Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 2/2019

Open Access 21.02.2019 | review

Resistance to chemotherapy and anti-angiogenic therapy in ovarian cancer

verfasst von: Verena Wieser, MD, PhD, Christian Marth, MD, PhD

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 2/2019

Summary

Ovarian cancer (OC) is the foremost lethal gynaecologic malignancy and among the top five deadliest cancers in women. Current treatment comprises a combination therapy of surgery, platinum-based chemotherapy and anti-vascular endothelial growth factor (VEGF) antibodies. However, patients typically experience a disease relapse within two years. Recurrent OC is incurable and resistance to platins and anti-VEGF treatment is a major determinant of prognosis. Understanding the molecular mechanisms that contribute to tumour metastasis and chemoresistance are essential to improve patient outcome and especially survival. In a current OC model, tumour metastasis and chemoresistance critically depend on the biology of cancer stem cells (CSCs). Recent studies also suggest that intratumour heterogeneity is the main cause of treatment failure due to chemoresistance. Furthermore, the proinflammatory tumour microenvironment seems to contribute to metastasis and chemoresistance. Despite an improved understanding of the complex interplay between classical mechanisms of drug inactivation or efflux, clonal selection and the tumour microenvironment, mechanisms of resistance in human OC are poorly understood. This review summarises current concepts in the treatment of OC, mechanisms of resistance to chemotherapy and angiogenic inhibitors and approaches to overcome drug resistance.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The frontline therapy of ovarian cancer (OC) consists of surgery and platinum-based chemotherapy (usually carboplatin area under the curve [AUC] 5–6 and paclitaxel 175 mg/m2 every 3 weeks for six cycles as demonstrated in the GOG-158 study) [1]. Patients with advanced disease (i. e. FIGO IIIb and higher) additionally receive anti-angiogenic therapy during chemotherapy (GOG-218: bevacizumab 15 mg/kg from second cycle carboplatin/paclitaxel; hazard ratio (HR) for progression or death 0.908 [95% confidence interval (CI), 0.795 to 1.040; P = 0.16]) and for maintenance (16 cycles after chemotherapy; HR 0.717 [95% CI, 0.625 to 0.824; P < 0.001]) [2]. Although response rates and complete responses after first-line treatment of advanced disease are >80% and 40–60%, respectively, most patients will relapse with a median progression-free survival (PFS) of 18 months [3]. In general, patients who respond to primary treatment and relapse within 6 months are considered “platinum-resistant”, and patients who relapse more than 6 months after completion of initial therapy are characterized as “platinum-sensitive” [4]. Interestingly, most of “platinum-sensitive” patients will respond to further platinum-based chemotherapy with response rates ranging from 30 to 90% [5]. In contrast, “platinum-resistant” patients typically have low response rates (15%) to subsequent chemotherapy and the outcome of these patients is poor with a median survival not exceeding 12 months [5]. Primary “platinum-refractory” OC patients (those that progress during treatment) are quite uncommon and usually seen with non-serous ovarian cancers such as clear cell or mucinous cancers rather than the more common high grade serous ovarian cancer (HGSOC). “Platinum-refractory” patients also exhibit a poor clinical outcome with comparable low median survival.
Tumour angiogenesis is an essential process of cancer growth and metastasis and influences the progression of ovarian cancer [6]. Vascular endothelial growth factor (VEGF) is an important promoter of the formation of new blood vessels that contribute to “feeding cancer” [6]. The expression of VEGF and its receptors in ovarian tumours directly correlate with poor prognosis, suggesting that angiogenesis, possibly mediated at least in part by VEGF, influences disease progression [7, 8]. To date, anti-angiogenic therapy has been identified as one of the most promising targeted therapies in OC. Bevacizumab, a recombinant, humanized, monoclonal VEGF antibody, has been implemented in the first-line treatment with a platinum-based chemotherapy but also in platinum-sensitive (OCEANS trial) and resistant (AURELIA trial) recurrent OC [2, 9, 10]. More specifically, in platinum-sensitive recurrent OC gemcitabine and carboplatin plus bevacizumab followed by bevacizumab until progression resulted in a better PFS compared with chemotherapy plus placebo (HR 0.484; 95% CI, 0.388 to 0.605; P < 0.0001) [9]. Combining bevacizumab with chemotherapy in platinum-resistant OC also improved PFS (HR 0.48; 95% CI, 0.38 to 0.60; P < 0.001) [10].

Influences on platinum sensitivity and mechanisms of platinum resistance

Different tumour-specific factors such as histological subtype, clonal selection, tumour mutations and microenvironment but also pharmacokinetic factors influence response and resistance to chemotherapy and anti-angiogenesis [11]. Platinum sensitivity and resistance is well known in patients with HGSOC. Endometrioid, clear cell, mucinous and low-grade serous OC are less common histotypes and differ from HGSOC in clinical course, tumour mutations, molecular aberrations and in response to chemotherapy. Mucinous, clear cell and low-grade serous cancers tend to be resistant to standard-of-care [12]. However, patients are still treated with platinum-based chemotherapy first-line due to the lack of proven alternatives. Since mucinous OC shares several common pathological and molecular features with gastrointestinal tumours, it has long been hypothesized that standard gastrointestinal treatments could be more effective for this histotype than the current standard-of-care. In all the phase I/II cohorts of platinum-refractory OC patients treated with some of these approaches (e. g., capecitabine, oxaliplatin, FOLFOX, gemcitabine + oxaliplatin), only a very small number of mucinous OC patients were included, meaning it was difficult to draw clear conclusions [13].
Germ-line mutations in BRCA1 and BRCA2 are well-known risk factors for developing HGSOC [14]. OC patients with germline (BReast CAncer) BRCA mutations exhibit a favourable outcome and higher responsiveness to platinum-based therapies [1517]. However, many sporadic HGSOCs exhibit phenotypic characteristics of germline BRCA mutated tumours. This so-called BRCAness can be defined as a defect in homologous recombination repair and can be, for example, caused by somatic mutation of BRCA1/2, epigenetic hypermethylation of the BRCA1 promoter, amplification of EMSY (also known as C11orf30) resulting in BRCA2 silencing, and loss of function mutations of the Fanconi anaemia complementation group family of genes [18]. Importantly, a BRCAness gene expression profile was shown to predict platinum responsiveness [19].
The cytotoxic effect of platins relies on single or double strand DNA breaks and may also cause mitochondrial damage and in turn cell death [19]. Platinum resistance may stem from reduced platinum uptake into the cell or increased efflux evoked by alterations of transport proteins. An increase in DNA repair by alterations of repair proteins such as nucleotide excision repair, mismatch repair, homologous recombination or base excision repair is also classically associated with platinum resistance. These various mechanisms may already exist at diagnosis or are acquired over time [12].
OC stem cells (OCSC) seem to play a potential role in OC recurrence following chemotherapy. Cancer stem cells typically exhibit a slow cycling rate which makes them inherently resistant to standard chemotherapy which, by definition, targets actively proliferating cells [20]. However, the underlying mechanisms that regulate the chemoresistance of OCSCs remain unclear [21]. Kryczek et al. and Silva et al. defined OSCSs via the presence of aldehyde dehydrogenase (ALDH) and CD133 [22, 23]. Furthermore, the presence of ALDH and CD133 positive cells in debulked primary tumour specimen correlated with reduced PFS and overall survival (OS) in ovarian cancer patients [23]. This may be because of the association of high ALDH1A1 expression/activity with platinum-resistant cells in vitro. In an in vivo orthotopic mouse model of ovarian cancer, ALDH1A1 silencing sensitized both taxane- and platinum-resistant tumours to chemotherapy [24]. Reimer et al. demonstrated that truncated isoform Vav3.1 is highly expressed in OCSCs and clinically relevant in predicting prognosis and platinum-response as Vav3.1 may be decisively involved in mechanisms causing genuine multidrug resistance [25]. In contrast, in the environment-mediated drug resistance (EMDR) model, cancer cells interact with their surrounding microenvironment and enter a quiescent state due to the complex interplay between tumour and its microenvironment. These surviving populations, which may or may not be OCSCs, can contribute to cancer relapse [26].
Patients who have an initial response to platinum-based chemotherapy are believed to have tumours with intratumour heterogeneity of both intrinsically platinum-resistant cells and also sensitive cells. The sensitive cells undergo apoptosis following chemotherapy (tumour response) but the resistant subpopulation of cells persist and expand, leading to early recurrence in “platinum-resistant” disease. “Platinum-sensitive” patients may respond to platinum, due to the regrowth of the sensitive population. Ultimately however, the “sensitive” cells may alter or mutate, rendering them resistant, or the resistant cell population will outgrow the sensitive population [5].

Mechanisms of anti-VEGF resistance

Evidence suggests that mechanisms of resistance to anti-VEGF therapy might be mediated by tumour cells and by members of the tumour microenvironment [11, 27, 28]. Tumour hypoxia is a major molecular controller of an “angiogenic switch” that determines a time-restricted event during tumour progression in which the balance between pro- and anti-angiogenic factors tilts towards a pro-angiogenic outcome [27, 29, 30]. Blocking the VEGF pathway inhibits vessel formation but also promotes recruitment of vascular progenitors and vascular modulators such as tumour-associated macrophages (TAMs), immature monocytes and hemangiocytes. Growing evidence indicates that inflammation controls angiogenesis as infiltrating tumour-associated macrophages have been linked to the escape from anti-angiogenic therapy [29]. Further, M2 polarized TAMs promote tumour vascularization by producing proangiogenic factors and growth factors, including transforming growth factor (TGF-β) and VEGF, and attracting leukocytes to further enhance angiogenesis [31].
Recent genomic interrogation of large numbers of HGSOC samples indicated high complexity in terms of genetic aberrations, intra- and intertumor heterogeneity and underscored their lack of targetable oncogenic mutations [3234]. Subclassifications of HGSOC based on expression profiles, termed “differentiated”, “immunoreactive”, “mesenchymal” and “proliferative”, were shown to have prognostic value. Proliferative and mesenchymal subtypes exhibit poorest survival but derive a comparably greater benefit from treatment with bevacizumab [35].

Concepts to overcome drug resistance

The response to cytotoxic chemotherapy remains the essential determinant of OC prognosis [36]. The lack of a detailed understanding of the mechanisms that underlie clinical drug resistance has not deterred investigators from initiating a range of clinical trials that aim to tackle the problem. Novel approaches include disruption of homologous recombination (HR) (i. e. poly-ADP-ribose-polymerase [PARP] inhibitors), reversing inflammation or tumour immune escape and simultaneous targeting of multiple angiogenic pathways using anti-angiogenics. Due to its inherent genomic heterogeneity, molecularly defined subgroups of HGSOC (“differentiated”, “immunoreactive”, “mesenchymal” and “proliferative”) may require different approaches.
BRCA1/2 mutated OCs and OCs with a BRCAness phenotype have demonstrated sensitivity to PARP inhibitors due to underlying deficiencies in DNA homologous recombination; however, clinical responses are often partial and highly dependent on platinum sensitivity. PARP inhibitors such as olaparib, niraparib and rucaparib are already approved for treatment of recurrent EOC and their indications are partially overlapping: niraparib [37] and olaparib [38] have been approved for maintenance therapy after partial or complete remission in recurrent ovarian cancer. Further, olaparib [39] and rucaparib [40] have been approved as monotherapy for advanced recurrent OC. More recently, olaparib has demonstrated impressive activity in BRCA-mutated OC as maintenance following first-line chemotherapy (SOLO-1 trial; HR for disease progression or death, 0.30; 95% CI, 0.23 to 0.41; P < 0.001). Today, probably another promising therapeutic approach in this context is the blockade of immune checkpoints, such as programmed cell death 1 (PD‑1), its ligand PD-L1 or cytotoxic T‑lymphocyte associated protein 4 (CTLA4), which demonstrated impressive response rates in malignant melanoma and non-small-cell lung carcinoma (NSCLC). Considering this and a positive expression of check point molecules in OC which is associated with clinical outcome [41] many clinical studies investigate check point inhibitors in OC, especially platinum-resistant or recurrent OC [42]. Various simultaneous anti-angiogenics may improve the therapeutic benefit and counteract compensatory escape mechanisms [43]. Additional studies are necessary to determine optimal combinations that could be either vertical (e. g., bevacizumab with other angiogenesis inhibitors like sorafenib, vandetanib, sunitinib), horizontal (e. g., inhibitors of phosphatidylinositol-4,5-bisphosphate 3‑kinase [PI3K] pathway, MAP kinse-ERK kinase [MEK], angiopoietin), or direct (e. g., bevacizumab with thrombospondin-1 or vascular disrupting agents such as combretastatin A1 phosphate [OXi4503]) [11].
Collectively, resistance to chemotherapy and anti-angiogenic approaches depends on multiple factors that are challenging to control in a clinical setting until today. Insights into tumour biology and the tumour microenvironment may help to overcome mechanisms of resistance to tackle OC progression. Increasing availability of novel (mechanistically distinct) treatment approaches in OC and the selection of patients that benefit from particular treatment modalities may improve OC survival. Addressing these issues will require further clinical investigations and identification of predictive biomarkers.

Conflict of interest

V. Wieser and C. Marth declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat 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;21:3194–200.CrossRef 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;21:3194–200.CrossRef
2.
Zurück zum Zitat Burger RA, Brady MF, Bookman MA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011;365:2473–83.CrossRef Burger RA, Brady MF, Bookman MA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011;365:2473–83.CrossRef
3.
Zurück zum Zitat Rubin SC, Randall TC, Armstrong KA, Chi DS, Hoskins WJ. Ten-year follow-up of ovarian cancer patients after second-look laparotomy with negative findings. Obstet Gynecol. 1999;93:21–4.PubMed Rubin SC, Randall TC, Armstrong KA, Chi DS, Hoskins WJ. Ten-year follow-up of ovarian cancer patients after second-look laparotomy with negative findings. Obstet Gynecol. 1999;93:21–4.PubMed
4.
Zurück zum Zitat Kim A, Ueda Y, Naka T, Enomoto T. Therapeutic strategies in epithelial ovarian cancer. J Exp Clin Cancer Res. 2012;31:14.CrossRef Kim A, Ueda Y, Naka T, Enomoto T. Therapeutic strategies in epithelial ovarian cancer. J Exp Clin Cancer Res. 2012;31:14.CrossRef
5.
Zurück zum Zitat Davis A, Tinker AV, Friedlander M. “Platinum resistant” ovarian cancer: what is it, who to treat and how to measure benefit? Gynecol Oncol. 2014;133:624–31.CrossRef Davis A, Tinker AV, Friedlander M. “Platinum resistant” ovarian cancer: what is it, who to treat and how to measure benefit? Gynecol Oncol. 2014;133:624–31.CrossRef
6.
Zurück zum Zitat Gavalas NG, Liontos M, Trachana SP, et al. Angiogenesis-related pathways in the pathogenesis of ovarian cancer. Int J Mol Sci. 2013;14:15885–909.CrossRef Gavalas NG, Liontos M, Trachana SP, et al. Angiogenesis-related pathways in the pathogenesis of ovarian cancer. Int J Mol Sci. 2013;14:15885–909.CrossRef
7.
Zurück zum Zitat Boocock CA, Charnock-Jones DS, Sharkey AM, et al. Expression of vascular endothelial growth factor and its receptors flt and KDR in ovarian carcinoma. J Natl Cancer Inst. 1995;87:506–16.CrossRef Boocock CA, Charnock-Jones DS, Sharkey AM, et al. Expression of vascular endothelial growth factor and its receptors flt and KDR in ovarian carcinoma. J Natl Cancer Inst. 1995;87:506–16.CrossRef
8.
Zurück zum Zitat Reynolds K, Farzaneh F, Collins WP, et al. Association of ovarian malignancy with expression of platelet-derived endothelial cell growth factor. J Natl Cancer Inst. 1994;86:1234–8.CrossRef Reynolds K, Farzaneh F, Collins WP, et al. Association of ovarian malignancy with expression of platelet-derived endothelial cell growth factor. J Natl Cancer Inst. 1994;86:1234–8.CrossRef
9.
Zurück zum Zitat Aghajanian C, Goff B, Nycum LR, Wang YV, Husain A, Blank SV. Final overall survival and safety analysis of OCEANS, a phase 3 trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent ovarian cancer. Gynecol Oncol. 2015;139:10–6.CrossRef Aghajanian C, Goff B, Nycum LR, Wang YV, Husain A, Blank SV. Final overall survival and safety analysis of OCEANS, a phase 3 trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent ovarian cancer. Gynecol Oncol. 2015;139:10–6.CrossRef
10.
Zurück zum Zitat Pujade-Lauraine E, Hilpert F, Weber B, et al. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial. J Clin Oncol. 2014;32:1302–8.CrossRef Pujade-Lauraine E, Hilpert F, Weber B, et al. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial. J Clin Oncol. 2014;32:1302–8.CrossRef
11.
Zurück zum Zitat Yang X, Shen F, Hu W, Coleman RL, Sood AK. New ways to successfully target tumor vasculature in ovarian cancer. Curr Opin Obstet Gynecol. 2015;27:58–65.CrossRef Yang X, Shen F, Hu W, Coleman RL, Sood AK. New ways to successfully target tumor vasculature in ovarian cancer. Curr Opin Obstet Gynecol. 2015;27:58–65.CrossRef
12.
Zurück zum Zitat Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat Rev Cancer. 2003;3:502–16.CrossRef Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance to chemotherapy. Nat Rev Cancer. 2003;3:502–16.CrossRef
14.
Zurück zum Zitat Alsop K, Fereday S, Meldrum C, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol. 2012;30:2654–63.CrossRef Alsop K, Fereday S, Meldrum C, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol. 2012;30:2654–63.CrossRef
15.
Zurück zum Zitat David BY, Chetrit A, Hirsh-Yechezkel G, et al. Effect of BRCA mutations on the length of survival in epithelial ovarian tumors. J Clin Oncol. 2002;20:463–6.CrossRef David BY, Chetrit A, Hirsh-Yechezkel G, et al. Effect of BRCA mutations on the length of survival in epithelial ovarian tumors. J Clin Oncol. 2002;20:463–6.CrossRef
16.
Zurück zum Zitat Tan DS, Rothermundt C, Thomas K, et al. “BRCAness” syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530–6.CrossRef Tan DS, Rothermundt C, Thomas K, et al. “BRCAness” syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530–6.CrossRef
17.
Zurück zum Zitat Vencken PM, Kriege M, Hoogwerf D, et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol. 2011;22:1346–52.CrossRef Vencken PM, Kriege M, Hoogwerf D, et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol. 2011;22:1346–52.CrossRef
18.
Zurück zum Zitat Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16:110–20.CrossRef Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16:110–20.CrossRef
19.
Zurück zum Zitat Konstantinopoulos PA, Spentzos D, Karlan BY, et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol. 2010;28:3555–61.CrossRef Konstantinopoulos PA, Spentzos D, Karlan BY, et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol. 2010;28:3555–61.CrossRef
20.
Zurück zum Zitat Boesch M, Sopper S, Zeimet AG, et al. Heterogeneity of cancer stem cells: rationale for targeting the stem cell niche. Biochim Biophys Acta. 2016;1866:276–89.PubMedPubMedCentral Boesch M, Sopper S, Zeimet AG, et al. Heterogeneity of cancer stem cells: rationale for targeting the stem cell niche. Biochim Biophys Acta. 2016;1866:276–89.PubMedPubMedCentral
21.
Zurück zum Zitat Zeimet AG, Reimer D, Sopper S, et al. Ovarian cancer stem cells. Neoplasma. 2012;59:747–55.CrossRef Zeimet AG, Reimer D, Sopper S, et al. Ovarian cancer stem cells. Neoplasma. 2012;59:747–55.CrossRef
22.
Zurück zum Zitat Kryczek I, Liu S, Roh M, et al. Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int J Cancer. 2012;130:29–39.CrossRef Kryczek I, Liu S, Roh M, et al. Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells. Int J Cancer. 2012;130:29–39.CrossRef
23.
Zurück zum Zitat Silva IA, Bai S, McLean K, et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011;71:3991–4001.CrossRef Silva IA, Bai S, McLean K, et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011;71:3991–4001.CrossRef
24.
Zurück zum Zitat Landen CN Jr., Goodman B, Katre AA, et al. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther. 2010;9:3186–99.CrossRef Landen CN Jr., Goodman B, Katre AA, et al. Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer. Mol Cancer Ther. 2010;9:3186–99.CrossRef
25.
Zurück zum Zitat Reimer D, Boesch M, Wolf D, et al. Truncated isoform Vav3.1 is highly expressed in ovarian cancer stem cells and clinically relevant in predicting prognosis and platinum-response. Int J Cancer. 2018;142:1640–51.CrossRef Reimer D, Boesch M, Wolf D, et al. Truncated isoform Vav3.1 is highly expressed in ovarian cancer stem cells and clinically relevant in predicting prognosis and platinum-response. Int J Cancer. 2018;142:1640–51.CrossRef
26.
Zurück zum Zitat Zahreddine H, Borden KL. Mechanisms and insights into drug resistance in cancer. Front Pharmacol. 2013;4:28.CrossRef Zahreddine H, Borden KL. Mechanisms and insights into drug resistance in cancer. Front Pharmacol. 2013;4:28.CrossRef
27.
Zurück zum Zitat Tomao F, Papa A, Rossi L, et al. Beyond bevacizumab: investigating new angiogenesis inhibitors in ovarian cancer. Expert Opin Investig Drugs. 2014;23:37–53.CrossRef Tomao F, Papa A, Rossi L, et al. Beyond bevacizumab: investigating new angiogenesis inhibitors in ovarian cancer. Expert Opin Investig Drugs. 2014;23:37–53.CrossRef
28.
Zurück zum Zitat Winiarski BK, Wolanska KI, Rai S, et al. Epithelial ovarian cancer-induced angiogenic phenotype of human omental microvascular endothelial cells may occur independently of VEGF signaling. Transl Oncol. 2013;6:703–14.CrossRef Winiarski BK, Wolanska KI, Rai S, et al. Epithelial ovarian cancer-induced angiogenic phenotype of human omental microvascular endothelial cells may occur independently of VEGF signaling. Transl Oncol. 2013;6:703–14.CrossRef
29.
Zurück zum Zitat Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.CrossRef Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61.CrossRef
30.
Zurück zum Zitat Baeriswyl V, Christofori G. The angiogenic switch in carcinogenesis. Semin Cancer Biol. 2009;19:329–37.CrossRef Baeriswyl V, Christofori G. The angiogenic switch in carcinogenesis. Semin Cancer Biol. 2009;19:329–37.CrossRef
31.
Zurück zum Zitat Wang X, Zhao X, Wang K, Wu L, Duan T. Interaction of monocytes/macrophages with ovarian cancer cells promotes angiogenesis in vitro. Cancer Sci. 2013;104:516–23.CrossRef Wang X, Zhao X, Wang K, Wu L, Duan T. Interaction of monocytes/macrophages with ovarian cancer cells promotes angiogenesis in vitro. Cancer Sci. 2013;104:516–23.CrossRef
32.
Zurück zum Zitat Welsh JB, Zarrinkar PP, Sapinoso LM, et al. Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc Natl Acad Sci U S A. 2001;98:1176–81.CrossRef Welsh JB, Zarrinkar PP, Sapinoso LM, et al. Analysis of gene expression profiles in normal and neoplastic ovarian tissue samples identifies candidate molecular markers of epithelial ovarian cancer. Proc Natl Acad Sci U S A. 2001;98:1176–81.CrossRef
33.
Zurück zum Zitat Iorio MV, Visone R, Di Leva G, et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007;67:8699–707.CrossRef Iorio MV, Visone R, Di Leva G, et al. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007;67:8699–707.CrossRef
34.
Zurück zum Zitat Nam EJ, Yoon H, Kim SW, et al. MicroRNA expression profiles in serous ovarian carcinoma. Clin Cancer Res. 2008;14:2690–5.CrossRef Nam EJ, Yoon H, Kim SW, et al. MicroRNA expression profiles in serous ovarian carcinoma. Clin Cancer Res. 2008;14:2690–5.CrossRef
35.
Zurück zum Zitat Kommoss S, Winterhoff B, Oberg AL, et al. Bevacizumab May differentially improve ovarian cancer outcome in patients with proliferative and mesenchymal molecular subtypes. Clin Cancer Res. 2017;23:3794–801.CrossRef Kommoss S, Winterhoff B, Oberg AL, et al. Bevacizumab May differentially improve ovarian cancer outcome in patients with proliferative and mesenchymal molecular subtypes. Clin Cancer Res. 2017;23:3794–801.CrossRef
36.
Zurück zum Zitat Colombo PE, Fabbro M, Theillet C, Bibeau F, Rouanet P, Ray-Coquard I. Sensitivity and resistance to treatment in the primary management of epithelial ovarian cancer. Crit Rev Oncol Hematol. 2014;89:207–16.CrossRef Colombo PE, Fabbro M, Theillet C, Bibeau F, Rouanet P, Ray-Coquard I. Sensitivity and resistance to treatment in the primary management of epithelial ovarian cancer. Crit Rev Oncol Hematol. 2014;89:207–16.CrossRef
37.
Zurück zum Zitat Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375:2154–64.CrossRef Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375:2154–64.CrossRef
38.
Zurück zum Zitat Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18:1274–84.CrossRef Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18:1274–84.CrossRef
39.
Zurück zum Zitat Matulonis UA, Penson RT, Domchek SM, et al. Olaparib monotherapy in patients with advanced relapsed ovarian cancer and a germline BRCA1/2 mutation: a multistudy analysis of response rates and safety. Ann Oncol. 2016;27:1013–9.CrossRef Matulonis UA, Penson RT, Domchek SM, et al. Olaparib monotherapy in patients with advanced relapsed ovarian cancer and a germline BRCA1/2 mutation: a multistudy analysis of response rates and safety. Ann Oncol. 2016;27:1013–9.CrossRef
40.
Zurück zum Zitat Balasubramaniam S, Beaver JA, Horton S, et al. FDA approval summary: rucaparib for the treatment of patients with deleterious BRCA mutation-associated advanced ovarian cancer. Clin Cancer Res. 2017;23:7165–70.CrossRef Balasubramaniam S, Beaver JA, Horton S, et al. FDA approval summary: rucaparib for the treatment of patients with deleterious BRCA mutation-associated advanced ovarian cancer. Clin Cancer Res. 2017;23:7165–70.CrossRef
41.
Zurück zum Zitat Wieser V, Gaugg I, Fleischer M, et al. BRCA1/2 and TP53 mutation status associates with PD-1 and PD-L1 expression in ovarian cancer. Oncotarget. 2018;9:17501–11.CrossRef Wieser V, Gaugg I, Fleischer M, et al. BRCA1/2 and TP53 mutation status associates with PD-1 and PD-L1 expression in ovarian cancer. Oncotarget. 2018;9:17501–11.CrossRef
42.
Zurück zum Zitat Marth C, Wieser V, Tsibulak I, Zeimet AG. Immunotherapy in ovarian cancer: fake news or the real deal? International Journal of Gynecologic Cancer. 2019;29(1):201–11.CrossRef Marth C, Wieser V, Tsibulak I, Zeimet AG. Immunotherapy in ovarian cancer: fake news or the real deal? International Journal of Gynecologic Cancer. 2019;29(1):201–11.CrossRef
43.
Zurück zum Zitat Moreno Garcia V, Basu B, Molife LR, Kaye SB. Combining antiangiogenics to overcome resistance: rationale and clinical experience. Clin Cancer Res. 2012;18:3750–61.CrossRef Moreno Garcia V, Basu B, Molife LR, Kaye SB. Combining antiangiogenics to overcome resistance: rationale and clinical experience. Clin Cancer Res. 2012;18:3750–61.CrossRef
Metadaten
Titel
Resistance to chemotherapy and anti-angiogenic therapy in ovarian cancer
verfasst von
Verena Wieser, MD, PhD
Christian Marth, MD, PhD
Publikationsdatum
21.02.2019
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 2/2019
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-019-0478-5

Weitere Artikel der Ausgabe 2/2019

memo - Magazine of European Medical Oncology 2/2019 Zur Ausgabe