Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Oct 1.
Published in final edited form as: Gynecol Oncol. 2011 Jul 12;123(1):19–26. doi: 10.1016/j.ygyno.2011.06.022

Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a Gynecologic Oncology Group study

Kian Behbakht 1, Michael W Sill 2,3, Kathleen M Darcy 2, Stephen C Rubin 4, Robert S Mannel 5, Steven Waggoner 6, Russell J Schilder 7, Kathy Q Cai 7, Andrew K Godwin 8,*, R Katherine Alpaugh 8
PMCID: PMC3336961  NIHMSID: NIHMS366988  PMID: 21752435

Abstract

Purpose

Patients with persistent/recurrent epithelial ovarian cancer/primary peritoneal cancer (EOC/PPC) have limited treatment options. AKT and PI3K pathway activation is common in EOC/PPC, resulting in constitutive activation of downstream mTOR. The GOG conducted a phase II evaluation of efficacy and safety for the mTOR inhibitor, temsirolimus in EOC/PPC and explored circulating tumor cells (CTC) and AKT/mTOR/downstream tumor markers.

Methods

Eligible women with measurable, persistent/recurrent EOC/PPC who had received 1–3 prior regimens were treated with 25 mg weekly IV temsirolimus until progression or intolerable toxicity. Primary endpoints were progression-free survival (PFS) ≥6-months, tumor response, and toxicity. CellSearch® system was used to examine CTC, and AKT/mTOR/downstream markers were evaluated by archival tumor immunohistochemistry. Kendall’s tau-b correlation coefficient (r) and Cox regression modeling were used to explore marker associations with baseline characteristics and outcome.

Results

Sixty patients were enrolled in a two-stage sequential design. Of 54 eligible and evaluable patients, 24.1% (90%CI 14.9%–38.6%) had PFS ≥6 months (median 3.1 months), 9.3% (90%CI 3.7%–23.4%) experienced a partial response. Grade 3/4 adverse events included metabolic(8), gastrointestinal(8), pain(6), constitutional(5) and pulmonary(4). Suggested associations were between cyclin D1 and PFS ≥6 months, PFS or survival; positive CTC pre-treatment and lack of response; and high CTC expression of M30 and PFS ≥6 months/longer PFS.

Conclusions

Temsirolimus appears to have modest activity in persistent/recurrent EOC/PPC; however, PFS is just below that required to warrant inclusion in phase III studies in unselected patients. Cyclin D1 as a selection marker and CTC measures merit further study.

Keywords: Circulating tumor cells, ovarian clinical trial, AKT/PI3K pathway, mTOR inhibitor

INTRODUCTION

Ovarian cancer is the second most common gynecologic malignancy and the fifth leading cause of cancer death among American women1. Most women are diagnosed in advanced stages and undergo surgical debulking followed by platinum\taxane-based chemotherapy. Initial complete response to therapy can be achieved; however, relapse is common and it is almost universally fatal2,3.

The mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that is expressed in all mammalian cells4. mTOR is a central switch balancing input from growth factors such as receptor tyrosine kinases with input from nutrient availability sensors to coordinate a series of downstream genes such as the p70-S6 protein kinase (p70-S6K) and 4E-BP1 family of proteins4. For example, phosphorylation of p70-S6K leads to phosphorylation of the S6 ribosomal protein which induces protein synthesis at the ribosome, stimulating mTOR activity, and regulating cell growth, whereas phosphorylation of 4E-BP1 results in dissociation from the eukaryotic translation initiation factor 4E (eIF4E) and translation of cell growth control genes, including cyclin D1. In this way, constitutive action of the mTOR pathway via overexpression of its components or inactivating mutations of its inhibitors can lead to carcinogenesis via uncontrolled growth.

Recent studies, both in laboratory models and in clinical trials, have demonstrated the phosphatidylinositol-3-kinase(PI3K)/AKT/mTOR pathway is activated in cancers and its inhibition may be an effective strategy514. Approximately 40% of ovarian cancers have increased AKT2 activity5,7. Downstream of AKT, phosphorylated (p)mTOR and pGSK-3β correlate with AKT activation status in >80% of ovarian cancers, validating activation of the PI3K/AKT pathway7. Thus, pharmacologic agents that inhibit downstream targets of the PI3K/AKT signaling pathway such as mTOR might preferentially kill tumor cells having AKT dependence for survival not shared by normal cells8. Specific small molecule inhibitors including rapamycin or rapamycin analogues such as temsirolimus, everolimus (RAD001) and ridaforlimus (formerly deferolimus, AP23573) prevent mTOR activity by binding to the mTORC1 complex (mTOR and the FK506 binding protein-12) but not the mTORC2 complex15. Temsirolimus 25 mg weekly IV (flat dose) was selected for this trial due to a favorable pharmacokinetic profile, reduced immunosuppressive properties as compared to rapamycin16, and extensive experience in phase I and phase II trials1012,1718.

Study of the specific pathway effects of targeted agents in ovarian cancer is hampered by the lack of easy access to tissue for repeat sampling. Circulating tumor cells (CTC) can be detected in the blood of cancer patients using the CellSearch® system whereas they are absent in healthy controls19. Furthermore, the presence of CTC is an independent prognostic marker for several cancer types including breast2022, prostate23,24 and colorectal cancers2527. Although CTC have been enriched from the blood of ovarian cancer patients by flow cytometry28,29, the prognostic/predictive value of CTC enumeration in epithelial ovarian cancer/primary peritoneal cancers (EOC/PPC) has not been assessed. Additionally, CTC can be queried for the effect of targeted agents30.

In this phase II trial of the efficacy and safety of temsirolimus in persisent/recurrent EOC/PPC, we also explored the AKT/mTOR pathway and downstream targets including p70-S6K, 4E-BP1 and cyclin D1 in archival tumor, and also recovered and characterized CTC for enumeration and expression of potential biomarkers indicative of temsirolimus therapeutic effects, including pS6 ribosomal protein via immunofluorescent techniques30 and apoptosis using M30, a biomarker indicating cytokeratin degradation31.

