Abstract
Introduction
While serine-threonine kinases (STK) are attractive therapeutic targets in epithelial ovarian cancer, clinical outcomes of STK inhibitors in the management of recurrent disease have not been completely described.
Areas covered
A systematic literature review of published clinical studies on STK inhibitors targeting mTOR, MAPK, and aurora kinase pathways in recurrent epithelial ovarian cancer was conducted, revealing 18 clinical trials (497 patients). Pooled analyses were performed to assess treatment response, survival time, and adverse events. Median progression-free survival was 3.4 months in STK inhibitor-based therapy, and the average response rate and clinical benefit rate were 13% and 67%, respectively. Among regimens comprised of only STK inhibitors (11 trials, 299 patients), median progression-free time was 2.7 months, response rate was 10%, and clinical benefit rate was 64%. Compared to single STK inhibitor monotherapy (52.5%), clinical benefit rates significantly improved when STK inhibitors were combined with a cytotoxic agent (71.4%), other class biological agent (74.2%), or an additional STK inhibitor (95.0%) (all, P ≤ 0.002).
Expert opinion
STK inhibitor-based therapy showed modest activity for recurrent epithelial ovarian cancer with reasonable clinical benefit rates, suggesting its potential utility for maintaining disease stability if supported by future studies. Efficacy appears greatly improved in appropriately selected patient populations, especially those with low-grade serous ovarian carcinoma, platinum-sensitive disease, cancers with somatic RAS or BRAF mutations, and when used in a combination regimen with a cytotoxic or biological agent.
Keywords: Aurora kinase inhibitor, MAPK inhibitor, MEK inhibitor, metformin, mTOR inhibitor, ovarian cancer, sorafenib, STK inhibitor
1. Introduction
Ovarian cancer ranks as the fifth most common cause of cancer death among women and the leading cause of gynecologic cancer mortality in the United States.[1] Standard treatment consists of cytoreductive surgery followed by platinum-based combination chemotherapy with carboplatin and paclitaxel. Though the majority of patients respond to initial therapy, cancer recurs in up to 70% of patients, and obtaining a cure is challenging.[2] While platinum compounds are often given to patients with platinum-sensitive recurrences, no standard treatment exists for platinum-resistant ovarian cancer, which often displays concomitant resistance to other cytotoxic agents. A meta-analysis of 40 clinical trials found an average overall response rate of only 16% among patients with platinum-resistant recurrences receiving any type of chemotherapy,[3] highlighting the need to uncover therapies that improve outcomes, especially for patients with relapsed disease.
Because response to currently available cytotoxic agents for recurrent ovarian cancer is limited, attention has shifted to a newer class of agents that target biological mechanisms underlying cancer development and progression. One such category includes drugs targeting serine–threonine kinases (STK), key components in signal transduction pathways controlling metabolism, cell-cycle regulation, cell division, and angiogenesis.[4] STK inhibitors can be grouped into their action on three main signal transduction pathways: (1) the PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) signaling pathway, (2) the MAPK (mitogen-activated protein kinase) pathway, and (3) the aurora kinase pathway.[5–7] Hyperactivation of these pathways have been linked to cancer development.
The PI3K/AKT/mTOR pathway regulates cell proliferation, survival, mRNA translation, and angiogenesis in response to growth factors, nutrients, hypoxia, and energy input. Other molecules implicated in this pathway include PTEN (phosphate and tensin homolog) and STK11.[5] Indeed, germline mutations in PTEN are responsible for autosomal dominant Cowden Syndrome in humans,[8] while mouse models that combine conditional deletion of PTEN within the ovarian surface epithelium with expression of oncogenic KRAS perfectly recapitulate invasive, widely metastatic endometrioid ovarian cancer.[9] STK11 is a serine threonine kinase tumor suppressor gene that controls the activity of AMP-activated protein kinase (AMPK) family members, and germline mutations in STK11 are responsible for Peutz–Jeghers syndrome,[10,11] although sporadic somatic mutations in STK11 are rare in ovarian tumors.[12] In ovarian cancers, mutations have been observed in several related proto-oncogenes, including PIK3CA, PIK3R1, and AKT1/2.[4]
In 2004, AKT and mTOR were found to be activated in 87% of epithelial ovarian cancers.[13] Since then, several other studies have shown a prevalence of activating mutations in genes encoding regulatory units for proteins in this pathway, including PIK3CA, PIK3R1, AKT, and PTEN.[14] Everolimus (for renal cell carcinoma, breast cancer, pancreatic cancer, and astrocytoma) and temsirolimus (for renal cell carcinoma) are the only two FDA-approved drugs acting on this pathway. However, a multitude of other inhibitors are in development including those acting on AKT, PI3K, and other pathway components. Using a combination of drugs acting at multiple stages of this pathway may help overcome resistance due to feedback inhibition.[4] For example, mTOR forms two complexes to enact downstream effects, mTORC1 and mTORC2. Currently available first-generation mTOR inhibitors selectively act on mTORC1, which may shift downstream effects to mTORC2. Furthermore, it is theorized that diminished output from mTORC1 feeds back to increase activity of AKT and PI3K, causing resistance to mTOR inhibitors. To overcome resistance, some suggest combining mTOR inhibitors with PI3K, AKT, or MAPK inhibitors.[4] While it is difficult to generalize across all drugs that modulate this pathway, the most common side effects include hyperglycemia, rash, stomatitis, myelosuppression, and gastrointestinal effects.[14,15] Metformin is also being studied as an adjunct to cancer therapy due to its indirect inhibition of mTOR and is extremely well tolerated.[14]
The MAPK pathway regulates cell proliferation, metabolism, apoptosis, and immune response. It involves both STK and tyrosine kinases, including BRAF (v-raf murine sarcoma viral oncogene homolog), downstream effectors MEK1/2 (mitogen-activated protein kinase kinase), and ERK1/2 (extracellular signal-regulated kinase).[16,17] Input to the pathway includes metabolic stress, DNA damage, and growth factors. ERK1/2 mediates cross talk with the PI3K/AKT/mTOR pathway.
Alteration of the MAPK pathway has been implicated in cancer survival, dissemination, and resistance to treatment. [17] Mutations in EGFR (epidermal growth factor receptor), RAS (v-ras viral oncogene), and BRAF, which are found frequently in melanoma and in low-grade serous ovarian carcinoma (LGSOC), lead to overactivation of the MAPK pathway. [18] Sorafenib targets the RAF/MEK/ERK pathway and is currently FDA-approved for use in renal cell carcinoma, and trametinib is a MEK inhibitor that is FDA-approved for BRAF-mutated melanoma.[19] Though multiple clinical trials have been conducted, to date, no drugs acting on these pathways have been approved for ovarian cancer.[20] Side effects of drugs interacting with this pathway generally include diarrhea, nausea/vomiting, rash, and fatigue.[21]
The aurora kinase pathway regulates the cell cycle and mitosis through the mitotic spindle, centrosome separation, and cytokinesis.[6] These kinases assist in monitoring the mitotic checkpoint and therefore play a critical role in chromosome distribution to daughter cells.[6] Dysregulation, however, can lead to unrestrained cell proliferation and is linked to poor prognosis and to resistance to chemotherapy, especially to microtubule-based therapy. Aurora kinase A subtype has been implicated in solid malignancies including breast, ovarian, colon, pancreas, and prostate cancers.[22] One study found overexpression of aurora kinase A in up to 91% of ovarian malignancies; higher levels of expression were correlated with worse overall survival (OS).[23] Inhibition of aurora kinase A leads to defective spindle formation and chromosome separation, causing cell cycle arrest and apoptosis.[24] In xenograft models of ovarian cancer, aurora kinase inhibitors induce growth arrest and tumor regression.[25] Though multiple aurora kinase inhibitors have been developed to treat cancer, none have been FDA-approved. Major side effects include neutropenia, mucositis, and alopecia.
The above findings have suggested STK pathway inhibitors as attractive anticancer therapies, though the efficacy of STK inhibitors as a class of medications for ovarian cancer has not been fully assessed. The objective of this study was to conduct a systematic review of clinical outcomes of STK inhibitor-based treatment in recurrent epithelial ovarian cancer.
2. Methods
2.1. Literature search and study eligibility
A systematic literature review was conducted using a public search engine, PubMed/MEDLINE. Forty-one target searches were conducted by entering each of the keywords listed in Table 1 and ‘ovarian cancer.’ The search was conducted without limits on date or language (24 September 2015). Included publications were required to evaluate recurrent ovarian cancer patient outcomes in a clinical study (randomized controlled trials and prospective or retrospective studies) using STK inhibitor-based treatment regimens and to include data on patient demographics, treatment regimen, response to therapy, survival outcomes, and adverse events. Publications were excluded from the analysis if they were duplicated between searches, if reports of individual subjects were duplicated between studies, if they lacked data specific to ovarian cancer treatment outcomes and adverse events, if no STK inhibitor was given, or if they were classified as review articles, case series, commentaries, or preclinical studies. Studies evaluating STK inhibitors for primary prevention or initial treatment of ovarian cancer were also excluded from the study. This literature review involved the use of published data only, was not considered a human subjects study, and was exempt from IRB review. ‘Meta-analysis of Observational Studies in Epidemiology (MOOSE)’ was consulted as a guideline for reporting methods and results.[26]
Table 1.
