Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 5.
Published in final edited form as: Mol Imaging. 2011 Apr 26;10(4):248–257. doi: 10.2310/7290.2011.00004

Intraoperative Imaging in Ovarian Cancer: Fact or Fiction?

Lucia MA Crane 1, Marleen van Oosten 1, Rick G Pleijhuis 1, Arash Motekallemi 1, Sean C Dowdy 1, William A Cliby 1, Ate GJ van der Zee 1, Gooitzen M van Dam 1
PMCID: PMC3763956  NIHMSID: NIHMS504272  PMID: 21521557

Abstract

Tumor-targeted fluorescence imaging for cancer diagnosis and treatment is an evolving field of research that is on the verge of clinical implementation. As each tumor has its unique biologic profile, selection of the most promising targets is essential. In this review, we focus on target finding in ovarian cancer, a disease in which fluorescence imaging may be of value in both adequate staging and in improving cytoreductive efforts, and as such may have a beneficial effect on prognosis. Thus far, tumor-targeted imaging for ovarian cancer has been applied only in animal models. For clinical implementation, the five most prominent targets were identified: folate receptor α, vascular endothelial growth factor, epidermal growth factor receptor, chemokine receptor 4, and matrix metalloproteinase. These targets were selected based on expression rates in ovarian cancer, availability of an antibody or substrate aimed at the target approved by the Food and Drug Administration, and the likelihood of translation to human use. The purpose of this review is to present requirements for intraoperative imaging and to discuss possible tumor-specific targets for ovarian cancer, prioritizing for targets with substrates ready for introduction into the clinic.


THE DETECTION AND MANAGEMENT OF CANCER have benefited greatly from the development of sensitive imaging modalities. Computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), single-photon emission computed tomography (SPECT), and ultrasonography all have unique advantages in visualizing tumors, and technical improvements such as multimodality systems (PET-CT, PET-MRI) have led to increasing efficiency and sensitivity and detailed two- and three-dimensional images. However, these modalities are not suitable for real-time feedback during surgery. In diseases with a peritoneal spreading pattern, such as ovarian cancer, cytoreduction is of eminent importance. Intraoperative fluorescence imaging aiding the surgeon in detection of (metastatic) tumor deposits may improve resection rates and thus positively influence prognosis. As tumor-targeted fluorescence imaging is on the verge of clinical implementation, we set out to provide an overview of useful imaging targets in ovarian cancer. In this review, the possible targets for intraoperative imaging in ovarian cancer are evaluated. First, we illustrate the importance of improved tumor visualization during ovarian cancer surgery and describe the basics of fluorescence imaging. Subsequently, the development of tumor-specific tracers for ovarian cancer toward clinical introduction is discussed.

Methods

PubMed was used as the primary database for search queries. The general search method was based on the full name and abbreviation for each target discussed and on combinations of the following search terms: gynecologic cancer, ovarian cancer or carcinoma, intraoperative imaging, and fluorescence imaging. To maximize the search results, we did not prioritize for the presence of the search terms in titles, abstracts, or key words. Relevant articles were selected and examined, including a screening identifying possibly relevant cited reports. Only articles written in English were considered for citation in this review. There was no limitation on the publication date, but emphasis was put on articles published in the last decade.

Imaging in Ovarian Cancer

Ovarian cancer, of which epithelial ovarian cancer (EOC) is the predominant histology, is a major cause of mortality in women, ranking the fifth most common cause of death from cancer.1 Because of late onset of symptoms, 75% of women are diagnosed with advanced disease, and 5-year survival rates for stage IV cancer are only about 30%.2 In advanced disease, the degree of cytoreduction is one of the most important prognostic factors.35 Given the improved survival associated with complete resection of all visible disease,6,7 debate is ongoing as to whether this is a more appropriate definition for optimal cytoreduction than the current standard of residual disease < 1 cm.8,9 The size of residual disease after cytoreduction is also of importance for penetration of adjuvant systemic or intraperitoneal chemotherapy into tumor nodules,1013 highlighting the importance of a thorough inspection of the abdomen for metastases.

Preoperative imaging offers a guideline for the multidisciplinary team and in particular the surgeon. However, the accuracy of computed tomography (CT) for adequate staging is around 75%1416 and depending solely on CT may thus result in over- or undertreatment. In discriminating benign from malignant masses, PET-CT is superior to other modalities, with an accuracy rate of 92%, compared to 75% and 83% for MRI and ultrasonography, respectively.16 For detection of large peritoneal metastases, CT and MRI are equally sensitive, up to 100%.17 Adding diffusion-weighted MRI to conventional MRI improves the detection sensitivity of peritoneal metastases.18,19 The greatest difficulty is the detection of very small tumor deposits in peritoneal carcinomatosis, which are missed by the above-mentioned modalities in 20 to 30% of cases but are pivotal for accurate staging.20 Laparoscopic staging is an attractive, less invasive option for staging and is considered safe in early-stage ovarian cancer.21 In more advanced stages, however, adhesions may impede thorough inspection of all abdominal quadrants.22 Therefore, current guidelines recommend exploratory laparotomy as the only fully reliable staging method.23,24 As an adjunct to this, intraoperative optical imaging for in vivo tumor detection could be of value for improving both adequate staging and cytoreduction. Real-time fluorescence imaging provides high sensitivity and resolution, illustrated by preclinical studies that report detection resolutions of 0.3 mm in an ovarian cancer xenograft model in mice.25

In Vivo Fluorescence Imaging

Intraoperative imaging is successful only if the tumor-specific signal derived from the targeted optical contrast agent is clearly discernible from the background, leading to low false-negative and false-positive rates of detection. As all tissues have some degree of autofluorescence, the signal from the fluorescent agent is impeded to some extent, especially in visible light wavelengths (400–750 nm). In this respect, the use of near-infrared (NIR) fluorescent dyes (750–1,000 nm) is advantageous for in vivo imaging because surrounding tissues generally absorb little of the emitted signal and light in the NIR spectrum does not compete with fluorescence in the visible light spectrum.26 Moreover, near-infrared fluorescence (NIRF) has reduced scattering properties compared to fluorescence detection in the visible light spectrum. NIRF imaging therefore gives a high signal at the target site, whereas surrounding tissues emit little to no signal. As this so-called high signal to background ratio is preferable, development of imaging agents is centered on the NIR wavelengths.27 The spectrum of fluorescent agents in the NIR range is broad, but only one has been approved by the Food and Drug Administration (FDA) for human use: indocyanine green (emission ≈820 nm).28,29 Although fluorescein (emission ≈518 nm) is also FDA approved,30 the emission wavelength of this agent is not within the range of the NIR spectrum. New promising dyes, such as infrared dye 800CW (emission ≈805 nm), are currently in preclinical testing for toxicity.31

With regard to the detection device, emitted NIRF signals cannot be perceived with the naked eye. Therefore, a sensitive multispectral NIR camera system is needed to simultaneously detect the fluorescence and the color signal and process it into one image. The ideal intraoperative camera system is light, portable, and easily operated with high data processing speed and detailed imaging properties. To date, only a few clinically approved intraoperative camera systems and laparoscopic fluorescence cameras have been described. These workable fluorescence camera systems have been extensively described.3235

Tumor-Specific Targets for Fluorescence Imaging

The ideal marker for tumor-targeted intraoperative NIRF imaging is tumor specific, clinically approved, safe, and nontoxic. Also, the marker needs to be easily conjugated to a NIRF dye.36 In the following paragraphs, we elaborate on five targets that are promising candidates for tumor-specific fluorescence imaging in ovarian cancer. This review is limited to agents that are already FDA approved for therapeutic or diagnostic imaging purposes, thus facilitating introduction into the clinic within the next 5 years.

Folate Receptor

The folate receptor (FR) is one of the most promising targets in EOC. The transmembrane receptor consists of four isoforms, of which FR-α is present on malignant cells, whereas FR-β is located on activated macrophages.37 FR-γ and FR-δ are reported to play a role in regulatory T cells.38 In general, FR-α is overexpressed by approximately 40% of human cancers, but expression rates vary greatly between tumor types.39 Given that FR-α is overexpressed in EOC in 72 to 97%, in both primary tumor tissue and metastatic tumor deposits,40,41 applications targeting FR-α can be applied in a large group of patients without prior screening for expression of the target. The advantages of folate as a biomarker are that it binds to FR with high affinity, is inexpensive, can be easily coupled to other substances, and has little or no toxicity.42

Folate and folate conjugates are quickly internalized in the cell via receptor-mediated endocytosis within 2 hours.43 The macromolecule inside the endosome remains intact and can therefore fully exert its function inside the cell.43,44 This led to the development of a broad variety of folate-targeted conjugates such as chemotherapeutic agents and radioactive tracers.4448 Studies with 111In-DTPA-folate as an imaging agent clearly show uptake in ovarian cancer, as well as in the kidneys, where FR-α is expressed on cells in the proximal tubule under physiologic conditions.49,50 Subsequently, a 99mTc-based folate tracer, EC20, was developed by Endocyte Inc. (West Lafayette, IN). This tracer is cleared more rapidly from the body.51,52

Farletuzumab (MORAb-003) is an antibody with high affinity for FR-α that has been evaluated preclinically and is currently being introduced in the clinic for therapeutic purposes.5355 Furthermore, an imaging study using radiolabeled 111-In-DOTA-MORAb-003 reports excellent biodistribution and tumor visualization, in both mice and patients.56

In short, FR-α is a promising target in EOC, and the above-mentioned FR-α-targeted agents provide sensitive visualization of tumor tissue. This leads us to believe that folate conjugated to a fluorescent dye could be suitable for intraoperative imaging in FR-α-expressing tumors, a concept that has already been reported in a preclinical study in mice.57

Hypoxia-Related Proteins

Hypoxia plays an important role in the development and aggressiveness of several types of solid tumors, including EOC.58 Tumors develop hypoxic areas when blood supply becomes inadequate in rapidly proliferating tissue. Moreover, hypoxic tumors show more aggressive behavior and appear to have a worse prognosis owing to a greater potential to metastasize, a decreased response to cell damage owing to inactivation of the tumor suppressor gene P53, and increased resistance to both radio- and chemotherapy.5961 In instances of low oxygen pressure, hypoxia-inducible-factor 1 (HIF-1) is expressed, of which the alpha subunit (HIF-1α) affects several regulatory processes, such as apoptosis and proliferation, through modulation of more than 100 hypoxia-related genes and their hypoxia- responsive elements (HREs).6265 Two of these are of special significance in the context of intraoperative imaging in EOC. Both epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor 1 (VEGFR-1) are upregulated by HIF-1α in EOC66,67 and could be suitable targets for imaging applications for two reasons. First, given that most ovarian cancers are diagnosed in a late stage, when the tumor is already of reasonable size, it is realistic to assume that there are areas of hypoxia and thus involvement of hypoxia-inducible genes. This is confirmed by genomic and proteomic testing of patient material.6870 Second, hypoxic tumors tend to metastasize more rapidly, and the associated hypoxia-inducible genes can be detected in the primary tumor as well as in metastatic tumor deposits.71 This implies that their protein products or, even at the cellular level, their HREs may serve as a target in intraoperative imaging to visualize both the primary tumor and foci of disseminated disease.

Vascular Endothelial Growth Factor

Vascular endothelial growth factor (VEGF), an important regulator of angiogenesis and vascular permeability, is involved in various steps of normal ovarian function as well as carcinogenesis.62,72,73 VEGF-related growth factors and receptors form primary signaling pathways in tumor angiogenesis and have been implicated as regulators of angiogenesis and disease progression in EOC through proliferation, cell survival, and metastasis.73,74 Triggering this so-called “angiogenic switch” by overexpression of a potent angiogenic growth factor is an important step in the growth and dissemination of EOC. VEGF activates two high-affinity transmembrane tyrosine kinase cell surface receptors, VEGFR-1 and VEGFR-2, which stimulate intracellular signaling cascades, leading to endothelial cell recruitment, activation, proliferation, and survival, as well as increasing vascular permeability.62 Of all VEGF subtypes, the VEGF-A ligand and VEGFR-2 receptor seem to play a particularly prominent role in EOC.75

Several studies are now focused on the value of VEGF inhibition in the treatment of EOC.76 Apart from therapeutic relevance, some of these agents could also be used for tumor-targeted imaging. In this light, we focus on bevacizumab (Avastin, F. Hoffmann-La Roche Ltd, Basel, Switzerland), a promising drug that has already been useful in several conventional imaging applications, such as PET. Bevacizumab is a humanized monoclonal antibody that binds to and neutralizes VEGF-A, preventing its association with endothelial receptors and, as a consequence, inhibiting angiogenesis. Inhibiting microvascular outgrowth is believed to slow down the growth of all tumor tissue, including metastatic sites. Recent clinical trials have reported that bevacizumab prolongs progression-free survival in EOC.7780

Bevacizumab itself can be conjugated to radioactive substances for application in PET. This was illustrated in a murine ovarian tumor xenograft model, in which bevacizumab conjugated to 111In or 89Zr showed clear tumor uptake on microPET.81 Subsequently, 111In-bevacizumab SPECT can clearly visualize tumor lesions in melanoma patients.82 These developments suggest that bevacizumab may also be conjugated to NIRF dyes and as such provide a fluorescent agent for intraoperative imaging of tumor activity.

Apart from targeting VEGF-A, fluorescence imaging of the VEGFR has also been shown, using a single-chain fusion protein (scVEGF) coupled to the fluorescent dye Cy5.5.83

Although VEGF-guided imaging seems promising, its specificity could be limited by the fact that VEGF is also scavenged in fluids and thus ascites and serves as an important survival factor for healthy cells experiencing chemical or physical stress.8488 Possibly, the presence of VEGF-A in ascites might hamper the detection of tumor spots in vivo. However, considering the high expression of VEGF in tumor tissue, the signal to background ratio may be sufficient to distinguish the strong tumor-related signal from the much weaker signal from healthy tissues. This assumption was illustrated in the above-mentioned murine model with radiolabeled bevacizumab,81 although it should be taken into account that the murine peritoneum does not excrete human VEGF-A. Moreover, during cytoreduction, the abdomen is thoroughly inspected, consecutively focusing on specific regions of interest—that is, the abdomen, pelvis, diaphragm, and liver capsule—and VEGF expression in other tissues will probably not interfere with imaging of this specific region. Furthermore, ascites can be drained during surgery for improving detection.

Epidermal Growth Factor Receptor

Members of the epidermal growth factor family and the associated receptor, EGFR, play a significant role in ovarian cancer as the majority of EOCs express high levels of EGFR.8991 This overexpression of EGFR is an independent factor for poor outcome and low survival rates.9294 The EGFR superfamily is made up of four distinct but structurally similar tyrosine kinase receptors. Upregulation leads to cell proliferation, differentiation, migration, adhesion, protection from apoptosis, and transformation.95

A few EGFR-targeted monoclonal antibodies are already available in the clinical setting, including cetuximab and panitumumab.96 These are used in therapeutic regimens but may also prove useful for imaging purposes. Panitumumab, either radiolabeled or in its pure form, conjugated to indocyanine green, has been shown to yield excellent optical imaging signals when used in a mouse model.97,98 Radiolabeled cetuximab for tumor-targeted imaging has also been successfully used in murine tumor models.99,100 To date, no clinical studies have been performed using an EGFR antibody for this type of tumor-targeted imaging. However, the existence of FDA-approved antibodies and promising preclinical data may lead to rapid translation into the clinic.

Because EGFR is expressed not only by cancer cells but also by healthy cells, one might suggest that tumor-specific imaging using EGFR as a target is impossible. However, expression rates in tumor tissue are 20 to 50 times higher than in the surrounding healthy tissue.101 This makes the signal to background ratio large enough for sensitive tumor-targeted imaging. As such, EGFR can also be regarded as a potential target for intraoperative imaging in ovarian cancer.

Chemokine Receptor 4 and Chemokine Ligand 12

Dissemination of disease in EOC starts with the loss of cell adhesion molecules and degradation of the extracellular matrix, after which migration of tumor cells commences.102 In this process, there is a key role for matrix metalloproteinases (MMPs) and several chemokine-receptor pairs, of which chemokine receptor 4 (CXCR4) and its chemokine, chemokine ligand 12 (CXCL12), or stromal cell–derived factor 1 (SDF-1) are among the most well studied.103 Many of the mechanisms governing the structure and migration of cells are influenced and disrupted by hypoxia and HIF-1 (for a comphrehensive review, see Gort and colleagues102). Although not specific for ovarian cancer, both MMP and CXCR4-CXCL12 indicate areas of disseminated disease and are therefore worth exploring as possible targets for intraoperative imaging.

Several chemokine-receptor pairs are expressed in tumors cells, as well as by the surrounding stroma.103,104 The CXCR4-CXCL12 axis supports chemotaxis of tumor cells to the metastatic site and subsequent hatching and proliferation. In ovarian cancer, the CXCR4-CXCL12 pathway seems to have a more prominent role than other chemokine-receptor pairs and portends a worse prognosis.105108 CXCR4 and CXCL12 staining was demonstrated in 59% and 91% of ovarian tumor cells, respectively, whereas no expression was seen in healthy tissue.109 On binding to CXCR4, CXCL12 is internalized and the receptor is recycled to the cell surface, where it is ready for further action within 15 minutes.107 Overexpression of CXCL12 was found in ascites, inducing the migration of tumor cells and contributing to peritoneal dissemination.109,110

Visualization of CXCR4 on the cell membrane was shown in vitro by using a CXCR4-directed fluorophore.111 For in vivo targeting, both the receptor and the chemokine can be targeted by a monoclonal antibody, which in itself can be conjugated to an imaging agent. In a mouse model, a CXCR4-radiolabeled antibody was successfully used for tumor visualization in SPECT-CT.112 For PET, the CXCR4 antagonist ADM3100 conjugated to 99mTc proved useful in locating tumor xenografts in mice.113

Matrix Metalloproteinases

MMPs degrade several components of the extracellular matrix and are associated with tumor progression and metastasis. Several subtypes, among which are MMP1, MMP2, MMP7, and MMP9, are overexpressed in EOC.114119 CXCL12 is known to activate MMP2 and MMP9 in ovarian cancer, further contributing to the dissemination of tumor cells.120

A few preclinical studies have been conducted on NIRF imaging of MMP activity in cancer, using so-called “smart activatable probes” that are activated by MMPs.121123 Although these probes are not yet suitable for human use, they could play an important role in the future development of ovarian cancer targeting.

These results indicate that substantial progress is being made in visualization of MMP and CXCR4 activation and expression in EOC. These factors could possibly find their way to the clinic as targets for tumor-specific imaging in ovarian cancer.

Discussion

The greatest potential of intraoperative fluorescence imaging in EOC lies in improved detection of metastatic tumor deposits, leading to more complete cytoreduction. Additionally, intraoperative fluorescence imaging may be valuable in other gynecologic malignancies and in a broader range of oncologic diseases. As many tumor types express the biomarkers mentioned in this review, albeit to different degrees, these tumor-targeted contrast agents could potentially be applied in a broad spectrum of diseases. Overexpression of FR-α is seen in 40% of human cancers, including breast, lung, renal, and brain cancer37; however, there is great variation in expression patterns between tumor types.39 Hypoxia has been shown to occur in several types of solid tumors, including gynecologic cancers such as cervical124 and endometrial125 carcinoma, and is in the vast majority of cases correlated with worse survival rates. HIF-1α, VEGF, and EGFR are all overexpressed in endometrial cancer.125 Furthermore, treatment with cetuximab has been shown to inhibit tumor growth and peritoneal dissemination in a mouse model for endometrial cancer.125,126 These findings suggest that contrast agents targeting hypoxia-induced gene products such as VEGF and EGFR could also be of value in the surgical treatment of endometrial cancer with peritoneal dissemination. In cervical cancer, peritoneal dissemination is rare, limiting the application of tumor-targeted contrast agents for the purposes of cytoreductive surgery. However, these agents may be successful in identifying parametrial disease, especially in the laparoscopic setting, as MRI and CT perform poorly in staging of advanced disease, with sensitivity rates of 53% and 42%, respectively.127

Although the concept of intraoperative tumor-targeted imaging is promising, more research is needed to define the value of each independent target. In the development of possible tumor-targeted agents, specificity and sensitivity of the agent for its target are of the utmost importance. The variable expression of biomarkers, not only between tumor types but also within a solid tumor, may call for a combination of tumor-targeted agents to obtain high sensitivity rates. In addition, fluorescent agents with varying wavelengths could be used to localize various active biomarkers using multispectral fluorescence imaging. Conversely, a panel of agents with the same wavelength may provide the most comprehensive image of tumor activity.

Apart from evaluating the value of each independent tumor-targeted contrast agent, attention needs to be focused on development of commercially available user-friendly and safe intraoperative camera systems and laparoscopic fluorescence cameras.

Conclusion

Treatment of advanced EOC primarily consists of surgical cytoreduction followed by chemotherapy. As the extent of cytoreduction greatly influences prognosis, detailed detection and resection of metastatic and residual tumor tissue during surgery are a necessary prerequisite to improve patient survival. New techniques such as intraoperative optical imaging using highly specific tumor-targeted fluorescent optical agents combined with sophisticated camera systems can help improve cytoreductive efforts. Preclinical and translational studies will determine the efficacy of such an approach in the near future.

Acknowledgment

Financial disclosure of authors and reviewers: None reported.

References

  • 1.Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71–96. doi: 10.3322/CA.2007.0010. doi:10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 2.Winter WE, III, Maxwell GL, Tian C, et al. Tumor residual after surgical cytoreduction in prediction of clinical outcome in stage IV epithelial ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2008;26:83–9. doi: 10.1200/JCO.2007.13.1953. doi:10.1200/JCO.2007.13.1953. [DOI] [PubMed] [Google Scholar]
  • 3.Zivanovic O, Aldini A, Carlson JW, et al. Advanced cytoreductive surgery: American perspective. Gynecol Oncol. 2009;114(2 Suppl):S3–9. doi: 10.1016/j.ygyno.2008.11.033. doi:10.1016/j.ygyno.2008.11.033. [DOI] [PubMed] [Google Scholar]
  • 4.Aletti GD, Dowdy SC, Gostout BS, et al. Aggressive surgical effort and improved survival in advanced-stage ovarian cancer. Obstet Gynecol. 2006;107:77–85. doi: 10.1097/01.AOG.0000192407.04428.bb. [DOI] [PubMed] [Google Scholar]
  • 5.Vergote I, van Gorp T, Amant F, et al. Timing of debulking surgery in advanced ovarian cancer. Int J Gynecol Cancer. 2008;18(Suppl 1):11–9. doi: 10.1111/j.1525-1438.2007.01098.x. doi:10.1111/j.1525-1438.2007.01098.x. [DOI] [PubMed] [Google Scholar]
  • 6.Wimberger P, Wehling M, Lehmann N, et al. Influence of residual tumor on outcome in ovarian cancer patients with FIGO stage IV disease: an exploratory analysis of the AGO-OVAR (Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group) Ann Surg Oncol. 2010;17:1642–8. doi: 10.1245/s10434-010-0964-9. doi:10.1245/s10434-010-0964-9. [DOI] [PubMed] [Google Scholar]
  • 7.Bristow RE, Tomacruz RS, Armstrong DK, et al. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol. 2002;20:1248–59. doi: 10.1200/JCO.2002.20.5.1248. doi:10.1200/JCO.20.5.1248. [DOI] [PubMed] [Google Scholar]
  • 8.Tian WJ, Jiang R, Cheng X, et al. Surgery in recurrent epithelial ovarian cancer: benefits on survival for patients with residual disease of 0.1-1 cm after secondary cytoreduction. J Surg Oncol. 2010;101:244–50. doi: 10.1002/jso.21491. [DOI] [PubMed] [Google Scholar]
  • 9.Chi DS, Eisenhauer EL, Zivanovic O, et al. Improved progression-free and overall survival in advanced ovarian cancer as a result of a change in surgical paradigm. Gynecol Oncol. 2009;114:26–31. doi: 10.1016/j.ygyno.2009.03.018. doi:10.1016/j.ygyno.2009.03.018. [DOI] [PubMed] [Google Scholar]
  • 10.Los G, Mutsaers PH, Lenglet WJ, et al. Platinum distribution in intraperitoneal tumors after intraperitoneal cisplatin treatment. Cancer Chemother Pharmacol. 1990;25:389–94. doi: 10.1007/BF00686048. doi:10.1007/BF00686048. [DOI] [PubMed] [Google Scholar]
  • 11.Los G, Mutsaers PH, van der Vijgh WJ, et al. Direct diffusion of cis-diamminedichloroplatinum(II) in intraperitoneal rat tumors after intraperitoneal chemotherapy: a comparison with systemic chemotherapy. Cancer Res. 1989;49:3380–4. [PubMed] [Google Scholar]
  • 12.Howell SB. Pharmacologic principles of intraperitoneal chemotherapy for the treatment of ovarian cancer. Int J Gynecol Cancer. 2008;18(Suppl 1):20–5. doi: 10.1111/j.1525-1438.2007.01099.x. doi:10.1111/j.1525-1438.2007.01099.x. [DOI] [PubMed] [Google Scholar]
  • 13.de Bree E, Rosing H, Michalakis J, et al. Intraperitoneal chemotherapy with taxanes for ovarian cancer with peritoneal dissemination. Eur J Surg Oncol. 2006;32:666–70. doi: 10.1016/j.ejso.2006.03.008. doi:10.1016/j.ejso.2006.03.008. [DOI] [PubMed] [Google Scholar]
  • 14.Forstner R. Radiological staging of ovarian cancer: imaging findings and contribution of CT and MRI. Eur Radiol. 2007;17:3223–35. doi: 10.1007/s00330-007-0736-5. doi:10.1007/s00330-007-0736-5. [DOI] [PubMed] [Google Scholar]
  • 15.Musto A, Rampin L, Nanni C, et al. Present and future of PET and PET/CT in gynaecologic malignancies. Eur J Radiol. 2010 Jan 28; doi: 10.1016/j.ejrad.2009.12.035. [Epub ahead of print], doi:10.1016/j.ejrad.2009.12.035. [DOI] [PubMed] [Google Scholar]
  • 16.Nam EJ, Yun MJ, Oh YT, et al. Diagnosis and staging of primary ovarian cancer: correlation between PET/CT, Doppler US, and CT or MRI. Gynecol Oncol. 2010;116:389–94. doi: 10.1016/j.ygyno.2009.10.059. doi:10.1016/j.ygyno.2009.10.059. [DOI] [PubMed] [Google Scholar]
  • 17.Tempany CM, Zou KH, Silverman SG, et al. Staging of advanced ovarian cancer: comparison of imaging modalities—report from the Radiological Diagnostic Oncology Group. Radiology. 2000;215:761–7. doi: 10.1148/radiology.215.3.r00jn25761. [DOI] [PubMed] [Google Scholar]
  • 18.Low RN, Sebrechts CP, Barone RM, et al. Diffusion-weighted MRI of peritoneal tumors: comparison with conventional MRI and surgical and histopathologic findings—a feasibility study. AJR Am J Roentgenol. 2009;193:461–70. doi: 10.2214/AJR.08.1753. doi:10.2214/AJR.08.1753. [DOI] [PubMed] [Google Scholar]
  • 19.Kyriazi S, Collins DJ, Morgan VA, et al. Diffusion-weighted imaging of peritoneal disease for noninvasive staging of advanced ovarian cancer. Radiographics. 2010;30:1269–85. doi: 10.1148/rg.305105073. doi:10.1148/rg.305105073. [DOI] [PubMed] [Google Scholar]
  • 20.Funicelli L, Travaini LL, Landoni F, et al. Peritoneal carcinomatosis from ovarian cancer: the role of CT and [(18)F]FDG-PET/ CT. Abdom Imaging. 2009 Sep 26; doi: 10.1007/s00261-009-9578-8. [Epub ahead of print], doi:10.1007/s00261-009-9578-8. [DOI] [PubMed] [Google Scholar]
  • 21.Cho JE, Liu C, Gossner G, et al. Laparoscopy and gynecologic oncology. Clin Obstet Gynecol. 2009;52:313–26. doi: 10.1097/GRF.0b013e3181b088d2. doi:10.1097/GRF.0b013e3181b088d2. [DOI] [PubMed] [Google Scholar]
  • 22.De Iaco P, Musto A, Orazi L, et al. FDG-PET/CT in advanced ovarian cancer staging: value and pitfalls in detecting lesions in different abdominal and pelvic quadrants compared with laparoscopy. Eur J Radiol. 2010 Aug 3; doi: 10.1016/j.ejrad.2010.07.013. [Epub ahead of print], doi:10.1016/j.ejrad.2010.07.013. [DOI] [PubMed] [Google Scholar]
  • 23.Benedet JL, Bender H, Jones H, III, et al. FIGO staging classifications and clinical practice guidelines in the management of gynecologic cancers. FIGO Committee on Gynecologic Oncology. Int J Gynaecol Obstet. 2000;70:209–62. doi:10.1016/S0020-7292(00)90001-8. [PubMed] [Google Scholar]
  • 24.Aebi S, Castiglione M. Newly and relapsed epithelial ovarian carcinoma: ESMO clinical recommendations for diagnosis, treatment and follow-up. Ann Oncol. 2009;20(Suppl 4):21–3. doi: 10.1093/annonc/mdp117. [DOI] [PubMed] [Google Scholar]
  • 25.Hama Y, Urano Y, Koyama Y, et al. In vivo spectral fluorescence imaging of submillimeter peritoneal cancer implants using a lectin-targeted optical agent. Neoplasia. 2006;8:607–12. doi: 10.1593/neo.06268. doi:10.1593/neo.06268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Troy T, Jekic-McMullen D, Sambucetti L, et al. Quantitative comparison of the sensitivity of detection of fluorescent and bioluminescent reporters in animal models. Mol Imaging. 2004;3:9–23. doi: 10.1162/15353500200403196. doi:10.1162/153535004773861688. [DOI] [PubMed] [Google Scholar]
  • 27.Sevick-Muraca EM, Lopez G, Reynolds JS, et al. Fluorescence and absorption contrast mechanisms for biomedical optical imaging using frequency-domain techniques. Photochem Photobiol. 1997;66:55–64. doi: 10.1111/j.1751-1097.1997.tb03138.x. doi:10.1111/j.1751-1097.1997.tb03138.x. [DOI] [PubMed] [Google Scholar]
  • 28.Mordon S, Devoisselle JM, Soulie-Begu S, et al. Indocyanine green: physicochemical factors affecting its fluorescence in vivo. Microvasc Res. 1998;55:146–52. doi: 10.1006/mvre.1998.2068. doi:10.1006/mvre.1998.2068. [DOI] [PubMed] [Google Scholar]
  • 29.Alford R, Simpson HM, Duberman J, et al. Toxicity of organic fluorophores used in molecular imaging: literature review. Mol Imaging. 2009;8:341–54. [PubMed] [Google Scholar]
  • 30.Butner RW, McPherson AR. Adverse reactions in intravenous fluorescein angiography. Ann Ophthalmol. 1983;15:1084–6. [PubMed] [Google Scholar]
  • 31.Kovar JL, Simpson MA, Schutz-Geschwender A, et al. A systematic approach to the development of fluorescent contrast agents for optical imaging of mouse cancer models. Anal Biochem. 2007;367:1–12. doi: 10.1016/j.ab.2007.04.011. doi:10.1016/j.ab.2007.04.011. [DOI] [PubMed] [Google Scholar]
  • 32.Themelis G, Yoo JS, Soh KS, et al. Real-time intraoperative fluorescence imaging system using light-absorption correction. J Biomed Opt. 2009;14:064012. doi: 10.1117/1.3259362. doi:10.1117/1.3259362. [DOI] [PubMed] [Google Scholar]
  • 33.Troyan SL, Kianzad V, Gibbs-Strauss SL, et al. The FLARE intraoperative near-infrared fluorescence imaging system: a first-in-human clinical trial in breast cancer sentinel lymph node mapping. Ann Surg Oncol. 2009;16:2943–52. doi: 10.1245/s10434-009-0594-2. doi:10.1245/s10434-009-0594-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fujiwara M, Mizukami T, Suzuki A, et al. Sentinel lymph node detection in skin cancer patients using real-time fluorescence navigation with indocyanine green: preliminary experience. J Plast Reconstr Aesthet Surg. 2009;62:e373–8. doi: 10.1016/j.bjps.2007.12.074. doi:10.1016/j.bjps.2007.12.074. [DOI] [PubMed] [Google Scholar]
  • 35.Ishizawa T, Bandai Y, Ijichi M, et al. Fluorescent cholangiography illuminating the biliary tree during laparoscopic cholecystectomy. Br J Surg. 2010;97:1369–77. doi: 10.1002/bjs.7125. doi:10.1002/bjs.7125. [DOI] [PubMed] [Google Scholar]
  • 36.Parungo CP, Ohnishi S, Kim SW, et al. Intraoperative identification of esophageal sentinel lymph nodes with near-infrared fluorescence imaging. J Thorac Cardiovasc Surg. 2005;129:844–50. doi: 10.1016/j.jtcvs.2004.08.001. doi:10.1016/j.jtcvs.2004.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Low PS, Kularatne SA. Folate-targeted therapeutic and imaging agents for cancer. Curr Opin Chem Biol. 2009;13:256–62. doi: 10.1016/j.cbpa.2009.03.022. doi:10.1016/j.cbpa.2009.03.022. [DOI] [PubMed] [Google Scholar]
  • 38.Yamaguchi T, Hirota K, Nagahama K, et al. Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity. 2007;27:145–59. doi: 10.1016/j.immuni.2007.04.017. doi:10.1016/j.immuni.2007.04.017. [DOI] [PubMed] [Google Scholar]
  • 39.Parker N, Turk MJ, Westrick E, et al. Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem. 2005;338:284–93. doi: 10.1016/j.ab.2004.12.026. doi:10.1016/j.ab.2004.12.026. [DOI] [PubMed] [Google Scholar]
  • 40.Kalli KR, Oberg AL, Keeney GL, et al. Folate receptor alpha as a tumor target in epithelial ovarian cancer. Gynecol Oncol. 2008;108:619–26. doi: 10.1016/j.ygyno.2007.11.020. doi:10.1016/j.ygyno.2007.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Markert S, Lassmann S, Gabriel B, et al. Alpha-folate receptor expression in epithelial ovarian carcinoma and non-neoplastic ovarian tissue. Anticancer Res. 2008;28:3567–72. [PubMed] [Google Scholar]
  • 42.Low PS, Antony AC. Folate receptor-targeted drugs for cancer and inflammatory diseases. Adv Drug Deliv Rev. 2004;56:1055–8. doi: 10.1016/j.addr.2004.02.003. doi:10.1016/j.addr.2004.02.003. [DOI] [PubMed] [Google Scholar]
  • 43.Leamon CP, Low PS. Delivery of macromolecules into living cells: a method that exploits folate receptor endocytosis. Proc Natl Acad Sci U S A. 1991;88:5572–6. doi: 10.1073/pnas.88.13.5572. doi:10.1073/pnas.88.13.5572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Low PS, Henne WA, Doorneweerd DD. Discovery and development of folic-acid-based receptor targeting for imaging and therapy of cancer and inflammatory diseases. Acc Chem Res. 2008;41:120–9. doi: 10.1021/ar7000815. doi:10.1021/ar7000815. [DOI] [PubMed] [Google Scholar]
  • 45.Jackman AL, Theti DS, Gibbs DD. Antifolates targeted specifically to the folate receptor. Adv Drug Deliv Rev. 2004;56:1111–25. doi: 10.1016/j.addr.2004.01.003. doi:10.1016/j.addr.2004.01.003. [DOI] [PubMed] [Google Scholar]
  • 46.Reddy JA, Dorton R, Westrick E, et al. Preclinical evaluation of EC145, a folate-vinca alkaloid conjugate. Cancer Res. 2007;67:4434–42. doi: 10.1158/0008-5472.CAN-07-0033. doi:10.1158/0008-5472.CAN-07-0033. [DOI] [PubMed] [Google Scholar]
  • 47.Lu Y, Low PS. Folate targeting of haptens to cancer cell surfaces mediates immunotherapy of syngeneic murine tumors. Cancer Immunol Immunother. 2002;51:153–62. doi: 10.1007/s00262-002-0266-6. doi:10.1007/s00262-002-0266-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lu Y, Sega E, Low PS. Folate receptor-targeted immunotherapy: induction of humoral and cellular immunity against haptendecorated cancer cells. Int J Cancer. 2005;116:710–9. doi: 10.1002/ijc.21126. doi:10.1002/ijc.21126. [DOI] [PubMed] [Google Scholar]
  • 49.Mathias CJ, Wang S, Waters DJ, et al. Indium-111-DTPA-folate as a potential folate-receptor-targeted radiopharmaceutical. J Nucl Med. 1998;39:1579–85. [PubMed] [Google Scholar]
  • 50.Siegel BA, Dehdashti F, Mutch DG, et al. Evaluation of 111In-DTPA-folate as a receptor-targeted diagnostic agent for ovarian cancer: initial clinical results. J Nucl Med. 2003;44:700–7. [PubMed] [Google Scholar]
  • 51.Leamon CP, Parker MA, Vlahov IR, et al. Synthesis and biological evaluation of EC20: a new folate-derived, (99m)Tc-based radio-pharmaceutical. Bioconjug Chem. 2002;13:1200–10. doi: 10.1021/bc0200430. doi:10.1021/bc0200430. [DOI] [PubMed] [Google Scholar]
  • 52.Fisher RE, Siegel BA, Edell SL, et al. Exploratory study of 99mTc-EC20 imaging for identifying patients with folate receptor-positive solid tumors. J Nucl Med. 2008;49:899–906. doi: 10.2967/jnumed.107.049478. doi:10.2967/jnumed.107.049478. [DOI] [PubMed] [Google Scholar]
  • 53.Ebel W, Routhier EL, Foley B, et al. Preclinical evaluation of MORAb-003, a humanized monoclonal antibody antagonizing folate receptor-alpha. Cancer Immun. 2007;7:6. [PMC free article] [PubMed] [Google Scholar]
  • 54.Spannuth WA, Sood AK, Coleman RL. Farletuzumab in epithelial ovarian carcinoma. Expert Opin Biol Ther. 2010;10:431–7. doi: 10.1517/14712591003592069. doi:10.1517/14712591003592069. [DOI] [PubMed] [Google Scholar]
  • 55.Armstrong DK, Bicher A, Coleman RL, et al. Exploratory phase II efficacy study of MORAb-003, a monoclonal antibody against folate receptor alpha, in platinum-sensitive ovarian cancer in first relapse [abstract[ J Clin Oncol. 2008;26:5500. doi:10.1200/JCO.2008.18.6064. [Google Scholar]
  • 56.Smith-Jones PM, Pandit-Taskar N, Cao W, et al. Preclinical radioimmunotargeting of folate receptor alpha using the monoclonal antibody conjugate DOTA-MORAb-003. Nucl Med Biol. 2008;35:343–51. doi: 10.1016/j.nucmedbio.2007.12.008. doi:10.1016/j.nucmedbio.2007.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kennedy MD, Jallad KN, Thompson DH, et al. Optical imaging of metastatic tumors using a folate-targeted fluorescent probe. J Biomed Opt. 2003;8:636–41. doi: 10.1117/1.1609453. doi:10.1117/1.1609453. [DOI] [PubMed] [Google Scholar]
  • 58.Birner P, Schindl M, Obermair A, et al. Expression of hypoxia-inducible factor 1alpha in epithelial ovarian tumors: its impact on prognosis and on response to chemotherapy. Clin Cancer Res. 2001;7:1661–8. [PubMed] [Google Scholar]
  • 59.Barnas C, Martel-Planche G, Furukawa Y, et al. Inactivation of the p53 protein in cell lines derived from human esophageal cancers. Int J Cancer. 1997;71:79–87. doi: 10.1002/(sici)1097-0215(19970328)71:1<79::aid-ijc14>3.0.co;2-4. doi:10.1002/(SICI)1097-0215(19970328)71:1<,79::AID-IJC14>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 60.Hockel M, Schlenger K, Hockel S, et al. Hypoxic cervical cancers with low apoptotic index are highly aggressive. Cancer Res. 1999;59:4525–8. [PubMed] [Google Scholar]
  • 61.Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist. 2004;9(Suppl 5):4–9. doi: 10.1634/theoncologist.9-90005-4. doi:10.1634/theoncologist.9-90005-4. [DOI] [PubMed] [Google Scholar]
  • 62.Harris AL. Hypoxia—a key regulatory factor in tumour growth. Nat Rev Cancer. 2002;2:38–47. doi: 10.1038/nrc704. doi:10.1038/nrc704. [DOI] [PubMed] [Google Scholar]
  • 63.Maynard MA, Ohh M. The role of hypoxia-inducible factors in cancer. Cell Mol Life Sci. 2007;64:2170–80. doi: 10.1007/s00018-007-7082-2. doi:10.1007/s00018-007-7082-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bacon AL, Harris AL. Hypoxia-inducible factors and hypoxic cell death in tumour physiology. Ann Med. 2004;36:530–9. doi: 10.1080/07853890410018231. doi:10.1080/07853890410018231. [DOI] [PubMed] [Google Scholar]
  • 65.Greijer AE, van der Groep P, Kemming D, et al. Up-regulation of gene expression by hypoxia is mediated predominantly by hypoxia-inducible factor 1 (HIF-1) J Pathol. 2005;206:291–304. doi: 10.1002/path.1778. doi:10.1002/path.1778. [DOI] [PubMed] [Google Scholar]
  • 66.Liu LZ, Hu XW, Xia C, et al. Reactive oxygen species regulate epidermal growth factor-induced vascular endothelial growth factor and hypoxia-inducible factor-1alpha expression through activation of AKT and P70S6K1 in human ovarian cancer cells. Free Radic Biol Med. 2006;41:1521–33. doi: 10.1016/j.freeradbiomed.2006.08.003. doi:10.1016/j.freeradbiomed.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 67.Kumaran GC, Jayson GC, Clamp AR. Antiangiogenic drugs in ovarian cancer. Br J Cancer. 2009;100:1–7. doi: 10.1038/sj.bjc.6604767. doi:10.1038/sj.bjc.6604767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhong H, De Marzo AM, Laughner E, et al. Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 1999;59:5830–5. [PubMed] [Google Scholar]
  • 69.Shimogai R, Kigawa J, Itamochi H, et al. Expression of hypoxia-inducible factor 1alpha gene affects the outcome in patients with ovarian cancer. Int J Gynecol Cancer. 2008;18:499–505. doi: 10.1111/j.1525-1438.2007.01055.x. doi:10.1111/j.1525-1438.2007.01055.x. [DOI] [PubMed] [Google Scholar]
  • 70.Lee S, Garner EI, Welch WR, et al. Over-expression of hypoxia-inducible factor 1 alpha in ovarian clear cell carcinoma. Gynecol Oncol. 2007;106:311–7. doi: 10.1016/j.ygyno.2007.03.041. doi:10.1016/j.ygyno.2007.03.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kim KS, Sengupta S, Berk M, et al. Hypoxia enhances lysophosphatidic acid responsiveness in ovarian cancer cells and lysophosphatidic acid induces ovarian tumor metastasis in vivo. Cancer Res. 2006;66:7983–90. doi: 10.1158/0008-5472.CAN-05-4381. doi:10.1158/0008-5472.CAN-05-4381. [DOI] [PubMed] [Google Scholar]
  • 72.Mor G, Visintin I, Lai Y, et al. Serum protein markers for early detection of ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:7677–82. doi: 10.1073/pnas.0502178102. doi:10.1073/pnas.0502178102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Voldborg BR, Damstrup L, Spang-Thomsen M, et al. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann Oncol. 1997;8:1197–206. doi: 10.1023/a:1008209720526. doi:10.1023/A:1008209720526. [DOI] [PubMed] [Google Scholar]
  • 74.Hefler LA, Mustea A, Konsgen D, et al. Vascular endothelial growth factor gene polymorphisms are associated with prognosis in ovarian cancer. Clin Cancer Res. 2007;13:898–901. doi: 10.1158/1078-0432.CCR-06-1008. doi:10.1158/1078-0432.CCR-06-1008. [DOI] [PubMed] [Google Scholar]
  • 75.Trinh XB, Tjalma WA, Vermeulen PB, et al. The VEGF pathway and the AKT/mTOR/p70S6K1 signalling pathway in human epithelial ovarian cancer. Br J Cancer. 2009;100:971–8. doi: 10.1038/sj.bjc.6604921. doi:10.1038/sj.bjc.6604921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Collinson F, Jayson G. New therapeutic agents in ovarian cancer. Curr Opin Obstet Gynecol. 2009;21:44–53. doi: 10.1097/GCO.0b013e32831ffe71. doi:10.1097/GCO.0b013e32831ffe71. [DOI] [PubMed] [Google Scholar]
  • 77.Sánchez-Muñoz A, Mendolia C, Rodriguez CA, et al. Bevacizumab plus low-dose metronomic oral cyclophosphamide in heavily pretreated recurrent ovarian cancer [abstract] J Clin Oncol. 2010;28:e15507. doi: 10.1159/000320602. Suppl:abstr. [DOI] [PubMed] [Google Scholar]
  • 78.Burger RA, Sill MW, Monk BJ, et al. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2007;25:5165–71. doi: 10.1200/JCO.2007.11.5345. doi:10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
  • 79.Tillmanns TD, Lowe MP, Schwartzberg LS, et al. A phase II study of bevacizumab with nab-paclitaxel in patients with recurrent, platinum-resistant primary epithelial ovarian or primary peritoneal carcinoma [abstract] J Clin Oncol. 2010;28(Suppl 15):5009. doi: 10.1016/j.ygyno.2012.08.039. abstr. [DOI] [PubMed] [Google Scholar]
  • 80.Burger RA, Liang SX, Boente M, et al. Phase III trial of bevacizumab (BEV) in the primary treatment of advanced epithelial ovarian cancer (EOC), primary peritoneal cancer (PPC), or fallopian tube cancer (FTC): a Gynecologic Oncology Group study [abstract] J Clin Oncol. 2010;28(Suppl 18) abstr LBA1. [Google Scholar]
  • 81.Nagengast WB, de Vries EG, Hospers GA, et al. In vivo VEGF imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft. J Nucl Med. 2007;48:1313–9. doi: 10.2967/jnumed.107.041301. doi:10.2967/jnumed.107.041301. [DOI] [PubMed] [Google Scholar]
  • 82.Nagengast WB, Lub-de Hooge MN, van Straten EME, et al. VEGF-SPECT with 111In-bevacizumab in stage III/IV melanoma patients [thesis] Rijksuniversiteit Groningen; Groningen (the Netherlands): 2009. [Google Scholar]
  • 83.Backer MV, Levashova Z, Patel V, et al. Molecular imaging of VEGF receptors in angiogenic vasculature with single-chain VEGF-based probes. Nat Med. 2007;13:504–9. doi: 10.1038/nm1522. doi:10.1038/nm1522. [DOI] [PubMed] [Google Scholar]
  • 84.Abu-Jawdeh GM, Faix JD, Niloff J, et al. Strong expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in ovarian borderline and malignant neoplasms. Lab Invest. 1996;74:1105–15. [PubMed] [Google Scholar]
  • 85.Bates DO, Harper SJ. Regulation of vascular permeability by vascular endothelial growth factors. Vascul Pharmacol. 2002;39:225–37. doi: 10.1016/s1537-1891(03)00011-9. doi:10.1016/S1537-1891(03)00011-9. [DOI] [PubMed] [Google Scholar]
  • 86.Kobold S, Hegewisch-Becker S, Oechsle K, et al. Intraperitoneal VEGF inhibition using bevacizumab: a potential approach for the symptomatic treatment of malignant ascites? Oncologist. 2009;14:1242–51. doi: 10.1634/theoncologist.2009-0109. doi:10.1634/theoncologist.2009-0109. [DOI] [PubMed] [Google Scholar]
  • 87.Pasquet M, Golzio M, Mery E, et al. Hospicells (ascites-derived stromal cells) promote tumorigenicity and angiogenesis. Int J Cancer. 2010;126:2090–101. doi: 10.1002/ijc.24886. [DOI] [PubMed] [Google Scholar]
  • 88.Giuntoli RL, Webb TJ, Zoso A, et al. Ovarian cancer-associated ascites demonstrates altered immune environment: implications for antitumor immunity. Anticancer Res. 2009;29:2875–84. [PubMed] [Google Scholar]
  • 89.Brustmann H. Epidermal growth factor receptor expression in serous ovarian carcinoma: an immunohistochemical study with galectin-3 and cyclin D1 and outcome. Int J Gynecol Pathol. 2008;27:380–9. doi: 10.1097/PGP.0b013e31815d060d. doi:10.1097/PGP.0b013e31815d060d. [DOI] [PubMed] [Google Scholar]
  • 90.Zeineldin R, Muller CY, Stack MS, et al. Targeting the EGF receptor for ovarian cancer therapy. J Oncol. 2010 doi: 10.1155/2010/414676. 414676. Epub 2009 Dec 28, doi:10.1155/2010/414676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Lafky JM, Wilken JA, Baron AT, et al. Clinical implications of the ErbB/epidermal growth factor (EGF) receptor family and its ligands in ovarian cancer. Biochim Biophys Acta. 2008;1785:232–65. doi: 10.1016/j.bbcan.2008.01.001. [DOI] [PubMed] [Google Scholar]
  • 92.Skirnisdottir I, Sorbe B, Seidal T. The growth factor receptors HER-2/neu and EGFR, their relationship, and their effects on the prognosis in early stage (FIGO I-II) epithelial ovarian carcinoma. Int J Gynecol Cancer. 2001;11:119–29. doi: 10.1046/j.1525-1438.2001.011002119.x. doi:10.1046/j.15251438.2001.011002119.x. [DOI] [PubMed] [Google Scholar]
  • 93.de Graeff P, Crijns AP, de Jong S, et al. Modest effect of p53, EGFR and HER-2/neu on prognosis in epithelial ovarian cancer: a meta-analysis. Br J Cancer. 2009;101:149–59. doi: 10.1038/sj.bjc.6605112. doi:10.1038/sj.bjc.6605112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Xia W, Wei Y, Du Y, et al. Nuclear expression of epidermal growth factor receptor is a novel prognostic value in patients with ovarian cancer. Mol Carcinog. 2009;48:610–7. doi: 10.1002/mc.20504. doi:10.1002/mc.20504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Salomon DS, Brandt R, Ciardiello F, et al. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 1995;19:183–232. doi: 10.1016/1040-8428(94)00144-i. doi:10.1016/10408428(94)00144-I. [DOI] [PubMed] [Google Scholar]
  • 96.Kim ES, Khuri FR, Herbst RS. Epidermal growth factor receptor biology (IMC-C225) Curr Opin Oncol. 2001;13:506–13. doi: 10.1097/00001622-200111000-00014. doi:10.1097/00001622-200111000-00014. [DOI] [PubMed] [Google Scholar]
  • 97.Ogawa M, Regino CA, Seidel J, et al. Dual-modality molecular imaging using antibodies labeled with activatable fluorescence and a radionuclide for specific and quantitative targeted cancer detection. Bioconjug Chem. 2009;20:2177–84. doi: 10.1021/bc900362k. doi:10.1021/bc900362k. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ogawa M, Kosaka N, Choyke PL, et al. In vivo molecular imaging of cancer with a quenching near-infrared fluorescent probe using conjugates of monoclonal antibodies and indocyanine green. Cancer Res. 2009;69:1268–72. doi: 10.1158/0008-5472.CAN-08-3116. doi:10.1158/0008-5472.CAN-08-3116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Milenic DE, Wong KJ, Baidoo KE, et al. Cetuximab: preclinical evaluation of a monoclonal antibody targeting EGFR for radio-immunodiagnostic and radioimmunotherapeutic applications. Cancer Biother Radiopharm. 2008;23:619–31. doi: 10.1089/cbr.2008.0493. doi:10.1089/cbr.2008.0493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nayak TK, Regino CA, Wong KJ, et al. PET imaging of HER1-expressing xenografts in mice with (86)Y-CHX-A”-DTPA-cetuximab. Eur J Nucl Med Mol Imaging. 2010;37:1368–76. doi: 10.1007/s00259-009-1370-z. doi:10.1007/s00259-009-1370-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Harding J, Burtness B. Cetuximab: an epidermal growth factor receptor chimeric human-murine monoclonal antibody. Drugs Today (Barc) 2005;41:107–27. doi: 10.1358/dot.2005.41.2.882662. doi:10.1358/dot.2005.41.2.882662. [DOI] [PubMed] [Google Scholar]
  • 102.Gort EH, Groot AJ, van der Wall E, et al. Hypoxic regulation of metastasis via hypoxia-inducible factors. Curr Mol Med. 2008;8:60–7. doi: 10.2174/156652408783565568. doi:10.2174/156652408783565568. [DOI] [PubMed] [Google Scholar]
  • 103.Balkwill F. The significance of cancer cell expression of the chemokine receptor CXCR4. Semin Cancer Biol. 2004;14:171–9. doi: 10.1016/j.semcancer.2003.10.003. doi:10.1016/j.semcancer.2003.10.003. [DOI] [PubMed] [Google Scholar]
  • 104.Arya M, Ahmed H, Silhi N, et al. Clinical importance and therapeutic implications of the pivotal CXCL12-CXCR4 (chemokine ligand-receptor) interaction in cancer cell migration. Tumour Biol. 2007;28:123–31. doi: 10.1159/000102979. doi:10.1159/000102979. [DOI] [PubMed] [Google Scholar]
  • 105.Barbieri F, Bajetto A, Florio T. Role of chemokine network in the development and progression of ovarian cancer: a potential novel pharmacological target. J Oncol. 2010 doi: 10.1155/2010/426956. 426956. Epub 2009 Dec 14, doi:10.1155/2010/426956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kajiyama H, Shibata K, Terauchi M, et al. Involvement of SDF-1alpha/CXCR4 axis in the enhanced peritoneal metastasis of epithelial ovarian carcinoma. Int J Cancer. 2008;122:91–9. doi: 10.1002/ijc.23083. doi:10.1002/ijc.23083. [DOI] [PubMed] [Google Scholar]
  • 107.Scotton CJ, Wilson JL, Milliken D, et al. Epithelial cancer cell migration: a role for chemokine receptors? Cancer Res. 2001;61:4961–5. [PubMed] [Google Scholar]
  • 108.Scotton CJ, Wilson JL, Scott K, et al. Multiple actions of the chemokine CXCL12 on epithelial tumor cells in human ovarian cancer. Cancer Res. 2002;62:5930–8. [PubMed] [Google Scholar]
  • 109.Jiang YP, Wu XH, Shi B, et al. Expression of chemokine CXCL12 and its receptor CXCR4 in human epithelial ovarian cancer: an independent prognostic factor for tumor progression. Gynecol Oncol. 2006;103:226–33. doi: 10.1016/j.ygyno.2006.02.036. doi:10.1016/j.ygyno.2006.02.036. [DOI] [PubMed] [Google Scholar]
  • 110.Jiang YP, Wu XH, Xing HY, et al. Role of CXCL12 in metastasis of human ovarian cancer. Chin Med J (Engl) 2007;120:1251–5. [PubMed] [Google Scholar]
  • 111.Nomura W, Tanabe Y, Tsutsumi H, et al. Fluorophore labeling enables imaging and evaluation of specific CXCR4-ligand interaction at the cell membrane for fluorescence-based screening. Bioconjug Chem. 2008;19:1917–20. doi: 10.1021/bc800216p. doi:10.1021/bc800216p. [DOI] [PubMed] [Google Scholar]
  • 112.Nimmagadda S, Pullambhatla M, Pomper MG. Immunoimaging of CXCR4 expression in brain tumor xenografts using SPECT/CT. J Nucl Med. 2009;50:1124–30. doi: 10.2967/jnumed.108.061325. doi:10.2967/jnumed.108.061325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhang J, Tian J, Li T, et al. 99mTc-AMD3100: a novel potential receptor-targeting radiopharmaceutical for tumor imaging. Chin Chem Lett. 2010;21:461–3. doi:10.1016/j.cclet.2009.12.018. [Google Scholar]
  • 114.Torng PL, Mao TL, Chan WY, et al. Prognostic significance of stromal metalloproteinase-2 in ovarian adenocarcinoma and its relation to carcinoma progression. Gynecol Oncol. 2004;92:559–67. doi: 10.1016/j.ygyno.2003.11.011. doi:10.1016/j.ygyno.2003.11.011. [DOI] [PubMed] [Google Scholar]
  • 115.Roomi MW, Monterrey JC, Kalinovsky T, et al. In vitro modulation of MMP-2 and MMP-9 in human cervical and ovarian cancer cell lines by cytokines, inducers and inhibitors. Oncol Rep. 2010;23:605–14. doi: 10.3892/or_00000675. [DOI] [PubMed] [Google Scholar]
  • 116.Zohny SF, Fayed ST. Clinical utility of circulating matrix metalloproteinase-7 (MMP-7), CC chemokine ligand 18 (CCL18) and CC chemokine ligand 11 (CCL11) as markers for diagnosis of epithelial ovarian cancer. Med Oncol. 2009 Nov 24; doi: 10.1007/s12032-009-9366-x. [Epub ahead of print], doi:10.1007/s12032-009-9366-x. [DOI] [PubMed] [Google Scholar]
  • 117.Moss NM, Barbolina MV, Liu Y, et al. Ovarian cancer cell detachment and multicellular aggregate formation are regulated by membrane type 1 matrix metalloproteinase: a potential role in I.p. metastatic dissemination. Cancer Res. 2009;69:7121–9. doi: 10.1158/0008-5472.CAN-08-4151. doi:10.1158/0008-5472.CAN-08-4151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kamat AA, Fletcher M, Gruman LM, et al. The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res. 2006;12:1707–14. doi: 10.1158/1078-0432.CCR-05-2338. doi:10.1158/1078-0432.CCR-05-2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Naylor MS, Stamp GW, Davies BD, et al. Expression and activity of MMPS and their regulators in ovarian cancer. Int J Cancer. 1994;58:50–6. doi: 10.1002/ijc.2910580110. doi:10.1002/ijc.2910580110. [DOI] [PubMed] [Google Scholar]
  • 120.Shen X, Wang S, Wang H, et al. The role of SDF-1/CXCR4 axis in ovarian cancer metastasis. J Huazhong Univ Sci Technolog Med Sci. 2009;29:363–7. doi: 10.1007/s11596-009-0320-0. doi:10.1007/s11596-009-0320-0. [DOI] [PubMed] [Google Scholar]
  • 121.von Burstin J, Eser S, Seidler B, et al. Highly sensitive detection of early-stage pancreatic cancer by multimodal near-infrared molecular imaging in living mice. Int J Cancer. 2008;123:2138–47. doi: 10.1002/ijc.23780. doi:10.1002/ijc.23780. [DOI] [PubMed] [Google Scholar]
  • 122.Olson ES, Aguilera TA, Jiang T, et al. In vivo characterization of activatable cell penetrating peptides for targeting protease activity in cancer. Integr Biol (Camb ) 2009;1:382–93. doi: 10.1039/b904890a. doi:10.1039/b904890a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Aguilera TA, Olson ES, Timmers MM, et al. Systemic in vivo distribution of activatable cell penetrating peptides is superior to that of cell penetrating peptides. Integr Biol (Camb) 2009;1:371–81. doi: 10.1039/b904878b. doi:10.1039/b904878b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hockel M, Schlenger K, Aral B, et al. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996;56:4509–15. [PubMed] [Google Scholar]
  • 125.Ozbudak IH, Karaveli S, Simsek T, et al. Neoangiogenesis and expression of hypoxia-inducible factor 1alpha, vascular endothelial growth factor, and glucose transporter-1 in endometrioid type endometrium adenocarcinomas. Gynecol Oncol. 2008;108:603–8. doi: 10.1016/j.ygyno.2007.11.028. doi:10.1016/j.ygyno.2007.11.028. [DOI] [PubMed] [Google Scholar]
  • 126.Takahashi K, Saga Y, Mizukami H, et al. Cetuximab inhibits growth, peritoneal dissemination, and lymph node and lung metastasis of endometrial cancer, and prolongs host survival. Int J Oncol. 2009;35:725–9. doi: 10.3892/ijo_00000385. [DOI] [PubMed] [Google Scholar]
  • 127.Hricak H, Gatsonis C, Chi DS, et al. Role of imaging in pretreatment evaluation of early invasive cervical cancer: results of the intergroup study American College of Radiology Imaging Network 6651-Gynecologic Oncology Group 183. J Clin Oncol. 2005;23:9329–37. doi: 10.1200/JCO.2005.02.0354. doi:10.1200/JCO.2005.02.0354. [DOI] [PubMed] [Google Scholar]

RESOURCES