MATERIALS AND METHODS

Patients

Women with recurrent/persistent EOC/PPC on review of the surgical pathology report and with RECIST measurable disease who had received 1–3 prior cytotoxic regimens were eligible. Patients must have had at least one prior platinum-based regimen and, if enrolled following primary treatment, must have had a platinum-free interval of <12 months. Cytostatic agents were allowed with primary treatment but not for recurrences. Patients must have had adequate bone marrow function with absolute neutrophil count ≥1,500/mcl, platelets ≥100,000/mcl as well as creatinine ≤1.5x institutional upper limit of normal (ULN), bilirubin ≤1.5xULN, SGOT and alkaline phosphatase ≤2.5xULN, neuropathy ≤ Common Criteria for Adverse Events (CTCAE) v3.0 grade 1, fasting cholesterol <350 mg/dl, and fasting triglycerides <400 mg/dl. Patients who had received radiation to more than 25% of marrow-bearing areas or a history of other invasive malignancies were excluded. This study was approved by the regulatory agencies and Institutional Review Boards at participating institutions. All patients provided written informed consent consistent with federal, state, and local requirements.

Treatment and Dose Modifications

Patients were treated by Gynecologic Oncology Group (GOG) member institutions with 25 mg weekly IV temsirolimus provided by the National Cancer Institute Cancer Therapy Evaluation Program (NCI-CTEP), in 28-day cycles until disease progression or toxicity. Subsequent cycles were delayed to a maximum of three weeks to allow bone marrow recovery and prophylactic granulocyte colony-stimulating factor (GCSF) was not allowed. Dose reductions to 15 mg IV (−1) and 10 mg IV (−2) were instituted with grade 4 thrombocytopenia. Treatment was discontinued if pneumonitis related to temsirolimus was suspected. Non-hematologic grade 2 or greater (peripheral neuropathy, renal and other) toxicities and grade 3 liver enzyme elevations required treatment delay for up to three weeks to allow recovery to ≤ grade 1 and one level dose reduction. Nausea, emesis, diarrhea, constipation, electrolyte abnormalities, hyperglycemia and hyperlipidemia were managed with supportive therapies without dose reduction.

Immunohistochemistry for AKT/ /mTOR/downstream biomarkers

Archival formalin-fixed and paraffin-embedded tumor were obtained from a previous surgery or biopsy and analyzed (see Supplemental Materials) by immunohistochemistry for pAKT (DAKO, clone 14-5, pS473), pmTOR (Cell Signaling, clone 49F9, pS2448), pp70-S6 (Cell Signaling, clone IA5, pT389), p4E-BP1 (Cell Signaling, clone 236B4, pT37/46), and cyclin D1 (DAKO, clone SP4) by blinded reviewers (KCQ, AKG).

Evaluation of Circulating Tumor Cells

CTC enrichment, enumeration and phenotypic characterization were performed (see Supplemental Materials) pre-cycle 1, 2 and 3 using the CellSearch® System (Veridex, Raritan NJ)32.

Statistical Considerations and End Points

Response was assessed by physical examination every 4 weeks and by radiologic studies every other cycle. Activity by either RECIST tumor response or progression-free survival ≥6 months (PFS at 6 months) was considered sufficient to declare the drug worthy of further investigation in a phase III trial. The probability of a type I error (i.e. alpha) was controlled with a bivariate 2-stage design33. The probability of early termination when the agent is inactive was approximately 50%, depending in part on the degree of association between response and PFS at 6 months.

The design targeted 23 patients for entry to the first stage but allowed deviation to account for non-eligible patients. The cumulative targeted sample size for the second stage was 52 patients but was allowed to deviate as before. Advancement to second stage occurred with rules as detailed in the results section. With 54 patients accrued, the design required more than 8 patients with tumor responses or more than 13 patients with PFS at 6 months before declaring the agent worthy of phase III investigation. The design had approximately 10% power when the regimen had πr ≤ 0.10 and πs ≤ 0.15 where πr and πs are the probabilities of a patient having a response and being PFS at 6 months respectively. The null probabilities were obtained from an analysis of historical controls3444. The design had approximately 90% power when πr = 0.25 or πs = 0.35, which were deemed to be minimally clinically significant values. The frequency/severity of adverse events were evaluated with CTCAEv3 and events deemed at least possibly related to the regimen were tabulated.

The correlations among the five tumor markers and three CTC measures, and associations between the eight biomarkers and six baseline characteristics (some data presented in Table S1 in Supplemental Materials) or two categorical measures of outcome were assessed with Kendall’s or Spearman’s correlation coefficient, Fisher’s Exact Test or an Exact Chi-Square test.4548 Kaplan-Meier method and Cox proportional hazards models were used to examine associations between the eight biomarkers and PFS and overall survival (OS)49,50. Suggested associations were assessed by any test with p<0.05 for the purpose of hypothesis generation and to prioritize further testing. The statistical power of these exploratory biomarker assessments was low due to small sample sizes.

RESULTS

Of the 60 patients enrolled, six were excluded due to improper prior treatment (n=3), inadequate tests (n=1), no measurable lesions (n=1) and patient refused all treatment (n=1) leaving 54 evaluable for efficacy and toxicity. Twenty-five patients were accrued during the first stage, and more than two responses or more than five PFS at 6 months were required to open to a second stage. First stage response/PFS criteria were met after interim analysis with three responses and seven PFS at 6 months, and the study completed second stage accrual. Patient characteristics are provided in Table 1. Median age for the group was 62 years and 90.7% were Caucasian. All patients had previous surgery, and the majority had recurrent serous ovarian cancer. Up to three courses of cytotoxic chemotherapy were allowed and 25.9% of patients had three courses of prior chemotherapy.

Table 1.

Characteristics of eligible and evaluable enrolled patients (n=54) as well as treatment cycles, response rates and follow up data.

Characteristic Category No %
Age 20–29
40–49
50–59
60–69
70–79
80–89
2
7
15
11
16
3
3.7
13.0
27.8
20.4
29.6
5.6
Race Unspecified
African American
Caucasian
1
4
49
1.9
7.4
90.7
Ethnicity Hispanic
Non-Hispanic
Unknown
0
41
13
0.0
75.9
24.1
Performance Status 0
1
37
17
68.5
31.5
Site of Disease Ovary
Peritoneum
46
8
85.2
14.8
Cell Type Adenocarcinoma, Unsp.
Clear Cell Carcinoma
Endometrioid Adenocarcinoma
Serous Adenocarcinoma
8
3
4
39
14.8
5.6
7.4
72.2
Grade 1: Well differentiated
2: Moderately differentiated
3: Poorly differentiated
2
11
41
3.7
20.4
75.9
Prior Chemotherapy 1
2
3
16
24
14
29.6
44.4
25.9
Prior Radiation No
Yes
50
4
92.6
7.4
Prior Surgery Yes 54 100.0
Response Partial response
Stable disease
Increase disease
Indeterminate
5
22
21
6
9.3
40.7
38.9
11.1
PFS > 6 Months No
Yes
Unknown (Pt. Lost)
40
13
1
74.1
24.1
1.9
Cycles of Treatment 1
2
3
4
5
6
9
10+
12
15
4
9
2
7
2
3
22.2
27.8
7.4
16.7
3.7
13.0
3.7
5.6
Alive Without progression
With progression
5
19
9.3
35.2
Dead From disease
From neither Rx nor disease
29
1
53.7
1.9

Proportion of patients with tumor responses was 9.3%. Approximate 90% marginal confidence interval (CI) was 3.7%, 23.4%. Proportion of patients with progression free survival (PFS) at 6 months was 24.1%. Approximate 90% marginal CI was 14.9%, 38.6%.

Distribution of completed cycles as well as response and follow-up data are reported in Table 1. Median number of cycles was 2.5 with 3 patients receiving 10 or more cycles (range 1–24 cycles). Dose reductions per protocol were necessary in 27/54 (50%) of patients. There were no dose re-escalations. Partial responses were seen in 5 of patients (9.3%; 90% CI 3.7%–23.4%). PFS at 6 months was also used as an efficacy endpoint and 13 (24.1%; 90% CI 14.9%–38.6%) achieved this outcome. Three patients had clear cell histology of which one had a partial response lasting 4 months. Kaplan-Meier Curves for PFS and OS are shown in Figure 1A. Median PFS and OS were 3.2 and 11.6 months respectively. The longest PFS was 21 months in a 53 year old with stage IIIC platinum-resistant ovarian cancer who received 24 courses at 25 mg/week with no dose reductions/delays. At 9.8 months of median follow-up, 44.4% of patients were alive, all with disease.

Figure 1.

Figure 1

Figure 1

Kaplan-Meier progression-free survival (PFS) and overall survival (OS) distributions for persistent/recurrent epithelial ovarian cancer or primary peritoneal cancer (A). Kaplan-Meier plots for progression-free survival (PFS)(B) and overall survival (OS)(C) by tumor expression of cyclin D1 categorized as negative or positive, and for progression-free survival by tumor expression of pAKT (D), pmTOR (E), pp70-S6K (F) categorized as negative or positive, or p4E-BP1 (G) categorized as low (<50% positive tumor cells) or positive (≥50% positive tumor cells) following temsirolimus treatment. Suggestive associations were assessed by logrank test with p<0.05.

Adverse events (AE) encountered on study were as expected for mTOR inhibitors and are detailed in Table 2. Notable grade 3 AE consisted of 4/54 constitutional (all fatigue), 6/54 gastrointestinal (2 mucositis, 2 dehydration and 2 bowel obstruction), and 8/54 metabolic (2 hypokalemia, 2 hypertriglyceridemia, 2 liver enzyme elevations, one hypophosphatemia and one hyperglycemia). One grade 3 renal failure was reported. Study drug was discontinued in 3 patients because of grade 3 pulmonary infiltrates thought to be related to study drug and to represent interstitial pneumonitis. The grade 4 constitutional AE consisted of asthenia and fatigue while the grade 4 vascular AE consisted of a pulmonary embolus.

Table 2.

Grade and number of patients experiencing Common Criteria or Adverse Events (CTCAE) v3 reported adverse events(n=54).

CTCAEv3 Grade*
1 2 3 4 5
Leukopenia 14 14 0 0 0
Thrombocytopenia 18 5 0 0 0
Neutropenia 6 12 1 0 0
Anemia 20 19 3 0 0
Other Hematologic 0 2 0 0 0
Allergy/Immunology 1 0 0 0 0
Auditory/Ear 1 0 0 0 0
Cardiac 3 1 1 0 0
Coagulation 2 1 0 0 0
Constitutional 23 15 4 1 0
Dermatologic 24 10 1 0 0
Nausea 19 2 2 0 0
Vomiting 2 3 1 0 0
Gastrointestinal 16 19 6 0 0
Genitourinary/Renal 4 0 1 0 0
Hemorrhage 5 0 0 0 0
Infection 0 5 2 0 0
Lymphatics 3 1 1 0 0
Metabolic 17 12 8 0 0
Musculoskeletal 2 0 1 0 0
Neurosensory 6 2 0 0 0
Other Neurological 5 1 0 0 0
Ocular/Visual 1 0 0 0 0
Pain 13 7 6 0 0
Pulmonary 17 5 4 0 0
Sexual/Reproductive 1 0 0 0 0
Vascular 0 0 0 1 0

Immunohistochemistry assays were performed for pAKTS473, pmTORS2448, pp70-S6KT389, p4E-BP1T37/46 and cyclin D1 in archival tumor to explore whether activation of the AKT/mTOR signaling pathway was associated with measures of clinical outcome following temsirolimus treatment (Table 3, Figure 1B–1G). Cyclin D1 expression was observed in 42% of the archival tumors and appeared to be correlated with PFS ≥6 months (r=0.281) but not tumor response (Table 3), and to be associated with a reduced risk of progression (Figure 1B; hazard ratio [HR]=0.495, 95% confidence interval [CI]=0.260–0.943) and longer OS (Figure 1C).

Table 3.

Relationship between pre-treatment biomarkers and tumor response or proportion experiencing progression-free survival (PFS) ≥6 months.

Biomarkers Tumor Response PFS ≥6 months

No Yes

ID+NE SD PR+CR No Yes
Tumor Expression

pAKT
  Negative 19 (51.4) 14 (37.8) 4 (10.8) 27 (75.0) 9 (25.0)
  Positive 5 (35.7) 8 (57.1) 1 (7.1) 11 (78.6) 3 (21.4)

pmTOR
  Negative 10 (58.8) 14 (41.2) 1 (5.9) 15 (88.2) 2 (11.8)
  Positive 14 (41.2) 16 (47.1) 4 (11.8) 23 (69.7) 10 (30.3)

pp70-S6K
  Negative 13 (44.8) 13 (44.8) 3 (10.3) 20 (69.0) 9 (31.0)
  Positive 11 (50.0) 9 (40.9) 2 (9.1) 18 (85.7) 3 (14.3)

p4E-BP1
  Low (≤ 50% +) 9 (36.0) 14 (56.0) 2 (8.0) 20 (80.0) 5 (20.0)
  High (> 50% +) 15 (57.7) 8 (30.8) 3 (11.5) 18 (72.0) 7 (28.0)

Cyclin D1 Suggested Association*
  Negative 15 (50.0) 13 (43.3) 2 (6.7) 25 (86.2) 4 (13.8)
  Positive 9 (42.9 9 (42.9) 3 (14.3) 13 (61.9) 8 (38.1)

CTC Count Suggested Association*
   Negative 7 (29.2) 14 (58.3) 3 (12.5) 17 (73.9) 6 (26.1)
   Positive 12 (63.2) 6 (32.6) 1 (5.2) 15 (78.9) 4 (21.1)

CTC Expression

M30 Suggested Association*
  Low (< 75% +) 10 (66.7) 4 (26.7) 1 (6.7) 14 (93.3) 1 (6.7)
  High (≥ 75% +) 2 (50.0) 2 (50.0) 0 (0.0) 1 (25.0) 3 (75.0)

pS6
  Low (< 75% +) 5 (50.0) 4(40.0) 1(10.0) 8 (80.0) 2 (20.0)
  High (≥ 75% +) 3 (42.9) 2 (28.6) 2 (28.6) 6 (85.7) 1 (14.3)

Number of cases (row percentages)

Immunohistochemistry assays were performed to examine expression of phosphorylated (p)AKTS273, pmTORS2448, pp70-S6KT389, p4E-BP1T37/46, and cyclin D1 in archival tumor. Circulating tumor cells (CTC) were enriched and characterized using the CellSearch® system (Veridex, Raitan NJ) for enumeration and expression of the apoptotic markers M30 and pS6.

Tumor response was categorized as no for increasing disease and not evaluable (ID+NE) plus stable disease (SD) versus yes for partial response and complete response (PR+CR).

*

Suggested Kendall’s tau-b correlations (r) were observed between cyclin D1 and PFS ≥ 6 months (r=0.281); and positive CTC and progressive disease (versus not) (r=0.340). The correlation between M30 and PFS ≥ 6 months was high (r=0.683). Fisher’s Exact Test suggested an association between M30 and PFS ≥ 6 months (odds ratio = 42; 90% CI 1.8 to 1150).

CTC were enriched in 19/43 (44%), 11/38 (29%) and 14/31 (45%) in pre-cycle1, 2 and 3 blood specimens with counts ranging from 1–11, 1–84, and 1–190, respectively. Positive CTC pre-cycle 1 appeared to be associated with increasing disease (r=0.340) but not with PFS ≥6 months (Table 3) or shorter PFS (Figure 2A). Positive CTCs persisted in 8 patients (4 increasing disease; 4 stable disease). High M30 and pS6 were both defined as ≥75% positive CTC. Of the cases with positive CTC counts pre-cycle1, high M30 (a marker of apoptosis) was observed in 10/19 with levels ranging from 17–100% and was suggestively correlated with PFS ≥6 months (r=0.683; Table 3). Figure 2B is consistent with the dichotomized PFS findings but the log-rank test was not suggestive. Of the cases with positive CTC counts pre-cycle1, high pS6 expression was observed in 12/17 patients but did not appear to be associated with any measure of clinical outcome (Table 3 and Figure 2C). Landmark analyses did not indicate that changes in CTC counts were associated with PFS (Figure 2D) or OS; however, the sample sizes were small, giving the tests low power. Paired pre-cycle1 and 2 assessments were available for four patients for M30 (all increasing disease) and three patients for pS6 (2 increasing disease; 1 stable disease), and it was not possible to explore the changes in M30 and pS6 with tumor response.

Figure 2.

Figure 2

Kaplan-Meier progression-free survival (PFS) distributions for pre-treatment circulating tumor cells (A), pre-treatment circulating tumor cell (CTC) expression of the apoptotic marker M30 (B) or pre-treatment CTC expression of pS6 (C), or change in CTC counts from pre-cycle 1 to pre-cycle 2 (D) following temsirolimus treatment. Circulating tumor cells (CTC) counts were categorized as negative or positive. M30 and pS6 were categorized as low (<75% positive CTC) or high (≥75% positive CTC). Serial changes in CTC were categorized as stable counts, decreasing counts or increasing counts. Suggestive associations were assessed by logrank test with p<0.05.

DISCUSSION

A variety of targeted agents have been tested in the phase II setting in ovarian cancer patients with multiple prior chemotherapy regimens51,52. Of these, only vascular endothelial growth factor (VEGF) inhibitors such as bevacizumab have been actively included in subsequent phase III trials of untreated patients. Bevacizumab was shown to have a 21% clinical response rate, including complete responses, and a 40% PFS at 6 months in a phase II single agent study with similar (albeit 2 prior chemotherapeutic agents as compared to 3 in this study) persistent/recurrent EOC/PPC patients as those reported here53. A PFS advantage was recently reported for carboplatin/paclitaxel with bevacizumab versus placebo from cycles 2 through 22 in a GOG phase III trial54. Meanwhile, the search continues for other biologic agents with single agent, additive or synergistic activity in EOC/PPC.

Results of other targeted agents in persistent/recurrent EOC/PPC including GOG phase II evaluations of single agent bortezomib55, trastuzumab56, gefitinib57, imatinib58, sorafenib59,60, and enzastaurin61 have not reached the level of interest seen with bevacizumab53,54,62. Given the preclinical activity, the paucity of activity of targeted agents in EOC/PPC other than those targeting VEGF is perplexing7. In this study, the 9.3% response rate and 24.1% PFS at 6 months of the mTOR inhibitor temsirolimus in EOC/PPC is second only to bevacizumab in the GOG Phase II series. However, this response is below and the PFS is just at the threshold for the response/PFS endpoint and is insufficient to warrant addition to upfront regimens in a phase III study. We recognize that the study population was unselected and subgroups may show greater benefit if selected by specific response markers. Additionally, such as seen in metastatic renal cell cancers treated with temsirolimus, an overall survival benefit may exist even without differences in response rates63. One purpose of translational research in the context of phase II studies may be to identify appropriate selection markers for targeted agents. In addition, since mTOR inhibitors can synergize with chemotherapy in ovarian cancer models13,14, phase II single agent activity may not be reflective of activity in combination with chemotherapy.

The current study suggested an association between cyclin D1 expression with high p4E-BP1 in tumors, a greater likelihood of being PFS ≥6 months, and overall longer PFS and survival following temsirolimus treatment. Several studies have shown that 4E-BP1 and cyclin D1 are downstream of mTOR activation6466, and that the anti-proliferative activity of mTOR inhibitors including temsirolimus are mediated by acceleration of cyclin D1 turnover, up-regulation of p27 expression, inhibition of cyclin-dependent kinase activation and retinoblastoma protein phosphorylation, as well as stabilization of 4E-BP1 binding to eIF4E preventing translation of genes stimulating cell growth including cyclin D16470. mTOR inhibitors also appear to exhibit anti-neoplastic activity via inhibition of translation of pro-angiogenic factors, pro-survival proteins, and proteases involved in tumor invasion and metastasis, and by disruption of metabolic processes that fuel tumor progression6470.