Terms for literature search.
mTOR pathway | MAPK pathway | Aurora kinase pathway |
---|---|---|
mTOR | MAPK | Aurora |
PI3K | MEK | Danusertib |
AKT | Raf | Alisertib |
STK11 | Refametinib | Hesperadin |
Metformin | Pimasertib | Barasertib |
Temsirolimus | Binimetinib | Tozasertib |
Everolimus | Trametinib | |
Ridaforolimus | Selumetinib | |
Pictilisib | Cometinib | |
Buparlisib | Vemurafenib | |
Idelalisib | Dabrafenib | |
Perifosine | Encorafenib | |
Apitolisib | Sorafenib | |
Voxtalisib | Tafinlar | |
Copanilsib | ||
Dactolisib | ||
Miltefosine | ||
Afuresertib | ||
Ipatasertib | ||
Trisiribine |
STK: serine–threonine kinase; PI3K: phosphatidylinositol 3-kinase; AKT: protein kinase B; mTOR: mammalian target of rapamycin; MEK: mitogen-activated protein kinase kinase; MAPK: mitogen-activated protein kinase.
2.2. Clinical information abstracted
For each study in the meta-analysis, we recorded drug regimens, year of publication, number of patients enrolled and evaluable for treatment, and type of study. Demographic data were compiled on individual study participants including the number of prior treatment regimens, platinum status, performance status, tumor histology, primary site (epithelial ovarian, primary peritoneal, or fallopian tubal), race, and median age of participants.
Treatment data were also abstracted including rates of overall response, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). Median progression-free survival (PFS) and OS were recorded where available. Adverse event information was collected, including incidences of overall and grade 3/4 events.
2.3. Study definitions
Responses were defined by the RECIST criteria at best response for all patients.[27] Both confirmed and unconfirmed responses were included. Overall response rate included patients with CR and PR, while clinical benefit rate was defined as the percentage of patients with CR, PR, and SD. PFS was defined as the time interval between the initiation of treatment and the date of first progression or last follow-up if the patient was alive. OS was defined as the time interval between the treatment initiation and the date of death due to disease or last follow-up if the patient was alive. Adverse events were graded per the CTCAE guidelines.[28]
2.4. Statistical analysis
The primary objective of the analysis was to examine treatment response and survival outcome of patients on STK inhibitor-based therapy. The secondary objective was to evaluate adverse events. A pooled analysis was conducted to tabulate response rates and clinical benefit rates of all STK inhibitors. Response rates among the various treatment regimens and STK pathways were compared with chi-square test or Fisher’s exact test as appropriate. The magnitude of statistical significance for treatment response was described with odds ratios (OR) and 95% confidence intervals (CI). The survival outcome and response rates were correlated among studies available for both outcome measurements, and treatment regimens above the median of PFS and treatment response (response and clinical benefit rates) were described. Frequency of adverse events per patient across the CTCAE system was compared with Mann–Whitney U test or Kruskal–Wallis test as appropriate. Statistical significance was set at a P value of less than 0.05 (two-tailed).
3. Results
3.1. Search results
Selection schema for the study is shown in Figure 1. The initial search terms returned 2020 publications. Of those, 632 were duplicated and excluded, leaving 1388 unique publications. Abstracts were then reviewed to eliminate 1351 publications not classified as clinical studies or not involving STK inhibitor therapy. This left 37 relevant studies, including 6 retrospective studies and 31 clinical trials.
Figure 1.
Selection schema. PubMed was searched on 9/24/2015. Abbreviation: STKi, serine-threonine kinase inhibitors.
3.2. Clinical trials cohort
3.2.1. Demographic data
Of the 31 qualifying clinical trials, 13 were excluded (Figure 1). Eight studies lacking ovarian cancer-specific outcomes were excluded in addition to three that did not use STK inhibitors as salvage therapy, one that did not specify treatment regimens used, and one in which patients were represented in another included study. The remaining 18 clinical trials represented 497 patients and 20 drug regimens.[29–46] Table 2 outlines demographic data. The median age of women in these trials was typically 50 years or older (77.8%). Only 6 studies (265 subjects representing 53% of the study population) reported data on race. Of these patients, the vast majority were Caucasian (96%). Only 3% were African-American and 1% was Asian. The majority of studies were published after 2010 (83.3%) and were phase II studies (52.6%). No phase III studies were reported. The majority (82.3%) of patients had performance status of 0–1, had serous histology (60%), had platinum resistant or refractory disease (73.2% among known platinum status), and had received at least two prior treatment regimens (55%). Notably, 63.6% of patients were given treatment with a MAPK pathway inhibitor, and 71% of these patients were treated with regimens that included sorafenib. Approximately one-fourth of patients (23.5%) were treated with mTOR-pathway inhibitors, and only 17% were given an aurora kinase inhibitor. Most patients (66%) received STK inhibitor therapy alone, while the remainder received a combination regimen. Nearly a quarter of patients (24.5%) received traditional cytotoxic chemotherapy with STK inhibitor.
Table 2.
Compiled demographics for clinical trial cohort.
N = 497 | % | |
---|---|---|
Year of publication | ||
Before 2000 | 0 | 0 |
2001–2010 | 81 (3 studies) | 16.3 |
2011 or later | 416 (15 studies) | 83.7 |
Study type | ||
Phase I | 126 (9 studiesa) | 25.4 |
Phase II | 371 (10 studiesa) | 74.6 |
Phase III | 0 (0 studies) | 0 |
Performance status | ||
0 | 243 | 48.9 |
1 | 166 | 33.4 |
2 | 0 | 0 |
Unknown | 88 | 17.7 |
Prior treatment regimens | ||
0 | 2 | 0.4 |
1 | 115 | 23.1 |
2 | 165 | 33.2 |
≥3 | 108 | 21.7 |
Unknown | 107 | 21.5 |
Platinum status | ||
Sensitive | 70 | 14.1 |
Resistant | 158 | 31.8 |
Refractory | 41 | 8.2 |
Platinum allergic | 3 | 0.6 |
Unknown | 225 | 45.3 |
Histology | ||
Serous | 298 | 60.0 |
Endometrioid | 20 | 4.0 |
Mucinous | 1 | 0.2 |
Clear cell | 11 | 2.2 |
Other or unknown | 167 | 33.6 |
Primary cancer | ||
Epithelial ovarian | 366 | 73.6 |
Fallopian tube | 1 | 0.2 |
Primary peritoneal | 47 | 9.5 |
Unknown | 83 | 16.7 |
STK inhibitor pathway type | ||
mTOR/AKT/PI3Kb | 117 | 23.5 |
MAPK/MEK/Rafb | 316 | 63.6 |
Aurora | 85 | 17.1 |
Drug regimens | ||
Single STK inhibition | 307 | 61.8 |
Multiple STK inhibitionb | 21 | 4.2 |
STK inhibitor + cytotoxic agent | 122 | 24.5 |
STK inhibitor + hormonal agent | 9 | 1.8 |
STK inhibitor + biologic agent | 38 | 7.6 |
One publication combined phase I and II trials.
Regimen of 21 pts taking both MEK 1/2 inhibitor trametinib and PI3K inhibitor buparlisib.[32]
mTOR: Mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase; STK: serine–threonine kinases.
3.2.2. Treatment and survival outcomes
Of the original 497 patients, 442 were analyzable for treatment outcome by the RECIST criteria. We first performed a focused analysis of the 299 patients (11 trials) receiving only STK inhibitors, revealing an overall response rate of 10% and a clinical benefit rate of 64% due to high rates of SD (Table 3). The overall response and clinical benefit rates were notably higher among PI3K inhibitors at 40% and 95%, respectively. These responses were seen in a single study of 20 patients with RAS or BRAF mutations given a multiple STK inhibitor regimen of buparlisib (PI3K inhibitor) and trametinib (MEK1/2 inhibitor), which also resulted in one of the only two CRs seen without cytotoxic chemotherapy.[32] Similarly, selumetinib therapy induced a single CR without an adjunct cytotoxic agent in a patient with recurrent LGSOC.[34] Data on PFS were available from eight studies and on OS from five studies (Table 3). For STK inhibitor monotherapy, median PFS was 2.7 months and median OS was 14.1 months. PI3K inhibitors resulted in the longest median PFS at 7 months, tested in a regimen involving multiple STK inhibitors in patients with RAS and BRAF mutations.[32] PFS and OS were lowest for patients on aurora kinase inhibitors (2.0 and 9.8 months, respectively).
Table 3.
Treatment outcomes by STK pathways for STK inhibitors only (clinical trial cohort).
mTOR/AKT/PI3K pathway |
Aurora pathway |
MAPK/MEK/Raf pathway |
||||||
---|---|---|---|---|---|---|---|---|
All STK inhibitors | All pathways | mTOR | AKT | PI3K | All pathways | All pathways | Sorafenib | |
Response rates (RECIST) | ||||||||
No. of subjects (n) | 299a | 74 | 54 | 0 | 20 | 82 | 163 | 76 |
Response rate (%) | 10 | 18 | 9 | n/a | 40 | 6 | 12 | 5 |
Clinical benefit rateb (%) | 64 | 95 | n/a | n/a | 95 | 69 | 63 | 47 |
Complete response (%) | 1 | 1 | 0 | n/a | 5 | 0 | 1 | 0 |
Partial responseb (%) | 9 | 16 | 9 | n/a | 35 | 6 | 11 | 5 |
Stable diseaseb (%) | 53 | 55 | n/a | n/a | 55 | 61 | 50 | 42 |
Progressive disease | 35 | 5 | n/a | n/a | 5 | 29 | 37 | 53 |
Response rates (CA125) | ||||||||
No. of subjects (n) | 98 | 0 | 0 | 0 | 0 | 51 | 47 | 0 |
Response rates (%) | 16 | n/a | n/a | n/a | n/a | 20 | 13 | n/a |
Survival time (months)c | ||||||||
PFS | 2.7 | 5.1 | 3.1 | n/a | 7.0 | 2.0 | 5.6 | 2.1 |
OS | 14.1 | 11.6 | 11.6 | n/a | n/a | 9.8 | 16.3 | 16.3 |
Response rate includes complete response and partial response. Clinical benefit rate includes complete response, partial response, and stable disease.
Not all subjects included in Table 2 were evaluable for treatment response so numbers differ.
Two trials (87 subjects) did not include data on rates of stable disease and progressive disease, so these subjects were removed from analysis of clinical benefit rate, stable disease, and progressive disease (for study with mTOR pathway inhibitor with temsirolims, subgroup of the study showed at least 53% of clinical benefit rate based on 51 cases reporting treatment response).
Survival time is described from study enrollment to final status, numbers are described as median of median reported in each study (not available for all studies).
STK: serine–threonine kinases; mTOR: mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase; PFS: Progression-free survival; OS: overall survival.
When all drug regimens were examined as a whole, the overall response rate was 13%, and the clinical benefit rate was 67% due to high rates of SD (Table 4). This result was similar among the three STK inhibitor pathways except for the previously mentioned study of buparlisib and trametinib.[32] Another notable exception was AKT inhibitor therapy with the lowest response rate (4%) of all drugs, the lowest clinical benefit rate (28%), and the highest rate of PD (72%). Data on PFS were available from 12 studies and on OS from 9 studies (Table 4). For all STK inhibitors combined, median PFS was 3.4 months and median OS was 13 months. The longest PFS, at 7.0 months, again resulted from the trial of multiple STK inhibitors.[32] PFS and OS were lower for patients on AKT inhibitors (1.9 and 4.5 months, respectively) than any other drug classes. For comparison, treatment response and survival outcome for only regimens combining STK inhibitors and cytotoxic agents are shown in Table 5.
Table 4.
Description of treatment outcomes by STK pathway (clinical trial cohort).
mTOR/AKT/PI3K pathway |
Aurora pathway |
MAPK/MEK/Raf pathway |
||||||
---|---|---|---|---|---|---|---|---|
All STK inhibitors | All pathways | mTOR | AKT | PI3K | All pathways | All pathways | Sorafenib | |
Response rates (RECIST) | ||||||||
No. of subjects (n) | 442a | 108 | 63 | 25 | 20 | 82 | 272 | 185 |
Response rate (%) | 13 | 13 | 8 | 4 | 40 | 6 | 17 | 17 |
Clinical benefit rateb (%) | 67 | 56 | 44 | 28 | 95 | 69 | 71 | 68 |
Complete response (%) | 1 | 1 | 0 | 0 | 5 | 0 | 2 | 2 |
Partial responseb (%) | 12 | 12 | 8 | 4 | 35 | 6 | 15 | 15 |
Stable disease (%) | 52 | 39 | 44 | 24 | 55 | 61 | 53 | 51 |
Progressive diseaseb (%) | 32 | 44 | 56 | 72 | 5 | 29 | 28 | 30 |
Response rates (CA125) | ||||||||
No. of subjects (n) | 175 | 21 | 0 | 21 | 0 | 51 | 103 | 56 |
Response rates (%) | 21 | 10 | 0 | 10 | 0 | 20 | 23 | 32 |
Survival time (months)c | ||||||||
PFS | 3.4 | 3.2 | 3.2 | 1.9 | 7.0 | 2.0 | 4.6 | 3.7 |
OS | 13.0 | 8.0 | 11.6 | 4.5 | n/a | 9.8 | 15.2 | 15.2 |
Response rate includes complete response and partial response. Clinical benefit rate includes complete response, partial response, and stable disease.
Not all subjects included in Table 2 were evaluable for treatment response so numbers differ.
Two trials (87 subjects) did not include data on rates of stable disease and progressive disease, so these subjects were removed from analysis of clinical benefit rate, stable disease, and progressive disease (for study with mTOR pathway inhibitor with temsirolims, subgroup of the study showed at least 53% of clinical benefit rate based on 51 cases reporting treatment response).
Survival time described from study enrollment to final status, numbers are described as median of median reported in each study (not available for all studies).
STK: Serine–threonine kinases; mTOR: mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase; PFS: progression-free survival; OS: overall survival; SD: stable disease; PD: progressive disease.
Table 5.
Treatment outcomes by STK pathway for STK inhibitor with cytotoxic chemotherapy (clinical trial cohort).
mTOR/AKT/PI3K pathway |
Aurora pathway |
MAPK/MEK/Raf pathway |
||||||
---|---|---|---|---|---|---|---|---|
All STK inhibitors | All pathways | mTOR | AKT | PI3K | All pathways | All pathways | Sorafenib | |
Response rates (RECIST) | ||||||||
No. of subjects (n) | 105 | 21 | 0 | 21 | 0 | 0 | 84 | 84 |
Response rate (%) | 19 | 5 | n/a | 5 | n/a | n/a | 23 | 23 |
Clinical benefit rate (%) | 71 | 24 | n/a | 24 | n/a | n/a | 83 | 83 |
Complete response (%) | 3 | 0 | n/a | 0 | n/a | n/a | 4 | 4 |
Partial response (%) | 16 | 5 | n/a | 5 | n/a | n/a | 19 | 19 |
Stable disease (%) | 52 | 19 | n/a | 19 | n/a | n/a | 61 | 61 |
Progressive disease (%) | 26 | 76 | n/a | 76 | n/a | n/a | 13 | 13 |
Response rates (CA125) | ||||||||
No. of subjects (n) | 77 | 21 | 0 | 21 | 0 | 0 | 56 | 56 |
Response rates (%) | 26 | 10 | n/a | 10 | n/a | n/a | 32 | 32 |
Survival time (months)a | ||||||||
PFS | 3.7 | 1.9 | n/a | 1.9 | n/a | n/a | 3.7 | 3.7 |
OS | 13.5 | 4.5 | n/a | 4.5 | n/a | n/a | 14.0 | 14.0 |
Only four trials included regimens with cytotoxic chemotherapy, represented above. All three studies in the MAPK/MEK/Raf pathway involved a combination of Sorafenib with traditional chemotherapy. The one study regimen involving an mTOR/Akt/PI3K pathway inhibitor involved the AKT inhibitor perifosine with docetaxel.
Survival time is described from study enrollment to final status; numbers are described as median of median reported in each study. PFS and OS were not available for all studies.
STK: Serine–threonine kinases; mTOR: mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase; PFS: progression-free survival; OS: overall survival.
Treatment responses were stratified by drug regimen classification (Figure 2(a)). Response rates differed significantly across the five study regimens (multiple STK inhibitors 40%, STK inhibitors with other class biological agents 25.8%, STK inhibitors with cytotoxic agents 19.0%, single STK inhibitors 7.9%, STK inhibitors with hormonal agents 0%; P < 0.001). STK inhibitor combination therapy resulted in significantly greater response rates than STK inhibitor monotherapy (STK inhibitors with cytotoxic agents vs. STK inhibitors alone, OR for response rate 2.75, 95%CI 1.43–5.28, P = 0.003; and STK inhibitors with other class biological agents vs. single STK inhibitors, OR 4.06, 95%CI 1.63–10.1, P = 0.005). Patients treated with multiple STK inhibitors responded at significantly higher rates than those taking single STK inhibitors (OR 7.79, 95%CI 2.88–21.1, P < 0.001).
Figure 2.
Treatment response based on study regimen and pathway. Response rates and clinical benefit rates broken down by (a) class of drug regimen and (b) STK pathway. mTOR inhibitors represent all STKi falling under the mTOR/AKT/PI3K pathway. Abbreviation: STKi, STK inhibitor.
Figure 2(a) also displays clinical benefit rates stratified by drug regimen class, demonstrating statistical significance across the five study regimens (multiple STK inhibitors 95%, STK inhibitors with other class biological agents 74.2%, STK inhibitors with cytotoxic agents 71.4%, single STK inhibitors alone 52.5%, STK inhibitors with hormonal agents 42.9%; P < 0.001). Clinical benefit rates with combination STK inhibitor therapy again were noteworthy for significantly greater response than single STK inhibitors (STK inhibitors with cytotoxic agents vs. single STK inhibitors, OR 2.26, 95%CI 1.39–3.67, P = 0.001; and STK inhibitors with other class biological agents vs. single STK inhibitors OR 2.60, 95%CI 1.12–6.01, P = 0.023). Multiple STK inhibitors improved the clinical benefit rate compared to STK inhibitor monotherapy (OR 17.2, 95%CI 2.27–130, P < 0.001).
Treatment responses were then compared among the three STK pathway types (Figure 2(b)). Response rates significantly differed across the pathways: inhibition for both mTOR and MAPK 40%, MAPK inhibitors 15.5%, mTOR pathway inhibitors 6.8%, and aurora kinase inhibitors 6.1%, P < 0.001. Patients receiving multiple STK inhibitors for the mTOR and MAPK pathways responded at higher rates than those given regimens based on a single STK pathway (mTOR/MAPK vs. mTOR, OR for response rate 9.11, 95%CI 2.69–30.8, P < 0.001; mTOR/MAPK vs. MAPK, OR 3.64, 95%CI 1.40–9.49, P = 0.011). Clinical benefit rate demonstrated statistical significance across the pathway types (mTOR and MAPK inhibitors 95.0%, MAPK inhibitors 68.7%, mTOR inhibitors 44.7%, and aurora kinase inhibitors 42.7%, P < 0.001). When compared with mTOR inhibitors alone, inhibition of both mTOR and MAPK pathways significantly improved clinical benefit rate (OR 23.5, 95%CI 3.01–184, P < 0.001), as it did when compared with use of MAPK inhibitors (OR 9.01, 95%CI 1.19–68.4, P = 0.01).
Treatment results (response and clinical benefit rates) for each regimen are displayed in Figure 3. The most effective treatment regimens included sorafenib with carboplatin/paclitaxel (response rate 60.9%),[31] sorafenib with bevacizumab response rate 42.1%),[36] and buparlisib and trametinib (response rate 40.0%).[32] Notably, the trial that showed the highest response rate (sorafenib with carboplatin/paclitaxel) used a regimen administered only to platinum-sensitive patients.[18] Similarly, when sorafenib was solely given to platinum-sensitive patients, the response rate was amongst the highest in STK inhibitor monotherapy (15.6%). The trial of buparlisib and trametinib included patients with somatic RAS or BRAF mutations (these upregulate MAPK/mTOR pathways), increasing the likelihood of response.[32] Lastly, the selumetinib regimen, which demonstrated the highest response rate among STK inhibitor monotherapy regimens (16.0%), was given only to patients with LGSOC.[34] These data indicate that the utility of STK inhibitors is maximized when combined with another class biological or cytotoxic agent in select patient populations.
Figure 3.
Treatment response per individual study regimen. Response rates and clinical benefit rates for each of 20 drug regimens represented in 18 clinical trials included in the analysis, listed in order of decreasing clinical benefit rates. Two trials randomized patients to different treatment regimens, which are listed separately in this table. *clinical benefit rate at least 53% based on best estimation among 51 cases. **SD and PD were not reported and clinical benefit rate is determined by the response rate.
Because clinical efficacy includes survival outcome, treatment responses were correlated with PFS (Figure 4). Twelve studies (42%) reported both treatment response and PFS. In this subset, the median response rate was 8.1% and the median clinical benefit rate was 57.6%. Median PFS was 3.4 months. The study regimens above the median for treatment response and PFS include sorafenib with carboplatin and paclitaxel,[31] selumetinib alone,[34] buparlisib and trametenib,[32] sorafenib alone,[31] and sorafenib with gemcitabine.[30]
Figure 4.
Correlation of treatment response with progression-free survival. Correlations of (a) response rate and (b) clinical benefit rate with PFS by individual drug regimen. Twelve studies with published PFS data were examined. Median PFS was 3.4 months demarcated horizontal lines. Median response rate was 8.1%, and median clinical benefit rate was 57.6% demarcated by vertical lines. Data points for drug regimens with both response rate and clinical benefit rate, and PFS above the median values for this analysis are labeled. Abbreviations: TC, paclitaxel and carboplatin; and PFS, progression-free survival.
3.2.3. Adverse events in clinical trials
Adverse events were analyzed in 13 clinical trials meeting the inclusion criteria. The most common grade 1–4 adverse effects per the CTCAE system were blood/lymphatic disorders, followed by gastrointestinal disorders, and skin/subcutaneous tissue disorders (Table 6). This pattern was similar for grade 3–4 adverse events. Adverse event rates between STK inhibitor monotherapy and STK inhibitors with chemotherapy were statistically similar (Table 6). Adverse event rates differed significantly across the three STK pathways with aurora kinase inhibitors having the highest adverse event rates and MAPK inhibitors having the lowest (Table 7).
Table 6.
Adverse event rate compared between STK inhibitor alone and STK inhibitor + cytotoxic agent regimens (clinical trial cohort).
Grades 1–4 |
Grades 3 and 4 |
|||||||
---|---|---|---|---|---|---|---|---|
All patients |
STK alone |
STK + cytotoxic agent |
All patients |
STK alone |
STK + cytotoxic agent |
|||
Subject no.(n) | 426 | 286 | 121 | P value | 426 | 286 | 121 | P value |
Blood/Lymphatic disorders | 1.6 | 1.7 | 1.8 | 0.62 | 0.5 | 0.4 | 0.7 | 0.83 |
Cardiac disorders | 0.1 | 0.1 | <0.1 | <0.1 | 0 | 0 | ||
Constitutional | 0.3 | 0.5 | <0.1 | <0.1 | 0 | 0 | ||
Endocrine disorders | <0.1 | <0.1 | 0 | 0 | 0 | 0 | ||
Eye disorders | <0.1 | <0.1 | <0.1 | <0.1 | 0 | 0 | ||
Gastrointestinal disorders | 1.4 | 1.3 | 1.7 | 0.2 | 0.1 | 0.3 | ||
General disorders/administration site conditions | 0.5 | 0.4 | 0.8 | 0.1 | 0.1 | 0.2 | ||
Immune system disorder | <0.1 | <0.1 | <0.1 | <0.1 | 0 | 0 | ||
Infections /Infestations | 0.1 | 0.1 | 0.1 | <0.1 | <0.1 | <0.1 | ||
Investigations | 0.3 | 0.2 | 0.4 | <0.1 | <0.1 | <0.1 | ||
Metabolism/Nutrition disorders | 0.7 | 0.7 | 0.8 | 0.1 | 92 (0.1) | 0.1 | ||
Musculoskeletal/Connective tissue disorders | 0.1 | 0.1 | 0.1 | <0.1 | <0.1 | <0.1 | ||
Neoplasms benign, malignant and unspecified | <0.1 | <0.1 | <0.1 | <0.1 | 0 | 0 | ||
Nervous system disorders | 0.3 | 0.3 | 0.3 | <0.1 | <0.1 | 0.1 | ||
Pain | 0.3 | 0.3 | 0.1 | <0.1 | <0.1 | <0.1 | ||
Renal/Urinary disorders | <0.1 | <0.1 | <0.1 | <0.1 | <0.1 | 0 | ||
Reproductive system/breast disorders | <0.1 | <0.1 | 0 | 0 | 0 | 0 | ||
Respiratory, thoracic, and mediastinal disorders | 0.1 | 0.2 | 0.1 | <0.1 | <0.1 | 0 | ||
Skin/Subcutaneous tissue disorders | 0.7 | 0.7 | 0.8 | 0.1 | 0.1 | 0.2 | ||
Syndromes | <0.1 | 0 | 0 | <0.1 | <0.1 | <0.1 | ||
Vascular disorders | 0.1 | <0.1 | 0.2 | <0.1 | <0.1 | <0.1 |
Mann–Whitney U test for P values for comparison between STK inhibitor monotherapy versus STK inhibitor with chemotherapy. Rates of adverse events per patient were calculated by adding all patients experiencing adverse events in a particular organ system class and dividing by the total number of patients exposed to the drug regimen.
STK: Serine–threonine kinases.
Table 7.
Adverse event rate per patient based on STK pathways (clinical trial cohort).
Grades 1–4 |
Grades 3 and 4 |
|||||||
---|---|---|---|---|---|---|---|---|
Aurora kinase |
mTOR-AKT-PI3K |
MAPK-MEK-Raf |
Aurora kinase |
mTOR-AKT-PI3K |
MAPK-MEK-Raf |
|||
Subject no. (n) | 65 | 75 | 286 | P value | 65 | 75 | 286 | P value |
Blood/Lymphatic disorders | 2.8 | 1.7 | 1.3 | 0.024 | 1.3 | <0.1 | 0.4 | 0.012 |
Cardiac disorders | 0 | <0.1 | 0.1 | 0 | <0.1 | <0.1 | ||
Constitutional | 0 | 0.6 | 0.3 | 0 | <0.1 | <0.1 | ||
Endocrine disorders | 0 | 0 | <0.1 | 0 | 0 | 0 | ||
Eye disorders | 0 | <0.1 | <0.1 | 0 | 0 | <0.1 | ||
Gastrointestinal disorders | 1.9 | 1.8 | 1.1 | 0.1 | 0.3 | 0.1 | ||
General disorders/administration site conditions | 0.8 | 0.3 | 0.5 | <0.1 | <0.1 | <0.1 | ||
Immune system disorder | 0 | <0.1 | <0.1 | 0 | 0 | <0.1 | ||
Infections and infestations | 0 | 0.1 | <0.1 | 0 | <0.1 | <0.1 | ||
Investigations | 0.7 | 0 | 0.2 | <0.1 | 0 | <0.1 | ||
Metabolism and nutrition disorders | 1.1 | 0.5 | 0.6 | <0.1 | 0.1 | 0.1 | ||
Musculoskeletal and connective tissue disorders | 0 | <0.1 | <0.1 | 0 | <0.1 | <0.1 | ||
Neoplasms benign, malignant, and unspecified | 0 | 0 | <0.1 | 0 | 0 | <0.1 | ||
Nervous system disorders | 0 | 0.2 | 0.3 | 0 | 0 | <0.1 | ||
Pain | 0 | 0.5 | 0.2 | 0 | 0.1 | <0.1 | ||
Renal and urinary disorders | 0 | <0.1 | <0.1 | 0 | <0.1 | <0.1 | ||
Reproductive system and breast disorders | 0 | <0.1 | <0.1 | 0 | 0 | 0 | ||
Respiratory, thoracic, and mediastinal disorders | 0 | 0.4 | <0.1 | 0 | <0.1 | <0.1 | ||
Skin/Subcutaneous tissue disorders | 0.4 | 0.5 | 0.9 | 0 | <0.1 | 0.2 | ||
Syndromes | 0 | 0 | <0.1 | 0 | 0 | <0.1 | ||
Vascular disorders | 0 | 0 | 0.2 | 0 | 0 | <0.1 |
Krsukal–Wallis test for P values (comparison across the three groups). Rates of adverse events per patient were calculated by adding all patients experiencing adverse events in a particular organ system class and dividing by the total number of patients exposed to the drug regimen.
STK: Serine–threonine kinases; mTOR: mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase.
3.3. Retrospective study cohort
All six retrospective studies involved treatment with metformin. Three described metformin’s impact on ovarian cancer incidence rather than treatment response or survival time; these were excluded from our analysis. Of the remaining three studies, published between 2012 and 2014, two were retrospective cohort studies, and one was a case-control study. While all three studies found that diabetes negatively affected survival, two studies reported that metformin significantly improved survival outcomes, even above those of nondiabetics.[47,48] One study reported no survival difference in diabetic patients based on metformin status [49]; however, authors noted the limitation of a small sample size.
4. Conclusion
In this predominantly platinum-resistant and heavily pretreated ovarian cancer population, the overall response rate of STK inhibitors was 13%. STK inhibitor-based therapy maintained a considerable proportion of SD (clinical benefit rate 67%), suggesting a possible utility for this purpose. When used as monotherapy, STK inhibitors seem to have minimal effect on recurrent ovarian cancer. However, response rates in combination regimens (sorafenib with carboplatin/paclitaxel, gemcitabine, or bevacizumab) were significantly greater. Lastly, certain conditions maximized the effectiveness of STK inhibitor-based therapy that warrant special attention. These include platinum-sensitive disease (sorafenib), LGSOC (selumetinib), and somatic RAS/BRAF mutations (buparlisib with trametnib). To elucidate the true benefit of STK inhibitors in ovarian cancer, future research should focus on the efficacy of STK inhibitors in these subtypes of ovarian cancer and on differentiating response rates in randomized controlled trials comparing chemotherapy with and without the addition of STK inhibitors.
5. Expert opinion
Responses to STK inhibitor-based therapy in this meta-analysis are comparable to published literature on overall response rates for all recurrent ovarian cancer therapies. Previously reported response rates of platinum-resistant disease to single-agent cytotoxic chemotherapy are around 10% with median PFS 3–4 months.[50] Combination cytotoxic regimens have higher antitumor activity than monotherapy with response rates ranging from 21.7 to 33.7%.[3] Our results for STK inhibitor-based therapy for recurrent ovarian cancer were on par with other available treatments currently used for platinum-resistant ovarian cancer. Combination therapy with STK inhibitors and cytotoxic agents resulted in higher response rates when compared with STK inhibitor monotherapy (19.0% vs. 7.9%, Figure 2(a)). Because adding cytotoxic chemotherapeutic agents to STKi does not seem to increase the risk of adverse events (Table 5), it may be a reasonable approach to add cytotoxic agents when considering STK inhibitor-based therapy for recurrent ovarian cancer. However, additional trials directly comparing STK inhibitor-based regimens with chemotherapy alone are needed to determine if STK inhibitors provide true additional benefit.
Notably, several studies not included in this meta-analysis found STK inhibitors ineffective when used as an adjunct to primary adjuvant, neoadjuvant, or maintenance therapy. One randomized control trial compared sorafenib to placebo as maintenance therapy following primary treatment with surgery and adjuvant chemotherapy but did not show significant benefit in PFS.[51] In another trial, patients were randomized to carboplatin and paclitaxel with or without sorafenib as primary therapy after surgical cytoreduction. Authors detected no difference in efficacy but substantially increased toxicity (skin reactions, hand–foot syndrome, mucositis, and hypertension).[52] Sorafenib was similarly studied as an adjunct to neoadjuvant carboplatin and paclitaxel; however, only four patients were enrolled before the study was terminated due to severe postoperative toxicities.[53] Though there does not seem to be a role for STK inhibitors in primary treatment, our study suggests possible efficacy in maintaining SD based on high clinical benefit rates. Additional research on alternate STK pathway inhibitors for maintenance therapy is warranted.
Our data suggest a possible use for STK inhibitors in a combination regimen for salvage therapy and for maintenance of disease stability, particularly in KRAS and BRAF mutant tumors, which are frequently seen in LGSOC. A recent meta-analysis showed rates of KRAS and BRAF mutations in LGSOC as high as 63% and 33%, respectively, which may explain susceptibility to MEK inhibitors.[54] In general, LGSOC and KRAS/BRAF mutations are associated with treatment resistance but longer survival.[54,55] We noted a unique 16% response with selumetinib monotherapy in LGSOC patients. In this trial, 47% of the tumor samples from 34 patients with tissue available for analysis harbored KRAS/BRAF mutations. However, response stratified by mutation status showed no difference between those with and without the mutation.[34] The single patient in this study experiencing CR was noted to have an ongoing response (>5 years), despite previous progression on multiple lines of intravenous and intraperitoneal chemotherapy. Molecular profiling of this patient’s tumor by Grisham et al. revealed a mutation in a negative regulatory region of MAP2K1, which encodes MEK1, a driver mutation deemed to explain her unique response. These authors then sequenced 48 LGSOC and serous borderline tumors and found that patients with exceptional responses to MEK inhibitors had multiple novel MAPK pathway alterations, rather than conventional hotspot KRAS/BRAF mutations.[56] Several clinical trials are currently in progress to further study STK inhibitors in LGSOC (Table 8).
Table 8.
Summary of ongoing trials for STKi for ovarian cancer.
Compound | Target | Study (NCT no.) | Phase | Country | Study start date | Status | Condition | Interventions |
---|---|---|---|---|---|---|---|---|
mTOR/AKT/PI3K pathway | ||||||||
Metformin | mTOR pathway | NCT01579812 | Phase II | USA | 10/2011 | Recruiting | Advanced (primary treatment) | Met + chemotherapy |
Metformin | mTOR pathway | NCT02437812 | Phase II | USA | 1/2014 | Recruiting | Newly diagnosed (primary treatment) | Met + TC |
Metformin | mTOR pathway | NCT02122185 | Phase II | USA | 6/2014 | Recruiting | Stage III–IV (primary therapy) | TC (docetaxel) ± met (followed by met vs. placebo maintenance) |
Metformin | mTOR pathway | NCT02312661 | Phase I | Netherlands | 10/2015 | Recruiting | Recurrent | Met + TC |
Everolimus | mTORC1 | NCT01031381 | Phase II | USA | 9/2010 | Completed 5/2013 | Recurrent | Everolimus + bevacizumab |
Everolimus | mTORC1 | NCT01281514 | Phase I | USA | 12/2010 | Recruiting | Platinum-sensitive recurrent | Everolimus + Carboplatin + PLD |
Everolimus | mTORC1 | NCT02188550 | Phase II | USA | 6/2014 | Recruiting | Platinum-resistant or -refractory recurrent or persistent | Everolimus + letrozole |
Everolimus | mTORC1 | NCT02283658 | Phase II | USA | 11/2014 | Ongoing Not recruiting | Recurrent hormone receptor positive | Everolimus Letrozole |
Ridaforolimus | mTORC1 | NCT01256268 | Phase I | USA | 6/2011 | Ongoing Not recruiting | Recurrent or metastatic platinum-sensitive | Ridaforolimus + TC |
Sirolimus | mTORC1 | NCT01536054 | Phase I | USA | 8/2012 | Ongoing Not recruiting | Recurrent | Sirolimus NY-ESO-1 |
Temsirolimus | mTORC1 | NCT01065662 | Phase I | USA | 2/2010 | Ongoing Not recruiting | Recurrent | Temsirolimus + cediranib |
Temsirolimus | mTORC1 | NCT01196429 | Phase II | USA, Japan, Korea | 8/2010 | Completed (1/2015) | Stages III and IV OCCC (primary treatment) | Temsirolimus + TC (then temsirolimus consolidation) |
Temsirolimus | mTORC1 | NCT01460979 | Phase II | Germany | 10/2011 | Completed (11/2015) | Platinum-refractory | Temsirolimus |
GSK2141795 | AKT | NCT01266954 | Phase I | UK | 6/2010 | Completed (12/2014)a | Recurrent or persistent | GSK2141795 |
MK2206 | AKT | NCT01283035 | Phase II | USA | 4/2011 | Completed (12/2014) | Recurrent platinum-resistant | MK2206 |
GSK2110183 | AKT | NCT01653912 | Phase I/II | Australia, Russia, UK | 12/2012 | Ongoing Not recruiting | Recurrent platinum-resistant | GSK2110183 + TC |
Triciribine | AKT | NCT01690468 | Phase I/II | USA | 9/2014 | Ongoing Not recruiting | Recurrent or persistent platinum-resistant | Triciribine + carboplatin |
COTI2 | AKT | NCT02433626 | Phase I | USA | 12/2015 | Recruiting | Recurrent platinum-resistant | COTI2 |
MAPK pathway | ||||||||
Sorafenib | MAPK | NCT00436215 | Phase II | USA | 9/2006 | Completed (6/2014) | Recurrent platinum-resistant/refractory | Sorafenib + bevacizumab |
Sorafenib | MAPK | NCT01047891 | phase II | Germany | 1/2010 | Completed (2/2015) | Recurrent platinum-resistant | Sorafenib |
Trametinib | MAPK | NCT02101788 | Phase II/III | USA, UK | 2/2014 | Recruiting | Recurrent or progressive LGSOC | Trametinib vs. letrozole, paclitaxel, PLD, tamoxifen, or topotecan |
LY2228820 | MAPK | NCT01663857 | Phase I/II | Multi-countriesc | 9/2012 | Recruiting | Recurrent platinum-sensitive | LY2228820 + gemcitabine + carboplatin |
MEK 162 | MEK | NCT01649336 | Phase I | USA | 7/2012 | Ongoing Not recruiting | Platinum-resistant or -refractory | MEK 162 + paclitaxel |
MEK 162 | MEK | NCT01849874 | Phase III | Multi-countriesd | 6/2013 | Recruiting | Recurrent, persistent, or progressive LGSOC | MEK 162 vs. PLD, paclitaxel, or topotecan |
Aurora kinase pathway | ||||||||
Alisertib | Aurora kinase | NCT01091428 | Phase I/II | USA | 5/2010 | Ongoing Not recruiting | Recurrent | Alisertib + paclitaxel |
Multiple STKi | ||||||||
Pimasertib + SAR245409 | MAPK, PI3K | NCT01936363 | Phase II | Multi-countriesb | 9/2013 | Ongoing Not recruiting | Previously treated unresectable LGSOC | Pimasertib + SAR245409 vs. placebo |
Table compiled through access to clinicaltrials.gov on 24 December 2015. Ongoing, recruiting, and completed studies involving only mullerian cancers are listed.
Results were available after our search on 24 September 2015.
USA, Australia, Belgium, Canada, France, Italy, Poland, and Spain.
USA, Australia, Belgium, Germany.
USA, Australia, Austria, Belgium, Canada, Czech Republic, Denmark, Finland, France, Germany, Hungary, Ireland, Italy, Luxembourg, Netherlands, Norway, Poland, Spain, Sweden, and the United Kingdom.
met: Metformin; TC: paclitaxel/carboplatin; PLD: pegylated liposomal doxorubicin; OCCC: ovarian clear cell carcinoma; LGSOC: low-grade serous ovarian carcinoma; STKi: STK inhibitor; STK: Serine–threonine kinases; mTOR: mammalian target of rapamycin; AKT: protein kinase B; PI3K: phosphatidylinositol 3-kinase; MAPK: mitogen-activated protein kinase; MEK: mitogen-activated protein kinase kinase.
Given that the Bedard et al. trial involving multiple STK inhibitors also focused on patients with somatic KRAS and BRAF mutations, we further examined the breakdown of included subjects’ tumor grades. Eighteen of 21 (85.7%) patients had well or moderately differentiated tumors, while only 2 (9.5%) were poorly differentiated. Of these, 90.5% had KRAS mutations while only one patient had a BRAF mutation. Responses were not stratified by tumor histology, but authors did note possible benefit for patients particularly with KRAS G12V-mutated ovarian cancers. They found no association between response and markers in the PI3K and MAPK pathways.[32] Several additional studies have noted an increased prevalence of mTOR pathway activation and PIK3CA mutations in ovarian clear cell carcinoma (OCCC).[14,57] One study published data on patients with somatic PIK3CA mutations treated on various study protocols involving mTOR pathway drugs. Of nine patients with ovarian cancer, two (22.2%) had PRs.[58] Furthermore, Grisham et al. suggested that MEK inhibitors may only be effective in patients with specific MAPK pathway alterations, similar to findings in the literature on melanoma. [56] These findings highlight the growing importance and complexity of biomarker identification and individualized cancer treatment, as the key to maximizing response is not a one drug fits all approach.
Another explanation for the efficacy of the multiple STK inhibitor regimen may lie in the ability of one pathway inhibitor to overcome resistance to other STK inhibitor therapies due to the existence of significant cross-talk and feedback inhibition between all three STK pathways. mTOR forms two complexes (mTORC1 and mTORC2) enacting downstream effects with feedback inhibition on pathway initiation.[14] First-generation mTOR inhibitors (including temsirolimus, everolimus, and ridaforolimus) only inhibit mTORC1, which may result in resistance via increased PI3K, AKT, and mTORC2 activation. The interaction between the MAPK and mTOR pathways has been extensively studied. Normally, RAS, RAF, MEK1/2, and ERK indirectly stimulate AKT activation of mTOR via inhibition of TSC1/2. As a result, inhibition of the MAPK pathway downregulates the mTOR pathway.[4] Moreover, studies have shown that mTOR inhibition leads to activation of the MAPK pathway via a PI3K-dependent feedback loop, reversible with MAPK inhibitors.[59] Potential mechanisms to overcome resistance include use of multiple mTOR and PI3K or AKT inhibitors. Other suggested combinations include mTOR inhibitors with MAPK inhibitors, VEGF inhibitors, and IGF-1 inhibitors. Examples include sorafenib with bevacizumab and ridaforolimus with dalotuzumab. [36,39,60] Furthermore, second-generation mTOR inhibitors that inhibit both mTORC1/2 are in development and may be of interest for ovarian cancer.[4] Further studies are needed to evaluate the efficacy of multiple mTOR and MAPK pathway inhibitor therapy.
Though less well understood, there is also evidence of interaction between the mTOR, MAPK, and aurora kinase pathways that may provide mechanisms for overcoming resistance. Preclinical studies have suggested that overexpression of aurora kinase A activates the mTOR pathway [61] and that cellular transformation induced by aurora kinase A is determined by co-activation of mTOR pathway.[62] Aurora kinase B phosphorylation of mTOR substrates has similarly been shown to promote cell-cycle progression in T lymphocytes.[63] Demonstrating cross-talk between the MAPK and aurora kinase pathway, inhibition of aurora kinases was indirectly shown to block ERK activity in renal cell carcinoma.[64] Additional research focused on ovarian cancer is needed to determine if combining aurora kinase inhibitors with mTOR or MAPK inhibitors may also assist in overcoming resistance.
Preclinical studies suggesting a potential use for PI3K/AKT/mTOR pathway inhibitors and MAPK pathway inhibitors in combating cancer stem cells may further explain the efficacy of multiple STK inhibitor therapy. Current theory on cancer stem cells proposes that stem cells comprise a small population of tumors that are relatively resistant to conventional chemotherapy. Recurrence following apparent complete remission may originate from these cells. This model seems relevant in ovarian cancer, given that the majority of patients relapse following complete remission.[65] Furthermore, presence of these cancer stem cells is associated with poor prognosis.[66] Studies have demonstrated that agents inhibiting the PI3K/AKT/mTOR pathway, especially newer class inhibitors that act on both mTORC1 and mTORC2 or on both the PI3K and mTOR molecules, preferentially target cancer stem cells. [67,68] A 2011 study by Alvero et al. demonstrated stem cell cytotoxicity using NV-128, which decreases adenosine triphosphate and indirectly inhibits both the mTOR and MAPK pathways.[69] Based on these findings, further preclinical investigation of the impact of second-generation mTOR inhibitors and multiple pathway inhibitors on ovarian cancer stem cells is warranted. Furthermore, this suggests a potential therapeutic role for these agents in patients whose tumors test positive for ovarian cancer stem cell markers such as aldehyde dehydrogenase and CD133.[66]
Metformin is a biguanide antidiabetic agent typically used as an insulin sensitizer. However, epidemiologic data show decreased incidence of ovarian cancer in diabetic women taking metformin.[70,71] Through multiple mechanisms, including AMPK activation and mTORC2 inhibition, metformin also inhibits the mTOR pathway.[72,73] Preclinical studies demonstrate that metformin inhibits proliferation of ovarian cancer cells.[74,75] Two of three studies in our analysis described a survival benefit for metformin.[47,48] The one study that did not detect a difference examined metformin effect as a secondary outcome and noted low numbers of patients on metformin.[49] Although no results are available from clinical trials examining the effects of metformin on ovarian cancer survival, the above studies provide the basis for an ongoing clinical trial evaluating the role of metformin as an adjunct to chemotherapy, even in nondiabetics.
Table 6 shows a list of ongoing, recruiting, and completed clinical trials as per a search of the clinical trial registration site (accessed 24 December 2015).[76] The most commonly examined target was the mTOR pathway. In addition to the trials mentioned previously, there are phase III trials underway for recurrent LGSOC. There is similarly a phase II study examining the efficacy of inhibition of MAPK and PI3K pathways, as well as a trial of temsirolimus specifically for OCCC.
This is the first meta-analysis in the literature examining the performance of STK inhibitors as a whole for ovarian cancer treatment. There were, however, several notable limitations. By necessity, the studies included in the analysis were comprised of a variety of different drug regimens at different doses in varying ovarian cancer histologies, contributing to a complex analysis. This study did not analyze the impact of differential dosing on response rates or survival. Due to the inclusion of multiple combination regimens, none of which compare regimens with and without an STK inhibitor, it is also difficult to estimate the relative contributions of STK inhibitors and chemotherapy to improved disease outcomes. Furthermore, the meta-analysis included studies with vastly different base patient populations, including variations in platinum status and tumor histology, which makes interpretation of our results difficult. That is, while platinum-sensitive disease was associated with high response rates in one particular trial, our study was not able to differentiate treatment outcomes between platinum-sensitive and -resistant cases. Though two studies notable for high response rates involved LGSOC, we were unable to fully stratify results by tumor histology, which would have greatly strengthened the analysis. The data on STK inhibitors in the select populations mentioned above each came from single trials. These data would be strengthened if combined in the future with data from the ongoing clinical trials mentioned in Table 6 and if supported by appropriate randomized control trials comparing the effects of STK inhibitors with chemotherapy.
Though Sorafenib is a dual-action inhibitor affecting both the RAF/MEK/ERK and VEGFR/PDGFR pathways, its antitumor effects are attributed more to VEGF pathway inhibition than MEK pathway inhibition.[77–79] Therefore, it is debatable if sorafenib can be included in this study. However, the clinical benefit rate of 12 studies excluding sorafenib was 53%, still supporting the role of STK inhibitors as potential agents to maintain SD in the management of recurrent ovarian cancer.
We were unable to stratify results by race, which may affect both mutational status and responsiveness to treatment. Though the majority of trials did not provide data on race, we were able to ascertain that at least 51%, but probably a much larger proportion, of the patients were Caucasian, likely a reflection of the patient populations in North America, Australia, and Europe, where the trials were conducted. In addition, data on adverse events were differentially classified between the studies included. For this reason, we were unable to calculate the actual proportion of patients experiencing side effects in each system organ class. Instead, this study calculated the frequency of adverse events per patient, enabling us to compare the relative occurrence of adverse events but not the proportion of patients that can expect to experience a given effect.
Overall, this study showed a modest impact of STK inhibitors in recurrent ovarian cancer. However, STK inhibitors may have potential to maintain SD in patients with ovarian cancer recurrence, especially in select populations with somatic mutations in STK pathway molecules and in patients with LGSOC. Additional research is needed to confirm this benefit and to separate the effects of STK inhibitors from those of cytotoxic chemotherapy. Further studies are expected to identify additional biomarkers for STK inhibitor efficacy. Potential directions for future inquiry in ovarian cancer include combination use of multiple STK inhibitors and second-generation mTOR inhibitors, combination regimens of STK inhibitors with other class biological agents, the effect of metformin on survival, the effect of STK inhibitors on ovarian cancer stem cells and the prevention of recurrent cancer, and the efficacy of these regimens in specific subsets of patients with LGSOC.
Article highlights.
Clinical outcomes of serine-threonine kinase inhibitors targeting mTOR, MAPK, and aurora kinase pathways for recurrent ovarian cancer were examined in 18 clinical trials.
Median progression-free survival was 3.4 months for STK inhibitor-based therapy with response rate being 13% in largely platinum-resistant study population.
The main benefit of STK inhibitor-based therapy seems to be its ability to maintain disease stability in ovarian cancer recurrence as the majority of patients undergoing this treatment (67%) attained clinical benefit; however, additional research is needed to support this idea.
Treatment responses improved when STK inhibitors were combined with other class biological agents or with cytotoxic agents. This was most notable in a single study of multiple STK inhibitors, where both agents may act together to overcome drug resistance. Randomized controlled trials are needed to directly compare the effects of STK inhibitors to cytotoxic chemotherapy.
Treatment response was significantly improved in selected patient populations with somatic mutations affecting STK inhibitor pathways, with platinum-sensitive disease, and with low-grade serous ovarian carcinoma, indicating the importance of appropriate patient selection when using these drugs.
This box summarizes key points contained in the article.
Acknowledgements
The authors thank Stephen B Gruber for his scientific advice in the preparation of this study.
Declaration of interest
MA Ciccone is supported by the ARCS Foundation Inc and the Los Angeles founder chapter-Margaret Kersten Ponty Postdoctoral Fellowship. Furthermore, K Matsuo is supported by the Ensign Endowment for Gynecologic Cancer Research. Additionally, A Maoz is supported by the Marita and Gary Robb Postdoctoral Fellowship in Oncology. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
References
Papers of special note have been highlighted as:
• of interest
•• of considerable interest
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5–29. [DOI] [PubMed] [Google Scholar]
- 2.Herzog TJ, Pothuri B. Ovarian cancer: a focus on management of recurrent disease. Nat Clin Prac Oncol. 2006;3(11):604–611. [DOI] [PubMed] [Google Scholar]
- 3.Matsuo K, Lin YG, Roman LD, et al. Overcoming platinum resistance in ovarian carcinoma. Expert Opin Investig Drugs. 2010;19 (11):1339–1354.Review discussing the state of therapies for platinum-resistant ovarian cancers. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dancey J mTOR signaling and drug development in cancer. Nat Rev Clin Oncol. 2010;7:209–219. Comprehensive review of the mTOR pathway and targeting drugs. [DOI] [PubMed] [Google Scholar]
- 5.Faivre S, Kroemer G, Raymond E. Current development of mTOR inhibitors as anticancer agents. Nat Rev Drug Discov. 2006;5 (8):671–688. [DOI] [PubMed] [Google Scholar]
- 6.Ducat D, Zheng Y. Aurora kinases in spindle assembly and chromosome segregation. Exp Cell Res. 2004;301(1):60–67. [DOI] [PubMed] [Google Scholar]
- 7.Capra M, Nuciforo PG, Confalonieri S, et al. Frequent alterations in the expression of serine/threonine kinases in human cancers. Cancer Res. 2006;66(16):8147–8154. [DOI] [PubMed] [Google Scholar]
- 8.Liaw D, Marsh DJ, Li J, et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet. 1997;16(1):64–67. [DOI] [PubMed] [Google Scholar]
- 9.Dinulescu DM, Ince TA, Quade BJ, et al. Role of K-ras and Pten in the development of mouse models of endometriosis and endometrioid ovarian cancer. Nat Med. 2005;11(1):63–70. [DOI] [PubMed] [Google Scholar]
- 10.Hemminki A, Tomlisnson I, Markie D, et al. Localization of a susceptibility locus for Peutz-Jeghers syndrome to 19p using comparative genomic hybridization and targeted linkage analysis. Nat Genet. 1997;15(1):87–90. [DOI] [PubMed] [Google Scholar]
- 11.Jenne DE, Reimann H, Nezu J, et al. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet. 1998;18(1):38–43. [DOI] [PubMed] [Google Scholar]
- 12.Connolly DC, Katabuchi H, Cliby WA, et al. Somatic mutations in the STK11/LKB1 gene are uncommon in rare gynecological tumor types associated with Peutz-Jegher’s syndrome. Am J Pathol. 2000;156(1):339–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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 (34):5853–5857. [DOI] [PubMed] [Google Scholar]
- 14.Mabuchi S, Sasano T, Kawano M, et al. Targeting mTOR Signaling in Ovarian Cancer. Curr Obstet Gynecol Rep. 2015;4(1):11–17. Recent comprehensive review of the mTOR pathway in ovarian cancer. [Google Scholar]
- 15.Pallet N, Legendre C. Adverse events associated with mTOR inhibitors. Expert Opin Drug Saf. 2013;12(2):177–186. [DOI] [PubMed] [Google Scholar]
- 16.Robinson MJ, Cobb MH. Mitogen-activated protein kinase pathways. Curr Opin Cell Biol. 1997;9(2):180–186. [DOI] [PubMed] [Google Scholar]
- 17.Burotto M, Chiou VL, Lee J-M, et al. The MAPK pathway across different malignancies: a new perspective. Cancer. 2014;120 (22):3446–3456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Singer G, Oldt R, Cohen Y, et al. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J Natl Cancer Inst. 2003;95(6):484–486. [DOI] [PubMed] [Google Scholar]
- 19.Long GV, Stroyakovskiy D, Gogas H, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet. 2015;386(9992):444–451. [DOI] [PubMed] [Google Scholar]
- 20.Miller CR, Oliver KE, Farley JH. MEK1/2 inhibitors in the treatment of gynecologic malignancies. Gynecol Oncol. 2014;133(1):128–137. [DOI] [PubMed] [Google Scholar]
- 21.Wang D, Boerner SA, Winkler JD, et al. Clinical experience of MEK inhibitors in cancer therapy. Biochim Biophys Acta (BBA) Mol Cell Res 2007;1773(8):1248–1255. [DOI] [PubMed] [Google Scholar]
- 22.Malumbres M, Pérez de Castro I. Aurora kinase A inhibitors: promising agents in antitumoral therapy. Expert Opin Ther Targets. 2014;18(12):1377–1393. [DOI] [PubMed] [Google Scholar]
- 23.Landen CN, Lin YG, Immaneni A, et al. Overexpression of the centrosomal protein aurora-a kinase is associated with poor prognosis in epithelial ovarian cancer patients. Clin Cancer Res. 2007;13 (14):4098–4104. [DOI] [PubMed] [Google Scholar]
- 24.Green MR, Woolery JE, Mahadevan D. Update on aurora kinase targeted therapeutics in oncology. Expert Opin Drug Discov. 2011;6(3):291–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Fu S, Hu W, Kavanagh JJ, et al. Targeting aurora kinases in ovarian cancer. Expert Opin Ther Targets. 2006;10(1):77–85. [DOI] [PubMed] [Google Scholar]
- 26.Stroup DF, Berlin JA, Morton SC, et al. Meta-analysis of observational studies in epidemiology: a proposal for reporting. JAMA. 2000;283(15):2008–2012. [DOI] [PubMed] [Google Scholar]
- 27.Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009. January;45(2):228–247. [DOI] [PubMed] [Google Scholar]
- 28.Common Terminology Criteria for Adverse Events (CTCAE) Guidelines National Cancer Institute; 2015. [cited 2015 Dec 8]. Available from: http://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_40 [Google Scholar]
- 29.Falchook GS, Long GV, Kurzrock R, et al. Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: a phase 1 dose-escalation trial. Lancet. 2012;379(9829):1893–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Welch SA, Hirte HW, Elit L, et al. Sorafenib in combination with gemcitabine in recurrent epithelial ovarian cancer: a study of the princess margaret hospital phase II consortium. Int J Gynecol Cancer. 2010;20(5):787–793. [DOI] [PubMed] [Google Scholar]
- 31.Schwandt A, von Gruenigen VE, Wenham RM, et al. Randomized phase II trial of sorafenib alone or in combination with carboplatin/paclitaxel in women with recurrent platinum sensitive epithelial ovarian, peritoneal, or fallopian tube cancer. Invest New Drugs. 2014;32(4):729–738. Study in meta-analysis with high response rates in patients with platinum-sensitive ovarian cancer. [DOI] [PubMed] [Google Scholar]
- 32.Bedard PL, Tabernero J, Janku F, et al. A phase Ib dose-escalation study of the oral pan-PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin Cancer Res. 2015;21(4):730–738. Study in meta-analysis with high response rates and clinical benefit rates in patients with BRAF/KRAS-mutated ovarian cancers treated with multiple STK inhibitors buparlisib and trametinib. [DOI] [PubMed] [Google Scholar]
- 33.Diamond JR, Bastos BR, Hansen RJ, et al. Phase I safety, pharmacokinetic, and pharmacodynamic study of ENMD-2076, a novel angiogenic and aurora kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2011;17(4):849–860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Farley J, Brady WE, Vathipadiekal V, et al. Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: an open-label, single-arm, phase 2 study. Lancet Oncol. 2013;14(2):134–140. Study in meta-analysis of low-grade serous ovarian carcinoma treated with single STK inhibitor, selumetinib, with a notably high response rate for STK inhibitor monotherapy, and a single patient experiencing a durable complete response. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ganesan P, Piha-Paul S, Naing A, et al. Phase I clinical trial of lenalidomide in combination with sorafenib in patients with advanced cancer. Invest New Drugs. 2014;32(2):279–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lee JM, Sarosy GA, Annunziata CM, et al. Combination therapy: intermittent sorafenib with bevacizumab yields activity and decreased toxicity. Br J Cancer. 2010;102(3):495–499. Study in meta-analysis noting particularly increased response rates and PFS for ovarian cancer patients indicating the possible benefit of combining STK inhibitors with biologic agents. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Matulonis UA, Sharma S, Ghamande S, et al. Phase II study of MLN8237 (alisertib), an investigational aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127(1):63–69. [DOI] [PubMed] [Google Scholar]
- 38.Ramasubbaiah R, Perkins SM, Schilder J, et al. Sorafenib in combination with weekly topotecan in recurrent ovarian cancer, a phase I/II study of the Hoosier oncology group. Gynecol Oncol. 2011;123 (3):499–504. [DOI] [PubMed] [Google Scholar]
- 39.Brana I, Berger R, Golan T, et al. A parallel-arm phase I trial of the humanised anti-IGF-1R antibody dalotuzumab in combination with the AKT inhibitor MK-2206, the mTOR inhibitor ridaforolimus, or the NOTCH inhibitor MK-0752, in patients with advanced solid tumours. Br J Cancer. 2014;111(10):1932–1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wheler JJ, Moulder SL, Naing A, et al. Anastrozole and everolimus in advanced gynecologic and breast malignancies: activity and molecular alterations in the PI3K/AKT/mTOR pathway. Oncotarget. 2014;5(10):3029–3038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.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(1):69–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bodnar L, Gornas M, Szczylik C. Sorafenib as a third line therapy in patients with epithelial ovarian cancer or primary peritoneal cancer: a phase II study. Gynecol Oncol. 2011;123(1):33–36. [DOI] [PubMed] [Google Scholar]
- 43.Oza AM, Elit L, Swenerton K, et al. Phase II study of CGP 69846A (ISIS 5132) in recurrent epithelial ovarian cancer: an NCIC clinical trials group study (NCIC IND.116). Gynecol Oncol. 2003;89(1):129–133. [DOI] [PubMed] [Google Scholar]
- 44.Fu S, Hennessy BT, Ng CS, et al. Perifosine plus docetaxel in patients with platinum and taxane resistant or refractory high-grade epithelial ovarian cancer. Gynecol Oncol. 2012;126(1):47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Behbakht K, Sill MW, Darcy KM, et al. 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. Gynecol Oncol. 2011;123(1):19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Schoffski P, Besse B, Gauler T, et al. Efficacy and safety of biweekly i. v. administrations of the aurora kinase inhibitor danusertib hydrochloride in independent cohorts of patients with advanced or metastatic breast, ovarian, colorectal, pancreatic, small-cell and non-small-cell lung cancer: a multi-tumour, multi-institutional phase II study. Ann Oncol. 2015;26(3):598–607. [DOI] [PubMed] [Google Scholar]
- 47.Romero IL, McCormick A, McEwen KA, et al. Relationship of type II diabetes and metformin use to ovarian cancer progression, survival, and chemosensitivity. Obstet Gynecol. 2012;119(1):61–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kumar S, Meuter A, Thapa P, et al. Metformin intake is associated with better survival in ovarian cancer: a case-control study. Cancer. 2013;119(3):555–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Shah MM, Erickson BK, Matin T, et al. Diabetes mellitus and ovarian cancer: more complex than just increasing risk. Gynecol Oncol. 2014;135(2):273–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Barakat RR, Berchuck A, Markman M, et al. Principles and practice of gynecologic oncology. 6th ed. Philadelphpia (PA): Lippincott Williams & Wilkins; 2013. [Google Scholar]
- 51.Herzog TJ, Scambia G, Kim BG, et al. A randomized phase II trial of maintenance therapy with Sorafenib in front-line ovarian carcinoma. Gynecol Oncol. 2013;130(1):25–30. [DOI] [PubMed] [Google Scholar]
- 52.Hainsworth JD, Thompson DS, Bismayer JA, et al. Paclitaxel/carboplatin with or without sorafenib in the first-line treatment of patients with stage III/IV epithelial ovarian cancer: a randomized phase II study of the Sarah cannon research institute. Cancer Med. 2015;4(5):673–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Polcher M, Eckhardt M, Coch C, et al. Sorafenib in combination with carboplatin and paclitaxel as neoadjuvant chemotherapy in patients with advanced ovarian cancer. Cancer Chemother Pharmacol. 2010;66(1):203–207. [DOI] [PubMed] [Google Scholar]
- 54.Della Pepa C, Tonini G, Santini D, et al. Low grade serous ovarian carcinoma: from the molecular characterization to the best therapeutic strategy. Cancer Treat Rev. 2015;41(2):136–143. [DOI] [PubMed] [Google Scholar]
- 55.Gershenson DM, Sun CC, Wong -K-K. Impact of mutational status on survival in low-grade serous carcinoma of the ovary or peritoneum. Br J Cancer. 2015;113(9):1254–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Grisham RN, Sylvester BE, Won H, et al. Extreme outlier analysis identifies occult mitogen-activated protein kinase pathway mutations in patients with low-grade serous ovarian cancer. J Clin Oncol. 2015;33(34):4099–4105. Mutational analysis of the single patient experiencing complete response on a trial of the single agent STK inhibitor, selumetinib, showing an inactivating mutation affecting MEK1 and investigating mutations that sensitize tumors to MEK inhibitors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mabuchi S, Kawase C, Altomare DA, et al. mTOR is a promising therapeutic target both in cisplatin-sensitive and cisplatin-resistant clear cell carcinoma of the ovary. Clin Cancer Res. 2009;15 (17):5404–5413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Janku F, Wheler JJ, Westin SN, et al. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol. 2012;30(8):777–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Carracedo A, Ma L, Teruya-Feldstein J, et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 2008;118(9):3065–3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Azad NS, Posadas EM, Kwitkowski VE, et al. Combination targeted therapy with sorafenib and bevacizumab results in enhanced toxicity and antitumor activity. J Clin Oncol. 2008;26(22):3709–3714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pal SK, He M, Tong T, et al. RNA-seq reveals aurora kinase driven-mTOR pathway activation in patients with sarcomatoid metastatic renal cell carcinoma. Mol Cancer Res. 2015;13(1):130–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Taga MHE, Ouchi T. Essential roles of mTOR/akt pathway in aurora-a cell transformation. Int J Biol Sci. 2009;5(5):444–450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Song J, Salek-Ardakani S, So T, et al. The kinases aurora B and mTOR regulate the G1-S cell cycle progression of T lymphocytes. Nat Immunol. 2007;8(1):64–73. [DOI] [PubMed] [Google Scholar]
- 64.Li Y, Zhou W, Wei L, et al. The effect of aurora kinases on cell proliferation, cell cycle regulation and metastasis in renal cell carcinoma. Int J Oncol. 2012;41(6):2139–2149. [DOI] [PubMed] [Google Scholar]
- 65.Zhang S, Balch C, Chan MW, et al. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008;68(11):4311–4320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Silva IA, Bai S, McLean K, et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011;71(11):3991–4001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kolev VN, Wright QG, Vidal CM, et al. PI3K/mTOR dual inhibitor VS-5584 preferentially targets cancer stem cells. Cancer Res. 2015;75(2):446–455. [DOI] [PubMed] [Google Scholar]
- 68.Francipane MG, Lagasse E. Therapeutic potential of mTOR inhibitors for targeting cancer stem cells. Br J Clin Pharmacol. 2015. November 26 [Epub ahead of print]. Recent comprehensive review of mTOR inhibitors targeting cancer stem cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Alvero AB, Montagna MK, Holmberg JC, et al. Targeting the mitochondria activates two independent cell death pathways in ovarian cancer stem cells. Mol Cancer Ther. 2011;10(8):1385–1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bodmer M, Becker C, Meier C, et al. Use of metformin and the risk of ovarian cancer: a case–control analysis. Gynecol Oncol. 2011;123 (2):200–204. [DOI] [PubMed] [Google Scholar]
- 71.Tseng C-H. Metformin reduces ovarian cancer risk in Taiwanese women with type 2 diabetes mellitus. Diabetes Metab Res Rev. 2015;31(6):619–626. [DOI] [PubMed] [Google Scholar]
- 72.Shi WY, Xiao D, Wang L, et al. Therapeutic metformin/AMPK activation blocked lymphoma cell growth via inhibition of mTOR pathway and induction of autophagy. Cell Death Dis. 2012;3:e275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vallianou NG, Evangelopoulos A, Kazazis C. Metformin and cancer. Rev Diabet Stud. 2013;10(4):228–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gotlieb WH, Saumet J, Beauchamp M-C, et al. In vitro metformin anti-neoplastic activity in epithelial ovarian cancer. Gynecol Oncol. 2008;110(2):246–250. [DOI] [PubMed] [Google Scholar]
- 75.Rattan R, Giri S, Hartmann LC, et al. Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensable manner. J Cell Mol Med. 2011;15(1):166–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.ClinicalTrials.gov. U.S. National Institutes of Health; [cited 2015 Dec 24]. Available from: https://clinicaltrials.gov [Google Scholar]
- 77.Koh YW, Shah MH, Agarwal K, et al. Sorafenib and Mek inhibition is synergistic in medullary thyroid carcinoma in vitro. Endocr Relat Cancer. 2012;19(1):29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Adnane LTP, Wilhelm SM. Sorafenib (BAY 43–9006, Nexvar), a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature. Methods Enzymol. 2006;407:597–612. [DOI] [PubMed] [Google Scholar]
- 79.Holderfield MNT, Stuart DD. Mechanism and consequences of RAF kinase activation by small-molecule inhibitors. Br J Cancer. 2014;111(4):640–645. [DOI] [PMC free article] [PubMed] [Google Scholar]