Abstract
Ovarian cancer is one of the gynecologic cancers with the highest mortality, wherein vascular endothelial growth factor (VEGF) is involved in regulating tumor vascularization, growth, migration, and invasion. VEGF-mediated angiogenesis in tumors has been targeted in various cancer treatments, and anti-VEGF therapy has been used clinically for treatment of several types of cancer. Paclitaxel is a natural antitumor agent in the standard front-line treatment that has significant efficiency to treat advanced cancers, including ovarian cancer. Although platinum/paclitaxel-based chemotherapy has good response rates, most patients eventually relapse because the disease develops drug resistance. We aim to review the recent advances in paclitaxel treatment of ovarian cancer via antiangiogenesis. Single-agent therapy may be used in selected cases of ovarian cancer. However, to prevent drug resistance, drug combinations should be identified for optimal effectiveness and existing therapies should be improved.
Keywords: Paclitaxel, ovarian cancer, vascular endothelial growth factor (VEGF), angiogenesis
Introduction
Ovarian cancer is one of the most common and lethal cancers in women. It is a significant public health burden all over the world. In the UK, 7,116 women were diagnosed with ovarian cancer in 2011, and 4,271 deaths from ovarian cancer occurred in 2012 (http://www.cancerresearchuk.org/cancer-info/cancerstats/types/ovary/incidence/). An estimated 21,290 new cases and 14,180 deaths attributed to ovarian cancer will be occurred in 2015 in the US (http://seer.cancer.gov/statfacts/html/ovary.html) [1]. Due to lack of a specific prodromal symptomatology and effective screening strategies, ovarian cancer is difficult to identify, so patients are already at the advanced stage of the disease upon diagnosis. The strongest known risk factors are old age and presence of certain gene mutations [2]. Moreover, age, performance status, tumor histology, optimal cytoreduction, and chemotherapy (e.g., platinum-based regimens) are key prognostic factors. In spite of numerous efforts in improving disease management and technology advances [3], the prognosis for patients with high-grade ovarian cancer has barely changed over the last 30 years, leaving a five-year survival rate of only 30% [4].
Conventional treatment strategy for ovarian cancer relies on surgical cytoreduction, followed by adjuvant chemotherapy [5-7]. A combination of maximal surgical debulking and platinum-based chemotherapy has been the standard treatment methods for advanced ovarian cancer since the mid-1990s [8,9]. This chemotherapy is widely accessible for patients with ovarian cancer, and approximately 70% to 80% of patients respond well to this regimen [10]. However, the remaining patients usually develop resistance to chemotherapy or ultimately suffer incurable recurrence; the average five-year survival rate of these patients is lower than 50% [4,11]. Given the limitations of a number of treatment options, identifying new effective drugs and establishing better treatment strategies are urgently needed. In the past decade, tremendous progress has been made on the discovery and development of novel therapeutic strategies parallel to our better understanding of the disease etiology [12,13]. Many molecule-targeted therapies are being passionately investigated for ovarian cancer treatment [13]. Many active cytotoxic drugs and an increasing number of biological agents are becoming available. For example, antiangiogenic agents (such as bevacizumab, cediranib, and pazopanib [14]) and PARP inhibitor olaparib have been demonstrated to effectively treat ovarian cancer [15,16]. The Bcl-2 family of apoptosis regulators, autotaxin, and mevalonate pathway have also been reported as therapeutic targets [1,13]. Novel therapies have provided new insights to target-based therapies. In this article, we aim to review the current advances of treatment on ovarian cancer via antiangiogenesis, focusing on paclitaxel.
VEGF-mediated angiogenesis in cancer treatment
Angiogenesis is a process that is critical for supplying oxygen and nutrients through the formation of new blood vessels, and thus, has an implication in cancer growth [17]. Vascular endothelial growth factor (VEGF) is the most notable pro-angiogenic factor in the microenvironment, which plays key roles in physiological angiogenesis during embryogenesis, skeletal growth, and reproductive function [18,19]. VEGF is overexpressed in the majority of solid tumors promoting tumor neovasculature [18]. It is also associated with tumor progression and poor prognosis for different cancers [20], including lung [21], colorectal [22], stomach [23], pancreatic [24], prostate [25], breast [26], and ovarian cancers [27,28]. For example, high expression level of VEGF-A is reportedly associated with poor survival in ovarian cancer patients [29], and VEGF-D participates in the process of lymphatic metastasis of epithelial ovarian cancer [30]. VEGF receptor (VEGFR) is aberrantly activated in subsets of ovarian tumors [31,32].
Recent studies have provided insights into the mechanism of VEGF in influencing tumor progression, such as targeting immune cells that are present in the tumor microenvironment [33], consequently affecting the host’s response to tumors [33]. In addition, VEGF receptors might regulate the function of fibroblasts in the tumor stroma [28]. VEGF secreted by tumor cells functions in an autocrine manner dominantly via VEGF receptor tyrosine kinases (RTKs) and neuropilin-mediated signaling pathways [34-36]. It can facilitate the function of cancer stem cells and promote dedifferentiation, which enhances growth, survival, migration, and invasion of cancer cells, as well as epithelial mesenchymal transition [35-37]. Furthermore, the phosphatidylinositol 3-kinase (PI3K)/AKT and MAPK pathways have been identified as the dominant signaling pathways involved in the mechanism through which VEGF exerts its influence [13,36].
Angiogenesis is a hallmark of cancer [34] in regulating tumor growth and lethality [38,39], it is targeted by various cancer therapies that focus on drugs that inhibit VEGF [40]. Angiogenesis via multiple RTK-related pathways promotes ovarian cancer growth and dissemination, which are rate-limiting steps in tumorigenesis and malignant progression [41]. Antiangiogenic agents could potentially be effective therapies in improving outcomes of the disease; bevacizumab, a monoclonal antibody, sequesters VEGF [42], whereas vinblastine, paclitaxel, and docetaxel are microtubule-interfering agents [43]. Inhibitors of VEGF receptor tyrosine kinase, such as pazopanib, cediranib, sorafenib, nintedanib, and anti-integrin antibody, which inhibit both angiogenesis and metastasis, are being intensely investigated [1,44,45].
Paclitaxel, its mechanisms, and drug-resistance
Paclitaxel (or taxol), a natural product extracted from the bark of the Pacific yew tree Taxus brevifolia, is a widely used agent for treatment of a variety of tumors, including lung, breast, head, and neck cancers, Kaposi’s sarcoma, and ovarian cancer [43,46-49]. Paclitaxel is a diterpene alkaloid drug that possesses a variety of pharmacological properties, such as destabilization of microtubules by disassembly and blockage of cell cycle, causing cell death and inducing apoptosis, autophagy, and antiangiogenesis [50].
Paclitaxel functions as a radiosensitizer targeting tubulin; it is also a cytoskeletal drug [51,52]. Paclitaxel treatment against KB cells might down-regulate the expression of polycomb repressive complex 1 and cyclin B2, resulting in microtubule polymer stabilization, and thus, preventing it from disassembly. It could further interfere with mitotic spindle formation, chromosome segregation, and cell division [50]. A mitotic arrest [53] characterized by blocking the G2/M phase of the cell cycle and modulating the radioresponsiveness of tumor cells, could be induced [46].
Paclitaxel acts against diseases through triggering various cancer cell deaths via apoptosis through several pathways, including activation of the death receptor pathway, mitochondrial pathway, and cysteine aspartic-specific protease (caspase) cascades [46]. Paclitaxel also induces upregulation of the proapoptotic proteins Bax [54] and Bak [48,55], as well as downregulation and inactivation of the antiapoptotic protein, Bcl-2 [56,57].
Paclitaxel induces autophagy to inhibit tumorigenesis by raising the expression levels of autophagy protein 5 and Beclin-1, which are required for autophagosome formation. It can enhance the expression of p53 and LC3B (an ingredient of the autophagosome), thus regulating initiation of autophagy [58]. Paclitaxel-induced autophagy is critical in modulating caspase-independent cancer cell death. In addition, paclitaxel has been reported to significantly diminish microvessel density and decrease VEGF synthesis in vivo. Moreover, antiangiogenic effects of paclitaxel might be due to its preferential accumulation in endothelial cells [59].
Interestingly, paclitaxel activates cell survival pathways, such as the Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase pathway and PI3K/AKT signaling pathway to antiapoptosis [60]. Furthermore, paclitaxel increases the expression of survival factors, such as apoptosis inhibitor survivin [61] and the cyclin-dependent kinase inhibitor protein, p21/WAF1/Cip1 [62,63], by inducing persistent activation of cyclin B-cdk1, which leads to the phosphorylation and stabilization of surviving proteins [64]. Thus, the combined use of paclitaxel with MEK inhibitors or paclitaxel with PI3K inhibitors might be promising strategies for chemotherapy.
Although paclitaxel presents significant anticancer effects, cytotoxic efficacy on normal cells and drug resistance exist. One of the major dose-limiting side effects of paclitaxel is distal axonal polyneuropathy (mainly sensory polyneuropathy) [65-67]. At present, the mechanism of paclitaxel resistance is not fully understood because of the complexity of tumor drug resistance, which is implicated in multiple genes and processes. However, according to recent studies, changes of the A-tubulin gene and its associated protein might lead to paclitaxel resistance in ovarian cancer patients [68]. Multidrug resistance 1 might also contribute to paclitaxel resistance [50,69,70]. Paclitaxel resistance has also been suggested to be involved with alterations in cell death response, deletion of multiple apoptotic factors by the upregulation of the autophagic pathway, and collateral sensitivity to platinum [71]. In addition, human melanoma A375 cells produce resistance to paclitaxel (0.001 µmol/L to 0.1 µmol/L) by activating the MAPK and PI3K/AKT signaling pathways [71].
Clinical evidence of paclitaxel and bevacizumab in treating ovarian cancer
In ovarian cancer, chemotherapy is needed in Stage I Grade 3 lesion or Stage II disease. A randomized trial concluded that three cycles of paclitaxel at 175 mg/m2 for 3 h plus carboplatin AUC of 7.5 is recommended [72].
In advanced ovarian cancer, after several trials, the patient showed excellent response rate to paclitaxel [73]. Platinum-paclitaxel doublets were tested in two trials [73,74], and established a front-line therapy in ovarian cancer. Although two subsequent trials have raised questions in the respect of benefit from paclitaxel [75,76], acknowledgement of the limitations of these trials [75,77] has led to the acceptance of this front-line therapy practice, which remains unchanged. Further trials [78-80] aim to decrease the side effects of cisplatin-supported carboplatin by combining cisplatin with paclitaxel, which is commonly used in current clinical practice. Recently publicized trials demonstrated that dose-dense treatment offered better survival or less toxicity than conventional treatment [81,82]. After its safety was confirmed, bevacizumab has been witnessed in several large clinical trials to bring significant progression-free survival and improvement after incorporating it into the paclitaxel-carboplatin doublet [83-87]. Another trial demonstrated a significant decrement of quality-of-life due to this drug [88], but another study showed that this decrement did not persist [89].
In recurrent ovarian cancer, paclitaxel demonstrates benefits in both platinum-sensitive and platinum-resistant patients. Regarding to platinum-sensitive recurrence, several studies do not support single-agent paclitaxel as an option [90,91] but prefer platinum-paclitaxel doublet because its advantages were justified in another set of trials [92-94]. In platinum-resistant recurrence, a response rate of about 20% is observed with single-agent paclitaxel [95]. Many trials have been conducted positively to determine which schedule and dose of paclitaxel could bring optimal benefit and least side effects [96]. In actuality, arguments never cease, weekly doses of paclitaxel proved to possess the same efficacy but attenuated toxicity [97-103], considered to be an optimal schedule. Although single-agent bevacizumab demonstrated an inspiring response rate of about 15%-20% in two trials [104,105], relative side effects paralleled. Bevacizumab has also been evaluated in combination with several other chemotherapy agents, including paclitaxel [106]; it turned out anti-tumor activity and manageable toxicity in recurrent ovarian cancer [107-119].
Paclitaxel and bevacizumab in treating other cancers
Paclitaxel has been used in treating several cancers, including breast cancer, lung cancer, head and neck cancer, and advanced stage of Kaposi’s sarcoma. In metastatic breast cancer, paclitaxel intervenes in both doxorubicin-naive and doxorubicin-refractory diseases, resulting in a response rate of 35%-55% and above 20%, respectively [120,121]. In platinum-resistant ovarian cancer recurrence, weekly regimen was superior to thrice-a-week schedule [122]. Paclitaxel also showed increasing rate of disease-free survival and overall survival in early breast cancer according to a meta-analysis [123]. Paclitaxel exhibited evident benefits in non-small cell lung cancer patients consulting a systematic review [124]. Interestingly, in advanced cases of ovarian cancer, bevacizumab exhibited apparent curative benefits when incorporated into a paclitaxel-carboplatin regimen in a large randomized trial and subsequent meta-analysis [125,126]. In recurrent or metastatic head and neck cancers (R/M HNSCC), paclitaxel showed 40% response rate and an overall nine-month survival in several phase II trials [127]. Nowadays, the combination of paclitaxel-cetuximab in treating patients with platinum-resistant R/M HNSCC is proposed, as supported by a recent study [128]. Moreover, Paclitaxel has also been effective in treating Kaposi’s sarcoma in several studies [129,130].
So far as to Bevacizumab, which is used in breast, lung, colorectal, glioblastoma, and renal cancer standard therapies, in breast cancer, the bevacizumab-paclitaxel combination was approved as a first-line treatment of HER-2-negative meditative breast cancer in 2008 by the US Food and Drug Administration after the significant progression-free survival confirmed in several studies [131]. However, in 2010, this approval was reversed because of side effects of using this drug [132]. Use of bevacizumab remains controversial in the treatment of breast cancer [133]. In lung cancer, bevacizumab is associated with improved survival benefits [134-136]. In colorectal cancer, bevacizumab is beneficial for progression-free survival or time-disease progression when integrated into fluoropyrimidine-based chemotherapy [137]. A meta-analysis also concluded that adding bevacizumab to first-line chemotherapy in advanced colorectal cancer proved magnifying efficacy [138]. As to recurrent glioblastoma, single-agent bevacizumab improved response and progression-free survival in several studies [139-142]. When combined with other chemotherapy drugs, bevacizumab amplified efficacy [143-146]. For renal cell carcinoma (RCC), bevacizumab combined with interferon-α is the first-line treatment for metastatic RCC in Europe and US, after showing significant clinical benefits [147].
Conclusion
Although molecule-targeted anticancer treatments for ovarian cancer are currently available, challenges still abound in the chemotherapy approaches, which combines either single-based or combination drugs with other diverse biological treatments, optimal timing, and treatment sequencing. Due to its notable effects, antiangiogenic therapies and paclitaxel-based combination therapies have presented an amplified synergy effect when used in targeting ovarian cancer. Furthermore, treatment based on radiation and accompanied by paclitaxel, which proved to be effective in inhibiting NSCLC tumor growth [48], is considered to be a treatment foundation against ovarian cancer. Paclitaxel combined with either MEK inhibitors or PI3K inhibitors might also enhance curative rate but might induce unacceptable levels of toxicity to normal cells in clinical settings. Thus, more investigations are left to be conducted to enhance effectiveness and reduce toxicity. From where we stand now, new drugs with acceptable non-cumulative toxicity, reduced risk of recurrence after positive clinical response, and improved survival constitution, will push great advances in curing ovarian cancer patients, especially those who developed drug resistance to normal chemotherapies.
Disclosure of conflict of interest
None.
References
- 1.Robinson E, Fisher N, Stamelos V, Redman C, Richardson A. New strategies for the treatment of ovarian cancer. Biochem Soc Trans. 2014;42:125–129. doi: 10.1042/BST20130203. [DOI] [PubMed] [Google Scholar]
- 2.Hunn J, Rodriguez GC. Ovarian cancer: etiology, risk factors, and epidemiology. Clin Obstet Gynecol. 2012;55:3–23. doi: 10.1097/GRF.0b013e31824b4611. [DOI] [PubMed] [Google Scholar]
- 3.Lheureux S, Karakasis K, Kohn EC, Oza AM. Ovarian cancer treatment: The end of empiricism? Cancer. 2015;121:3203–11. doi: 10.1002/cncr.29481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bast RC Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009;9:415–428. doi: 10.1038/nrc2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Poveda A, Ray-Coquard I, Romero I, Lopez-Guerrero JA, Colombo N. Emerging treatment strategies in recurrent platinum-sensitive ovarian cancer: Focus on trabectedin. Cancer Treat Rev. 2014;40:366–375. doi: 10.1016/j.ctrv.2013.08.001. [DOI] [PubMed] [Google Scholar]
- 6.Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J. Clin. Oncol. 2002;20:1248–1259. doi: 10.1200/JCO.2002.20.5.1248. [DOI] [PubMed] [Google Scholar]
- 7.Wright JD, Ananth CV, Tsui J, Glied SA, Burke WM, Lu YS, Neugut AI, Herzog TJ, Hershman DL. Comparative effectiveness of upfront treatment strategies in elderly women with ovarian cancer. Cancer. 2014;120:1246–54. doi: 10.1002/cncr.28508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Cannistra SA, Bast RC Jr, Berek JS, Bookman MA, Crum CP, DePriest PD, Garber JE, Koh WJ, Markman M, McGuire WP 3rd, Rose PG, Rowinsky EK, Rustin GJ, Skates SJ, Vasey PA, King L. Progress in the management of gynecologic cancer: consensus summary statement. J. Clin. Oncol. 2003;21:129s–132s. doi: 10.1200/JCO.2003.04.003. [DOI] [PubMed] [Google Scholar]
- 9.Collinson FJ, Seligmann J, Perren TJ. Ovarian cancer: advances in first-line treatment strategies with a particular focus on anti-angiogenic agents. Curr Oncol Rep. 2012;14:509–518. doi: 10.1007/s11912-012-0274-4. [DOI] [PubMed] [Google Scholar]
- 10.Harter P, Hilpert F, Mahner S, Heitz F, Pfisterer J, du Bois A. Systemic therapy in recurrent ovarian cancer: current treatment options and new drugs. Expert Rev Anticancer Ther. 2010;10:81–88. doi: 10.1586/era.09.165. [DOI] [PubMed] [Google Scholar]
- 11.Armstrong DK. Relapsed ovarian cancer: challenges and management strategies for a chronic disease. Oncologist. 2002;7(Suppl 5):20–28. doi: 10.1634/theoncologist.7-suppl_5-20. [DOI] [PubMed] [Google Scholar]
- 12.Puistola U, Leminen A. [Treatment strategies in ovarian cancer] . Duodecim. 2013;129:1917–1924. [PubMed] [Google Scholar]
- 13.Banerjee S, Kaye SB. New strategies in the treatment of ovarian cancer: current clinical perspectives and future potential. Clin Cancer Res. 2013;19:961–968. doi: 10.1158/1078-0432.CCR-12-2243. [DOI] [PubMed] [Google Scholar]
- 14.McLachlan J, Banerjee S. Pazopanib in ovarian cancer. Expert Rev Anticancer Ther. 2015;15:995–1005. doi: 10.1586/14737140.2015.1081383. [DOI] [PubMed] [Google Scholar]
- 15.Li J, Li S, Chen R, Yu H, Lu X. The prognostic significance of anti-angiogenesis therapy in ovarian cancer: a meta-analysis. J Ovarian Res. 2015;8:54. doi: 10.1186/s13048-015-0181-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pignata S, Lorusso D, Scambia G, Sambataro D, Tamberi S, Cinieri S, Mosconi AM, Orditura M, Brandes AA, Arcangeli V, Panici PB, Pisano C, Cecere SC, Di Napoli M, Raspagliesi F, Maltese G, Salutari V, Ricci C, Daniele G, Piccirillo MC, Di Maio M, Gallo C, Perrone F. Pazopanib plus weekly paclitaxel versus weekly paclitaxel alone for platinum-resistant or platinum-refractory advanced ovarian cancer (MITO 11): a randomised, open-label, phase 2 trial. Lancet Oncol. 2015;16:561–568. doi: 10.1016/S1470-2045(15)70115-4. [DOI] [PubMed] [Google Scholar]
- 17.Jayson GC, Hicklin DJ, Ellis LM. Antiangiogenic therapy--evolving view based on clinical trial results. Nat Rev Clin Oncol. 2012;9:297–303. doi: 10.1038/nrclinonc.2012.8. [DOI] [PubMed] [Google Scholar]
- 18.Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer. 2013;13:871–882. doi: 10.1038/nrc3627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Finley SD, Popel AS. Effect of tumor microenvironment on tumor VEGF during anti-VEGF treatment: systems biology predictions. J Natl Cancer Inst. 2013;105:802–811. doi: 10.1093/jnci/djt093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hata K, Watanabe Y, Nakai H, Hata T, Hoshiai H. Expression of the vascular endothelial growth factor (VEGF) gene in epithelial ovarian cancer: an approach to anti-VEGF therapy. Anticancer Res. 2011;31:731–737. [PubMed] [Google Scholar]
- 21.Yuan A, Yu CJ, Chen WJ, Lin FY, Kuo SH, Luh KT, Yang PC. Correlation of total VEGF mRNA and protein expression with histologic type, tumor angiogenesis, patient survival and timing of relapse in non-small-cell lung cancer. Int J Cancer. 2000;89:475–483. doi: 10.1002/1097-0215(20001120)89:6<475::aid-ijc2>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 22.Lee JC, Chow NH, Wang ST, Huang SM. Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. Eur J Cancer. 2000;36:748–753. doi: 10.1016/s0959-8049(00)00003-4. [DOI] [PubMed] [Google Scholar]
- 23.Maeda K, Chung YS, Ogawa Y, Takatsuka S, Kang SM, Ogawa M, Sawada T, Sowa M. Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer. 1996;77:858–863. doi: 10.1002/(sici)1097-0142(19960301)77:5<858::aid-cncr8>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 24.Ikeda N, Adachi M, Taki T, Huang C, Hashida H, Takabayashi A, Sho M, Nakajima Y, Kanehiro H, Hisanaga M, Nakano H, Miyake M. Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer. 1999;79:1553–1563. doi: 10.1038/sj.bjc.6690248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.George DJ, Halabi S, Shepard TF, Vogelzang NJ, Hayes DF, Small EJ, Kantoff PW Cancer and Leukemia Group B 9480. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin Cancer Res. 2001;7:1932–1936. [PubMed] [Google Scholar]
- 26.Berns EM, Klijn JG, Look MP, Grebenchtchikov N, Vossen R, Peters H, Geurts-Moespot A, Portengen H, van Staveren IL, Meijer-van Gelder ME, Bakker B, Sweep FC, Foekens JA. Combined vascular endothelial growth factor and TP53 status predicts poor response to tamoxifen therapy in estrogen receptor-positive advanced breast cancer. Clin Cancer Res. 2003;9:1253–1258. [PubMed] [Google Scholar]
- 27.Cheng D, Liang B, Li Y. Serum vascular endothelial growth factor (VEGF-C) as a diagnostic and prognostic marker in patients with ovarian cancer. PLoS One. 2013;8:e55309. doi: 10.1371/journal.pone.0055309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Musrap N, Diamandis EP. Revisiting the complexity of the ovarian cancer microenvironment--clinical implications for treatment strategies. Mol Cancer Res. 2012;10:1254–1264. doi: 10.1158/1541-7786.MCR-12-0353. [DOI] [PubMed] [Google Scholar]
- 29.van der Bilt AR, Terwisscha van Scheltinga AG, Timmer-Bosscha H, Schröder CP, Pot L, Kosterink JG, van der Zee AG, Lub-de Hooge MN, de Jong S, de Vries EG, Reyners AK. Measurement of tumor VEGF-A levels with 89Zr-bevacizumab PET as an early biomarker for the antiangiogenic effect of everolimus treatment in an ovarian cancer xenograft model. Clin Cancer Res. 2012;18:6306–6314. doi: 10.1158/1078-0432.CCR-12-0406. [DOI] [PubMed] [Google Scholar]
- 30.He L, He J, Zhao X. Expression of VEGF-D in epithelial ovarian cancer and its relationship to lymphatic metastasis. Asia Pac J Clin Oncol. 2016;12:e161–6. doi: 10.1111/ajco.12086. [DOI] [PubMed] [Google Scholar]
- 31.Becker MA, Farzan T, Harrington SC, Krempski JW, Weroha SJ, Hou X, Kalli KR, Wong TW, Haluska P. Dual HER/VEGF receptor targeting inhibits in vivo ovarian cancer tumor growth. Mol Cancer Ther. 2013;12:2909–2916. doi: 10.1158/1535-7163.MCT-13-0547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Spannuth WA, Nick AM, Jennings NB, Armaiz-Pena GN, Mangala LS, Danes CG, Lin YG, Merritt WM, Thaker PH, Kamat AA, Han LY, Tonra JR, Coleman RL, Ellis LM, Sood AK. Functional significance of VEGFR-2 on ovarian cancer cells. Int J Cancer. 2009;124:1045–1053. doi: 10.1002/ijc.24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ziogas AC, Gavalas NG, Tsiatas M, Tsitsilonis O, Politi E, Terpos E, Rodolakis A, Vlahos G, Thomakos N, Haidopoulos D, Antsaklis A, Dimopoulos MA, Bamias A. VEGF directly suppresses activation of T cells from ovarian cancer patients and healthy individuals via VEGF receptor Type 2. Int J Cancer. 2012;130:857–864. doi: 10.1002/ijc.26094. [DOI] [PubMed] [Google Scholar]
- 34.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 35.Barr MP, Bouchier-Hayes DJ, Harmey JJ. Vascular endothelial growth factor is an autocrine survival factor for breast tumour cells under hypoxia. Int J Oncol. 2008;32:41–48. [PubMed] [Google Scholar]
- 36.Parikh AA, Liu WB, Fan F, Stoeltzing O, Reinmuth N, Bruns CJ, Bucana CD, Evans DB, Ellis LM. Expression and regulation of the novel vascular endothelial growth factor receptor neuropilin-1 by epidermal growth factor in human pancreatic carcinoma. Cancer. 2003;98:720–729. doi: 10.1002/cncr.11560. [DOI] [PubMed] [Google Scholar]
- 37.Duncan TJ, Al-Attar A, Rolland P, Scott IV, Deen S, Liu DT, Spendlove I, Durrant LG. Vascular endothelial growth factor expression in ovarian cancer: a model for targeted use of novel therapies? Clin Cancer Res. 2008;14:3030–3035. doi: 10.1158/1078-0432.CCR-07-1888. [DOI] [PubMed] [Google Scholar]
- 38.Pralhad T, Madhusudan S, Rajendrakumar K. Concept, mechanisms and therapeutics of angiogenesis in cancer and other diseases. J Pharm Pharmacol. 2003;55:1045–1053. doi: 10.1211/0022357021819. [DOI] [PubMed] [Google Scholar]
- 39.Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257. doi: 10.1038/35025220. [DOI] [PubMed] [Google Scholar]
- 40.Jayson G. VEGF inhibitors and advanced ovarian cancer. Lancet Oncol. 2011;12:1082–1083. doi: 10.1016/S1470-2045(11)70269-8. [DOI] [PubMed] [Google Scholar]
- 41.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 42.Barnett JC, Alvarez Secord A, Cohn DE, Leath CA 3rd, Myers ER, Havrilesky LJ. Cost effectiveness of alternative strategies for incorporating bevacizumab into the primary treatment of ovarian cancer. Cancer. 2013;119:3653–3661. doi: 10.1002/cncr.28283. [DOI] [PubMed] [Google Scholar]
- 43.Wang H, Liu B, Zhang C, Peng G, Liu M, Li D, Gu F, Chen Q, Dong JT, Fu L, Zhou J. Parkin regulates paclitaxel sensitivity in breast cancer via a microtubule-dependent mechanism. J Pathol. 2009;218:76–85. doi: 10.1002/path.2512. [DOI] [PubMed] [Google Scholar]
- 44.Blankenberg FG, Levashova Z, Sarkar SK, Pizzonia J, Backer MV, Backer JM. Noninvasive assessment of tumor VEGF receptors in response to treatment with pazopanib: a molecular imaging study. Transl Oncol. 2010;3:56–64. doi: 10.1593/tlo.09271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ledermann JA, Raja FA. Clinical trials and decision-making strategies for optimal treatment of relapsed ovarian cancer. Eur J Cancer. 2011;47(Suppl 3):S104–115. doi: 10.1016/S0959-8049(11)70154-X. [DOI] [PubMed] [Google Scholar]
- 46.Hsiao JR, Leu SF, Huang BM. Apoptotic mechanism of paclitaxel-induced cell death in human head and neck tumor cell lines. J Oral Pathol Med. 2009;38:188–197. doi: 10.1111/j.1600-0714.2008.00732.x. [DOI] [PubMed] [Google Scholar]
- 47.Honore S, Kamath K, Braguer D, Horwitz SB, Wilson L, Briand C, Jordan MA. Synergistic suppression of microtubule dynamics by discodermolide and paclitaxel in non-small cell lung carcinoma cells. Cancer Res. 2004;64:4957–4964. doi: 10.1158/0008-5472.CAN-04-0693. [DOI] [PubMed] [Google Scholar]
- 48.Li G, Zhao J, Peng X, Liang J, Deng X, Chen Y. Radiation/paclitaxel treatment of p53-abnormal non-small cell lung cancer xenograft tumor and associated mechanism. Cancer Biother Radiopharm. 2012;27:227–233. doi: 10.1089/cbr.2011.1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Collins TS, Lee LF, Ting JP. Paclitaxel up-regulates interleukin-8 synthesis in human lung carcinoma through an NF-kappaB- and AP-1-dependent mechanism. Cancer Immunol Immunother. 2000;49:78–84. doi: 10.1007/s002620050605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang X, Chen LX, Ouyang L, Cheng Y, Liu B. Plant natural compounds: targeting pathways of autophagy as anti-cancer therapeutic agents. Cell Prolif. 2012;45:466–476. doi: 10.1111/j.1365-2184.2012.00833.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mollinedo F, Gajate C. Microtubules, microtubule-interfering agents and apoptosis. Apoptosis. 2003;8:413–450. doi: 10.1023/a:1025513106330. [DOI] [PubMed] [Google Scholar]
- 52.Abal M, Andreu JM, Barasoain I. Taxanes: microtubule and centrosome targets, and cell cycle dependent mechanisms of action. Curr Cancer Drug Targets. 2003;3:193–203. doi: 10.2174/1568009033481967. [DOI] [PubMed] [Google Scholar]
- 53.Wilson L, Jordan MA. New microtubule/tubulin-targeted anticancer drugs and novel chemotherapeutic strategies. J Chemother. 2004;16(Suppl 4):83–85. doi: 10.1179/joc.2004.16.Supplement-1.83. [DOI] [PubMed] [Google Scholar]
- 54.Liu SY, Song SX, Lin L, Liu X. Molecular mechanism of cell apoptosis by paclitaxel and pirarubicin in a human osteosarcoma cell line. Chemotherapy. 2010;56:101–107. doi: 10.1159/000305257. [DOI] [PubMed] [Google Scholar]
- 55.Li J, Wang WL, Yang XK, Yu XX, Hou YD, Zhang J. Inducible overexpression of Bak sensitizes HCC-9204 cells to apoptosis induced by doxorubicin. Acta Pharmacol Sin. 2000;21:769–776. [PubMed] [Google Scholar]
- 56.Ferlini C, Raspaglio G, Mozzetti S, Distefano M, Filippetti F, Martinelli E, Ferrandina G, Gallo D, Ranelletti FO, Scambia G. Bcl-2 down-regulation is a novel mechanism of paclitaxel resistance. Mol Pharmacol. 2003;64:51–58. doi: 10.1124/mol.64.1.51. [DOI] [PubMed] [Google Scholar]
- 57.Sgadari C, Toschi E, Palladino C, Barillari G, Carlei D, Cereseto A, Ciccolella C, Yarchoan R, Monini P, Sturzl M, Ensoli B. Mechanism of paclitaxel activity in Kaposi’s sarcoma. J Immunol. 2000;165:509–517. doi: 10.4049/jimmunol.165.1.509. [DOI] [PubMed] [Google Scholar]
- 58.Lanni JS, Lowe SW, Licitra EJ, Liu JO, Jacks T. p53-independent apoptosis induced by paclitaxel through an indirect mechanism. Proc Natl Acad Sci U S A. 1997;94:9679–9683. doi: 10.1073/pnas.94.18.9679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Merchan JR, Jayaram DR, Supko JG, He X, Bubley GJ, Sukhatme VP. Increased endothelial uptake of paclitaxel as a potential mechanism for its antiangiogenic effects: potentiation by Cox-2 inhibition. Int J Cancer. 2005;113:490–498. doi: 10.1002/ijc.20595. [DOI] [PubMed] [Google Scholar]
- 60.MacKeigan JP, Taxman DJ, Hunter D, Earp HS 3rd, Graves LM, Ting JP. Inactivation of the antiapoptotic phosphatidylinositol 3-kinase-Akt pathway by the combined treatment of taxol and mitogen-activated protein kinase kinase inhibition. Clin Cancer Res. 2002;8:2091–2099. [PubMed] [Google Scholar]
- 61.Fortugno P, Wall NR, Giodini A, O’Connor DS, Plescia J, Padgett KM, Tognin S, Marchisio PC, Altieri DC. Survivin exists in immunochemically distinct subcellular pools and is involved in spindle microtubule function. J Cell Sci. 2002;115:575–585. doi: 10.1242/jcs.115.3.575. [DOI] [PubMed] [Google Scholar]
- 62.Mitsuuchi Y, Johnson SW, Selvakumaran M, Williams SJ, Hamilton TC, Testa JR. The phosphatidylinositol 3-kinase/AKT signal transduction pathway plays a critical role in the expression of p21WAF1/CIP1/SDI1 induced by cisplatin and paclitaxel. Cancer Res. 2000;60:5390–5394. [PubMed] [Google Scholar]
- 63.Heliez C, Baricault L, Barboule N, Valette A. Paclitaxel increases p21 synthesis and accumulation of its AKT-phosphorylated form in the cytoplasm of cancer cells. Oncogene. 2003;22:3260–3268. doi: 10.1038/sj.onc.1206409. [DOI] [PubMed] [Google Scholar]
- 64.Greenberg VL, Zimmer SG. Paclitaxel induces the phosphorylation of the eukaryotic translation initiation factor 4E-binding protein 1 through a Cdk1-dependent mechanism. Oncogene. 2005;24:4851–4860. doi: 10.1038/sj.onc.1208624. [DOI] [PubMed] [Google Scholar]
- 65.Yang IH, Siddique R, Hosmane S, Thakor N, Hoke A. Compartmentalized microfluidic culture platform to study mechanism of paclitaxel-induced axonal degeneration. Exp Neurol. 2009;218:124–128. doi: 10.1016/j.expneurol.2009.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bennett GJ, Liu GK, Xiao WH, Jin HW, Siau C. Terminal arbor degeneration--a novel lesion produced by the antineoplastic agent paclitaxel. Eur J Neurosci. 2011;33:1667–1676. doi: 10.1111/j.1460-9568.2011.07652.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Melli G, Jack C, Lambrinos GL, Ringkamp M, Hoke A. Erythropoietin protects sensory axons against paclitaxel-induced distal degeneration. Neurobiol Dis. 2006;24:525–530. doi: 10.1016/j.nbd.2006.08.014. [DOI] [PubMed] [Google Scholar]
- 68.Mozzetti S, Ferlini C, Concolino P, Filippetti F, Raspaglio G, Prislei S, Gallo D, Martinelli E, Ranelletti FO, Ferrandina G, Scambia G. Class III beta-tubulin overexpression is a prominent mechanism of paclitaxel resistance in ovarian cancer patients. Clin Cancer Res. 2005;11:298–305. [PubMed] [Google Scholar]
- 69.Zhang J, Zhao J, Zhang W, Liu G, Yin D, Li J, Zhang S, Li H. Establishment of paclitaxel-resistant cell line and the underlying mechanism on drug resistance. Int J Gynecol Cancer. 2012;22:1450–1456. doi: 10.1097/IGC.0b013e31826e2382. [DOI] [PubMed] [Google Scholar]
- 70.Luo SX, Deng WY, Wang XF, Lu HF, Han LL, Chen BB, Chen XB, Li N. Molecular mechanism of indirubin-3’-monoxime and Matrine in the reversal of paclitaxel resistance in NCI-H520/TAX25 cell line. Chin Med J (Engl) 2013;126:925–929. [PubMed] [Google Scholar]
- 71.Zhang XJ, Zhang L, Liu YP, Xu HM, Sun P, Song JG, Luo YH. [Molecular mechanism of chemosensitization to paclitaxel in human melanoma cells induced by targeting the EGFR signaling pathway] . Zhonghua Zhong Liu Za Zhi. 2013;35:181–186. doi: 10.3760/cma.j.issn.0253-3766.2013.03.005. [DOI] [PubMed] [Google Scholar]
- 72.Bell J, Brady MF, Young RC, Lage J, Walker JL, Look KY, Rose GS, Spirtos NM Gynecologic Oncology Group. Randomized phase III trial of three versus six cycles of adjuvant carboplatin and paclitaxel in early stage epithelial ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2006;102:432–439. doi: 10.1016/j.ygyno.2006.06.013. [DOI] [PubMed] [Google Scholar]
- 73.McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look KY, Clarke-Pearson DL, Davidson M. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer. N Engl J Med. 1996;334:1–6. doi: 10.1056/NEJM199601043340101. [DOI] [PubMed] [Google Scholar]
- 74.Piccart MJ, Bertelsen K, James K, Cassidy J, Mangioni C, Simonsen E, Stuart G, Kaye S, Vergote I, Blom R, Grimshaw R, Atkinson RJ, Swenerton KD, Trope C, Nardi M, Kaern J, Tumolo S, Timmers P, Roy JA, Lhoas F, Lindvall B, Bacon M, Birt A, Andersen JE, Zee B, Paul J, Baron B, Pecorelli S. Randomized intergroup trial of cisplatin-paclitaxel versus cisplatin-cyclophosphamide in women with advanced epithelial ovarian cancer: three-year results. J Natl Cancer Inst. 2000;92:699–708. doi: 10.1093/jnci/92.9.699. [DOI] [PubMed] [Google Scholar]
- 75.Muggia FM, Braly PS, Brady MF, Sutton G, Niemann TH, Lentz SL, Alvarez RD, Kucera PR, Small JM. Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: a gynecologic oncology group study. J. Clin. Oncol. 2000;18:106–115. doi: 10.1200/JCO.2000.18.1.106. [DOI] [PubMed] [Google Scholar]
- 76.Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial. Lancet. 2002;360:505–515. doi: 10.1016/S0140-6736(02)09738-6. [DOI] [PubMed] [Google Scholar]
- 77.Sandercock J, Parmar MK, Torri V, Qian W. First-line treatment for advanced ovarian cancer: paclitaxel, platinum and the evidence. Br J Cancer. 2002;87:815–824. doi: 10.1038/sj.bjc.6600567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Neijt JP, Engelholm SA, Tuxen MK, Sorensen PG, Hansen M, Sessa C, de Swart CA, Hirsch FR, Lund B, van Houwelingen HC. 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]
- 79.du Bois A, Luck HJ, Meier W, Adams HP, Mobus V, Costa S, Bauknecht T, Richter B, Warm M, Schroder W, Olbricht S, Nitz U, Jackisch C, Emons G, Wagner U, Kuhn W, Pfisterer J. A randomized clinical trial of cisplatin/paclitaxel versus carboplatin/paclitaxel as first-line treatment of ovarian cancer. J Natl Cancer Inst. 2003;95:1320–1329. doi: 10.1093/jnci/djg036. [DOI] [PubMed] [Google Scholar]
- 80.Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R. 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]
- 81.Katsumata N, Yasuda M, Isonishi S, Takahashi F, Michimae H, Kimura E, Aoki D, Jobo T, Kodama S, Terauchi F, Sugiyama T, Ochiai K. Long-term results of dose-dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): a randomised, controlled, open-label trial. Lancet Oncol. 2013;14:1020–1026. doi: 10.1016/S1470-2045(13)70363-2. [DOI] [PubMed] [Google Scholar]
- 82.Abaid LN, Micha JP, Rettenmaier MA, Brown JV, Mendivil AA, Lopez KL, Goldstein BH. A phase II study of modified dose-dense paclitaxel and every 4-week carboplatin for the treatment of advanced-stage primary epithelial ovarian, fallopian tube, or peritoneal carcinoma. Cancer Chemother Pharmacol. 2013;72:101–107. doi: 10.1007/s00280-013-2173-2. [DOI] [PubMed] [Google Scholar]
- 83.Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, Mannel RS, Homesley HD, Fowler J, Greer BE, Boente M, Birrer MJ, Liang SX. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 2011;365:2473–2483. doi: 10.1056/NEJMoa1104390. [DOI] [PubMed] [Google Scholar]
- 84.Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, Carey MS, Beale P, Cervantes A, Kurzeder C, du Bois A, Sehouli J, Kimmig R, Stahle A, Collinson F, Essapen S, Gourley C, Lortholary A, Selle F, Mirza MR, Leminen A, Plante M, Stark D, Qian W, Parmar MK, Oza AM. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365:2484–2496. doi: 10.1056/NEJMoa1103799. [DOI] [PubMed] [Google Scholar]
- 85.Penson RT, Dizon DS, Cannistra SA, Roche MR, Krasner CN, Berlin ST, Horowitz NS, Disilvestro PA, Matulonis UA, Lee H, King MA, Campos SM. Phase II study of carboplatin, paclitaxel, and bevacizumab with maintenance bevacizumab as first-line chemotherapy for advanced mullerian tumors. J. Clin. Oncol. 2010;28:154–159. doi: 10.1200/JCO.2009.22.7900. [DOI] [PubMed] [Google Scholar]
- 86.Micha JP, Goldstein BH, Rettenmaier MA, Genesen M, Graham C, Bader K, Lopez KL, Nickle M, Brown JV 3rd. A phase II study of outpatient first-line paclitaxel, carboplatin, and bevacizumab for advanced-stage epithelial ovarian, peritoneal, and fallopian tube cancer. Int J Gynecol Cancer. 2007;17:771–776. doi: 10.1111/j.1525-1438.2007.00886.x. [DOI] [PubMed] [Google Scholar]
- 87.Burger RA, Brady MF, Rhee J, Sovak MA, Kong G, Nguyen HP, Bookman MA. Independent radiologic review of the Gynecologic Oncology Group Study 0218, a phase III trial of bevacizumab in the primary treatment of advanced epithelial ovarian, primary peritoneal, or fallopian tube cancer. Gynecol Oncol. 2013;131:21–26. doi: 10.1016/j.ygyno.2013.07.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Stark D, Nankivell M, Pujade-Lauraine E, Kristensen G, Elit L, Stockler M, Hilpert F, Cervantes A, Brown J, Lanceley A, Velikova G, Sabate E, Pfisterer J, Carey MS, Beale P, Qian W, Swart AM, Oza A, Perren T. Standard chemotherapy with or without bevacizumab in advanced ovarian cancer: quality-of-life outcomes from the International Collaboration on Ovarian Neoplasms (ICON7) phase 3 randomised trial. Lancet Oncol. 2013;14:236–243. doi: 10.1016/S1470-2045(12)70567-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Monk BJ, Huang HQ, Burger RA, Mannel RS, Homesley HD, Fowler J, Greer BE, Boente M, Liang SX, Wenzel L. Patient reported outcomes of a randomized, placebo-controlled trial of bevacizumab in the front-line treatment of ovarian cancer: a Gynecologic Oncology Group Study. Gynecol Oncol. 2013;128:573–578. doi: 10.1016/j.ygyno.2012.11.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Piccart MJ, Green JA, Lacave AJ, Reed N, Vergote I, Benedetti-Panici P, Bonetti A, Kristeller-Tome V, Fernandez CM, Curran D, Van Glabbeke M, Lacombe D, Pinel MC, Pecorelli S. Oxaliplatin or paclitaxel in patients with platinum-pretreated advanced ovarian cancer: A randomized phase II study of the European Organization for Research and Treatment of Cancer Gynecology Group. J. Clin. Oncol. 2000;18:1193–1202. doi: 10.1200/JCO.2000.18.6.1193. [DOI] [PubMed] [Google Scholar]
- 91.Cantu MG, Buda A, Parma G, Rossi R, Floriani I, Bonazzi C, Dell’Anna T, Torri V, Colombo N. Randomized controlled trial of single-agent paclitaxel versus cyclophosphamide, doxorubicin, and cisplatin in patients with recurrent ovarian cancer who responded to first-line platinum-based regimens. J. Clin. Oncol. 2002;20:1232–1237. doi: 10.1200/JCO.2002.20.5.1232. [DOI] [PubMed] [Google Scholar]
- 92.Gronlund B, Hogdall C, Hansen HH, Engelholm SA. Results of reinduction therapy with paclitaxel and carboplatin in recurrent epithelial ovarian cancer. Gynecol Oncol. 2001;83:128–134. doi: 10.1006/gyno.2001.6364. [DOI] [PubMed] [Google Scholar]
- 93.Gonzalez-Martin AJ, Calvo E, Bover I, Rubio MJ, Arcusa A, Casado A, Ojeda B, Balana C, Martinez E, Herrero A, Pardo B, Adrover E, Rifa J, Godes MJ, Moyano A, Cervantes A. Randomized phase II trial of carboplatin versus paclitaxel and carboplatin in platinum-sensitive recurrent advanced ovarian carcinoma: a GEICO (Grupo Espanol de Investigacion en Cancer de Ovario) study. Ann Oncol. 2005;16:749–755. doi: 10.1093/annonc/mdi147. [DOI] [PubMed] [Google Scholar]
- 94.Parmar MK, Ledermann JA, Colombo N, du Bois A, Delaloye JF, Kristensen GB, Wheeler S, Swart AM, Qian W, Torri V, Floriani I, Jayson G, Lamont A, Trope C. Paclitaxel plus platinum-based chemotherapy versus conventional platinum-based chemotherapy in women with relapsed ovarian cancer: the ICON4/AGO-OVAR-2.2 trial. Lancet. 2003;361:2099–2106. doi: 10.1016/s0140-6736(03)13718-x. [DOI] [PubMed] [Google Scholar]
- 95.Coleman RL, Brady WE, McMeekin DS, Rose PG, Soper JT, Lentz SS, Hoffman JS, Shahin MS. A phase II evaluation of nanoparticle, albumin-bound (nab) paclitaxel in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2011;122:111–115. doi: 10.1016/j.ygyno.2011.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Omura GA, Brady MF, Look KY, Averette HE, Delmore JE, Long HJ, Wadler S, Spiegel G, Arbuck SG. Phase III trial of paclitaxel at two dose levels, the higher dose accompanied by filgrastim at two dose levels in platinum-pretreated epithelial ovarian cancer: an intergroup study. J. Clin. Oncol. 2003;21:2843–2848. doi: 10.1200/JCO.2003.10.082. [DOI] [PubMed] [Google Scholar]
- 97.Markman M, Hall J, Spitz D, Weiner S, Carson L, Van Le L, Baker M. Phase II trial of weekly single-agent paclitaxel in platinum/paclitaxel-refractory ovarian cancer. J. Clin. Oncol. 2002;20:2365–2369. doi: 10.1200/JCO.2002.09.130. [DOI] [PubMed] [Google Scholar]
- 98.Markman M, Blessing J, Rubin SC, Connor J, Hanjani P, Waggoner S. Phase II trial of weekly paclitaxel (80 mg/m2) in platinum and paclitaxel-resistant ovarian and primary peritoneal cancers: a Gynecologic Oncology Group study. Gynecol Oncol. 2006;101:436–440. doi: 10.1016/j.ygyno.2005.10.036. [DOI] [PubMed] [Google Scholar]
- 99.Kita T, Kikuchi Y, Takano M, Suzuki M, Oowada M, Konno R, Yamamoto K, Inoue H, Seto H, Yamamoto T, Shimizu K. The effect of single weekly paclitaxel in heavily pretreated patients with recurrent or persistent advanced ovarian cancer. Gynecol Oncol. 2004;92:813–818. doi: 10.1016/j.ygyno.2003.12.002. [DOI] [PubMed] [Google Scholar]
- 100.Kaern J, Baekelandt M, Trope CG. A phase II study of weekly paclitaxel in platinum and paclitaxel-resistant ovarian cancer patients. Eur J Gynaecol Oncol. 2002;23:383–389. [PubMed] [Google Scholar]
- 101.Rosenberg P, Andersson H, Boman K, Ridderheim M, Sorbe B, Puistola U, Paro G. Randomized trial of single agent paclitaxel given weekly versus every three weeks and with peroral versus intravenous steroid premedication to patients with ovarian cancer previously treated with platinum. Acta Oncol. 2002;41:418–424. doi: 10.1080/028418602320404998. [DOI] [PubMed] [Google Scholar]
- 102.Havrilesky LJ, Alvarez AA, Sayer RA, Lancaster JM, Soper JT, Berchuck A, Clarke-Pearson DL, Rodriguez GC, Carney ME. Weekly low-dose carboplatin and paclitaxel in the treatment of recurrent ovarian and peritoneal cancer. Gynecol Oncol. 2003;88:51–57. doi: 10.1006/gyno.2002.6859. [DOI] [PubMed] [Google Scholar]
- 103.van der Burg ME, Vergote I, Onstenk W, Boere IA, Leunen K, van Montfort CA, van Doorn HC. Long-term results of weekly paclitaxel carboplatin induction therapy: an effective and well-tolerated treatment in patients with platinum-resistant ovarian cancer. Eur J Cancer. 2013;49:1254–1263. doi: 10.1016/j.ejca.2012.11.027. [DOI] [PubMed] [Google Scholar]
- 104.Burger RA, Sill MW, Monk BJ, Greer BE, Sorosky JI. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. J. Clin. Oncol. 2007;25:5165–5171. doi: 10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
- 105.Cannistra SA, Matulonis UA, Penson RT, Hambleton J, Dupont J, Mackey H, Douglas J, Burger RA, Armstrong D, Wenham R, McGuire W. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J. Clin. Oncol. 2007;25:5180–5186. doi: 10.1200/JCO.2007.12.0782. [DOI] [PubMed] [Google Scholar]
- 106.Poveda AM, Selle F, Hilpert F, Reuss A, Savarese A, Vergote I, Witteveen P, Bamias A, Scotto N, Mitchell L, Pujade-Lauraine E. Bevacizumab Combined With Weekly Paclitaxel, Pegylated Liposomal Doxorubicin, or Topotecan in Platinum-Resistant Recurrent Ovarian Cancer: Analysis by Chemotherapy Cohort of the Randomized Phase III AURELIA Trial. J. Clin. Oncol. 2015;33:3836–8. doi: 10.1200/JCO.2015.63.1408. [DOI] [PubMed] [Google Scholar]
- 107.Tillmanns TD, Lowe MP, Walker MS, Stepanski EJ, Schwartzberg LS. Phase II clinical trial of bevacizumab with albumin-bound paclitaxel in patients with recurrent, platinum-resistant primary epithelial ovarian or primary peritoneal carcinoma. Gynecol Oncol. 2013;128:221–228. doi: 10.1016/j.ygyno.2012.08.039. [DOI] [PubMed] [Google Scholar]
- 108.Garcia AA, Hirte H, Fleming G, Yang D, Tsao-Wei DD, Roman L, Groshen S, Swenson S, Markland F, Gandara D, Scudder S, Morgan R, Chen H, Lenz HJ, Oza AM. Phase II clinical trial of bevacizumab and low-dose metronomic oral cyclophosphamide in recurrent ovarian cancer: a trial of the California, Chicago, and Princess Margaret Hospital phase II consortia. J. Clin. Oncol. 2008;26:76–82. doi: 10.1200/JCO.2007.12.1939. [DOI] [PubMed] [Google Scholar]
- 109.McGonigle KF, Muntz HG, Vuky J, Paley PJ, Veljovich DS, Greer BE, Goff BA, Gray HJ, Malpass TW. Combined weekly topotecan and biweekly bevacizumab in women with platinum-resistant ovarian, peritoneal, or fallopian tube cancer: results of a phase 2 study. Cancer. 2011;117:3731–3740. doi: 10.1002/cncr.25967. [DOI] [PubMed] [Google Scholar]
- 110.Kudoh K, Takano M, Kouta H, Kikuchi R, Kita T, Miyamoto M, Watanabe A, Kato M, Goto T, Kikuchi Y. Effects of bevacizumab and pegylated liposomal doxorubicin for the patients with recurrent or refractory ovarian cancers. Gynecol Oncol. 2011;122:233–237. doi: 10.1016/j.ygyno.2011.04.046. [DOI] [PubMed] [Google Scholar]
- 111.O’Malley DM, Richardson DL, Rheaume PS, Salani R, Eisenhauer EL, McCann GA, Fowler JM, Copeland LJ, Cohn DE, Backes FJ. Addition of bevacizumab to weekly paclitaxel significantly improves progression-free survival in heavily pretreated recurrent epithelial ovarian cancer. Gynecol Oncol. 2011;121:269–272. doi: 10.1016/j.ygyno.2011.01.009. [DOI] [PubMed] [Google Scholar]
- 112.Wenham RM, Lapolla J, Lin HY, Apte SM, Lancaster JM, Judson PL, Gonzalez-Bosquet J, Herschberger A, Havrilesky LJ, Secord AA. A phase II trial of docetaxel and bevacizumab in recurrent ovarian cancer within 12 months of prior platinum-based chemotherapy. Gynecol Oncol. 2013;130:19–24. doi: 10.1016/j.ygyno.2013.04.049. [DOI] [PubMed] [Google Scholar]
- 113.Ikeda Y, Takano M, Oda K, Kouta H, Goto T, Kudoh K, Sasaki N, Kita T, Kikuchi Y. Weekly administration of bevacizumab, gemcitabine, and oxaliplatin in patients with recurrent and refractory ovarian cancer: a preliminary result of 19 cases. Int J Gynecol Cancer. 2013;23:355–360. doi: 10.1097/IGC.0b013e31827de69e. [DOI] [PubMed] [Google Scholar]
- 114.Moroney J, Fu S, Moulder S, Falchook G, Helgason T, Levenback C, Hong D, Naing A, Wheler J, Kurzrock R. Phase I study of the antiangiogenic antibody bevacizumab and the mTOR/hypoxia-inducible factor inhibitor temsirolimus combined with liposomal doxorubicin: tolerance and biological activity. Clin Cancer Res. 2012;18:5796–5805. doi: 10.1158/1078-0432.CCR-12-1158. [DOI] [PubMed] [Google Scholar]
- 115.Verschraegen CF, Czok S, Muller CY, Boyd L, Lee SJ, Rutledge T, Blank S, Pothuri B, Eberhardt S, Muggia F. Phase II study of bevacizumab with liposomal doxorubicin for patients with platinum- and taxane-resistant ovarian cancer. Ann Oncol. 2012;23:3104–3110. doi: 10.1093/annonc/mds172. [DOI] [PubMed] [Google Scholar]
- 116.del Carmen MG, Micha J, Small L, Street DG, Londhe A, McGowan T. A phase II clinical trial of pegylated liposomal doxorubicin and carboplatin plus bevacizumab in patients with platinum-sensitive recurrent ovarian, fallopian tube, or primary peritoneal cancer. Gynecol Oncol. 2012;126:369–374. doi: 10.1016/j.ygyno.2012.05.028. [DOI] [PubMed] [Google Scholar]
- 117.Aghajanian C, Blank SV, Goff BA, Judson PL, Teneriello MG, Husain A, Sovak MA, Yi J, Nycum LR. OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J. Clin. Oncol. 2012;30:2039–2045. doi: 10.1200/JCO.2012.42.0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Matulonis UA, Pereira L, Liu J, Lee H, Lee J, Whalen C, Campos S, Atkinson T, Hill M, Berlin S. Sequential bevacizumab and oral cyclophosphamide for recurrent ovarian cancer. Gynecol Oncol. 2012;126:41–46. doi: 10.1016/j.ygyno.2012.04.003. [DOI] [PubMed] [Google Scholar]
- 119.Sanchez-Munoz A, Mendiola C, Perez-Ruiz E, Rodriguez-Sanchez CA, Jurado JM, Alonso-Carrion L, Ghanem I, de Velasco G, Quero-Blanco C, Alba E. Bevacizumab plus low-dose metronomic oral cyclophosphamide in heavily pretreated patients with recurrent ovarian cancer. Oncology. 2010;79:98–104. doi: 10.1159/000320602. [DOI] [PubMed] [Google Scholar]
- 120.Seidman AD, Tiersten A, Hudis C, Gollub M, Barrett S, Yao TJ, Lepore J, Gilewski T, Currie V, Crown J, et al. Phase II trial of paclitaxel by 3-hour infusion as initial and salvage chemotherapy for metastatic breast cancer. J. Clin. Oncol. 1995;13:2575–2581. doi: 10.1200/JCO.1995.13.10.2575. [DOI] [PubMed] [Google Scholar]
- 121.Holmes FA, Walters RS, Theriault RL, Forman AD, Newton LK, Raber MN, Buzdar AU, Frye DK, Hortobagyi GN. Phase II trial of taxol, an active drug in the treatment of metastatic breast cancer. J Natl Cancer Inst. 1991;83:1797–1805. doi: 10.1093/jnci/83.24.1797-a. [DOI] [PubMed] [Google Scholar]
- 122.Seidman AD, Berry D, Cirrincione C, Harris L, Muss H, Marcom PK, Gipson G, Burstein H, Lake D, Shapiro CL, Ungaro P, Norton L, Winer E, Hudis C. Randomized phase III trial of weekly compared with every-3-weeks paclitaxel for metastatic breast cancer, with trastuzumab for all HER-2 overexpressors and random assignment to trastuzumab or not in HER-2 nonoverexpressors: final results of Cancer and Leukemia Group B protocol 9840. J. Clin. Oncol. 2008;26:1642–1649. doi: 10.1200/JCO.2007.11.6699. [DOI] [PubMed] [Google Scholar]
- 123.De Laurentiis M, Cancello G, D’Agostino D, Giuliano M, Giordano A, Montagna E, Lauria R, Forestieri V, Esposito A, Silvestro L, Pennacchio R, Criscitiello C, Montanino A, Limite G, Bianco AR, De Placido S. Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J. Clin. Oncol. 2008;26:44–53. doi: 10.1200/JCO.2007.11.3787. [DOI] [PubMed] [Google Scholar]
- 124.Clegg A, Scott DA, Hewitson P, Sidhu M, Waugh N. Clinical and cost effectiveness of paclitaxel, docetaxel, gemcitabine, and vinorelbine in non-small cell lung cancer: a systematic review. Thorax. 2002;57:20–28. doi: 10.1136/thorax.57.1.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:2542–2550. doi: 10.1056/NEJMoa061884. [DOI] [PubMed] [Google Scholar]
- 126.Yang K, Wang YJ, Chen XR, Chen HN. Effectiveness and safety of bevacizumab for unresectable non-small-cell lung cancer: a meta-analysis. Clin Drug Investig. 2010;30:229–241. doi: 10.2165/11532260-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 127.Forastiere AA, Shank D, Neuberg D, Taylor SG 4th, DeConti RC, Adams G. Final report of a phase II evaluation of paclitaxel in patients with advanced squamous cell carcinoma of the head and neck: an Eastern Cooperative Oncology Group trial (PA390) Cancer. 1998;82:2270–2274. [PubMed] [Google Scholar]
- 128.Hitt R, Irigoyen A, Cortes-Funes H, Grau JJ, Garcia-Saenz JA, Cruz-Hernandez JJ. Phase II study of the combination of cetuximab and weekly paclitaxel in the first-line treatment of patients with recurrent and/or metastatic squamous cell carcinoma of head and neck. Ann Oncol. 2012;23:1016–1022. doi: 10.1093/annonc/mdr367. [DOI] [PubMed] [Google Scholar]
- 129.Brambilla L, Romanelli A, Bellinvia M, Ferrucci S, Vinci M, Boneschi V, Miedico A, Tedeschi L. Weekly paclitaxel for advanced aggressive classic Kaposi sarcoma: experience in 17 cases. Br J Dermatol. 2008;158:1339–1344. doi: 10.1111/j.1365-2133.2008.08517.x. [DOI] [PubMed] [Google Scholar]
- 130.Gill PS, Tulpule A, Espina BM, Cabriales S, Bresnahan J, Ilaw M, Louie S, Gustafson NF, Brown MA, Orcutt C, Winograd B, Scadden DT. Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi’s sarcoma. J. Clin. Oncol. 1999;17:1876–1883. doi: 10.1200/JCO.1999.17.6.1876. [DOI] [PubMed] [Google Scholar]
- 131.Montero AJ, Escobar M, Lopes G, Gluck S, Vogel C. Bevacizumab in the treatment of metastatic breast cancer: friend or foe? Curr Oncol Rep. 2012;14:1–11. doi: 10.1007/s11912-011-0202-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kumler I, Nielsen DL. Trials of bevacizumab in breast cancer--a safety review. Expert Opin Drug Saf. 2012;11(Suppl 1):S37–48. doi: 10.1517/14740338.2011.594038. [DOI] [PubMed] [Google Scholar]
- 133.Dieras V, Campone M, Yardley DA, Romieu G, Valero V, Isakoff SJ, Koeppen H, Wilson TR, Xiao Y, Shames DS, Mocci S, Chen M, Schmid P. Randomized, phase II, placebo-controlled trial of onartuzumab and/or bevacizumab in combination with weekly paclitaxel in patients with metastatic triple-negative breast cancer. Ann Oncol. 2015;26:1904–10. doi: 10.1093/annonc/mdv263. [DOI] [PubMed] [Google Scholar]
- 134.Planchard D. Bevacizumab in non-small-cell lung cancer: a review. Expert Rev Anticancer Ther. 2011;11:1163–1179. doi: 10.1586/era.11.80. [DOI] [PubMed] [Google Scholar]
- 135.Gridelli C, Maione P, Rossi A, De Marinis F. The role of bevacizumab in the treatment of non-small cell lung cancer: current indications and future developments. Oncologist. 2007;12:1183–1193. doi: 10.1634/theoncologist.12-10-1183. [DOI] [PubMed] [Google Scholar]
- 136.Enomoto Y, Kenmotsu H, Watanabe N, Baba T, Murakami H, Yoh K, Ogura T, Takahashi T, Goto K, Kato T. Efficacy and Safety of Combined Carboplatin, Paclitaxel, and Bevacizumab for Patients with Advanced Non-squamous Non-small Cell Lung Cancer with Pre-existing Interstitial Lung Disease: A Retrospective Multi-institutional Study. Anticancer Res. 2015;35:4259–4263. [PubMed] [Google Scholar]
- 137.McCormack PL, Keam SJ. Bevacizumab: a review of its use in metastatic colorectal cancer. Drugs. 2008;68:487–506. doi: 10.2165/00003495-200868040-00009. [DOI] [PubMed] [Google Scholar]
- 138.Macedo LT, da Costa Lima AB, Sasse AD. Addition of bevacizumab to first-line chemotherapy in advanced colorectal cancer: a systematic review and meta-analysis, with emphasis on chemotherapy subgroups. BMC Cancer. 2012;12:89. doi: 10.1186/1471-2407-12-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009;27:4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
- 140.Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, Garren N, Mackey M, Butman JA, Camphausen K, Park J, Albert PS, Fine HA. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J. Clin. Oncol. 2009;27:740–745. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Chamberlain MC, Johnston SK. Salvage therapy with single agent bevacizumab for recurrent glioblastoma. J Neurooncol. 2010;96:259–269. doi: 10.1007/s11060-009-9957-6. [DOI] [PubMed] [Google Scholar]
- 142.Raizer JJ, Grimm S, Chamberlain MC, Nicholas MK, Chandler JP, Muro K, Dubner S, Rademaker AW, Renfrow J, Bredel M. A phase 2 trial of single-agent bevacizumab given in an every-3-week schedule for patients with recurrent high-grade gliomas. Cancer. 2010;116:5297–5305. doi: 10.1002/cncr.25462. [DOI] [PubMed] [Google Scholar]
- 143.Vredenburgh JJ, Desjardins A, Herndon JE 2nd, Dowell JM, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Wagner M, Bigner DD, Friedman AH, Friedman HS. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253–1259. doi: 10.1158/1078-0432.CCR-06-2309. [DOI] [PubMed] [Google Scholar]
- 144.Vredenburgh JJ, Desjardins A, Herndon JE 2nd, Marcello J, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Sampson J, Wagner M, Bailey L, Bigner DD, Friedman AH, Friedman HS. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J. Clin. Oncol. 2007;25:4722–4729. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
- 145.Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, Ross GL, Ciampa AS, Ebbeling LG, Levy B, Drappatz J, Kesari S, Wen PY. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70:779–787. doi: 10.1212/01.wnl.0000304121.57857.38. [DOI] [PubMed] [Google Scholar]
- 146.Nghiemphu PL, Liu W, Lee Y, Than T, Graham C, Lai A, Green RM, Pope WB, Liau LM, Mischel PS, Nelson SF, Elashoff R, Cloughesy TF. Bevacizumab and chemotherapy for recurrent glioblastoma: a single-institution experience. Neurology. 2009;72:1217–1222. doi: 10.1212/01.wnl.0000345668.03039.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.McDermott DF, George DJ. Bevacizumab as a treatment option in advanced renal cell carcinoma: an analysis and interpretation of clinical trial data. Cancer Treat Rev. 2010;36:216–223. doi: 10.1016/j.ctrv.2009.12.003. [DOI] [PubMed] [Google Scholar]