Temsirolimus was not active in >75% of persistent/recurrent EOC/PPC in this trial. Resistance to the targeted agent is a possible cause for the low response rate and proportion of women PFS at 6 months. For example, mTORC1 is sensitive whereas mTORC2 is resistant to rapamycin6470. Indeed, development of rapid resistance to mTORC1 inhibition (with rapamycin) has been shown in glioblastoma mutliforme via reflex upregulation of pAKTS473, an activating substrate of mTORC271. Second generation inhibitors of mTOR, referred to mTOR kinase domain (mTORK) inhibitors, are now available that target rapamycin-resistant pathways by inhibiting the ATP site of both mTORC1 and mTORC26870. mTORK inhibitors have yet to be evaluated in EOC/PPC. Additionally, there is redundancy in signaling pathways and one site of inhibition may be insufficient to block cellular processes driving tumor progression. Funding was not available for a comprehensive analysis of the mTOR pathway in this multi-institutional phase II trial of single agent temsirolimus72.

The informed development and evaluation of targeted agents would benefit from the availability of serially assayed non-invasive predictive biomarkers. CTC have been documented in breast cancers where their enumeration is prognostic2022,73, but have also been documented in colorectal cancers2527, prostate cancers2324, and other cancers7477 but not in healthy subjects or patients with non-malignant diseases78. CTC have been identified in ovarian cancers28,29, but their relation to prognosis has not been reported. In the current study, the CellSearch® system was employed due to the improved accuracy and sensitivity compared with flow cytometry methods19. At least one tumor cell was identified from blood in up to 45% of patients and a suggested associated was seen between pre-cyle1 CTC and progressive disease

Studies to detect molecular events in CTC are accumulating for many malignancies31,76,79. We examined a selective marker of apoptosis using M30 which recognizes a caspase-cleaved epitope of cytokeratin-1827 and pS62626 a substrate of p70-S6K. Suggested associations were observed pre-treatment between positive CTC and progressive disease, high M30 and PFS ≥6 months, and high pS6 with fewer prior regimens. In the absence of robust anti-tumor activity, it is not possible to definitively state that these associations are due to treatment and not disease predictors of prognosis. However, these exploratory findings support the continued development of CTC analysis and phenotypic characterization of CTC as translational endpoints in EOC/PPC. In the current study, serial CTC assessments did not appear to change notably following treatment; however, serial CTCs were not acquired in the majority of patients. Emphasis on the ability to phenotypically characterize CTC may increase enthusiasm for specimen submission. Additional CTC studies and further optimization and standardization of techniques (CellSearch® vs. other methods) are needed to define the clinical value of predictive/prognostic CTC assessments in EOC/PPC.

In conclusion, temsirolimus appears to have modest activity, but insufficient to justify further study in a phase III study in unselected EOC/PPC. Exploratory findings regarding cyclin D1 expression in tumors as a selection marker for mTOR inhibitor treatment and CTC measures in blood merit further study in this disease.

Research Highlights.

  • Single agent temsirolimus exhibited modest activity in unselected patients with persistent or recurrent epithelial ovarian/peritoneal cancer with 24% of women progression-free ≥6 months and 9% experiencing a partial response.

  • Positive tumor expression of cyclin D1 appeared to be associated with high p4E-BP1 in tumors, a greater likelihood of being PFS ≥6 months, longer PFS and longer survival following temsirolimus treatment.

  • Detectable circulating tumor cells pre-treatment appeared to be associated with lack of response to temsirolimus.

Supplementary Material

01

ACKNOWLEDGEMENTS

The authors thank Mr. Frank Zambito for excellent technical support, Anne Reardon for preparing this manuscript for publication, Sandra Dascomb for data management, and Meg Colahan for protocol administration. We also thank the GOG Publications Subcommittee for their critical review of and thoughtful suggestions for the manuscript.

The following GOG member institutions participated in this protocol: Abington Memorial Hospital; University of Colorado-Anschutz Cancer Pavilion; University of Pennsylvania Cancer Center; Indiana University Cancer Center; Rush University Medical Center; Cleveland Clinic Foundation; Cooper Hospital/University Medical Center; Columbus Cancer Council/Ohio State; University of Oklahoma; Case Western Reserve University; University of Wisconsin Hospital; Women and Infants Hospital; and Community Clinical Oncology Program.

This study was supported by National Cancer Institute grants to the Gynecologic Oncology Group (GOG) Administrative Office (CA 27469), the GOG Tissue Bank (CA 27469 and CA 11479), the GOG Statistical and Data Center (CA 37517), to AKG (CA 140323 and Ovarian Cancer Research Fund) and the Ovarian Cancer SPORE at FCCC/U Penn (P50 CA083638).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Temsirolimus was provided by the Cancer Therapy Evaluation Program of the National Cancer Institute (NCI-CTEP). The study involves original work which was presented in part at the 41st Annual Meeting of the Society of Gynecologic Oncologists, San Francisco, CA, March 13–17, 2010.

CONFLICT OF INTEREST

The authors wish to report that there are no conflicts of interest.

REFERENCES

  • 1.Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96. doi: 10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 2.Guppy AE, Nathan PD, Rustin GJ. Epithelial ovarian cancer: a review of current management. Clin Oncol (R Coll Radiol) 2005;17:399–341. doi: 10.1016/j.clon.2005.05.009. [DOI] [PubMed] [Google Scholar]
  • 3.Ozols RF. Systemic therapy for ovarian cancer: current status and new treatments. Semin Oncol. 2006;33:S3–S11. doi: 10.1053/j.seminoncol.2006.03.011. [DOI] [PubMed] [Google Scholar]
  • 4.Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature. 2006;441:424–430. doi: 10.1038/nature04869. [DOI] [PubMed] [Google Scholar]
  • 5.Sun M, Wang G, Paciga JE, et al. AKT1/PKBalpha kinase is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIH3T3 cells. Am J Pathol. 2001;159:431–437. doi: 10.1016/s0002-9440(10)61714-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Altomare DA, Tanno S, De Rienzo A, et al. Frequent activation of AKT2 kinase in human pancreatic carcinomas. J Cell Biochem. 2002;87:470–476. doi: 10.1002/jcb.10287. [DOI] [PubMed] [Google Scholar]
  • 7.Altomare DA, Wang HQ, Skele KL, et al. AKT and mTOR phosphorylation is frequently detected in ovarian cancer and can be targeted to disrupt ovarian tumor cell growth. Oncogene. 2004;23:5853–5857. doi: 10.1038/sj.onc.1207721. [DOI] [PubMed] [Google Scholar]
  • 8.Brognard J, Clark AS, Ni Y, et al. Akt/protein kinase B is constitutively active in non-small cell lung cancer cells and promotes cellular survival and resistance to chemotherapy and radiation. Cancer Res. 2001;61:3986–3997. [PubMed] [Google Scholar]
  • 9.Schmelzle T, Hall MN. TOR, a central controller of cell growth. Cell. 2000;103:253–262. doi: 10.1016/s0092-8674(00)00117-3. [DOI] [PubMed] [Google Scholar]
  • 10.Yu K, Toral-Barza L, Discafani C, et al. mTOR, a novel target in breast cancer: the effect of CCI-779, an mTOR inhibitor, in preclinical models of breast cancer. Endocr Relat Cancer. 2001;8:249–258. doi: 10.1677/erc.0.0080249. [DOI] [PubMed] [Google Scholar]
  • 11.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;22:909–918. doi: 10.1200/JCO.2004.08.185. [DOI] [PubMed] [Google Scholar]
  • 12.Dancey JE. Clinical development of mammalian target of rapamycin inhibitors. Hematol Oncol Clin North Am. 2002;16:1101–1114. doi: 10.1016/s0889-8588(02)00051-5. [DOI] [PubMed] [Google Scholar]
  • 13.Mabuchi S, Altomare DA, Poulikakos PI, et al. The mTOR inhibitor RAD001 inhibits the proliferation of p53-deficient human ovarian cancer cells, enhances the efficacy of cisplatin in vitro, and prolongs survival in an in vivo ovarian cancer model. Clin Cancer Res. 2007;13:4261–4270. doi: 10.1158/1078-0432.CCR-06-2770. [DOI] [PubMed] [Google Scholar]
  • 14.Mabuchi S, Kawase C, Altomare DA, et al. mTOR is a promising target both in cisplatin-sensitive and cisplatin-resistant clear cell carcinoma of the ovary. Clin Cancer Res. 2009;15:5404–5413. doi: 10.1158/1078-0432.CCR-09-0365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yuan R, Kay A, Berg WJ, et al. Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol. 2009;2:45. doi: 10.1186/1756-8722-2-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Alexandre J, Raymond E, Armand JP. Rapamycin and CCI-779. Bull Cancer. 1999;86:808–811. [PubMed] [Google Scholar]
  • 17.Hidalgo M, Buckner JC, Erlichman C, et al. A phase I and pharmacokinetic study of temsirolimus (CCI-779) administered intravenously daily for 5 days every 2 weeks to patients with advanced cancer. Clin Cancer Res. 2006;12:5755–5763. doi: 10.1158/1078-0432.CCR-06-0118. [DOI] [PubMed] [Google Scholar]
  • 18.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;22:2336–2347. doi: 10.1200/JCO.2004.08.116. [DOI] [PubMed] [Google Scholar]
  • 19.Balic M, Dandachi N, Hofmann G, et al. Comparison of two methods for enumerating circulating tumor cells in carcinoma patients. Cytometry B Clin Cytom. 2005;68:25–30. doi: 10.1002/cyto.b.20065. [DOI] [PubMed] [Google Scholar]
  • 20.Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791. doi: 10.1056/NEJMoa040766. [DOI] [PubMed] [Google Scholar]
  • 21.Dawood S, Cristofanilli M. Integrating circulating tumor cell assays into the management of breast cancer. Curr Treat Options Oncol. 2007;8:89–95. doi: 10.1007/s11864-007-0018-0. [DOI] [PubMed] [Google Scholar]
  • 22.Dawood S, Broglio K, Valero V, et al. Circulating tumor cells in metastatic breast cancer: from prognostic stratification to modification of the staging system? Cancer. 2008;113:2422–2430. doi: 10.1002/cncr.23852. [DOI] [PubMed] [Google Scholar]
  • 23.Goodman OB, Jr, Fink LM, Symanowski JT, et al. Circulating tumor cells in patients with castration-resistant prostate cancer baseline values and correlation with prognostic factors. Cancer Epidemiol Biomarkers Prev. 2009;18:1904–1913. doi: 10.1158/1055-9965.EPI-08-1173. [DOI] [PubMed] [Google Scholar]
  • 24.Danila DC, Heller G, Gignac GA, et al. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:7053–7058. doi: 10.1158/1078-0432.CCR-07-1506. [DOI] [PubMed] [Google Scholar]
  • 25.Cohen SJ, Alpaugh RK, Gross S, et al. Isolation and characterization of circulating tumor cells in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2006;6:125–132. doi: 10.3816/CCC.2006.n.029. [DOI] [PubMed] [Google Scholar]
  • 26.Cohen SJ, Punt CJ, Iannotti N, et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:3213–3221. doi: 10.1200/JCO.2007.15.8923. [DOI] [PubMed] [Google Scholar]
  • 27.Cohen SJ, Punt CJ, Iannotti N, et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann Oncol. 2009;20:1223–1229. doi: 10.1093/annonc/mdn786. [DOI] [PubMed] [Google Scholar]
  • 28.He W, Wang H, Hartmann LC, et al. In vivo quantitation of rare circulating tumor cells by multiphoton intravital flow cytometry. Proc Natl Acad Sci U S A. 2007;104:11760–11765. doi: 10.1073/pnas.0703875104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.He W, Kularatne SA, Kalli KR, et al. Quantitation of circulating tumor cells in blood samples from ovarian and prostate cancer patients using tumor-specific fluorescent ligands. Int J Cancer. 2008;123:1968–1973. doi: 10.1002/ijc.23717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shaffer DR, Leversha MA, Danila DC, et al. Circulating tumor cell analysis in patients with progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:2023–2029. doi: 10.1158/1078-0432.CCR-06-2701. [DOI] [PubMed] [Google Scholar]
  • 31.de Haas EC, di Pietro A, Simpson KL, et al. Clinical evaluation of M30 and M65 ELISA cell death assays as circulating biomarkers in a drug-sensitive tumor, testicular cancer. Neoplasia. 2008;10:1041–1048. doi: 10.1593/neo.08620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kagan M, Howard D, Bendele T, et al. A sample preparation and analysis system for identification of circulating tumor cells. J Clin Ligand Assay. 2002;25:104–110. [Google Scholar]
  • 33.Sill MW, Yothers G. A method for utilizing bivariate efficacy outcome measures to screen agents for activity in 2-stage phase II clinical trials. State University of New York at Buffalo Department of Biostatistics Technical Report 2006–2008. 2008 [Google Scholar]
  • 34.Manetta A, Blessing JA, Hurteau JA. Evaluation of cisplatin and cyclosporin A in recurrent platinum-resistant ovarian cancer: a phase II study of the Gynecologic Oncology Group. Gynecol Oncol. 1998;68:45–46. doi: 10.1006/gyno.1997.4887. [DOI] [PubMed] [Google Scholar]
  • 35.Markman M, Blessing JA, Moore D, et al. Altretamine (hexamethylmelamine) in platinum-resistant and platinum-refractory ovarian cancer: a Gynecologic Oncology Group phase II trial. Gynecol Oncol. 1998;69:226–229. doi: 10.1006/gyno.1998.5016. [DOI] [PubMed] [Google Scholar]
  • 36.Plaxe SC, Blessing JA, Morgan MA, et al. Phase II trial of pyrazoloacridine in recurrent platinum-resistant ovarian cancer: a Gynecologic Oncology Group study. Am J Clin Oncol. 2002;25:45–47. doi: 10.1097/00000421-200202000-00009. [DOI] [PubMed] [Google Scholar]
  • 37.Fracasso PM, Brady MF, Moore DH, et al. Phase II study of paclitaxel and valspodar (PSC 833) in refractory ovarian carcinoma: a Gynecologic Oncology Group study. J Clin Oncol. 2001;19:2975–2982. doi: 10.1200/JCO.2001.19.12.2975. [DOI] [PubMed] [Google Scholar]
  • 38.Hoffman MA, Blessing JA, Morgan M. Phase II trial of CI-958 in recurrent platinum-refractory ovarian carcinoma. A Gynecologic Oncology Group Study. Gynecol Oncol. 2000;79:463–465. doi: 10.1006/gyno.2000.5984. [DOI] [PubMed] [Google Scholar]
  • 39.Markman M, Blessing JA, DeGeest K, et al. Lack of efficacy of 24-h infusional topotecan in platinum-refractory ovarian cancer: A Gynecologic Oncology Group trial. Gynecol Oncol. 1999;75:444–446. doi: 10.1006/gyno.1999.5640. [DOI] [PubMed] [Google Scholar]
  • 40.Fracasso PM, Blessing JA, Morgan MA, et al. Phase II study of oxaliplatin in platinum-resistant and refractory ovarian cancer: a gynecologic group study. J Clin Oncol. 2003;21:2856–2859. doi: 10.1200/JCO.2003.03.077. [DOI] [PubMed] [Google Scholar]
  • 41.Muggia FM, Blessing JA, Homesley HD, et al. Tomudex (ZD1694, NSC 639186) in platinum-pretreated recurrent epithelial ovarian cancer: a phase II study by the Gynecologic Oncology Group. Cancer Chemother Pharmacol. 1998;42:68–70. doi: 10.1007/s002800050786. [DOI] [PubMed] [Google Scholar]
  • 42.Hoffman MA, Blessing JA, Nunez ER. A phase II trial of CI-958 in recurrent platinum-sensitive ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2001;81:433–435. doi: 10.1006/gyno.2001.6182. [DOI] [PubMed] [Google Scholar]
  • 43.Markman M, Blessing JA, Alvarez RD, et al. Phase II evaluation of 24-h continuous infusion topotecan in recurrent, potentially platinum-sensitive ovarian cancer: A Gynecologic Oncology Group study. Gynecol Oncol. 2000;77:112–115. doi: 10.1006/gyno.2000.5755. [DOI] [PubMed] [Google Scholar]
  • 44.Hoffman MA, Blessing JA, Lentz SS. A phase II trial of dolastatin-10 in recurrent platinum-sensitive ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2003;89:95–98. doi: 10.1016/s0090-8258(03)00007-6. [DOI] [PubMed] [Google Scholar]
  • 45.Kendall MG. The treatment of ties in rank problems. Biometrika. 1945;33:239–251. doi: 10.1093/biomet/33.3.239. [DOI] [PubMed] [Google Scholar]
  • 46.Spearman C. The proof and measurement of association between two things. Amer J Psychol. 1904;15:72–101. [PubMed] [Google Scholar]
  • 47.Fisher RA. Statistical methods for research workers. Edinburgh, Oliver and Boyd, 1934. 1970 [Google Scholar]
  • 48.Mehta CR, Patel NR. A netwrok algorithm for performing Fisher's Exact test in r x c contingency tables. J Amer Stat Assn. 1983;78:427–434. [Google Scholar]
  • 49.Kaplan EL, Meier P. Nonparemetric estimation from incomplete observations. Am J Statis Assn. 1958;53:457–481. [Google Scholar]
  • 50.Cox DR. Regression models and life tables. J Royal Stat Soc. 1972;Series B 34:187–220. [Google Scholar]
  • 51.Han ES, Lin P, Wakabayashi M. Current status on biologic therapies in the treatment of epithelial ovarian cancer. Curr Treat Options Oncol. 2009;10:54–66. doi: 10.1007/s11864-009-0100-x. [DOI] [PubMed] [Google Scholar]
  • 52.Markman M. Antiangiogenic drugs in ovarian cancer. Expert Opin Pharmacother. 2009;10:2269–2277. doi: 10.1517/14656560903120907. [DOI] [PubMed] [Google Scholar]
  • 53.Burger RA, Sill MW, Monk BJ, et al. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. J Clin Oncol. 2007;25:5165–5171. doi: 10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
  • 54.Burger RA, Brady MF, Bookman MA, et al. Phase III trial of bevacizumab (BEV) in the primary treatment of advanced epithelial ovarian cancer (EOC), primary peritoneal cancer (PPC) or fallopian tube cancer (FTC): A Gynecologic Oncology Group study. J Clin Oncol. 2010;28:7s. doi: 10.1016/j.ygyno.2013.07.100. (Supplemental ASCO Abst #LBA1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aghajanian C, Blessing JA, Darcy KM, et al. A phase II evaluation of bortezomib in the treatment of recurrent platinum-sensitive ovarian or primary peritoneal cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2009;115:215–220. doi: 10.1016/j.ygyno.2009.07.023. [DOI] [PubMed] [Google Scholar]
  • 56.Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody (Trastuzumab) in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J.Clin.Oncol. 2003;21(2):283–290. doi: 10.1200/JCO.2003.10.104. [DOI] [PubMed] [Google Scholar]
  • 57.Schilder R, Sill M, Chen X, et al. Phase II study of gefitinib in patients with relapsed or persistent ovarian or primary peritoneal carcinoma and evaluation of epidermal growth factor receptor mutations and immunohistochemical expression: A Gynecologic Oncology Group study. Clin Cancer Res. 2005;11:5539–5548. doi: 10.1158/1078-0432.CCR-05-0462. [DOI] [PubMed] [Google Scholar]
  • 58.Schilder RJ, Sill MW, Lee RB, et al. Phase II evaluation of imatinib methylate in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. J Clin Oncol. 2008;26:3418–3425. doi: 10.1200/JCO.2007.14.3420. [DOI] [PubMed] [Google Scholar]
  • 59.Matei D, Sill MW, DeGeest K, Bristow RE. Phase II trial of sorafenib in persistent or recurrent epithelial ovarian cancer (EOC) or primary peritoneal cancer (PPC): a Gynecologic Oncology Group (GOG) study. J Clin Oncol. 2008;26:15S. 301s (ASCO Abst#5537) [Google Scholar]
  • 60.Matei D, Sill MW, Lankes HA, et al. Activity of sorafenib in recurrent ovarian cancer and primary peritoneal carcinomatosis: a Gynecologic Oncology Group trial. J Clin Oncol. 2011;29:69–75. doi: 10.1200/JCO.2009.26.7856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Usha L, Sill M, Darcy K, Benbrook D, Hurteau J, Michelin D, Mannel R, Hanjani P, De Geest K, Godwin A. A Gynecologic Oncology Group phase II trial of the protein kinase C-beta inhibitor, enzastaurin and evaluation of markers with potential predictive and prognostic value in persistent or recurrent epithelial ovarian and primary peritoneal malignancies. Gynecol Oncol. 2010;116:S16. doi: 10.1016/j.ygyno.2011.02.013. (SGO Abst#37) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol. 2007;25:5180–5186. doi: 10.1200/JCO.2007.12.0782. [DOI] [PubMed] [Google Scholar]
  • 63.Pal SK, Figlin RA. Treatment Options in Metastatic Renal Cell Carcinoma: Focus on mTOR inhibitors. Clinical Medicine Insights: Clin Med Insights Oncol. 2010;4:43–53. doi: 10.4137/cmo.s1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes & Develop. 2004;18:1926–1945. doi: 10.1101/gad.1212704. [DOI] [PubMed] [Google Scholar]
  • 65.Frost P, Moatamed F, Hoang B. In vivo antitumor effects of the mTOR inhibitor CCI-779 against human multiple myeloma cells ina xenograft model. Blood. 2004;104:4181–4187. doi: 10.1182/blood-2004-03-1153. [DOI] [PubMed] [Google Scholar]
  • 66.Giles FJ, Albitar M. Mammalian target of rapamycin as a therapeutic target in leukemia. Curr Mol Med. 2005;5:653–661. doi: 10.2174/156652405774641034. [DOI] [PubMed] [Google Scholar]
  • 67.Gibbons JJ, Abraham RT, Yu K. Mammalian target of rapamycin: discovery of rapamycin reveals a signaling pathway important for normal and cancer cell growth. Semin Oncol. 2009;(Suppl 3):S3–S17. doi: 10.1053/j.seminoncol.2009.10.011. [DOI] [PubMed] [Google Scholar]
  • 68.Feldman ME, Apsel B, Uotila A, et al. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 2009;7:e38. doi: 10.1371/journal.pbio.1000038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yu K, Toral-Barza L, Shi C. Z. Biochemical, cellular, and in vivo activity of novel ATP-competitive and selective inhibitors of the mammalian target of rapamycin. Cancer Res. 2009;69:6232–6240. doi: 10.1158/0008-5472.CAN-09-0299. [DOI] [PubMed] [Google Scholar]
  • 70.Targeting mTOR globally in cancer: thinking beyond rapamycin. Cell Cycle. 2009;8:3831–3837. doi: 10.4161/cc.8.23.10070. [DOI] [PubMed] [Google Scholar]
  • 71.Gulati N, Karsy M, Albert L, et al. Involvement of mTORC1 and mTORC2 in regulation of glioblastoma multiforme growth and motility. Int J Oncol. 2009;35:731–740. doi: 10.3892/ijo_00000386. [DOI] [PubMed] [Google Scholar]
  • 72.Frost P, Shi Y, Hoang B, Gera J, Lichtenstein A. Regulation of D-cyclin translation inhibition in myeloma cells treated with mammalian target of rapamycin inhibitors: rationale for combined treatment with extracellular signal-regulated kinase inhibitors and rapamycin. Mol Cancer Ther. 2009;8:83–93. doi: 10.1158/1535-7163.MCT-08-0254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bauernhofer T, Zenahlik S, Hofmann G, et al. Association of disease progression and poor overall survival with detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer. Oncol Rep. 2005;13:179–184. [PubMed] [Google Scholar]
  • 74.Nezos A, Pissimisis N, Lembessis P, et al. Detection of circulating tumor cells in bladder cancer patients. Cancer Treat Rev. 2009;35:272–279. doi: 10.1016/j.ctrv.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 75.Nezos A, Lembessis P, Sourla A, et al. Molecular markers detecting circulating melanoma cells by reverse transcription polymerase chain reaction: methodological pitfalls and clinical relevance. Clin Chem Lab Med. 2009;47:1–11. doi: 10.1515/CCLM.2009.009. [DOI] [PubMed] [Google Scholar]
  • 76.Maheswaran S, Sequist LV, Nagrath S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med. 2008;359:366–377. doi: 10.1056/NEJMoa0800668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Blumke K, Bilkenroth U, Schmidt U, et al. Detection of circulating tumor cells from renal carcinoma patients: experiences of a two-center study. Oncol Rep. 2005;14:895–899. [PubMed] [Google Scholar]
  • 78.Allard WJ, Matera J, Miller MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res. 2004;10:6897–6904. doi: 10.1158/1078-0432.CCR-04-0378. [DOI] [PubMed] [Google Scholar]
  • 79.Hayes DF, Walker TM, Singh B, et al. Monitoring expression of HER-2 on circulating epithelial cells in patients with advanced breast cancer. Int J Oncol. 2002;21:1111–1117. doi: 10.3892/ijo.21.5.1111. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES