3-G4
`<35%
`35%
`<5%
`5%
`<65%
` 65%
`
`0
`1
`T1-T2
`T3-T4
`N0
`N 1
`G1-G2
`G3-G4
`<35%
`35%
`<35%
`35%
`<70%
`70%
`
`1
`2.87[1.37, 6.00]
`1
`3.52 [1.72, 7.20]
`1
`1.69 [0.79, 3.58]
`1
`0.36 [0.12, 1.10]
`1
`0.66 [0.31, 1.38]
`1
`3.14 [1.50, 6.58
`
`1
`1.46 [0.80, 2.63]
`1
`1.82 [1.15, 2.90]
`1
`1.75 [1.15, 2.68]
`1
`1.14 [0.75, 1.74]
`1
`1.31 [0.83, 2.04]
`1
`0.74 [0.43, 1.28]
`1
`1.55 [1.02, 2.35]
`
`.005
`
`.001
`
`.175
`
`.072
`
`.268
`
`.002
`
`.215
`
`.011
`
`.010
`
`.532
`
`.245
`
`.285
`
`.042
`
`RCC indicates renal cell carcinoma; 95% CI, 95% confidence interval; ECOG PS, Eastern Cooperative
`Oncology Group performance status.
`
`and formulation. Rapamycin and its derivatives first
`bind to FK506 binding protein 12 (FKBP12). The
`FKBP12/rapamycin complex then binds mTOR, indu-
`cing a G1 growth arrest rather than apoptosis. Com-
`pleted clinical
`trials show safety and efficacy of
`mTOR targeting therapy in patients with RCC. In a
`randomized phase 2 trial of the mTOR inhibitor tem-
`sirolimus, Atkins et al.14 observed an objective
`response rate (complete and partial responses) of 7%
`in patients with advanced RCC and a clinical benefit
`rate (complete, partial, and minor responses and
`stable disease for at least 24 weeks) of 51%. As noted
`above, mTOR inhibitors induce G1 arrest rather than
`apoptosis, which may explain why rates of disease
`stabilization were higher than objective response
`rates. More recently, Hudes et al.15 presented the
`results of a randomized phase 3 trial of temsirolimus
`comparing temsirolimus alone with IFN alone and
`both combined. All enrolled patients were selected
`with poor prognostic factors. This trial demonstrated
`a statistically significant 49% improvement in median
`
`mTOR Pathway in RCC/Pantuck et al.
`
`2265
`
`in advanced RCC patients treated
`overall survival
`with this mTOR inhibitor when compared with IFN
`in the first-line setting.
`this study need to be
`Some limitations of
`acknowledged. The nature of this study was retro-
`spective and therefore only limited conclusions can
`be drawn. The herein reported expressions, cutoffs,
`and their associations with survival have to be vali-
`dated independently by other groups, if possible in a
`prospective setting. Also, it is reasonable to evaluate
`the proteins of the mTOR pathway in conjunction
`with other well-known prognostic relevant
`factors
`such as p21,24 p53,23 and CAIX47 to identify the
`marker with the greatest prognostic accuracy.
`In conclusion, the mTOR pathway appears to be
`very important in patients with RCC. We were able to
`demonstrate highly significant associations with
`pathological parameters and, more important, with
`survival. Further, our analysis suggests that not all
`RCC tumor types are equally amenable to treatment
`strategies targeting the mTOR pathway, but that a
`majority of patients have at least 1 component of the
`mTOR pathway impacted. The results also suggest
`that the mTOR pathway is more significantly altered
`in clear-cell RCC, high-grade tumors, and tumors
`with poor prognostic features. Thus, these patients
`with a highly activated mTOR pathway may benefit
`most from therapy specifically targeting this pathway.
`However, patients with nonclear-cell
`tumors and
`highly activated pathway may also be candidates for
`targeted therapy. Of the different biomarkers, pS6
`showed the highest associations with pathology and
`survival; however, it should be studied in prospective
`trials as a predictive marker for response and survival
`to mTOR targeted therapy.
`
`2.
`
`REFERENCES
`1. Rini BI, Campbell SC, Rathmell WK. Renal cell carcinoma.
`Curr Opin Oncol. 2006;18:289–296.
`Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006. CA
`Cancer J Clin. 2006;56:106–130.
`3. Zisman A, Pantuck AJ, Wieder J, et al. Risk group assess-
`ment and clinical outcome algorithm to predict the natural
`history of patients with surgically resected renal cell carci-
`noma. J Clin Oncol. 2002;20:4559–4566.
`4. Zisman A, Pantuck AJ, Dorey F, et al. Mathematical model
`to predict individual survival for patients with renal cell
`carcinoma. J Clin Oncol. 2002;20:1368–1374.
`5. Motzer RJ, Rini BI, Bukowski RM, et al. Sunitinib in pa-
`tients with metastatic renal cell carcinoma. JAMA. 2006;295:
`2516–2524.
`6. Motzer RJ, Michaelson MD, Redman BG, et al. Activity of
`SU11248, a multitargeted inhibitor of vascular endothelial
`growth factor receptor and platelet-derived growth factor
`receptor, in patients with metastatic renal cell carcinoma.
`J Clin Oncol. 2006;24:16–24.
`
`

`

`2266
`
`CANCER June 1, 2007 / Volume 109 / Number 11
`
`8.
`
`7. Hainsworth JD, Sosman JA, Spigel DR, Edwards DL, Baugh-
`man C, Greco A. Treatment of metastatic renal cell carci-
`noma with a combination of bevacizumab and erlotinib.
`J Clin Oncol. 2005;23:7889–7896.
`Yang JC, Haworth L, Sherry RM, et al. A randomized trial of bev-
`acizumab, an anti-vascular endothelial growth factor antibody,
`for metastatic renal cancer. N Engl J Med. 2003;349:427–434.
`9. Ratain MJ, Eisen T, Stadler WM, et al. Phase II placebo-
`controlled randomized discontinuation trial of sorafenib in
`patients with metastatic renal cell carcinoma. J Clin Oncol.
`2006;24:2505–2512.
`10. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in
`advanced clear-cell renal-cell carcinoma. N Engl J Med.
`2007;356:125–134.
`11. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus
`interferon alfa in metastatic renal-cell carcinoma. N Engl
`J Med. 2007;356:115–124.
`12. Pantuck AJ, Zeng G, Belldegrun AS, Figlin RA. Pathobiology,
`prognosis, and targeted therapy for renal cell carcinoma:
`exploiting the h

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket