Abstract
Resistance to chemotherapy is among the most important issues in the management of ovarian cancer. Unlike cancer cells, which are heterogeneous as a result of remarkable genetic instability, stromal cells are considered relatively homogeneous. Thus, targeting the tumor microenvironment is an attractive approach for cancer therapy. Arguably, anti-vascular endothelial growth factor (anti-VEGF) therapies hold great promise, but their efficacy has been modest, likely owing to redundant and complementary angiogenic pathways. Components of platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), and other pathways may compensate for VEGF blockade and allow angiogenesis to occur despite anti-VEGF treatment. In addition, hypoxia induced by antiangiogenesis therapy modifies signaling pathways in tumor and stromal cells, which induces resistance to therapy. Because of tumor cell heterogeneity and angiogenic pathway redundancy, combining cytotoxic and targeted therapies or combining therapies targeting different pathways can potentially overcome resistance. Although targeted therapy is showing promise, much more work is needed to maximize its impact, including the discovery of new targets and identification of individuals most likely to benefit from such therapies.
Keywords: Ovarian cancer, targeted therapy, angiogenesis, anti-vascular agent, resistance to anti-VEGF therapy
1. Introduction
The evolution of surgical techniques and chemotherapy regimens over the past three decades has resulted in improvements in the survival of women with ovarian cancer [1, 2]. Debulking surgery remains a cornerstone of ovarian cancer treatment, and the platinum–paclitaxel combination regimen is established as a first-line treatment for advanced ovarian cancer that yields response rates of over 80% and complete response rates of 40–60% [1, 3–6]. However, most of the patients who respond to treatment eventually experience a relapse, with a median progression-free survival (PFS) of 18 months [7]. At relapse, patients with platinum-sensitive disease might be treated with the same drugs when the treatment-free interval is greater than 6 months. Patients who develop platinum-resistant or refractory disease are treated with a range of other drugs [8]. However, improving cure rates remains a critical unmet need.
Improvements in the understanding of cancer biology and the underlying mechanisms governing the cancer process have facilitated the development of targeted therapies, including smallmolecule inhibitors and monoclonal antibodies (MoAbs). These agents target tumor cells, surrounding stroma, tumor vasculature, and aberrant cellular signaling mechanisms. However, because of the lack of predictive markers, identifying individual patients who are most likely to benefit from specific targeted drugs has had limited success. Moreover, cancer cells can adapt to various therapies due to their remarkable genetic instability. This trait is especially important for ovarian cancer therapy. In contrast, stromal cells are non-malignant and thought to be genetically stable. Some studies, however, do suggest possible adaptive mechanisms in stromal populations as well [9–11].
Efforts to target tumor angiogenesis have focused on the VEGF pathway. Angiogenesis is one of the hallmarks of cancer. This feature contributes not only to tumor growth but also to tumor cell invasion [12]. Indirectly killing tumors by compromising their vasculature is an attractive anticancer treatment approach because resistance is theoretically less likely to appear in endothelial cells (ECs). Bevacizumab, a MoAb targeting vascular endothelial growth factor-A (VEGF-A), is a good example of a tumor vasculature-targeting agent. Bevacizumab has been approved by the Food and Drug Administration (FDA) for treating colon, lung, kidney, and brain cancers. Anti-VEGF therapy has shown potential for the treatment of some cancers but has not been as efficacious as expected [13]. Moreover, resistance to anti-VEGF treatment has been observed in tumor cells and components of the tumor microenvironment.
Besides VEGF, several other growth factors have a significant proangiogenic effect, including platelet-derived growth factors (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), and angiopoietins [14]. These pathways are taken into consideration when developing combination therapies to overcome resistance to anti-VEGF therapy. In this article, we will review the basic mechanisms of tumor microenvironment interactions in ovarian cancer angiogenesis and ongoing clinical trials of second-generation vascular targeting drugs that target pathways other than VEGF.
2. Vascular disrupting agents (VDAs)
VDAs target ECs and pericytes of the resident or established tumor vasculature, which results in tumor ischemia and necrosis. Selective vascular shutdown suggests a structural difference between the endothelium of tumor vessels and that of non-tumor vessels. Indeed, tumor vasculature is marked by a high rate of EC proliferation, a reduction in the extent of pericyte coverage, abnormalities in the basement membrane, and often, increased vascular permeability. VDA treatment results in structurally disorganized, tortuous, thin-walled vessels that lack smooth muscle and have reduced physiological regulation [15]. VDAs can be divided into two categories: tubulin-destabilizing agents and flavonoids.
2. A. Tubulin destabilizers
ECs are highly dependent on their tubulin cytoskeleton for their function [16]. Most VDAs induce changes in EC shape by disrupting the cytoskeleton and cell-to-cell junctions. This results in increased permeability to proteins and increased interstitial fluid pressure, which can be sufficient to reduce vessel diameter. Plasma leakage also leads to increased blood viscosity, which results in decreased blood flow and rouleaux formation. Another factor contributing to vascular shutdown is the activation of platelets through contact with basement membrane components, which are exposed. This cascade of events results in vascular occlusion more selectively in tumor endothelium than in normal endothelium [17].
Combretastatins, structurally related to colchicine, destabilize the endothelial cytoskeleton by binding to tubulin and induce microtubule depolymerization. Combretastatin A4 phosphate (CA4P) is a lead compound in its class. It has antivascular and antitumor activities in preclinical models [18]. Animal studies have revealed that CA4P induces a high level of blood flow reduction in tumors compared to its effects in normal organs such as the spleen, skeletal muscle, and brain; and causes no significant decrease in heart, kidney, and intestine[19]. However, preclinical studies suggest that following exposure to a VDA, only the center of the tumor becomes necrotic, with a viable rim remaining in the periphery. This rim of viable tumor cells is highly proliferative and has potential for tumor growth [20]. This phenomenon provides the rationale for developing combination therapy with VDAs and anti-angiogenesis or conventional cytotoxic drugs [21]. Both CA4P and bevacizumab can cause hypertension, but a significant increase in blood pressure was not observed when the drugs were used in combination. Other combretastatin derivatives include the serine-linked amino-derivative AVE8062 and the combretastatin A-1 derivative OXi4503.
Bevacizumab and CA4P combination therapy was tested in advanced solid tumor patients (63 mg/m2 CA4P + 10 mg/kg bevacizumab q14) [21]. A total of 15 patients were enrolled. Nine of 14 patients experienced disease stabilization. A patient with ovarian cancer had a CA125 response lasting for more than a year. Dynamic contrast-enhanced magnetic resonance imaging showed statistically significant reductions in tumor perfusion/vascular permeability, which reversed after CA4P alone but which were sustained following bevacizumab. A randomized phase II trial of CA4P plus bevacizumab versus bevacizumab alone has completed accrual in patients with recurrent, persistent ovarian cancer (NCT01305213). CA4P is also being tested in combination with paclitaxel and carboplatin in phase II trials. Patients with ovarian cancer that had relapsed and who could start trial therapy within 6 months of their last platinum chemotherapy were given 63 mg/m2 CA4P at a minimum of 18 hours before 175 mg/m2 paclitaxel and carboplatin area under the concentration curve (AUC ) 5, repeated every 3 weeks. Six (13.5%) of the 44 patients had disease response by Response Evaluation Criteria in Solid Tumors criteria, and 15 (34%) by Gynecologic Cancer InterGroup CA 125 criteria. Hypertension (23% of patients) was controlled by glyceryl trinitrate or prophylactic amlodipine. CA4P followed by paclitaxel and carboplatin is well tolerated and appears to produce a higher response rate than chemotherapy alone group.[22]
2. B. Flavonoids
5, 6-Dimethylxanthenone-4-acetic acid (ASA-404) is a flavonoid derivative that damages DNA and induces apoptosis in ECs in preclinical models. The exact mechanism that leads to tumor cell death remains unknown but involves nuclear factor kappa B (NF-κB), serotonin, tumor necrosis factor alpha (TNF-α), and nitric oxide [23]. To our knowledge, no clinical studies of flavonoids for ovarian cancer therapy have been carried out.
3. PDGF receptor (PDGFR) pathway
PDGF molecules are key regulatory molecules in oncogenesis and angiogenesis and play an important role in ovarian cancer. Four isoforms of the PDGF molecule have been identified: PDGF A-D [24, 25]. PDGF is a dimeric molecule composed of two disulfide-bound chains [26], and it binds to specific receptor isoforms to exert their effects. PDGF-A and PDGF-C bind to PDGFR-α, whereas PDGF-B and PDGF-D bind to PDGFR-β [27]. Thus, PDGF-BB can bind PBGF-β, and PDGF-AB can bind both PDGFR-α and PDGFR-β [27]. At the site of vessel sprouting, ECs secrete PDGF-BB as a chemoattractant for surrounding pericytes to stabilize endothelial channels. Newly formed vessels must be mature and covered by pericytes to function properly. Several growth-factor families, such as PDGFs, angiopoietins, and TGF-β, contribute to this maturation process [28]. Secreted PDGF-BB interacts with heparan sulfate at the EC surface or in the periendothelial matrix. This deposition creates a concentration gradient of PDGF-BB, which, in turn, is critical for the correct investment of pericytes in the vessel wall [29]. Disruption of endothelial-pericyte associations results in excessive regression of vascular loops and abnormal remodeling [30].
Pericytes respond to stimulation created by PDGF-BB concentration gradients and are activated by the dimerization of PDGFR-β [31, 32]. Dimerization of PDGFR leads to autophosphorylation of tyrosine residues in the intracellular domain [25]. Upon activation of the PDGF pathway, signaling occurs via the phosphatidylinositol 3-kinase/protein kinase-B (PI3K/Akt) pathway, and mitogen-activated protein kinase (MAPK) molecules are involved alongside proteins of the Src family and phospholipase C-γ [33]. Other molecules related to PDGF signaling include Ras [34], signal transducer and activator of transcription (STAT), and guanine-5'-triphosphatase (GTPase) activating protein [35]. PDGF also induces cell growth and survival [36], transformation [37], migration, vascular permeability, and wound healing [38]. In the tumor vasculature, pericytes express PDGFRs, which play an important role in angiogenesis by recruiting more pericytes and also influence resistance to anti-VEGF therapy. PDGF-BB chemoattracts pericytes that express PDGFR-β [39, 40]. Therefore, pericyte deficiency after PDGF-B ablation causes vessel leakage, tortuosity, microaneurysm formation, and bleeding [41].
The PDGF/PDGFR axis contributes to resistance to anti-VEGF therapy through several mechanisms. Pericytes support EC survival during anti-VEGF therapy in a paracrine manner. PDGF-BB-related activation of PDGFR-β can stimulate pericytes to produce VEGF [32], and VEGF production from pericytes can protect ECs from VEGF withdrawal and confer resistance to VEGF blockade by close EC–pericyte interaction. PDGF-BB also acts on pericytes that express PDGFR-β to expand the stromal compartment and activate erythropoietin expression, which leads to enhanced tumor angiogenesis [42]. Increased expression of PDGF-CC by tumor-associated fibroblasts can also confer resistance against anti-VEGF treatment. PDGF-CC stimulates vessel growth and maturation and attenuates the response to anti-VEGF therapy [43]. The effect on tumor angiogenesis of PDGF-CC is mediated by its receptors, PDGFR-α and PDGFR-β, which are expressed by ECs, tumor-associated fibroblasts, and bone marrow-derived cells [43].
PDGFR inhibition decreases tumor growth by causing pericyte detachment, which leads to immature vessels that are prone to regression [44]. Anti-PDGF/PDGFR drugs (e.g., imatinib, an anti-PDGFR antibody and aptamers) are largely ineffective in tumors as monotherapy because these drugs can potentially make the tumor vasculature more immature, a state that is characterized by decreased pericyte coverage [32]. PDGFR inhibitors are shown in Tables 1–7. However, these drugs might enhance the efficacy of anti-VEGF drugs by making the ECs more sensitive [44–46]. Initial studies using multitargeted receptor tyrosine kinase inhibitors (TKIs) showed that blocking PDGF-BB increased sensitivity to anti-VEGF therapy by depleting the mature vessels of pericytes [39]. In xenograft models of melanoma and pancreatic cancer, VEGFR and PDGFR inhibition by tyrosine kinase resulted in detachment of pericytes and decreased tumor burden and vascularization [45, 47]. However, there are also potential disadvantages to PDGFR blockade for cancer therapy. Inhibition of vessel maturation can promote malignancy. In primary tumors, pericytes are a barrier to cancer cell intravasation. Because leaky vessels that are not covered by pericytes are not sufficient barriers, tumor cell dissemination can be facilitated by inhibiting PDGFR [48]
Table 1.
Drug | Targets | FDA-approved indication |
---|---|---|
Cediranib | VEGFR-1, -2 and -3, PDGFR, and c-kit | N/A |
Sorafenib | VEGFR, PDGFR, c-kit, FLT-3, and -RAF | HCC, RCC |
Imatinib | PDGFRs and c-kit | Leukemia, GIST |
Nintedanib | VEGFR, PDGFR, and FGFR | N/A |
Sunitinib | VEGFR, PDGFR, c-kit, c-FLT-3, and c-RET | GIST, RCC, HCC, neuroendocrine tumors |
Pazopanib | VEGFR, PDGFR, FGFR, and c-kit | Sarcoma, RCC |
VEGFR: vascular endothelial growth factor receptor, PDGFR: platelet-derived growth factor receptor, FGFFR: fibroblast growth factor receptor, HCC: hepatocellular carcinoma, RCC: renal cell carcinoma, GIST: gastrointestinal stromal tumor
Table 7.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
PC±pazopanib | I, II | Resistance or refractory EOC, PPC, or FC | Recruiting (NCT01402271) | |
P±pazopanib | II | Resistance or refractory | Ongoing (NCT01468909) | |
Pazopanib | II | Recurrent EOC, PPC, or FC | 3 PR (CA125 response, n=18) | [65] |
Gemcitabine±pazopanib | II | Persistent or recurrent EOC,PPC, or FC | Recruiting (NCT01610206) | |
wP±papzopanib (MITO-11) | II | Platinum resistance or refractory ovarian cancer | Ongoing (NCT01644825) | |
Pazopanib+PLD | I, II | Platinum resistance or sensitive EOC, PPC, or FC | Ongoing (NCT01035658) | |
Pazopanib | II | Platinum-resistant EOC, PPC, or FC | Ongoing (NCT01262014) | |
Pazopanib+P (PAZPET-1) | I | Platinum resistance ovarian cancer | Recruiting (NCT01608009) | |
Pazopanib+cyclophosphamide | I,II | Platinum resistance or refractory EOC, PPC, or FC | Recruiting (NCT01238770) | |
Pazopanib+weekly topotecan (TOPAZ) | I,II | Platinum resistance/intermedia te sensitivity EOC PPC,or FC | Recruiting (NCT01600573) | |
Pazopanib±Fosbreta bulin (PAZOFOS) | I, II | Recurrent EOC, PPC, or FC | Not yet recruiting (NCT02055690) | |
Pazopanib(maintenance) | III | After first-line chemotherapy | Ongoing (NCT00866697) |
Phase II trials using imatinib, c-Abl, Abl-related gene (Arg/Abl2), PDGFR, and c-kit inhibitors all showed minimal activity in ovarian cancer patients [49–51]. Because of its limited effect on ovarian cancer, imatinib was tested with cytotoxic agents. Combining imatinib with docetaxel did not improve efficacy over expected outcomes with docetaxel alone but the toxicity of that regimen was tolerable. Another study of imatinib in combination with weekly paclitaxel demonstrated 50% of patients were free of disease progression at 12 weeks and 5 of 12 patients had a PFS of more than 6 months, including 2 of the 12 with a PFS of more than 12 months [52]. Again, the improvement in clinical activity over weekly paclitaxel alone remains to be determined. (Tables 1, 2)
Table 2.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Imatinib+docetaxel | II | Platinum-refractory EOC, FT, or PPC | 4PR, 3SD(n=23) | [214] |
Imatinib | II | Recurrent resistant EOC, FT, or PPC | Completed(NCT00510653) | |
Imatinib+gemcitabine | II | Persistent, refractory EOC, FT, or PPC | Completed(NCT00928642) | |
Imatinib | II | Refractory or relapsed EOC | 2DS(n=23) | [51] |
Imatinib+paclitaxel | II | Recurrent EOC, Mullerian origin cancer | Terminated( NCT00840450) | |
Imatinib | II | Progressive refractory non-epithelial ovarian cancer (germ-cell tumor) | Terminated(NCT00042952) | |
Imatinib | II | Taxane-refractory EOC or PPC | Completed(NCT00036751) | |
Imatinib | II | Persistent recurrent EOC or PPC | Completed(NCT00041041) | |
Imatinib + wP | II | Recurrent persistent EOC, FT, or PPC | 4PR(n=12) | [52] |
Imatinib (maintenance) | II | CR patient | [215] |
EOC: epithelial ovarian carcinoma, PPC: primary peritoneal carcinoma, FC: fallopian tube carcinoma, PR: partial response, SD: stable disease, CR: complete response. P: Paclitaxel, wP: weekly paclitaxel
Multiple TKIs besides imatinib are used to target the PDGFR pathway. Cediranib is a TKI of VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-α and c-kit. In a phase II study of cediranib monotherapy for recurrent epithelial ovarian cancer(EOC), primary peritoneal cancer(PPC), and fallopian tube cancer(FC), partial responses were documented in 17%, and 13% of patients had stable disease; there were no complete responses [53]. Median PFS was 5.2 months, and 17% of patients were free of progression at 6 months. At the dose administered in this trial (initially 45 mg orally, daily, reduced to 30 mg orally, daily), 11 patients (23%) were removed from the study because of toxicities before 2 cycles. Grade 3 toxicities (>20% of patients) included hypertension (46%), fatigue (24%), and diarrhea (13%). Grade 2 hypothyroidism occurred in 43% of patients. Grade 4 toxicities included central nervous system hemorrhage (n = 1), hypertriglyceridemia/hypercholesterolemia (n = 1), and dehydration/elevated creatinine (n = 1). No bowel perforations or fistulas occurred [53]. In a designed phase III trial (ICON6) [54], cediranib was investigated in combination with platinum-based chemotherapy. The three-arm trial randomized platinum-sensitive patients 2:3:3 to chemotherapy plus placebo followed by 18 months of placebo maintenance (reference arm), chemotherapy plus cediranib followed by 18 months of placebo maintenance (concurrent arm) and chemotherapy plus cediranib followed by 18 months of cediranib (maintenance arm). Acceptable chemotherapy regimens included single agent carboplatin, carboplatin/paclitaxel or carboplatin/gemcitabine for 6 cycles followed by regular monitoring for at least 18 months or until progression of daily cediranib for recurrent platinum-sensitive ovarian cancer. The stage I analysis of ICON6 demonstrated that it is feasible to add cediranib (initially 30 mg orally, daily) to platinum-based therapy but was better tolerated at 20 mg orally, daily, used in the randomized phase II stage II to evaluate the treatments’ effects of the trial. Due to a decision to suspend development of cediranib, the phase III program was halted and the trial data based on 456 patients was recently reported. The primary endpoint was changed from OS to PFS and the primary analysis was the maintenance arm versus the reference arm. Approximately 70% of the treatment cohort had a platinum-free interval of at least 12 months and 90% of patients received one of the two allowable combination platinum regimens. The primary endpoint (PFS) was significantly longer in the maintenance arm (median 11.1 vs. 8.7 months, HR: 0.57, 95% CI: 0.45–0.74). Surprisingly, OS was also significantly longer in this arm relative to reference, at a median 26.3 months versus 20.3 months (HR: 0.70, 95% CI: 0.51–0.99). Hypertension, nausea and diarrhea were each experienced more frequently in the cediranib arms relative to placebo, but only the latter was significant more commonly in the maintenance setting. Grade 3 and grade 4 events were uncommon. [54] (Table 3)
Table 3.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Olaparib±cediranib | I,II | Recurrent ovarian or breast cancer | Ongoing (NCT01116648) | |
Cediranib | I | Recurrent EOC, PPC, or FC | 7 PR (n = 20) | [216] |
Cediranib | II | Recurrent EOC, PPC, or FC | 8 PR (n = 47) | [217] |
Cediranib | II | Persistent, recurrent, or refractory EOC, PPC, or FC | Ongoing (NCT00278343) | |
Cediranib maleate + RO4929097 | I | Advanced solid tumors (including ovarian cancer) | Ongoing (NCT01131234) | |
Cediranib + temsirolimus | I | Advanced gynecologic cancers | Ongoing (NCT01065662) | |
Cediranib + AZD0530 | I | Advanced solid tumors (including ovarian cancer) | Completed(NCT00475956) | |
platinum based chemotherapy±cediranib ± maintenance cediranib (ICON6) | II, III | Platinum sensitive, recurrent EOC, PPC, or FC | Ongoing (NCT00532194) |
Sorafenib is an inhibitor of VEGFR-1, VEGFR-2, and VEGFR-3, PDGFR-β, and Raf-1 tyrosine kinase activity [55]. It is approved for the treatment of unresectable hepatocellular carcinoma and advanced renal cell carcinoma by the FDA [56, 57]. Matei and colleagues evaluated sorafenib alone (400 mg orally twice daily) in patients with recurrent ovarian cancer or primary peritoneal cancer; 24% of the patients had stable disease for 6 months, and 3.4% of patients had a partial response [58]. This modest response was further hindered by substantial toxicity. Sorafenib has been evaluated in a phase II trial in combination with chemotherapeutic agents (gemcitabine and topotecan) in recurrent ovarian cancer and showed modest effect [59, 60]. Currently, a phase II trial of sorafenib in combination with carboplatin and paclitaxel for first-line treatment of ovarian cancer and in combination with bevacizumab for refractory ovarian cancer is under way (Table 4). A double-blind, randomized, placebo-controlled, phase II study to assess the efficacy and safety of maintenance therapy with sorafenib was performed. In this trial, 246 patients were randomized 1:1 to receive either sorafenib or placebo; the primary endpoint was PFS. There was no significant difference between sorafenib and placebo arms for PFS (median 12.7 vs. 15.7 months; hazard ratio 1.09; 95% CI 0.72–1.63). More patients receiving sorafenib versus placebo required dose reductions of sorafenib (67.5% vs. 30.1%), resulting in a lower than planned median daily dose (median 584.6 vs. 800.0 mg). Treatment with sorafenib was of shorter duration (median 17.6 vs. 51.9 weeks) with more frequent discontinuations due to adverse effects (37.4% vs. 6.5%). Although assessment of efficacy was limited by the high rate of dose reductions and early discontinuations, sorafenib 400 mg BID cannot be recommended as maintenance therapy for patients with OC in complete remission [61].
Table 4.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Sorafenib + topotecan | I,II | Platinum-resistant EOC or PPC | 14 SD, 5 PR (n = 30) | Terminated (NCT00526799) [60] |
Sorafenib+topotecan(TRIAS 2009) | I,II | Platinum resistant EOC, PPC or FC | (NCT01047891) | |
PC±Sorafenib | II | First-line chemotherapy in FIGO stage III-IV EOC, PPC, or FC | Ongoing (NCT00390611) | |
Sorafenib + gemcitabine | II | recurrent or refractory EOC, PPC | 2 PR, 10 SD (n = 33) | [59] |
Sorafenib | At least the second remission in EOC, PPC, or FC | Terminated (NCT00522301) | ||
PC ± sorafenib | II | Recurrent EOC or PPC | Suspended (NCT00096200) | |
Sorafenib + bevacizumab | II | Refractory EOC or PPC | Ongoing (NCT00436215) | |
Sorafenib (maintenance) | II | CR with EOC or PPC | No effect | [61] |
Sorafenib | II | Persistent recurrent EOC or PPC | 20 SD, 2 PR (n=59) | [58] |
Sorafenib | II | Two prior cytotoxic treatments; recurrent, refractory EOC or PPC | 4 SD (n = 11) | [218] |
PC: paclitaxel and carboplatin
Nintedanib (BIBF 1120) is a potent inhibitor of VEGFR, PDGFR, and FGFR. In a randomized, phase II, placebo-controlled trial, patients who had just completed chemotherapy for relapsed ovarian cancer with evidence of response but at high risk of early recurrence were treated with Nintedanib. The progression free rates were 16.3% and 5.0% in the nintedanib and placebo groups, respectively (hazard ratio [HR] 0.65; 95% confidence interval [CI], 0.42 to 1.02; P = .06) [62]. This prompted a phase III trial (NCT01015118) in women with newly diagnosed advanced stage (stage II-IV) ovarian cancer following primary cytoreduction (Table 5). The trial randomized 1,366 women at a 2:1 ratio to paclitaxel/carboplatin plus concomitant and 120 weeks of nintedanib maintenance or paclitaxel/carboplatin plus placebo. Approximately 51% of the population had no residual disease after primary debulking. The experimental arm was associated with a 16% reduction in the hazard for progression (primary endpoint measured as RECIST and/or CA125) with a median PFS of 17.3 months versus 16.6 months (HR: 0.84 95% CI: 0.72–0.98). The combination was associated with higher frequencies of non-hematologic and hematological toxicity and led to significantly higher rate of treatment discontinuation although the time to treatment discontinuation was not shorter in the experimental arm.
Table 5.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Cyclophosphamid e±nintedanib | II | Recurrent EOC, PPC, or FC | Not yet recruiting (NCT01610869) | |
Nintedanib | II | Bevacizumab-resistant, persistent, or recurrent EOC, PPC, or FC | Recruiting (NCT01669798) | |
Nintedanib+PLD | I,II | Platinum-resistant, refractory EOC | Recruiting (NCT01485874) | |
Nintedanib+PLD +C | I | Advanced, platinum sensitive relapsed EOC, PPC, or FC | Ongoing (NCT01314105) | |
Nintedanib B+PLD+C | I | Advanced, platinum-sensitive relapsed | Completed (NCT01329549) | |
Nintedanib | II | Addition to first-line chemotherapy with interval debulking surgery in patients of EOC, PPC, or FC | Recruiting (NCT01583322) | |
Nintedanib (maintenance) | II | Following chemotherapy in patients with relapsed EOC, PPC, or FC | PFR: 16.3% (nintedanib) vs. 5.0% (placebo) | [62] |
PC±nintedanib | III | First-line therapy in advanced EOC, PPC, or FC | Ongoing (NCT01015118) |
C: carboplatin, PLD: pegylated liposomal doxorubicin, PFR: progression free survival
Sunitinib, also a multikinase inhibitor, blocks VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-β, and RET [63]. It is currently FDA-approved for advanced renal cell carcinoma and gastrointestinal stromal tumors [63]. A phase II trial of sunitinib monotherapy for the treatment of patients with recurrent EOC and PPC resulted in a partial response rate of 3.3%; 53% of patients had stable disease [64] (Table 6).
Table 6.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Sunitinib | II | Recurrent ovarian clear cell carcinoma | Recruiting (NCT01824615) | |
Sunitinib | II | Persistent or recurrent clear cell ovarian cancer | Ongoing (NCT0979992) | |
Sunitinib | II | Recurrent EOC, FC, or PPC | 1 PR, 16 SD (n = 30) | [64] |
Sunitinib | II | Refractory or relapsed germ cell tumors | Completed (NCT00453310) | |
Sunitinib | II | Recurrent and refractory EOC, FC, or PPC | 3 PR (n = 36) | [219] |
Sunitinib | II | Recurrent platinum-resistant ovarian cancer | Not continuous: 6 PR(n = 36) Continuous: 2 PR (n = 37) | [220] |
Pazopanib is an inhibitor of VEGFR-1, VEGFR-2, VEGFR-3, PDGFR-α, PDGFR-β, and c-kit. Pazopanib is approved for the treatment of patients with advanced renal cell carcinoma and advanced sarcoma. A phase II trial tested activity in patients with recurrent EOC, PPC, and FC, and the CA-125 response rate was 31%. No patients with measurable disease had a partial or complete response. The PFS at 6 months was 17%. The most common adverse events leading to discontinuation of the study drug were grade 3 alanine aminotransferase (8%) and aspartate transaminase (8%) elevation. Only one grade 4 toxicity (peripheral edema) was reported [65]. Recently, a double-blind, placebo-controlled, phase III clinical study of pazopanib was reported in women with ovarian cancer who achieved a partial or complete response to primary platinum-based adjuvant chemotherapy. In this trial, 940 women (90% stage III-IV) were randomized 1:1 to pazopanib or placebo for up to 24 months. The primary endpoint was PFS. The majority of the women (>85%) had achieved a complete clinical response prior to enrolling into the study. Pazopanib use significantly reduced the risk of progression (RECIST) by 23% (HR: 0.77, 95% CI: 0.64–0.91) represented by a difference in the median PFS of 4.6 months (12.3 months to 17.9 months). The toxicity assessment was similar to other trials with the agent and was associated with a treatment discontinuation rate of 14%. OS was immature but did not appear to be prolonged at the time of the report. (NCT00866697) (Table 7)
4. EGF receptor (EGFR) pathway
The EGFR family consists of four members: EGFR (erbB1), HER2/neu (erbB2), HER3 (erbB3), and HER4 (erbB4). More than 30 ligands to EGFR family proteins have been identified in humans [66]. Dimerization of EGFR proteins is a critical step for transmission of signals, and activation of EGFR and HER2/neu induces a cascade of downstream signaling through several pathways (e.g., Ras/Raf/MAPK, PI3K/Akt/mTOR, Src, and JAK/STAT), resulting in cellular proliferation, differentiation, survival, motility, adhesion, and repair [67].
The EGFR pathway has a role in generating resistance to anti-VEGF therapy. Cascone et al. showed that increased activated EGFR was detected on pericytes of xenografts that acquired resistance to anti-VEGF therapy. They also showed that dual targeting of the VEGF and EGFR pathways reduced pericyte coverage and increased PFS in mouse xenograft model of human lung adenocarcinoma [68]. On the other hand, blockade with an anti-EGFR antibody resulted in the selection of tumor cell subpopulations with increased angiogenic potential [69]. Based on this knowledge, a phase II clinical trial was performed in patients with recurrent or refractory EOC, PPC, and FC without any prior treatment with anti-VEGF or anti-EGFR agents [68]. A total of 13 patients were enrolled. Bevacizumab 15 mg/kg was administered intravenously every 21 days, and the EGFR inhibitor erlotinib, 150 mg orally, was given daily. One complete response was observed for more than 16 months, and one partial response was observed for more than 11 months. Seven patients showed stable disease as a best response. Two patients had fatal gastrointestinal perforation, and the study was therefore stopped. [70]. The largest trial of an EGFR inhibitor in ovarian cancer was conducted by the EORTC, which randomized 835 patients 1:1 to erlotinib (150 mg daily for 24 months) or observation. The primary endpoint, PFS was similar between the groups at a median 12.7 months versus 12.4 months. Similarly, no difference in OS was observed 51 months versus 59 months. Activating EGFR mutations were present in less than 1% of the study population and downstream pathway activators were present in about 7% of cases [70].
Finally, small molecule inhibitors targeting VEGFR and EGFR have been developed and at least one (vandetanib) has been reported in combination with chemotherapy in patients with recurrent platinum-resistant disease. Coleman and colleagues randomized 129 patients with platinum-resistant recurrent ovarian cancer 1:1 to vandetanib in combination with docetaxel versus docetaxel alone. Allowance for crossover to single agent vandetanib was optional. The primary endpoint was PFS and was not significantly improved in the combination arm (median 3.0 months versus 3.5 months, respectively). Similar findings were observed relative to OS. The regimen was well tolerated and reflective of the relatively low dose of the agent (100 mg orally, daily). [71].
5. FGF receptor (FGFR) pathway
FGFs that signal through FGF receptors (FGFRs) regulate development, controlling many events from mesoderm patterning in the early embryo [72], all the way to the development of multiple organ systems [73]. FGF signaling extends to many physiological roles in the adult organism as well, including the regulation of angiogenesis and wound healing [74]. The family of FGFs consists of at least 23 secreted glycoproteins, of which 18 are true ligands (FGFs 1–20 and 16–23) [74]. FGFs are sequestered to the extracellular matrix (ECM) by heparan sulphate proteoglycans. Upon release from the ECM by haparanase, proteases, or specific FGF-binding proteins, FGFs bind to FGFRs. The FGFRs comprise the four transmembrane receptor tyrosine kinases FGFR1, FGFR2, FGFR3, and FGFR4 [75]. FGF triggers the autophosphorylation of FGFR at a key tyrosine residue in an activation loop of the tyrosine kinase domain [76]. Activated FGFR phosphorylates FGF receptor substrate 2 to recruit the growth factor receptor-bound protein 2 (GRB2) adaptor molecule [74, 77]. FGF signals can be transduced to the RAS/MAPK or PI3K/Akt signaling cascades, Src tyrosine kinase, and STATs [78]. FGF increases cellular proliferation through the RAS/MAPK pathway, whereas FGF supports cellular survival through the PI3K/Akt pathway [79].
FGFs activate their receptors on ECs or indirectly stimulate angiogenesis by inducing the release of angiogenic factors from other cell types [74]. Among the molecules of the FGF/FGFR pathway, FGF1 and FGF2 are potent proangiogenic factors [80]. FGFR1 and FGFR2 are found in ECs [81]. Released FGFs from ECM stimulate tumor cells in an autocrine manner, whereas ECs are stimulated in a paracrine manner. During tumor angiogenesis, FGFs increase EC proliferation by activation of MAPK and long-lasting activation of protein kinase C, whereas migration is caused by activation of MAPK and downregulation of protein kinase C [82, 83].
The FGF pathway has crosstalk with other angiogenesis pathways, and this crosstalk may confer resistance to anti-VEGF therapy [84]. FGF2 shows synergistic effects with VEGF and PDGF-BB [85–87]. FGF2 upregulates the expression of PDGFR to increase responsiveness to PDGF-BB; likewise, PDGF-BB-treated vascular smooth muscle cells may lead to upregulation for FGFR1 to increase responsiveness to FGF2 [88]. FGF2 also increases the expression of other proangiogenic factors including angiopoietin-2 and VEGF [89, 90]. In the clinical setting, a study of cytokine and growth factor profiles at response and upon progression after a single dose of bevacizumab-containing therapy demonstrated that FGF2 levels were elevated after tumor progression in patients with colorectal cancer and glioblastoma [91, 92]. Pazopanib and nintedanib inhibit FGFR’s role as a small molecule tyrosine kinase inhibitor, however other agents with selective FGFR targeting are under development.
6. Angiopoietin/Tie pathway
The angiopoietin/Tie family is an on/off switch for angiogenesis. The angiopoietin family consists of two receptors, Tie1 and Tie2, and three ligands, Ang1 and Ang2 and the orthologous Ang3 in mice or Ang4 in humans. Ang1 has agonistic functions to Tie2, and Ang2 has a competitive antagonist function that is controlled in a context-dependent manner [93]. Because no ligand for Tie1 has been identified, this orphan receptor may act as a negative regulator of Tie2, but its precise role remains elusive [94]. Ang1 is produced by pericytes and smooth muscle cells; it activates endothelial Tie2. Ang1 strengthens interactions between ECs and pericytes in a paracrine manner, is expressed behind the leading edge of angiogenic vessels, and plays an important role in vessel maturation [95]. In contrast, Ang2 is released by angiogenic tip cells. At the cue of angiogenesis, pericytes first detach from the vessel wall (in response to Ang2) and liberate themselves from the basement membrane by proteolytic degradation, which is mediated by matrix metalloproteinases [96]. Ang2 expression is confined to the vascular remodeling area to destabilize the growing edge of ECs and works as a competitive antagonist of Ang1. At this point of EC destabilization, Ang2 binds to Tie2 in an autocrine manner without inducing signal transduction [97, 98]. However, the role of Ang2 appears to be more contextual. When VEGF is absent, Ang2 destabilizes vessels by inhibiting Ang1 signaling, but in the presence of VEGF, Ang2 facilitates vascular sprouting [39]. The relative Ang1:Ang2 ratio may also be important. When Ang1 expression is greater, the vasculature remains quiescent. Conversely, when Ang2 expression is dominant, angiogenesis is favored [99]. Tumor-derived Ang2 promotes angiogenesis by recruiting monocytes expressing Tie2 [100]. Overexpression of Ang1 compared with Ang2 results in dense hypervascularization, with large vessels, and excess Ang2 binding to Tie2 leads to destabilized, leaky vessels [94].
For anti-angiopoietin cancer treatment, selective Ang2-inhibiting agents seem to be more effective and safe. The overall effects of the Ang–Tie system on tumors are context-dependent [94]. Ang1 stimulates tumor growth by promoting EC survival and vessel maturation, but it also inhibits tumor cell migration across vessel walls. These conflicting biological activities warrant caution when considering Ang1 as an anticancer target. Instead, Ang2 may be a more desirable target because it stimulates tumor angiogenesis and recruits proangiogenic monocytes, and Ang2 inhibition promotes vessel regression and normalization [101]. Given that Ang2 and VEGF increase angiogenesis cooperatively, simultaneous blocking of VEGF and angiopoietins is superior to anti-angiopoietin therapy alone for inhibiting tumor angiogenesis, metastasis, and vessel leakage and represents a viable clinical treatment paradigm [102].
Various agents that block either Tie2 or Ang2 are being evaluated in early-phase clinical trials. Among the drugs targeting this pathway, trebananib (AMG386) is a novel, investigational angiopoietin antagonist peptide-Fc fusion protein (peptibody) that selectively binds Ang1 and Ang2, prevents their interaction with Tie2, and inhibits tumor EC proliferation and tumor growth [103]. A randomized, placebo controlled phase II trial of combination weekly trebananib and weekly paclitaxel demonstrated clinical efficacy relative to weekly paclitaxel alone in women with recurrent EOC, FC or PPC. Trebananib was infused at 2 dose levels (3 mg/kg or 10 mg/kg) and was associated with a median PFS of 5.7 months (95%CI: 4.6–8.0 months) and 7.2 months (95%CI: 5.3 to 8.1 months), respectively. This compared favorably to weekly paclitaxel alone (median PFS: 4.6 months, 95% CI: 1.9 vs. 6.7 months). The combined HR of the trebananib arms relative to weekly paclitaxel was 0.76 (95% CI: 0.52–1.12, P=0.165) and further analysis suggested a dose-response effect on PFS [103]. Adverse effects included peripheral edema, hypokalemia, thromboembolic events, and hypertension. One of the two ongoing phase III clinical trials has been reported for the combination of trebananib and paclitaxel in recurrent ovarian cancer. TRINOVA-1, (NCT01204749) mirrored the phase II design and combined weekly paclitaxel with a higher dose (15 mg/kg) of trebananib (or placebo) in patients with up to 3 prior anticancer regimens and a platinum-free interval of 12 months or less. The 919 patients were randomized 1:1 to the treatment arms. The primary endpoint, PFS, was significantly improved in the combination arm (median 7.2 vs. 5.4, HR: 0.66, 95%CI: 0.57–0.77). The effect was persistent, albeit lower, among the 7–8% of patients who were previously treated with bevacizumab. Similarly, approximately half of the treatment cohort had a platinum-free interval between 6 and 12 months but the effect appeared consistent in this stratum. The combination was associated with more edema, like the phase II, but did not reflect other anti-VEGF based agents. TRINOVA-2 is being run in a similar population but in combination with pegylated liposomal doxorubicin (NCT01281254) in patients with recurrent or resistant EOC, FTC, or PPC. Finally, a phase III clinical trial of trebananib in combination with paclitaxel and carboplatin as a first-line chemotherapy (TRINOVA-3, NCT01493505) has completed enrollment. Clinical trials for angiopoietin-based therapies are listed in Table 8.
Table 8.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Trebananib | I | Advanced solid tumor (including ovarian cancer) | 1 PR(n=3) | [103] |
Trebananib +PLD or topotecan | Ib | Advanced recurrent ovarian cancer | Ongoing (NCT00770536) | |
Trebananib+PC | I | First line chemo therapy in ovarian cancer | Ongoing (NCT01253681) | |
P±trebananib | II | Advanced recurrent ovarian cancer | Ongoing (NCT00479807) | |
wP (a)±trebananib | II | Recurrent ovarian cancer | Arm A: 37% PR Arm B: 19% PR Arm C: 27% PR |
[221] |
wP±trebananib(TRINO VA-1) | III | Recurrent, partially platinum sensitive ovarian cancer | Ongoing (NCT01204749) | |
PLD± trebananib (TRINOVA-2) | III | Recurrent or resistant ovarian cancer | Ongoing (NCT01281254) | |
PC± trebananib (TRINOVA -3) | III | First line chemo therapy in FIGO stage III-IV ovarian cancer | Ongoing (NCT01493505) |
Arm A: QW (3 weeks on/1 week off) plus intravenous trevananib10 mg/kg QW, Arm B: trebananib 3 mg/kg QW, Arm C: placebo QW (QW:paclitaxel (80 mg/m2) once weekly).
7. HGF and the c-MET pathway
The HGF/c-MET axis is implicated in a wide variety of epithelial, mesenchymal, and hematological malignancies and angiogenesis [104]. The c-MET proto-oncogene is essential for embryonic development, epithelial–mesenchymal transition, angiogenesis, and wound healing [104, 105]. HGF is the only known ligand for c-MET [106]. Upon HGF binding, c-MET autophosphorylates and recruits several downstream effectors, including Grb2, Gab1, PI3k, phospholipase C-γ, Shc, Src, Shp2, Shp1, and STAT3. Grb2 and Gab1 interact directly with c-MET and are critical in HGF/c-MET signaling [107]. MET is normally expressed by epithelial cells, and it is also seen in ECs, neurons, hepatocytes, hematopoietic cells, and melanocytes. HGF is usually expressed in cells of mesenchymal origin. Because of these characteristics, HGF and MET are the principal mediators of paracrine epithelial-mesenchymal interaction. C-MET receptor expression is regulated by the MET proto-oncogene. Dysregulation of HGF/c-MET results from amplification and/or rearrangement of MET mutations, ligand and/or receptor overexpression, abnormal paracrine stimulation, or autocrine loop formation [108, 109].
HGF/c-MET also increases angiogenesis by promoting the growth, movement, and morphogenesis of ECs [110, 111]. HGF has a direct effect on ECs via enhancement of cancer cell-EC contact induced by focal adhesion kinase (FAK) phosphorylation [112]. HGF also decreases endothelial occludin, a cell–cell adhesion molecule, which results in reduced trans-endothelial resistance of tumor vessels and allows cancer cells to migrate across the EC barrier. Increased expression of HGF/c-MET is related to high vascular density in tumors, and overexpression of this pathway leads to increased angiogenesis in experimental xenograft models. HGF and c-MET regulate angiogenesis directly or indirectly through VEGF signaling. MET signaling can induce VEGFA expression and angiogenesis through common signaling intermediates such as SRC homology 2 domain-containing proteins (SHCs). HGF/c–MET and VEGF/VEGFR2 cooperate to induce angiogenesis although MET and VEGFR do not physically associate or transphosphorylate each other. They do, however, activate common signaling mediators ERK/MAPK, AKT/mTOR, and FAK [113]. MET kinase inhibitors and a peptide that contained the MET bidentate docking site blocked cancer growth and decreased the number of blood vessels in experimental models [114, 115].
Another interesting aspect of MET biology in tumors is its regulation and activation by hypoxia [116]. Hypoxia induces the expression of the transcription factor hypoxia-inducible factor 1α, and hypoxia-inducible factor-dependent expression of MET occurs in several types of carcinoma cells [117–119]. Anti-angiogenic therapy reduces tumor vascularization, thus causing tumor hypoxia. Anti-angiogenic therapy alone may reduce tumor mass but may also promote MET-dependent spreading of cancer cells, and these observations argue for combination therapies that target both angiogenesis and HGF/c–MET. Preclinical studies with low-molecular-mass compounds that inhibit both VEGFR2 and MET kinases have validated this concept in mouse xenograft models [120, 121], and these inhibitors may prove valuable in controlling metastatic cancer by concurrently blocking angiogenesis and invasion [122].
The HGF/c-MET pathway plays a role in sunitinib resistance by supporting angiogenesis. Sunitinib-resistant tumors show increased HGF expression compared to sunitinib-sensitive tumors, and in these tumors, c-MET expression is elevated in ECs but not tumor cells. Immunohistochemical analysis from tissue that was treated with a combination of sunitinib and c-MET inhibitors revealed significant decreases in microvascular density [123]. Several approaches have been developed to target this pathway, including MoAbs against human HGF (AMG102 or rilotumumab) and NK4 (a four-kringle antagonist). NK4 inhibits not only invasive growth but also tumor angiogenesis [124]. Rilotumumab (AMG102) is a fully human IgG2 MoAb directed against HGF. Rilotumumab suppressed subcutaneous growth of glioblastoma (U-87) in a preclinical model through enhanced apoptosis and reduced mitogenesis [125], but the drug had minimal effect on angiogenesis. Rilotumumab has been tested in patients with persistent or recurrent EOC, PPC, and FC [126]. Single agent rilotumumab showed limited benefit in this patient population in this study. Cabozantinib, a small molecule inhibitor of c-MET, ret, and VEGFR2 has been studied in recurrent ovarian cancer. In a 68 patient, randomized phase II discontinuation trial, women with recurrent platinum sensitive and resistant ovarian cancer were given cabozantinib 100 mg orally, daily for 12 weeks. Patients responding to therapy continued on treatment until progression or intolerance. Patients achieving stable disease were then randomized to either continued cabozantinib or placebo until progression. Fifty-one patients were evaluable for response. The overall response rate was 24%, with 58% having stable disease at 12 weeks. Responses were observed in platinum refractory, resistant and sensitive patients. Toxicities included 10% with grade 3 hand-foot syndrome and 8% with diarrhea. Approximately 10% discontinued treatment due to adverse events, including 2 fatal GI perforations [127]. A GOG trial randomizing recurrent ovarian cancer patients to either cabozantinib or weekly paclitaxel is ongoing. (NCT01716715)
8. Ephrin/Eph receptor pathway
The erythropoietin-producing hepatocellular (Eph) family of receptor tyrosine kinases regulates various physiologic and pathologic processes, including insulin secretion, bone homeostasis, immune function, blood clotting, pathological forms of angiogenesis, and cancer [128]. The erythropoietin-producing hepatocellular (Eph) family of receptor tyrosine kinases regulates various physiologic and pathologic processes including the regulation of insulin secretion, bone homeostasis, immune function, blood clotting, pathological forms of angiogenesis, and cancer [128]. EphA2 plays a role in axonal migration developmentally and is largely absent from most normal adult tissues except some epithelial cells [129].
EphA2 belongs to the tyrosine kinase Eph receptor family. The Eph receptors can be divided into two subgroups according to their ligands. EphA receptors interact with 5 ligands of the glycosylphosphatidylinostitol-linked ephrin-A subclass, whereas EphB receptors interact with 3 transmembrane ligands of the ephrin-B subclass [130]. Among the various Eph family members, EphA2 is of particular interest because it is overexpressed in ovarian cancer and is related to disease severity and clinical outcome [131].
Several studies have investigated the impact of EphA2 on tumor angiogenesis. EphA2 receptor activation plays an essential role in VEGF-induced EC migration [132]. The EphA2/FAK/paxillin axis has been implicated in vasculogenic mimicry [133, 134], a process by which tumor cells mimic endothelial-derived vasculogenic networks [135]. In ovarian cancer, EphA2 is overexpressed in a substantial fraction of tumor cells as well as the associated vasculature [131]. EphA2 targeting with either a MoAb or small interfering RNA (siRNA) results in anti-tumor effects, mediated by reduced angiogenesis. Some studies have suggested that increased EphA2 expression is associated with resistance to anti-VEGF therapy. Casanova and co-workers showed increased expression of ephrin A1 and EphA2 in a pancreatic cancer xenograft model. Ephrin A1 functions as a proangiogenic factor when blockade of the VEGFR pathway induces tissue hypoxia [84]. EphA2-targeted siRNA incorporated into dioleyol phosphatidylcholine (DOPC) nanoliposomes caused a significant decrease in tumor growth in combination with paclitaxel in an ovarian cancer xenograft model when compared to nonsilencing siRNA [136]. A subsequent clinical study using siRNA-EphA2-DOPC was based on those successful preclinical studies, and a phase I clinical trial is planned to assess the safety of siRNA-EphA2-DOPC in patients with advanced, recurrent cancers (NCT01591356).
9. Delta-like 4 (Dll4)/Notch pathway
The Notch signaling pathway has been implicated in tumor angiogenesis, including vessel maturation, pericyte recruitment, and branching as well as cell differentiation, proliferation, survival, and apoptosis. In mammalian cells, this pathway consists of five transmembrane Notch ligands (Jagged1, Jagged2, and Delta-like ligands [Dll] 1, 3, and 4 [137] and four Notch receptors [Notch 1–4]). Ligand receptor binding leads to cleavage via intramembranous proteolysis by ɣ-secretase and subsequent translocation from the cell membrane to the nucleus. The Notch intracellular domain interacts with transcription factors to regulate transcription of the basic helix-loop-helix proteins hairy/enhancer of split (Hes) and related proteins (Hey) [138]. ECs express Notch1 and Notch4 receptors and the Jagged1, Dll1, and Dll4 ligands. Among the various Notch ligands, Dll4 is expressed specifically in the endothelium at sites of vascular development and angiogenesis. Experiments involving gene disruption in mice have shown that knockout of only one Dll4 allele is lethal to the embryo and results in various defects in arterial development and venous circulation, enlargement of the pericardial sac, and failure to remodel the yolk sac vasculature [139]. This suggests that the Dll4/Notch signaling system is a major stimulator of angiogenesis.
VEGF can increase Dll4 expression in the developing ECs, and Dll4 acts as a negative feedback mechanism. Consequently, inhibition of this pathway actually increases angiogenesis, but most of the newly formed vessels are abnormal and functionally compromised. As a result, tumor hypoxia increases, which retards tumor growth [140, 141]. Dll4 expression is also an independent predictor of poor patient survival and a predictor of response to anti-VEGF therapy. Interestingly, tumors that are resistant to anti-VEGF treatment may be sensitive to anti-Dll4 therapy, and combination treatment with anti-VEGF and anti-Dll4 drugs appears to have an additive inhibitory effect on tumor growth [142, 143]. In a mouse model of human glioblastoma, Dll4 targeting along with anti-VEGF therapy resulted in greater inhibition of tumor growth than controls or treatment with an anti-VEGF drug alone [138].
A phase I trial in patients with advanced solid malignancies showed that RO4929097, a γ-secretase inhibitor of Notch signaling, is tolerable in combination with temsirolomus, gemcitabine, and cediranib [144–146]. A phase II study was performed in patients with metastatic colorectal cancer, but single-agent RO4929097 did not show efficacy [147]. A phase II clinical trial of RO4929097 is ongoing in ovarian cancer patients (NCT01195343). Demcizumab (OMP-21M18), monoclonal antibody targeting Dll4, is under phase Ib/II clinical development in ovarian cancer patients (NCT01952249). In this Phase 1b/2 trial, demcizumab is being tested in combination with paclitaxel in patients with platinum-resistant EOC, PPC and FC. Following a phase 1b safety evaluation, a Phase 2 clinical trial will proceed in these patients.
10. Src family kinases (SFKs)
SFKs are a family of nonreceptor protein tyrosine kinases that are around 60 kD in weight and include 9 genes: Src, Blk, Fgr, Fyn, Hcy, Lck, Lyn, Yes, and Yrk. Src is rarely mutated in human tumors, and amplifications and rearrangements of the gene in tumors are even rarer[148]. Src mediates mitogenic signals between growth factor receptors like VEGF, EGFR, c-MET and insulin-like growth factor 1R and downstream signaling cascades such as FAK, MAPK, and PI3K/AKT/mTOR [149]. Accumulating data suggest that Src plays an important role in many steps in malignancy, including cancer cell mitosis, adhesion, invasion, motility, survival, angiogenesis, and progression [150].
The antiangiogenic effect of Src is mediated by interleukin 8 (IL-8) and VEGF [151]. Src is associated with VEGF signaling in two aspects. One aspect is as a regulator of activation of the VEGFR pathway upstream; the other is as a signal transduction molecule downstream of VEGFR2. SFKs can control the expression of angiogenic growth factors and cytokines, including VEGF, by regulating their gene expression [150, 152]. Src activation also leads to increased IL-8 expression, and Src inhibition can reduce IL-8 expression levels [151]. SFKs also mediate ligand-dependent VEGFR2 signal transduction. SFKs can cooperate with angiogenic growth factor receptors, such as VEGFR, to elicit signaling in ECs or tumor cells [152]. VEGF-induced Src phosphorylation can promote formation of the FAK/αvβ5 signaling complex and caveolin/VEGFR2 complex [153], which are required for vascular permeability response and neovascularization. In c-Src-deficient mice, there was reduced vascular permeability in response to VEGF, suggesting an important role for Src in VEGF-induced angiogenesis [154]. Src is also required for either VEGF-induced ERK1/2 or FAK activation, which leads to increased cell proliferation. Inhibition of Src kinase activity can suppress cell proliferation and migration in human umbilical vein ECs [155].
There are several Src-targeted inhibitors in clinical development. Dasatinib (BMS-354825) is an orally available Src and Abl kinase inhibitor with antiproliferative activity against a broad spectrum of hematological and solid cancer cell lines [156]. As a multikinase inhibitor, dasatinib is being evaluated in breast, lung, colorectal, pancreatic, uterine and ovarian cancers. Bosutinib (SKI-606) is another potent oral Src inhibitor with anti-Abl activity. This compound demonstrated antitumor activity in preclinical models, and clinical development in hematological and solid malignancies is under way. The Src inhibitors AZD-0530, XL-999, and XL-228 are also undergoing early-phase testing. One randomized phase II trial in combination with paclitaxel and carboplatin has been conducted with saracatinib in recurrent platinum sensitive patients [157] and a second in combination with weekly paclitaxel (platinum-resistant patients) is completed [158]. In those two randomized phase II trial, the addition of saracatinib to chemotherapeutic agents did not improve response rate or PFS in ovarian cancer patients. Most of these small molecules have activities against other kinases as well [159] (Table 10).
Table 10.
mTOR inhibitors | Temsirolimus, sirolimus (rapamycin), everolimus, deforolimus |
---|---|
Akt inhibitors | Perifosine, GSK-690693, |
PDK1 inhibitor | UCN-01 |
PI3K inhibitors | PI-103, BGT-226, BEZ-235, XL-765, XL-147 |
11. PI3K/AKT/mTOR pathway
PI3K is a lipid kinase that generates 3′-phosphoinositides at the cell membrane when activated by receptor kinases [160]. This leads to recruitment of phosphoinositide-dependent kinase 1 (PDK1) and Akt to the cell membrane. The generation of 3′-phosphoinositides is negatively regulated by phosphatase and tensin homologue. Akt is activated by several enzymes, including PDK1, mTORC2, and IRS-1, and this activation allows Akt to then inhibit tuberous sclerosis protein 2. The PI3K/Akt/mTOR pathway is responsible for survival of ECs during stimulation with VEGF. The binding of VEGF to VEGFR results in dimerization of the receptor followed by activation of the PI3K/Akt/mTOR pathway, which leads to increased EC survival.
Akt is also important for EC survival when new blood vessels are being formed. During new vessel formation, angiogenic cells need to degrade the extracellular matrix. ECs must therefore reinforce the mechanisms of apoptosis inhibition to avoid the risk of anoikis, apoptosis induced by lack of adhesion to the substrate [161]. ECs repress their apoptogenic program through two main signaling pathways initiated from integrin-mediated attachment to the extracellular matrix and from survival factors such as VEGF and FGF2. Akt is a convergence point for both pathways [162]. Increased expression of Akt-1 in ECs phosphorylates apoptogenic proteins such as Bad, Bax, and caspase-9 [162, 163], which induces apoptosis. At the same time, it increases the levels of the antiapoptotic proteins A1 and Bcl-2 [162]. Other tyrosine kinase receptors (FGFRs, Tie2, insulin receptor, insulin-like growth factor receptor, and MET) and integrins can also activate the PI3K pathway in ECs [164].
Rapamycin analogues inhibit downstream mTOR signaling. Everolimus (RAD001), an oral rapamycin analogue, has been approved for progressive neuroendocrine pancreatic cancer treatment, subependymal giant cell astrocytoma, hormone receptor positive-HER2 negative breast cancer, and advanced renal cell carcinoma. Everolimus showed anti-angiogenic effects in EGFR-resistant cancer cell lines and everolimus partially restored the ability of EGFR inhibitors of EGFR-related signaling effectors and VEGF production, which inhibits proliferation and capillary tube formation of ECs, both alone and in combination with gefitinib [165]. Another mTOR inhibitor, temsirolomus, is approved for renal cell carcinoma.
12. Integrins/focal adhesion-associated proteins
ECs express at least 11 integrins. Integrins are heterodimeric cell surface adhesion receptors for the ECM comprising an α subunit and a β subunit [166]. Among them, α5β1 and αvβ3 are upregulated during angiogenesis [167]. VEGFA expression in ECs is regulated in an α3β1-dependent manner [168]. Integrin αv supports EC survival in tumors by pericyte-induced control of autocrine VEGFA. PDGFR also interacts with integrins, which enhances cell proliferation, migration, and survival [161, 169]. However, integrins lack intrinsic enzymatic activity. The proteins transduce proliferative, survival, migratory, and angiogenic signals by clustering together with kinases and adaptor proteins to form focal adhesion complexes. Focal adhesions are the origin of signals that activate or inhibit cellular processes such as proliferation, survival, and migration of ECs. For example, shear stress induces endothelial migration through a signal initiated by α5β1 binding to fibronectin and involving the adaptor proteins Shc, PI3K, and ERK-MAPK1/2 [170]. Integrin αvβ3 promotes adhesion, migration, and phosphorylation of Akt, MAPK, and FAK.
FAK, a non-receptor cytoplasmic tyrosine kinase, and can be activated by VEGFR2. FAK regulates the organization of the cytoskeleton and activates diverse signaling pathways involved in the localized adhesion of the cell surface and in cell motility [171]. Activated FAK recruits SH2 domain-containing proteins such as Src, which phosphorylates FAK at additional sites; this phosphorylation induces the association between signaling molecules (such as the Ras-activating protein Sos, PI3K, p130Cas, and paxillin) and focal adhesions. FAK also activates ERK-MAPK and the PI3K/Akt pathways. Intracellular activation of the RAF/MEK/MAPK pathway induces subsequent initiation of DNA synthesis and cell growth. VEGF/VEGFR2 signaling and integrin signaling converge to RAF/MEK/MAPK through FAK.
Endostatin binds to the α5β1 integrin, which leads to focal adhesion and actin stress fiber disassembly mediated by Src and dependent on tyrosyl phosphatase [159]. Interestingly, the inhibition of EC migration occurs without interference with pathways mediated by PLCγ, Akt, MAPK, Rac, or Pak [160], which implies that there is a specific integrin signaling pathway involved in migration and mediated by Src. However, endostatin blocks VEGF binding, VEGFR2 phosphorylation, and ERK, p38 MAPK, and FAK activation in ECs, which suggests a direct interaction between endostatin and VEGFR2 [161].
Integrins αvβ3 and αvβ5 are upregulated in both tumor cells and angiogenic ECs, making them attractive therapeutic targets. They are involved in angiogenesis and expressed in malignancies such as melanoma, gliomas, and cancers of the breast, prostate, and colon. Function-blocking MoAbs were among the first integrin antagonists developed and showed considerable anti-angiogenic activity in preclinical models [172, 173]. Cilengitide (EMD-121974) is a synthetic cyclic pentapeptide small-molecule inhibitor of αvβ3 and αvβ5 integrins [174].
Integrin α5β1 is expressed mainly on vascular ECs and upregulated together with fibronectin in tumor neovasculature. In ovarian cancer, peritoneal dissemination is facilitated by α5β1 through increased expression of MMP-9 and fibronectin [175]. It was shown that upregulated α5β1 expression was also related to poor prognosis [176]. Strategies that target β1 integrins, particularly α5β1, have shown efficacy in reducing tumor burden in preclinical models. An integrin β1 inhibitory antibody significantly affected the in vitro and in vivo growth of human breast cancer tumor cells [177]. Volociximab is a chimeric human immunoglobulin G4 and a function-blocking MoAb against integrin α5β1 [178, 179]. It inhibits angiogenesis independently of VEGF/VEGFR and induced apoptosis in proliferating, but not quiescent, ECs in preclinical experiments [180]. A multicenter phase II study tested volociximab in 16 patients with platinum-resistant ovarian cancer or primary peritoneal cancer. There was one patient with stable disease out of 14 evaluable patients, and weekly volociximab was well tolerated [181]. A phase I clinical trial using volociximab in combination with liposomal doxorubicin (NCT00635193) has just been completed.
13. Inflammatory cytokines
13. A. TNF-α
One of the most prominent cytokines implicated in inflammation is TNF-α, which is constitutively expressed in ovarian cancer tumors and stromal cells such as macrophages [182]. TNF-α plays a multifaceted role in cancer by stimulating other cytokines and angiogenic factors by paracrine and autocrine signaling [183, 184]. TNF-α induces angiogenic factors, including chemokine (C-C motif) ligand 2 (CCL2), IL-6, chemokine (C-X-C motif) ligand 12 (CXCL12), VEGF, IL-8, bFGF, and migration inhibitory factor, and their receptors [184]. TNF-α is an inducer of VEGF, and VEGF in turn induces CXCL12 [185, 186]. TNFα also directly induces CXCL12, and CXCL12 and VEGF synergize in the stimulation of blood vessel formation in ovarian cancer [187, 188]. TNFα-depleted cancer cells showed reduced tumor growth and vascularization [184]. TNFα regulates IL-8 transcriptionally via NF-κB, regulates VEGF via Sp1 and NF-κB, and regulates bFGF via c-Jun[188]. Administration of anti-IL-8, anti-VEGF, and anti-bFGF antibodies abrogates EC function. In another study, TNF-α signaling was responsible for tumor growth. TNF-α/TNF receptor-1 signaling in CD4+ cells is essential for tumor growth and was linked to increased IL-17 levels in malignant ascites. In a mouse ovarian cancer model, TNF-α neutralizing antibody treatment resulted in decreased tumor burden and leukocyte infiltration. The concentration of IL-17 in ascites and the levels of IL-6 in plasma were decreased as well [189].
Infliximab, a chimeric MoAb against TNF-α, led to decreased levels of CXCL12 and IL-6 and resulted in reduced tumor growth, vascularization, and infiltration of myeloid cells [190]. A TNF-α antagonist, etanercept, which is a soluble p75 TNF receptor that inhibits the TNF-α pathway by competitive binding, was also assessed in a phase I trial to evaluate its efficacy in treating recurrent ovarian cancer. In that study, two cohorts of 17 and 13 patients were treated with a dose of 25 mg twice a week (cohort 1) or 3 times a week (cohort 2) [191]. Six of 18 patients who received a minimum of 12 weeks of therapy reached disease stabilization. The trial demonstrated the feasibility of anti-TNF-α therapy for epithelial ovarian cancer treatment.
13. B. Other inflammatory cytokines
In addition to TNF-α, high levels of several other proinflammatory cytokines have been identified in ascites fluid from ovarian cancer patients, including IL-6, IL-8, CCL2, and macrophage inflammatory protein-1β [192, 193]. IL-6 induces VEGF to increase angiogenesis; VEGF also increases IL-6 in a positive feedback loop [194]. IL-6 also activates STAT3 and MAPK in ECs [195]. It is believed that STAT3 upregulates MMP-9 in these cells, thus contributing to angiogenesis and metastasis [196]. To evaluate the prognostic significance of IL-6 and IL-8 levels in ascites, Lane and colleagues related those levels to a number of clinical measures, including PFS. Using multivariate analyses, the authors concluded that high levels of IL-6 could be related to shorter PFS [193]. Similarly, in another study, high serum IL-6 levels were also correlated with poor prognosis [197].
Blockade of IL-6 using an antibody, siltuximab, was shown to effectively block IL-6 signaling pathways by suppressing Stat3 phosphorylation, which led to decreases in downstream antiapoptotic factors [198]. Siltuximab enhanced paclitaxel sensitivity and cytotoxicity in a paclitaxel-resistant cell line, SKOV3-TR; however, these observations require additional work [198]. The therapeutic efficacy of this agent was assessed in a phase II clinical trial with 20 patients who had advanced platinum-resistant ovarian cancer [199]. One of the patients had a partial response, whereas seven patients experienced disease stabilization in addition to exhibiting decreased plasma levels of several cytokines including CCL2, CXCL12, and VEGF, which suggests that these cytokines are regulated by IL-6 [199].
14. RNA interference
The discovery of RNA interference (RNAi), including miRNA and siRNA-mediated gene silencing, is considered one of the most important advancements in biology in the last decade [200–202]. A specifically designed siRNA can bind the target gene (mRNA) in a sequence-specific manner and induce degradation of mRNA translation [202]. SiRNA is now commonly used as a powerful tool for silencing post-transcriptional gene expression and investigating gene function.
The main issues related to systemic use of siRNA-based therapeutics are development of an efficient delivery system to enhance the stability of siRNA and avoid unintended effects like immune response and off-target effects. Early studies with siRNA-based therapies relied on local administration for specific diseases, such as age-related macular degeneration, diabetic macular edema, respiratory virus infection, and pachyonychia congenita [203, 204]. To overcome obstacles to the systemic use of siRNA-based therapeutics, various nanoparticles made of biodegradable nanomaterials such as natural or synthetic lipids (e.g., liposomes and micelles) and polymers (e.g., chitosan, poly(lactic-co-glycolic) acid [PLGA], polylactic acid [PLA], polyethilenimine [PEI], and atelocollagen), carbon nanotubes, quantum dots, gold nanoshells, or magnetic iron oxide have been used [205–210]. Chemical modification to increase stability have also been tested [211].
An RNAi approach can also be used to target tumor angiogenesis. Several clinical trials based on systemic delivery of siRNA therapies have progressed into the clinic and are currently being tested in phase I/II clinical trials for viral diseases (hepatitis B) and acute renal failure. Currently, there are eight RNAi-based clinical trials in solid tumors and chronic myeloid leukemia in the United States. The first demonstration of siRNA-mediated effects in a clinical trial was for melanoma in 2008; this study targeted ribonuclease reductase using a cyclodextrin-based polymer-conjugated siRNA following a study in non-human primates [212, 213]. Preliminary data showed that this approach was well tolerated, although dose-limiting toxicity was observed in several patients. Targeting EphA2 using siRNA-DOPC showed antitumor activity via antiangiogenic effects in ovarian cancer xenografts [142]. As mentioned above [136], a phase I clinical trial will be started to target EphA2 using neutral nanoliposomal EphA2 siRNA at the University of Texas MD Anderson Cancer Center (NCT01591356).
15. Conclusions
Recurrent and refractory ovarian cancer remains a significant clinical challenge. Even though tumor stromal cells are more stable compared to tumor cells, stromal cells can still adapt. Furthermore, pathways related to angiogenesis are redundant and complementary. These characteristics enable tumors and their supportive vasculature to develop resistance to antiangiogenic therapy. Growing understanding of the mechanisms underlying such resistance is leading to new anti-angiogenesis strategies. Maintenance of complete remission and overcoming chemoresistance in patients who do not achieve complete remission are important goals for targeted therapy in ovarian cancer. Combination therapies that target multiple pathways relevant for tumor angiogenesis have the potential to overcome adaptive resistance and lead to improved patient outcomes
Table 9.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Dasatinib+PC | I | Recurrent EOC, PPC, FC | 3CR, 5PR, 10SD(n=20) | [222] |
Dasatinib | II | Recurrent persistent EOC, PPC, FC | No response | [223] |
Dasatinib +carboplatin +ifosfamide +etoposide phosphate | I,II | Young patients(1–25yrs) with Metastatic recurrent solid tumors | Ongoing (NCT00788125) | |
PC± Saracatinib (OVERT1) | II | Recurrent, resistant ovarian cancer | No additional benefit | [157](NCT00610714) |
wP± Saracatinib | II, III | Platinum resistance EOC, PPC, or FC | No additional benefit | [158] (NCT01196741) |
Table 11.
Treatment | Phase | Indication | Efficacy | Reference |
---|---|---|---|---|
Everolimus (Certican) + bevacizumab | II | Recurrent EOC, PPC, or FC | Recruiting (NCT01031381) | |
Sirolimus + ALVAC(2)-NY-ESO-1 (M)/TRICOM vaccine + sargramostim | I | Primary or recurrent EOC, PPC, and FC | Recruiting (NCT01536054) | |
Deforolimus (AP-23573, MK-8669, ridaforolimus) + carboplatin + paclitaxel | I | Advanced, recurrent solid Endometrial cancer EOC | Recruiting (NCT01256268) | |
Deforolimus (AP-23573, MK-8669, Redaforolimus) + MK0752 + MK2206 | Advanced solid tumors | Ongoing (NCT01295632) | ||
Everolimus + carboplatin + PLD | I | first relapse must occur >= 6 months | Recruiting (NCT01281514) | |
Everolimus + bevacizumab | II | Recurrent, persistent EOC, PPC, FC | Ongoing (NCT00886691) | |
Temsirolimus + carboplatin + docetaxel + paclitaxel | II | Stage III-IV ovarian clear cell carcinoma | Recruiting (NCT01196429) | |
Temsirolimus + PLD | Ib | Refractory Breast cancer, Endometrial, ovarian cancer | Recruiting (NCT00982631) | |
Temsirolimus | II | refractory or recurrent EOC, PPC, or FC | 5 PR,13 SD (n=54) | [224] |
Temsirolimus | II | Recurrent EOC, PPC, or FC | ||
Temsirolimus + topotecan | I | Recurrent, refractory gynecologic cancer | No response | [225] |
Temsirolimus + carboplatin + paclitaxel | I | Advanced solid tumor (including ovarian cancer) | 1 PR (n=6) | [226] |
Temsirolimus + AZD2171 | I | Advanced Gynecologic cancer | Ongoing (NCT01065662) | |
Temsirolimus + PLD | I | Resistant solid tumors | Completed (NCT00703170) | |
Temsirolimus + docetaxel | I | Resistant solid tumors | Completed (NCT00703625) |
Acknowledgements
Portions of this work were supported by grants from the US National Institutes of Health (P50CA083639, P50CA098258, CA109298, U54 CA151668, CA177909, UH2TR000943, T32CA101642, and CA16672), the Department of Defense (OC120547 and OC093416), a Program Project Development Grant and an Ann Schreiber mentored Investigators Award from the Ovarian Cancer Research Fund, CPRIT RP110595, the Bettyann Asche Murray Distinguished Professorship, the Chapman Foundation, the Meyer and Ida Gordon Foundation, the Gilder Foundation, the RGK Foundation, the Judi A. Rees Ovarian Cancer Research Fund, and the Blanton-Davis Ovarian Cancer Research Program. We thank Zachary S. Bohannan, Dawn Chalaire, and Arthur Gelmis for editorial review.
Reference
- 1.Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat Rev Cancer. 2003;3:502–516. doi: 10.1038/nrc1123. [DOI] [PubMed] [Google Scholar]
- 2.Balvert-Locht HR, Coebergh JW, Hop WC, et al. Improved prognosis of ovarian cancer in The Netherlands during the period 1975–1985: a registry-based study. Gynecol Oncol. 1991;42:3–8. doi: 10.1016/0090-8258(91)90222-q. [DOI] [PubMed] [Google Scholar]
- 3.Ozols RF, Bundy BN, Greer BE, et al. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2003;21:3194–3200. doi: 10.1200/JCO.2003.02.153. [DOI] [PubMed] [Google Scholar]
- 4.du Bois A, Neijt JP, Thigpen JT. First line chemotherapy with carboplatin plus paclitaxel in advanced ovarian cancer--a new standard of care? Ann Oncol. 1999;10(Suppl 1):35–41. doi: 10.1023/a:1008355317514. [DOI] [PubMed] [Google Scholar]
- 5.Biagi JJ, Eisenhauer EA. Systemic treatment policies in ovarian cancer: the next 10 years. Int J Gynecol Cancer. 2003;13(Suppl 2):231–240. doi: 10.1111/j.1525-1438.2003.13356.x. [DOI] [PubMed] [Google Scholar]
- 6.Neijt JP, Engelholm SA, Tuxen MK, et al. Exploratory phase III study of paclitaxel and cisplatin versus paclitaxel and carboplatin in advanced ovarian cancer. J Clin Oncol. 2000;18:3084–3092. doi: 10.1200/JCO.2000.18.17.3084. [DOI] [PubMed] [Google Scholar]
- 7.Greenlee RT, Hill-Harmon MB, Murray T, Thun M. Cancer statistics, 2001. CA Cancer J Clin. 2001;51:15–36. doi: 10.3322/canjclin.51.1.15. [DOI] [PubMed] [Google Scholar]
- 8.Gore ME, Fryatt I, Wiltshaw E, Dawson T. Treatment of relapsed carcinoma of the ovary with cisplatin or carboplatin following initial treatment with these compounds. Gynecol Oncol. 1990;36:207–211. doi: 10.1016/0090-8258(90)90174-j. [DOI] [PubMed] [Google Scholar]
- 9.Wernert N, Locherbach C, Wellmann A, Behrens P, Hugel A. Presence of genetic alterations in microdissected stroma of human colon and breast cancers. Anticancer Res. 2001;21:2259–2264. [PubMed] [Google Scholar]
- 10.Allinen M, Beroukhim R, Cai L, et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell. 2004;6:17–32. doi: 10.1016/j.ccr.2004.06.010. [DOI] [PubMed] [Google Scholar]
- 11.Fukino K, Shen L, Patocs A, Mutter GL, Eng C. Genomic instability within tumor stroma and clinicopathological characteristics of sporadic primary invasive breast carcinoma. JAMA. 2007;297:2103–2111. doi: 10.1001/jama.297.19.2103. [DOI] [PubMed] [Google Scholar]
- 12.Folkman J. What is the evidence that tumors are angiogenesis dependent? J Natl Cancer Inst. 1990;82:4–6. doi: 10.1093/jnci/82.1.4. [DOI] [PubMed] [Google Scholar]
- 13.Eskander RN, Randall LM. Bevacizumab in the treatment of ovarian cancer. Biologics. 2011;5:1–5. doi: 10.2147/BTT.S13071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967–974. doi: 10.1038/nature04483. [DOI] [PubMed] [Google Scholar]
- 15.Konerding MA, Fait E, Gaumann A. 3D microvascular architecture of pre-cancerous lesions and invasive carcinomas of the colon. Br J Cancer. 2001;84:1354–1362. doi: 10.1054/bjoc.2001.1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Denekamp J. Endothelial cell proliferation as a novel approach to targeting tumour therapy. Br J Cancer. 1982;45:136–139. doi: 10.1038/bjc.1982.16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hinnen P, Eskens FA. Vascular disrupting agents in clinical development. Br J Cancer. 2007;96:1159–1165. doi: 10.1038/sj.bjc.6603694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Holwell SE, Cooper PA, Thompson MJ, et al. Anti-tumor and anti-vascular effects of the novel tubulin-binding agent combretastatin A-1 phosphate. Anticancer Res. 2002;22:3933–3940. [PubMed] [Google Scholar]
- 19.Tozer GM, Prise VE, Wilson J, et al. Combretastatin A-4 phosphate as a tumor vascular-targeting agent: early effects in tumors and normal tissues. Cancer Res. 1999;59:1626–1634. [PubMed] [Google Scholar]
- 20.Marysael T, Ni Y, Lerut E, de Witte P. Influence of the vascular damaging agents DMXAA and ZD6126 on hypericin distribution and accumulation in RIF-1 tumors. J Cancer Res Clin Oncol. 2011;137:1619–1627. doi: 10.1007/s00432-011-1032-y. [DOI] [PubMed] [Google Scholar]
- 21.Nathan P, Zweifel M, Padhani AR, et al. Phase I trial of combretastatin A4 phosphate (CA4P) in combination with bevacizumab in patients with advanced cancer. Clin Cancer Res. 2012;18:3428–3439. doi: 10.1158/1078-0432.CCR-11-3376. [DOI] [PubMed] [Google Scholar]
- 22.Zweifel M, Jayson GC, Reed NS, et al. Phase II trial of combretastatin A4 phosphate, carboplatin, and paclitaxel in patients with platinum-resistant ovarian cancer. Ann Oncol. 2011;22:2036–2041. doi: 10.1093/annonc/mdq708. [DOI] [PubMed] [Google Scholar]
- 23.Ching LM, Cao Z, Kieda C, Zwain S, Jameson MB, Baguley BC. Induction of endothelial cell apoptosis by the antivascular agent 5,6-Dimethylxanthenone-4-acetic acid. Br J Cancer. 2002;86:1937–1942. doi: 10.1038/sj.bjc.6600368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fredriksson L, Li H, Fieber C, Li X, Eriksson U. Tissue plasminogen activator is a potent activator of PDGF-CC. EMBO J. 2004;23:3793–3802. doi: 10.1038/sj.emboj.7600397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kazlauskas A, Cooper JA. Autophosphorylation of the PDGF receptor in the kinase insert region regulates interactions with cell proteins. Cell. 1989;58:1121–1133. doi: 10.1016/0092-8674(89)90510-2. [DOI] [PubMed] [Google Scholar]
- 26.Antoniades HN, Hunkapiller MW. Human platelet-derived growth factor (PDGF): amino-terminal amino acid sequence. Science. 1983;220:963–965. doi: 10.1126/science.6844921. [DOI] [PubMed] [Google Scholar]
- 27.Andrae J, Gallini R, Betsholtz C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 2008;22:1276–1312. doi: 10.1101/gad.1653708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9:685–693. doi: 10.1038/nm0603-685. [DOI] [PubMed] [Google Scholar]
- 29.Abramsson A, Kurup S, Busse M, et al. Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development. Genes and Development. 2007;21:316–331. doi: 10.1101/gad.398207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Benjamin LE, Hemo I, Keshet E. A plasticity window for blood vessel remodelling is defined by pericyte coverage of the preformed endothelial network and is regulated by PDGF-B and VEGF. Development. 1998;125:1591–1598. doi: 10.1242/dev.125.9.1591. [DOI] [PubMed] [Google Scholar]
- 31.Oikawa T, Onozawa C, Sakaguchi M, Morita I, Murota S. Three isoforms of platelet-derived growth factors all have the capability to induce angiogenesis in vivo. Biol Pharm Bull. 1994;17:1686–1688. doi: 10.1248/bpb.17.1686. [DOI] [PubMed] [Google Scholar]
- 32.Lu C, Thaker PH, Lin YG, et al. Impact of vessel maturation on antiangiogenic therapy in ovarian cancer. Am J Obstet Gynecol. 2008;198:477, e471–e479. doi: 10.1016/j.ajog.2007.12.028. discussion 477 e479-410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Valius M, Kazlauskas A. Phospholipase C-gamma 1 and phosphatidylinositol 3 kinase are the downstream mediators of the PDGF receptor's mitogenic signal. Cell. 1993;73:321–334. doi: 10.1016/0092-8674(93)90232-f. [DOI] [PubMed] [Google Scholar]
- 34.Coughlin SR, Escobedo JA, Williams LT. Role of phosphatidylinositol kinase in PDGF receptor signal transduction. Science. 1989;243:1191–1194. doi: 10.1126/science.2466336. [DOI] [PubMed] [Google Scholar]
- 35.Heldin CH, Ostman A, Ronnstrand L. Signal transduction via platelet-derived growth factor receptors. Biochim Biophys Acta. 1998;1378:F79–F113. doi: 10.1016/s0304-419x(98)00015-8. [DOI] [PubMed] [Google Scholar]
- 36.Yao R, Cooper GM. Requirement for phosphatidylinositol-3 kinase in the prevention of apoptosis by nerve growth factor. Science. 1995;267:2003–2006. doi: 10.1126/science.7701324. [DOI] [PubMed] [Google Scholar]
- 37.Huang JS, Huang SS, Deuel TF. Transforming protein of simian sarcoma virus stimulates autocrine growth of SSV-transformed cells through PDGF cell-surface receptors. Cell. 1984;39:79–87. doi: 10.1016/0092-8674(84)90193-4. [DOI] [PubMed] [Google Scholar]
- 38.Greenhalgh DG, Sprugel KH, Murray MJ, Ross R. PDGF and FGF stimulate wound healing in the genetically diabetic mouse. Am J Pathol. 1990;136:1235–1246. [PMC free article] [PubMed] [Google Scholar]
- 39.Hellberg C, Ostman A, Heldin CH. PDGF and vessel maturation. Recent Results Cancer Res. 2010;180:103–114. doi: 10.1007/978-3-540-78281-0_7. [DOI] [PubMed] [Google Scholar]
- 40.Gaengel K, Genove G, Armulik A, Betsholtz C. Endothelial-mural cell signaling in vascular development and angiogenesis. Arterioscler Thromb Vasc Biol. 2009;29:630–638. doi: 10.1161/ATVBAHA.107.161521. [DOI] [PubMed] [Google Scholar]
- 41.Quaegebeur A, Segura I, Carmeliet P. Pericytes: blood-brain barrier safeguards against neurodegeneration? Neuron. 2010;68:321–323. doi: 10.1016/j.neuron.2010.10.024. [DOI] [PubMed] [Google Scholar]
- 42.Ribatti D, Vacca A, Roccaro AM, Crivellato E, Presta M. Erythropoietin as an angiogenic factor. Eur J Clin Invest. 2003;33:891–896. doi: 10.1046/j.1365-2362.2003.01245.x. [DOI] [PubMed] [Google Scholar]
- 43.Crawford Y, Kasman I, Yu L, et al. PDGF-C mediates the angiogenic and tumorigenic properties of fibroblasts associated with tumors refractory to anti-VEGF treatment. Cancer Cell. 2009;15:21–34. doi: 10.1016/j.ccr.2008.12.004. [DOI] [PubMed] [Google Scholar]
- 44.Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D. Benefits of targeting both pericytes and endothelial cells in the tumor vasculature with kinase inhibitors. J Clin Invest. 2003;111:1287–1295. doi: 10.1172/JCI17929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Erber R, Thurnher A, Katsen AD, et al. Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte-mediated endothelial cell survival mechanisms. FASEB J. 2004;18:338–340. doi: 10.1096/fj.03-0271fje. [DOI] [PubMed] [Google Scholar]
- 46.Jo N, Mailhos C, Ju M, et al. Inhibition of platelet-derived growth factor B signaling enhances the efficacy of anti-vascular endothelial growth factor therapy in multiple models of ocular neovascularization. Am J Pathol. 2006;168:2036–2053. doi: 10.2353/ajpath.2006.050588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hasumi Y, Klosowska-Wardega A, Furuhashi M, Ostman A, Heldin CH, Hellberg C. Identification of a subset of pericytes that respond to combination therapy targeting PDGF and VEGF signaling. Int J Cancer. 2007;121:2606–2614. doi: 10.1002/ijc.22999. [DOI] [PubMed] [Google Scholar]
- 48.Gerhardt H, Semb H. Pericytes: gatekeepers in tumour cell metastasis? J Mol Med (Berl) 2008;86:135–144. doi: 10.1007/s00109-007-0258-2. [DOI] [PubMed] [Google Scholar]
- 49.Alberts DS, Liu PY, Wilczynski SP, et al. Phase II trial of imatinib mesylate in recurrent, biomarker positive, ovarian cancer (Southwest Oncology Group Protocol S0211) Int J Gynecol Cancer. 2007;17:784–788. doi: 10.1111/j.1525-1438.2007.00882.x. [DOI] [PubMed] [Google Scholar]
- 50.Coleman RL, Broaddus RR, Bodurka DC, et al. Phase II trial of imatinib mesylate in patients with recurrent platinum- and taxane-resistant epithelial ovarian and primary peritoneal cancers. Gynecol Oncol. 2006;101:126–131. doi: 10.1016/j.ygyno.2005.09.041. [DOI] [PubMed] [Google Scholar]
- 51.Posadas EM, Kwitkowski V, Kotz HL, et al. A prospective analysis of imatinib-induced c-KIT modulation in ovarian cancer: a phase II clinical study with proteomic profiling. Cancer. 2007;110:309–317. doi: 10.1002/cncr.22757. [DOI] [PubMed] [Google Scholar]
- 52.Safra T, Andreopoulou E, Levinson B, et al. Weekly paclitaxel with intermittent imatinib mesylate (Gleevec): tolerance and activity in recurrent epithelial ovarian cancer. Anticancer Res. 2010;30:3243–3247. [PubMed] [Google Scholar]
- 53.Matulonis UA, Berlin S, Ivy P, et al. Cediranib, an oral inhibitor of vascular endothelial growth factor receptor kinases, is an active drug in recurrent epithelial ovarian, fallopian tube, and peritoneal cancer. J Clin Oncol. 2009;27:5601–5606. doi: 10.1200/JCO.2009.23.2777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Raja FA, Griffin CL, Qian W, et al. Initial toxicity assessment of ICON6: a randomised trial of cediranib plus chemotherapy in platinum-sensitive relapsed ovarian cancer. Br J Cancer. 2011;105:884–889. doi: 10.1038/bjc.2011.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wilhelm S, Carter C, Lynch M, et al. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5:835–844. doi: 10.1038/nrd2130. [DOI] [PubMed] [Google Scholar]
- 56.Kane RC, Farrell AT, Saber H, et al. Sorafenib for the treatment of advanced renal cell carcinoma. Clin Cancer Res. 2006;12:7271–7278. doi: 10.1158/1078-0432.CCR-06-1249. [DOI] [PubMed] [Google Scholar]
- 57.Kane RC, Farrell AT, Madabushi R, et al. Sorafenib for the treatment of unresectable hepatocellular carcinoma. Oncologist. 2009;14:95–100. doi: 10.1634/theoncologist.2008-0185. [DOI] [PubMed] [Google Scholar]
- 58.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]
- 59.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;20:787–793. doi: 10.1111/IGC.0b013e3181e273a8. [DOI] [PubMed] [Google Scholar]
- 60.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;123:499–504. doi: 10.1016/j.ygyno.2011.08.033. [DOI] [PubMed] [Google Scholar]
- 61.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;130:25–30. doi: 10.1016/j.ygyno.2013.04.011. [DOI] [PubMed] [Google Scholar]
- 62.Ledermann JA, Hackshaw A, Kaye S, et al. Randomized phase II placebo-controlled trial of maintenance therapy using the oral triple angiokinase inhibitor BIBF 1120 after chemotherapy for relapsed ovarian cancer. J Clin Oncol. 2011;29:3798–3804. doi: 10.1200/JCO.2010.33.5208. [DOI] [PubMed] [Google Scholar]
- 63.Izzedine H, Buhaescu I, Rixe O, Deray G. Sunitinib malate. Cancer Chemother Pharmacol. 2007;60:357–364. doi: 10.1007/s00280-006-0376-5. [DOI] [PubMed] [Google Scholar]
- 64.Biagi JJ, Oza AM, Chalchal HI, et al. A phase II study of sunitinib in patients with recurrent epithelial ovarian and primary peritoneal carcinoma: an NCIC Clinical Trials Group Study. Ann Oncol. 2011;22:335–340. doi: 10.1093/annonc/mdq357. [DOI] [PubMed] [Google Scholar]
- 65.Friedlander M, Hancock KC, Rischin D, et al. A Phase II, open-label study evaluating pazopanib in patients with recurrent ovarian cancer. Gynecol Oncol. 2010;119:32–37. doi: 10.1016/j.ygyno.2010.05.033. [DOI] [PubMed] [Google Scholar]
- 66.Normanno N, De Luca A, Bianco C, et al. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366:2–16. doi: 10.1016/j.gene.2005.10.018. [DOI] [PubMed] [Google Scholar]
- 67.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
- 68.Cascone T, Herynk MH, Xu L, et al. Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor-resistant human lung adenocarcinoma. J Clin Invest. 2011;121:1313–1328. doi: 10.1172/JCI42405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Viloria-Petit A, Crombet T, Jothy S, et al. Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: a role for altered tumor angiogenesis. Cancer Res. 2001;61:5090–5101. [PubMed] [Google Scholar]
- 70.Vergote IB, Jimeno A, Joly F, et al. Randomized Phase III Study of Erlotinib Versus Observation in Patients With No Evidence of Disease Progression After First-Line Platin-Based Chemotherapy for Ovarian Carcinoma: A European Organisation for Research and Treatment of Cancer-Gynaecological Cancer Group, and Gynecologic Cancer Intergroup Study. J Clin Oncol. 2014;32:320–326. doi: 10.1200/JCO.2013.50.5669. [DOI] [PubMed] [Google Scholar]
- 71.Pakkala S, Ramalingam SS. Combined inhibition of vascular endothelial growth factor and epidermal growth factor signaling in non-small-cell lung cancer therapy. Clin Lung Cancer. 2009;10(Suppl 1):S17–S23. doi: 10.3816/CLC.2009.s.003. [DOI] [PubMed] [Google Scholar]
- 72.Kimelman D, Kirschner M. Synergistic induction of mesoderm by FGF and TGF-beta and the identification of an mRNA coding for FGF in the early Xenopus embryo. Cell. 1987;51:869–877. doi: 10.1016/0092-8674(87)90110-3. [DOI] [PubMed] [Google Scholar]
- 73.De Moerlooze L, Spencer-Dene B, Revest JM, Hajihosseini M, Rosewell I, Dickson C. An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Development. 2000;127:483–492. doi: 10.1242/dev.127.3.483. [DOI] [PubMed] [Google Scholar]
- 74.Beenken A, Mohammadi M. The FGF family: biology, pathophysiology and therapy. Nat Rev Drug Discov. 2009;8:235–253. doi: 10.1038/nrd2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res. 1993;60:1–41. doi: 10.1016/s0065-230x(08)60821-0. [DOI] [PubMed] [Google Scholar]
- 76.Bae JH, Schlessinger J. Asymmetric tyrosine kinase arrangements in activation or autophosphorylation of receptor tyrosine kinases. Mol Cells. 2010;29:443–448. doi: 10.1007/s10059-010-0080-5. [DOI] [PubMed] [Google Scholar]
- 77.Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005;16:139–149. doi: 10.1016/j.cytogfr.2005.01.001. [DOI] [PubMed] [Google Scholar]
- 78.Cunningham DL, Sweet SM, Cooper HJ, Heath JK. Differential phosphoproteomics of fibroblast growth factor signaling: identification of Src family kinase-mediated phosphorylation events. J Proteome Res. 2010;9:2317–2328. doi: 10.1021/pr9010475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Klint P, Claesson-Welsh L. Signal transduction by fibroblast growth factor receptors. Front Biosci. 1999;4:D165–D177. doi: 10.2741/klint. [DOI] [PubMed] [Google Scholar]
- 80.Presta M, Dell'Era P, Mitola S, Moroni E, Ronca R, Rusnati M. Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 2005;16:159–178. doi: 10.1016/j.cytogfr.2005.01.004. [DOI] [PubMed] [Google Scholar]
- 81.Cross MJ, Claesson-Welsh L. FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci. 2001;22:201–207. doi: 10.1016/s0165-6147(00)01676-x. [DOI] [PubMed] [Google Scholar]
- 82.Presta M, Tiberio L, Rusnati M, Dell'Era P, Ragnotti G. Basic fibroblast growth factor requires a long-lasting activation of protein kinase C to induce cell proliferation in transformed fetal bovine aortic endothelial cells. Cell Regul. 1991;2:719–726. doi: 10.1091/mbc.2.9.719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Shono T, Kanetake H, Kanda S. The role of mitogen-activated protein kinase activation within focal adhesions in chemotaxis toward FGF-2 by murine brain capillary endothelial cells. Exp Cell Res. 2001;264:275–283. doi: 10.1006/excr.2001.5154. [DOI] [PubMed] [Google Scholar]
- 84.Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell. 2005;8:299–309. doi: 10.1016/j.ccr.2005.09.005. [DOI] [PubMed] [Google Scholar]
- 85.Compagni A, Wilgenbus P, Impagnatiello MA, Cotten M, Christofori G. Fibroblast growth factors are required for efficient tumor angiogenesis. Cancer Res. 2000;60:7163–7169. [PubMed] [Google Scholar]
- 86.Giavazzi R, Sennino B, Coltrini D, et al. Distinct role of fibroblast growth factor-2 and vascular endothelial growth factor on tumor growth and angiogenesis. Am J Pathol. 2003;162:1913–1926. doi: 10.1016/S0002-9440(10)64325-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Nissen LJ, Cao R, Hedlund EM, et al. Angiogenic factors FGF2 and PDGF-BB synergistically promote murine tumor neovascularization and metastasis. J Clin Invest. 2007;117:2766–2777. doi: 10.1172/JCI32479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lieu C, Heymach J, Overman M, Tran H, Kopetz S. Beyond VEGF: inhibition of the fibroblast growth factor pathway and antiangiogenesis. Clin Cancer Res. 2011;17:6130–6139. doi: 10.1158/1078-0432.CCR-11-0659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Fujii T, Kuwano H. Regulation of the expression balance of angiopoietin-1 and angiopoietin-2 by Shh and FGF-2. In Vitro Cell Dev Biol Anim. 2010;46:487–491. doi: 10.1007/s11626-009-9270-x. [DOI] [PubMed] [Google Scholar]
- 90.Pepper MS, Ferrara N, Orci L, Montesano R. Potent synergism between vascular endothelial growth factor and basic fibroblast growth factor in the induction of angiogenesis in vitro. Biochem Biophys Res Commun. 1992;189:824–831. doi: 10.1016/0006-291x(92)92277-5. [DOI] [PubMed] [Google Scholar]
- 91.Kopetz S, Hoff PM, Morris JS, et al. Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance. J Clin Oncol. 2010;28:453–459. doi: 10.1200/JCO.2009.24.8252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–307. doi: 10.1038/nature10144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Augustin HG, Koh GY, Thurston G, Alitalo K. Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol. 2009;10:165–177. doi: 10.1038/nrm2639. [DOI] [PubMed] [Google Scholar]
- 95.Sundberg C, Kowanetz M, Brown LF, Detmar M, Dvorak HF. Stable expression of angiopoietin-1 and other markers by cultured pericytes: phenotypic similarities to a subpopulation of cells in maturing vessels during later stages of angiogenesis in vivo. Lab Invest. 2002;82:387–401. doi: 10.1038/labinvest.3780433. [DOI] [PubMed] [Google Scholar]
- 96.Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell. 2004;6:553–563. doi: 10.1016/j.ccr.2004.10.011. [DOI] [PubMed] [Google Scholar]
- 97.Maisonpierre PC, Suri C, Jones PF, et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science. 1997;277:55–60. doi: 10.1126/science.277.5322.55. [DOI] [PubMed] [Google Scholar]
- 98.Scharpfenecker M, Fiedler U, Reiss Y, Augustin HG. The Tie-2 ligand angiopoietin-2 destabilizes quiescent endothelium through an internal autocrine loop mechanism. J Cell Sci. 2005;118:771–780. doi: 10.1242/jcs.01653. [DOI] [PubMed] [Google Scholar]
- 99.Bach F, Uddin FJ, Burke D. Angiopoietins in malignancy. Eur J Surg Oncol. 2007;33:7–15. doi: 10.1016/j.ejso.2006.07.015. [DOI] [PubMed] [Google Scholar]
- 100.Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 2011;10:417–427. doi: 10.1038/nrd3455. [DOI] [PubMed] [Google Scholar]
- 101.Falcon BL, Hashizume H, Koumoutsakos P, et al. Contrasting actions of selective inhibitors of angiopoietin-1 and angiopoietin-2 on the normalization of tumor blood vessels. Am J Pathol. 2009;175:2159–2170. doi: 10.2353/ajpath.2009.090391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Koh YJ, Kim HZ, Hwang SI, et al. Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage. Cancer Cell. 2010;18:171–184. doi: 10.1016/j.ccr.2010.07.001. [DOI] [PubMed] [Google Scholar]
- 103.Herbst RS, Hong D, Chap L, et al. Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. J Clin Oncol. 2009;27:3557–3565. doi: 10.1200/JCO.2008.19.6683. [DOI] [PubMed] [Google Scholar]
- 104.Birchmeier C, Birchmeier W, Gherardi E, Vande Woude GF. Met, metastasis, motility and more. Nat Rev Mol Cell Biol. 2003;4:915–925. doi: 10.1038/nrm1261. [DOI] [PubMed] [Google Scholar]
- 105.Funakoshi H, Nakamura T. Hepatocyte growth factor: from diagnosis to clinical applications. Clin Chim Acta. 2003;327:1–23. doi: 10.1016/s0009-8981(02)00302-9. [DOI] [PubMed] [Google Scholar]
- 106.Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science. 1991;251:802–804. doi: 10.1126/science.1846706. [DOI] [PubMed] [Google Scholar]
- 107.Bolanos-Garcia VM. MET meet adaptors: functional and structural implications in downstream signalling mediated by the Met receptor. Mol Cell Biochem. 2005;276:149–157. doi: 10.1007/s11010-005-3696-6. [DOI] [PubMed] [Google Scholar]
- 108.Yu J, Miehlke S, Ebert MP, et al. Frequency of TPR-MET rearrangement in patients with gastric carcinoma and in first-degree relatives. Cancer. 2000;88:1801–1806. [PubMed] [Google Scholar]
- 109.Dharmawardana PG, Giubellino A, Bottaro DP. Hereditary papillary renal carcinoma type I. Curr Mol Med. 2004;4:855–868. doi: 10.2174/1566524043359674. [DOI] [PubMed] [Google Scholar]
- 110.Bussolino F, Di Renzo MF, Ziche M, et al. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell motility and growth. J Cell Biol. 1992;119:629–641. doi: 10.1083/jcb.119.3.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kitajima Y, Ide T, Ohtsuka T, Miyazaki K. Induction of hepatocyte growth factor activator gene expression under hypoxia activates the hepatocyte growth factor/c-Met system via hypoxia inducible factor-1 in pancreatic cancer. Cancer Sci. 2008;99:1341–1347. doi: 10.1111/j.1349-7006.2008.00828.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kubota T, Taiyoh H, Matsumura A, et al. NK4, an HGF antagonist, prevents hematogenous pulmonary metastasis by inhibiting adhesion of CT26 cells to endothelial cells. Clin Exp Metastasis. 2009;26:447–456. doi: 10.1007/s10585-009-9244-0. [DOI] [PubMed] [Google Scholar]
- 113.Sulpice E, Ding S, Muscatelli-Groux B, et al. Cross-talk between the VEGF-A and HGF signalling pathways in endothelial cells. Biol Cell. 2009;101:525–539. doi: 10.1042/BC20080221. [DOI] [PubMed] [Google Scholar]
- 114.Puri N, Khramtsov A, Ahmed S, et al. A selective small molecule inhibitor of c-Met, PHA665752, inhibits tumorigenicity and angiogenesis in mouse lung cancer xenografts. Cancer Res. 2007;67:3529–3534. doi: 10.1158/0008-5472.CAN-06-4416. [DOI] [PubMed] [Google Scholar]
- 115.Cantelmo AR, Cammarota R, Noonan DM, et al. Cell delivery of Met docking site peptides inhibit angiogenesis and vascular tumor growth. Oncogene. 2010;29:5286–5298. doi: 10.1038/onc.2010.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Gherardi E, Birchmeier W, Birchmeier C, Woude GV. Targeting MET in cancer: Rationale and progress. Nature Reviews Cancer. 2012;12:89–103. doi: 10.1038/nrc3205. [DOI] [PubMed] [Google Scholar]
- 117.Hara S, Nakashiro K, Klosek SK, Ishikawa T, Shintani S, Hamakawa H. Hypoxia enhances c-Met/HGF receptor expression and signaling by activating HIF-1alpha in human salivary gland cancer cells. Oral Oncol. 2006;42:593–598. doi: 10.1016/j.oraloncology.2005.10.016. [DOI] [PubMed] [Google Scholar]
- 118.Ide T, Kitajima Y, Miyoshi A, et al. Tumor-stromal cell interaction under hypoxia increases the invasiveness of pancreatic cancer cells through the hepatocyte growth factor/c-Met pathway. Int J Cancer. 2006;119:2750–2759. doi: 10.1002/ijc.22178. [DOI] [PubMed] [Google Scholar]
- 119.Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003;3:347–361. doi: 10.1016/s1535-6108(03)00085-0. [DOI] [PubMed] [Google Scholar]
- 120.Qian F, Engst S, Yamaguchi K, et al. Inhibition of tumor cell growth, invasion, and metastasis by EXEL-2880 (XL880, GSK1363089), a novel inhibitor of HGF and VEGF receptor tyrosine kinases. Cancer Res. 2009;69:8009–8016. doi: 10.1158/0008-5472.CAN-08-4889. [DOI] [PubMed] [Google Scholar]
- 121.Nakagawa T, Tohyama O, Yamaguchi A, et al. E7050: a dual c-Met and VEGFR-2 tyrosine kinase inhibitor promotes tumor regression and prolongs survival in mouse xenograft models. Cancer Sci. 2010;101:210–215. doi: 10.1111/j.1349-7006.2009.01343.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.You WK, McDonald DM. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008;41:833–839. doi: 10.5483/bmbrep.2008.41.12.833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Shojaei F, Lee JH, Simmons BH, et al. HGF/c-Met acts as an alternative angiogenic pathway in sunitinib-resistant tumors. Cancer Res. 2010;70:10090–10100. doi: 10.1158/0008-5472.CAN-10-0489. [DOI] [PubMed] [Google Scholar]
- 124.Tomioka D, Maehara N, Kuba K, et al. Inhibition of growth, invasion, and metastasis of human pancreatic carcinoma cells by NK4 in an orthotopic mouse model. Cancer Res. 2001;61:7518–7524. [PubMed] [Google Scholar]
- 125.Burgess T, Coxon A, Meyer S, et al. Fully human monoclonal antibodies to hepatocyte growth factor with therapeutic potential against hepatocyte growth factor/c-Met-dependent human tumors. Cancer Res. 2006;66:1721–1729. doi: 10.1158/0008-5472.CAN-05-3329. [DOI] [PubMed] [Google Scholar]
- 126.Martin LP, Sill M, Shahin MS, et al. A phase II evaluation of AMG 102 (rilotumumab) in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2014;132:526–530. doi: 10.1016/j.ygyno.2013.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Buckanovich RJ BR, Sella A, et al. Results from pahse II randomized duscontinuation trial. Journal of clinical oncology ASCO Annual Meeting. 2011;29 abstract 5008. [Google Scholar]
- 128.Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell. 2008;133:38–52. doi: 10.1016/j.cell.2008.03.011. [DOI] [PubMed] [Google Scholar]
- 129.Walker-Daniels J, Hess AR, Hendrix MJ, Kinch MS. Differential regulation of EphA2 in normal and malignant cells. Am J Pathol. 2003;162:1037–1042. doi: 10.1016/S0002-9440(10)63899-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Pasquale EB. The Eph family of receptors. Curr Opin Cell Biol. 1997;9:608–615. doi: 10.1016/s0955-0674(97)80113-5. [DOI] [PubMed] [Google Scholar]
- 131.Thaker PH, Deavers M, Celestino J, et al. EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res. 2004;10:5145–5150. doi: 10.1158/1078-0432.CCR-03-0589. [DOI] [PubMed] [Google Scholar]
- 132.Cheng N, Brantley DM, Liu H, et al. Blockade of EphA receptor tyrosine kinase activation inhibits vascular endothelial cell growth factor-induced angiogenesis. Mol Cancer Res. 2002;1:2–11. [PubMed] [Google Scholar]
- 133.Spannuth WA, Sood AK, Coleman RL. Angiogenesis as a strategic target for ovarian cancer therapy. Nat Clin Pract Oncol. 2008;5:194–204. doi: 10.1038/ncponc1051. [DOI] [PubMed] [Google Scholar]
- 134.Lu XS, Sun W, Ge CY, Zhang WZ, Fan YZ. Contribution of the PI3K/MMPs/Ln-5gamma2 and EphA2/FAK/Paxillin signaling pathways to tumor growth and vasculogenic mimicry of gallbladder carcinomas. Int J Oncol. 2013;42:2103–2115. doi: 10.3892/ijo.2013.1897. [DOI] [PubMed] [Google Scholar]
- 135.Hess AR, Seftor EA, Gardner LM, et al. Molecular regulation of tumor cell vasculogenic mimicry by tyrosine phosphorylation: role of epithelial cell kinase (Eck/EphA2) Cancer Res. 2001;61:3250–3255. [PubMed] [Google Scholar]
- 136.Landen CN, Jr, Chavez-Reyes A, Bucana C, et al. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 2005;65:6910–6918. doi: 10.1158/0008-5472.CAN-05-0530. [DOI] [PubMed] [Google Scholar]
- 137.Adam MG, Berger C, Feldner A, et al. Synaptojanin-2 binding protein stabilizes the Notch ligands DLL1 and DLL4 and inhibits sprouting angiogenesis. Circ Res. 2013;113:1206–1218. doi: 10.1161/CIRCRESAHA.113.301686. [DOI] [PubMed] [Google Scholar]
- 138.Hu W, Lu C, Dong HH, et al. Biological roles of the Delta family Notch ligand Dll4 in tumor and endothelial cells in ovarian cancer. Cancer Res. 2011;71:6030–6039. doi: 10.1158/0008-5472.CAN-10-2719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Gale NW, Dominguez MG, Noguera I, et al. Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci U S A. 2004;101:15949–15954. doi: 10.1073/pnas.0407290101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Lobov IB, Renard RA, Papadopoulos N, et al. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative regulator of angiogenic sprouting. Proc Natl Acad Sci U S A. 2007;104:3219–3224. doi: 10.1073/pnas.0611206104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Thurston G, Noguera-Troise I, Yancopoulos GD. The Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth. Nat Rev Cancer. 2007;7:327–331. doi: 10.1038/nrc2130. [DOI] [PubMed] [Google Scholar]
- 142.Ridgway J, Zhang G, Wu Y, et al. Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature. 2006;444:1083–1087. doi: 10.1038/nature05313. [DOI] [PubMed] [Google Scholar]
- 143.Li JL, Sainson RC, Shi W, et al. Delta-like 4 Notch ligand regulates tumor angiogenesis, improves tumor vascular function, and promotes tumor growth in vivo. Cancer Res. 2007;67:11244–11253. doi: 10.1158/0008-5472.CAN-07-0969. [DOI] [PubMed] [Google Scholar]
- 144.Tolcher AW, Messersmith WA, Mikulski SM, et al. Phase I study of RO4929097, a gamma secretase inhibitor of notch signaling, in patients with refractory metastatic or locally advanced solid tumors. Journal of Clinical Oncology. 2012;30:2348–2353. doi: 10.1200/JCO.2011.36.8282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sahebjam S, Bedard PL, Castonguay V, et al. A phase i study of the combination of ro4929097 and cediranib in patients with advanced solid tumours (PJC-004/NCI 8503) Br J Cancer. 2013;109:943–949. doi: 10.1038/bjc.2013.380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Diaz-Padilla I, Hirte H, Oza AM, et al. A phase Ib combination study of RO4929097, a gamma-secretase inhibitor, and temsirolimus in patients with advanced solid tumors. Invest New Drugs. 2013;31:1182–1191. doi: 10.1007/s10637-013-0001-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Strosberg JR, Yeatman T, Weber J, et al. A phase II study of RO4929097 in metastatic colorectal cancer. European Journal of Cancer. 2012;48:997–1003. doi: 10.1016/j.ejca.2012.02.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Summy JM, Gallick GE. Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev. 2003;22:337–358. doi: 10.1023/a:1023772912750. [DOI] [PubMed] [Google Scholar]
- 149.Ma WW, Adjei AA. Novel Agents on the Horizon for Cancer Therapy. CA: A Cancer Journal for Clinicians. 2009;59:111–137. doi: 10.3322/caac.20003. [DOI] [PubMed] [Google Scholar]
- 150.Frame MC. Src in cancer: deregulation and consequences for cell behaviour. Biochim Biophys Acta. 2002;1602:114–130. doi: 10.1016/s0304-419x(02)00040-9. [DOI] [PubMed] [Google Scholar]
- 151.Trevino JG, Summy JM, Gray MJ, et al. Expression and activity of SRC regulate interleukin-8 expression in pancreatic adenocarcinoma cells: implications for angiogenesis. Cancer Res. 2005;65:7214–7222. doi: 10.1158/0008-5472.CAN-04-3858. [DOI] [PubMed] [Google Scholar]
- 152.Kanda S, Miyata Y, Kanetake H, Smithgall TE. Non-receptor protein-tyrosine kinases as molecular targets for antiangiogenic therapy (Review) Int J Mol Med. 2007;20:113–121. [PubMed] [Google Scholar]
- 153.Labrecque L, Royal I, Surprenant DS, Patterson C, Gingras D, Béliveau R. Regulation of vascular endothelial growth factor receptor-2 activity by caveolin-1 and plasma membrane cholesterol. Mol Biol Cell. 2003;14:334–347. doi: 10.1091/mbc.E02-07-0379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Eliceiri BP, Puente XS, Hood JD, et al. Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol. 2002;157:149–160. doi: 10.1083/jcb.200109079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kim YM, Lee YM, Kim HS, et al. TNF-related activation-induced cytokine (TRANCE) induces angiogenesis through the activation of Src and phospholipase C (PLC) in human endothelial cells. J Biol Chem. 2002;277:6799–6805. doi: 10.1074/jbc.M109434200. [DOI] [PubMed] [Google Scholar]
- 156.Laird AD, Li G, Moss KG, et al. Src family kinase activity is required for signal tranducer and activator of transcription 3 and focal adhesion kinase phosphorylation and vascular endothelial growth factor signaling in vivo and for anchorage-dependent and -independent growth of human tumor cells. Mol Cancer Ther. 2003;2:461–469. [PubMed] [Google Scholar]
- 157.Bankhead C. ESMO: Failed Trials Dominate Gyn Cancer Session. 2010. [Accessed OCT, 14, 2010]. [Google Scholar]
- 158.McNeish IA, Ledermann JA, Webber LC, et al. A randomized placebo-controlled trial of saracatinib (AZD0530) plus weekly paclitaxel in platinum-resistant ovarian, fallopian-tube, or primary peritoneal cancer (SaPPrOC) J Clin Oncol. 2013;31 doi: 10.1093/annonc/mdu363. [DOI] [PubMed] [Google Scholar]
- 159.Hermann C, Assmus B, Urbich C, Zeiher AM, Dimmeler S. Insulin-mediated stimulation of protein kinase Akt: A potent survival signaling cascade for endothelial cells. Arterioscler Thromb Vasc Biol. 2000;20:402–409. doi: 10.1161/01.atv.20.2.402. [DOI] [PubMed] [Google Scholar]
- 160.Granville CA, Memmott RM, Gills JJ, Dennis PA. Handicapping the race to develop inhibitors of the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway. Clin Cancer Res. 2006;12:679–689. doi: 10.1158/1078-0432.CCR-05-1654. [DOI] [PubMed] [Google Scholar]
- 161.Frisch SM, Ruoslahti E. Integrins and anoikis. Curr Opin Cell Biol. 1997;9:701–706. doi: 10.1016/s0955-0674(97)80124-x. [DOI] [PubMed] [Google Scholar]
- 162.Gerber HP, McMurtrey A, Kowalski J, et al. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem. 1998;273:30336–30343. doi: 10.1074/jbc.273.46.30336. [DOI] [PubMed] [Google Scholar]
- 163.Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96:857–868. doi: 10.1016/s0092-8674(00)80595-4. [DOI] [PubMed] [Google Scholar]
- 164.Yang D, Sun Y, Hu L, et al. Integrated analyses identify a master microRNA regulatory network for the mesenchymal subtype in serous ovarian cancer. Cancer Cell. 2013;23:186–199. doi: 10.1016/j.ccr.2012.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Bianco R, Garofalo S, Rosa R, et al. Inhibition of mTOR pathway by everolimus cooperates with EGFR inhibitors in human tumours sensitive and resistant to anti-EGFR drugs. Br J Cancer. 2008;98:923–930. doi: 10.1038/sj.bjc.6604269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Jin H, Varner J. Integrins: roles in cancer development and as treatment targets. Br J Cancer. 2004;90:561–565. doi: 10.1038/sj.bjc.6601576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ruegg C, Mariotti A. Vascular integrins: pleiotropic adhesion and signaling molecules in vascular homeostasis and angiogenesis. Cell Mol Life Sci. 2003;60:1135–1157. doi: 10.1007/s00018-003-2297-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.da Silva RG, Tavora B, Robinson SD, et al. Endothelial alpha3beta1-integrin represses pathological angiogenesis and sustains endothelial-VEGF. Am J Pathol. 2010;177:1534–1548. doi: 10.2353/ajpath.2010.100043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Assoian RK. Anchorage-dependent cell cycle progression. J Cell Biol. 1997;136:1–4. doi: 10.1083/jcb.136.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Urbich C, Dernbach E, Reissner A, Vasa M, Zeiher AM, Dimmeler S. Shear stress-induced endothelial cell migration involves integrin signaling via the fibronectin receptor subunits alpha(5) and beta(1) Arterioscler Thromb Vasc Biol. 2002;22:69–75. doi: 10.1161/hq0102.101518. [DOI] [PubMed] [Google Scholar]
- 171.Abedi H, Zachary I. Vascular endothelial growth factor stimulates tyrosine phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and paxillin in endothelial cells. J Biol Chem. 1997;272:15442–15451. doi: 10.1074/jbc.272.24.15442. [DOI] [PubMed] [Google Scholar]
- 172.Brooks PC, Stromblad S, Klemke R, Visscher D, Sarkar FH, Cheresh DA. Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest. 1995;96:1815–1822. doi: 10.1172/JCI118227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Gutheil JC, Campbell TN, Pierce PR, et al. Targeted antiangiogenic therapy for cancer using Vitaxin: a humanized monoclonal antibody to the integrin alphavbeta3. Clin Cancer Res. 2000;6:3056–3061. [PubMed] [Google Scholar]
- 174.Stupp R, Ruegg C. Integrin inhibitors reaching the clinic. J Clin Oncol. 2007;25:1637–1638. doi: 10.1200/JCO.2006.09.8376. [DOI] [PubMed] [Google Scholar]
- 175.Shibata K, Kikkawa F, Nawa A, Suganuma N, Hamaguchi M. Fibronectin secretion from human peritoneal tissue induces Mr 92,000 type IV collagenase expression and invasion in ovarian cancer cell lines. Cancer Res. 1997;57:5416–5420. [PubMed] [Google Scholar]
- 176.Sawada K, Mitra AK, Radjabi AR, et al. Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res. 2008;68:2329–2339. doi: 10.1158/0008-5472.CAN-07-5167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Park CC, Zhang H, Pallavicini M, et al. Beta1 integrin inhibitory antibody induces apoptosis of breast cancer cells, inhibits growth, and distinguishes malignant from normal phenotype in three dimensional cultures and in vivo. Cancer Res. 2006;66:1526–1535. doi: 10.1158/0008-5472.CAN-05-3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Bhaskar V, Zhang D, Fox M, et al. A function blocking anti-mouse integrin alpha5beta1 antibody inhibits angiogenesis and impedes tumor growth in vivo. J Transl Med. 2007;5:61. doi: 10.1186/1479-5876-5-61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Bhaskar V, Fox M, Breinberg D, et al. Volociximab, a chimeric integrin alpha5beta1 antibody, inhibits the growth of VX2 tumors in rabbits. Invest New Drugs. 2008;26:7–12. doi: 10.1007/s10637-007-9078-z. [DOI] [PubMed] [Google Scholar]
- 180.Ramakrishnan V, Bhaskar V, Law DA, et al. Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. J Exp Ther Oncol. 2006;5:273–286. [PubMed] [Google Scholar]
- 181.Bell-McGuinn KM, Matthews CM, Ho SN, et al. A phase II, single-arm study of the anti-alpha5beta1 integrin antibody volociximab as monotherapy in patients with platinum-resistant advanced epithelial ovarian or primary peritoneal cancer. Gynecol Oncol. 2011;121:273–279. doi: 10.1016/j.ygyno.2010.12.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Naylor MS, Stamp GW, Foulkes WD, Eccles D, Balkwill FR. Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression. J Clin Invest. 1993;91:2194–2206. doi: 10.1172/JCI116446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wu S, Boyer CM, Whitaker RS, et al. Tumor necrosis factor alpha as an autocrine and paracrine growth factor for ovarian cancer: monokine induction of tumor cell proliferation and tumor necrosis factor alpha expression. Cancer Res. 1993;53:1939–1944. [PubMed] [Google Scholar]
- 184.Kulbe H, Thompson R, Wilson JL, et al. The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res. 2007;67:585–592. doi: 10.1158/0008-5472.CAN-06-2941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Jin DK, Shido K, Kopp HG, et al. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nat Med. 2006;12:557–567. doi: 10.1038/nm1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–827. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kryczek I, Lange A, Mottram P, et al. CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res. 2005;65:465–472. [PubMed] [Google Scholar]
- 188.Aguayo A, Kantarjian H, Manshouri T, et al. Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood. 2000;96:2240–2245. [PubMed] [Google Scholar]
- 189.Charles KA, Kulbe H, Soper R, et al. The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest. 2009;119:3011–3023. doi: 10.1172/JCI39065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Kulbe H, Chakravarty P, Leinster DA, et al. A dynamic inflammatory cytokine network in the human ovarian cancer microenvironment. Cancer Res. 2012;72:66–75. doi: 10.1158/0008-5472.CAN-11-2178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Madhusudan S, Muthuramalingam SR, Braybrooke JP, et al. Study of etanercept, a tumor necrosis factor-alpha inhibitor, in recurrent ovarian cancer. J Clin Oncol. 2005;23:5950–5959. doi: 10.1200/JCO.2005.04.127. [DOI] [PubMed] [Google Scholar]
- 192.Giuntoli RL, 2nd, Webb TJ, Zoso A, et al. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res. 2009;29:2875–2884. [PubMed] [Google Scholar]
- 193.Lane D, Matte I, Rancourt C, Piche A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer. 2011;11:210. doi: 10.1186/1471-2407-11-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Dankbar B, Padro T, Leo R, et al. Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood. 2000;95:2630–2636. [PubMed] [Google Scholar]
- 195.Nilsson MB, Langley RR, Fidler IJ. Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Res. 2005;65:10794–10800. doi: 10.1158/0008-5472.CAN-05-0623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Rabinovich A, Medina L, Piura B, Segal S, Huleihel M. Regulation of ovarian carcinoma SKOV-3 cell proliferation and secretion of MMPs by autocrine IL-6. Anticancer Res. 2007;27:267–272. [PubMed] [Google Scholar]
- 197.Scambia G, Testa U, Benedetti Panici P, et al. Prognostic significance of interleukin 6 serum levels in patients with ovarian cancer. Br J Cancer. 1995;71:354–356. doi: 10.1038/bjc.1995.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Guo Y, Nemeth J, O'Brien C, et al. Effects of siltuximab on the IL-6-induced signaling pathway in ovarian cancer. Clin Cancer Res. 2010;16:5759–5769. doi: 10.1158/1078-0432.CCR-10-1095. [DOI] [PubMed] [Google Scholar]
- 199.Coward J, Kulbe H, Chakravarty P, et al. Interleukin-6 as a therapeutic target in human ovarian cancer. Clin Cancer Res. 2011;17:6083–6096. doi: 10.1158/1078-0432.CCR-11-0945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391:806–811. doi: 10.1038/35888. [DOI] [PubMed] [Google Scholar]
- 201.Hammond SM, Bernstein E, Beach D, Hannon GJ. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature. 2000;404:293–296. doi: 10.1038/35005107. [DOI] [PubMed] [Google Scholar]
- 202.Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411:494–498. doi: 10.1038/35078107. [DOI] [PubMed] [Google Scholar]
- 203.Fattal E, Bochot A. Ocular delivery of nucleic acids: antisense oligonucleotides, aptamers and siRNA. Adv Drug Deliv Rev. 2006;58:1203–1223. doi: 10.1016/j.addr.2006.07.020. [DOI] [PubMed] [Google Scholar]
- 204.Bitko V, Musiyenko A, Shulyayeva O, Barik S. Inhibition of respiratory viruses by nasally administered siRNA. Nat Med. 2005;11:50–55. doi: 10.1038/nm1164. [DOI] [PubMed] [Google Scholar]
- 205.Ozpolat B, Sood AK, Lopez-Berestein G. Nanomedicine based approaches for the delivery of siRNA in cancer. J Intern Med. 2010;267:44–53. doi: 10.1111/j.1365-2796.2009.02191.x. [DOI] [PubMed] [Google Scholar]
- 206.Zhou J, Shum KT, Burnett JC, Rossi JJ. Nanoparticle-Based Delivery of RNAi Therapeutics: Progress and Challenges. Pharmaceuticals (Basel) 2013;6:85–107. doi: 10.3390/ph6010085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Tan WB, Jiang S, Zhang Y. Quantum-dot based nanoparticles for targeted silencing of HER2/neu gene via RNA interference. Biomaterials. 2007;28:1565–1571. doi: 10.1016/j.biomaterials.2006.11.018. [DOI] [PubMed] [Google Scholar]
- 208.Lee JH, Lee K, Moon SH, Lee Y, Park TG, Cheon J. All-in-one target-cell-specific magnetic nanoparticles for simultaneous molecular imaging and siRNA delivery. Angew Chem Int Ed Engl. 2009;48:4174–4179. doi: 10.1002/anie.200805998. [DOI] [PubMed] [Google Scholar]
- 209.Yu D, Peng P, Dharap SS, et al. Antitumor activity of poly(ethylene glycol)-camptothecin conjugate: the inhibition of tumor growth in vivo. J Control Release. 2005;110:90–102. doi: 10.1016/j.jconrel.2005.09.050. [DOI] [PubMed] [Google Scholar]
- 210.Maeda H. The enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targeting. Adv Enzyme Regul. 2001;41:189–207. doi: 10.1016/s0065-2571(00)00013-3. [DOI] [PubMed] [Google Scholar]
- 211.Nikitenko NA, Prassolov VS. Non-Viral Delivery and Therapeutic Application of Small Interfering RNAs. Acta Naturae. 2013;5:35–53. [PMC free article] [PubMed] [Google Scholar]
- 212.Davis ME, Zuckerman JE, Choi CH, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464:1067–1070. doi: 10.1038/nature08956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Heidel JD, Yu Z, Liu JY, et al. Administration in non-human primates of escalating intravenous doses of targeted nanoparticles containing ribonucleotide reductase subunit M2 siRNA. Proc Natl Acad Sci U S A. 2007;104:5715–5721. doi: 10.1073/pnas.0701458104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Matei D, Emerson RE, Schilder J, et al. Imatinib mesylate in combination with docetaxel for the treatment of patients with advanced, platinum-resistant ovarian cancer and primary peritoneal carcinomatosis : a Hoosier Oncology Group trial. Cancer. 2008;113:723–732. doi: 10.1002/cncr.23605. [DOI] [PubMed] [Google Scholar]
- 215.Juretzka M, Hensley ML, Tew W, et al. A phase 2 trial of oral imatinib in patients with epithelial ovarian, fallopian tube, or peritoneal carcinoma in second or greater remission. Eur J Gynaecol Oncol. 2008;29:568–572. [PubMed] [Google Scholar]
- 216.Liu JF, Tolaney SM, Birrer M, et al. A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer. 2013;49:2972–2978. doi: 10.1016/j.ejca.2013.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Hjalmarson A. Heart rate and beta-adrenergic mechanisms in acute myocardial infarction. Basic Res Cardiol. 1990;85(Suppl 1):325–333. doi: 10.1007/978-3-662-11038-6_26. [DOI] [PubMed] [Google Scholar]
- 218.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;123:33–36. doi: 10.1016/j.ygyno.2011.06.019. [DOI] [PubMed] [Google Scholar]
- 219.Campos SM, Penson RT, Matulonis U, et al. A phase II trial of Sunitinib malate in recurrent and refractory ovarian, fallopian tube and peritoneal carcinoma. Gynecol Oncol. 2013;128:215–220. doi: 10.1016/j.ygyno.2012.07.126. [DOI] [PubMed] [Google Scholar]
- 220.Baumann KH, du Bois A, Meier W, et al. A phase II trial (AGO 2.11) in platinum-resistant ovarian cancer: a randomized multicenter trial with sunitinib (SU11248) to evaluate dosage, schedule, tolerability, toxicity and effectiveness of a multitargeted receptor tyrosine kinase inhibitor monotherapy. Ann Oncol. 2012;23:2265–2271. doi: 10.1093/annonc/mds003. [DOI] [PubMed] [Google Scholar]
- 221.Karlan BY, Oza AM, Richardson GE, et al. Randomized, double-blind, placebo-controlled phase II study of AMG 386 combined with weekly paclitaxel in patients with recurrent ovarian cancer. J Clin Oncol. 2012;30:362–371. doi: 10.1200/JCO.2010.34.3178. [DOI] [PubMed] [Google Scholar]
- 222.Secord AA, Teoh DK, Barry WT, et al. A phase I trial of dasatinib, an SRC-family kinase inhibitor, in combination with paclitaxel and carboplatin in patients with advanced or recurrent ovarian cancer. Clin Cancer Res. 2012;18:5489–5498. doi: 10.1158/1078-0432.CCR-12-0507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Schilder RJ, Brady WE, Lankes HA, et al. Phase II evaluation of dasatinib in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2012;127:70–74. doi: 10.1016/j.ygyno.2012.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.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;123:19–26. doi: 10.1016/j.ygyno.2011.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Temkin SM, Yamada SD, Fleming GF. A phase I study of weekly temsirolimus and topotecan in the treatment of advanced and/or recurrent gynecologic malignancies. Gynecol Oncol. 2010;117:473–476. doi: 10.1016/j.ygyno.2010.02.022. [DOI] [PubMed] [Google Scholar]
- 226.Kollmannsberger C, Hirte H, Siu LL, et al. Temsirolimus in combination with carboplatin and paclitaxel in patients with advanced solid tumors: a NCIC-CTG, phase I, open-label dose-escalation study (IND 179) Ann Oncol. 2012;23:238–244. doi: 10.1093/annonc/mdr063. [DOI] [PMC free article] [PubMed] [Google Scholar]