Glycosylated antitumor ether lipids: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Mark William Nachtigal.

Recurrent epithelial ovarian cancer (EOC) coincident with chemotherapy resistance remains the main contributor to patient mortality. There is an ongoing investigation to enhance patient progression-free and overall survival with novel chemotherapeutic delivery, such as the utilization of antiangiogenic medications, PARP inhibitors, or immune modulators. OuResearchers' preclinical studies highlight a novel tool to combat chemotherapy-resistant human EOC. Glycosylated antitumor ether lipids (GAELs) are synthetic glycerolipids capable of killing established human epithelial cell lines from a wide variety of human cancers, including EOC cell lines representative of different EOC histotypes. Importantly, GAELs kill high-grade serous ovarian cancer (HGSOC) cells isolated from the ascites of chemotherapy-sensitive and chemotherapy-resistant patients grown as monolayers of spheroid cultures. In addition, GAELs were well tolerated by experimental animals (mice) and were capable of reducing tumor burden and blocking ascites formation in an OVCAR-3 xenograft model. Overall, GAELs show great promise as adjuvant therapy for EOC patients with or without chemotherapy resistance.

  • epithelial ovarian cancer (EOC)
  • chemotherapy resistance
  • methuosis
  • glycosylated antitumor ether lipid

1. Introduction

Epithelial ovarian cancer (EOC) continues to be a lethal malignancy in women worldwide, and resistance to chemotherapy remains one of the main contributors to EOC patient morbidity and mortality [1,2][1][2]. The early disease has a relatively good prognosis, and this fact led to screening and symptomatic-based studies [3,4,5,6,7][3][4][5][6][7] in the hopes of detecting disease earlier and improving outcomes. Unfortunately, none of these studies have proven useful for increasing survival. Other studies examined the role of time-to-diagnosis and time-to-care and once again found no significant benefit for survival [8,9][8][9]. Improved surgical techniques resulting in optimal tumor debulking, improved cytotoxic delivery methods, and novel non-cytotoxic chemotherapeutics are treatment modalities that have contributed to improved progression-free and overall survival in the EOC patient population in the past several decades. Herein, wresearchers review the development and utility of a novel glycolipid compound belonging to the glycosylated antitumor ether lipid (GAEL) class of drugs for the potential treatment of EOCs.
Historically, EOC was treated with multiple different agents until the late 1990s, when several studies confirmed the superiority of cisplatin plus paclitaxel as the basis of EOC chemotherapeutic treatments [10,11][10][11]. Subsequent trials showed the equivalence of carboplatin to cisplatin, with decreased toxicity, leading to the fundamental base of modern cytotoxic therapy for EOC [12,13][12][13]. While the majority of high-grade serous ovarian cancer (HGSOC) patients respond to initial chemotherapy (6–9 cycles of a platinum agent (carboplatin) and a taxane (paclitaxel)), up to 75% of EOC patients will relapse within 18 months with chemotherapy-resistant disease [14,15][14][15]. Moreover, mucinous, clear cell and low-grade serous EOC histotypes are characterized by a poor response to chemotherapy. EOC therapy has continued to develop with changes in surgical approaches [16[16][17][18],17,18], different approaches for dosing of chemotherapy (e.g., dose dense and dose intense) [19[19][20],20], chemotherapy delivery (e.g., intraperitoneal and hyperthermic intraperitoneal) [21[21][22],22], additional adjuvant agents (e.g., Bevacizumab) [23,24,25[23][24][25][26],26], and maintenance therapy [27,28][27][28]. The most promising recent advances in EOC management have surrounded advancements in poly-ADP ribose (PARP) inhibitor therapies, specifically for BRCA1/2 and homologous recombination-deficient (HRD) patients [29,30,31,32,33,34][29][30][31][32][33][34]. Ongoing trials with immune modulators have not yet shown benefits for this patient population [35]. Despite great advances in care, overall survival, although improved slightly, has not yet made drastic changes in several decades [36,37,38,39,40,41][36][37][38][39][40][41]. Thus, additional treatments capable of preventing tumor growth and effectively killing chemoresistant EOC cells are desirable. The connection between translational research and clinical work will be critical in the advancement of gynecologic oncology and cancer therapy.
As part of ouresearchers' ongoing search for novel treatments for human epithelial cancers, weresearchers developed glycosylated antitumor ether lipids (GAELs; Figure 1) and demonstrated that GAELs exhibit anticancer activities in a diverse group of human cancer types in vitro. GAEL compounds are structurally related to antitumor ether lipids (AELs), including edelfosine [42], miltefosine [43], erucylphosphocholine [44] and perifosine [45] that have been tested as anticancer agents in clinical studies. GAELs would fall under the class of cytotoxic rather than targeted agents. GAELs are able to kill chemosensitive and chemoresistant human high-grade serous ovarian cancer (HGSOC) cell lines and patient samples grown as adherent or 3D spheroid cultures [46,47,48,49,50,51,52][46][47][48][49][50][51][52]. Importantly, the GAEL mechanism of action is apoptosis-independent. This characteristic distinguishes GAELs from most of the currently used anticancer drugs that rely on apoptosis to kill tumor cells. Because cancer cells often overcome apoptosis induced by many cytotoxic agents, the addition of GAELs would be an excellent complementary treatment to overcome drug-resistance mechanisms. Moreover, as cellular genetic heterogeneity is observed in EOCs, the ability of GAELs to kill chemotherapy-sensitive and resistant cells would be an additional benefit of including GAELs as adjuvant treatment. This entreviewy introduces how GAELs were developed from observations of the biological activity of lysophosphatidylcholine (LPC), provides a brief overview of structure–activity relationship (SAR) studies, and highlights ourresearchers' studies testing GAEL activity to kill chemotherapy-sensitive and -resistant human EOC cells and the likely mechanism of action contributing to GAEL-induced cell death, namely methuosis. WeResearchers postulate that induction of methuosis in chemotherapy-resistant EOC cells identifies a novel cell death pathway for the further development of new treatments for EOC patients.
Cancers 14 03318 g001
Figure 1.
 Different classes of antitumor ether lipids (AELs); alkyllysophospholipids (ALP), alkylphospholipids (APL), and glycosylated antitumor ether lipids (GAEL).

2. GAELs: How Did We Get Here?

Antitumor ether lipids (AELs) are broadly defined as a group of synthetic ether lipid analogs with antitumor activity. They are comprised of three subclasses including alkyllysophospholipids (ALPs), alkylphospholipids (APLs), and GAELs (Figure 1). The discovery of the antitumor activity of ether lipids originated from studies at the Max-Planck-Institut für Immunobiologie in Freiburg, Germany, in the 1960s. LPC, which is generated from hydrolysis of phosphatidylcholine by phospholipase A2, was found to strongly potentiate the phagocytic activity of peritoneal macrophages in vitro and in vivo [53,54,55][53][54][55]. LPC is not a viable drug because it is rapidly metabolized and inactivated in cells by acyltransferases and lysophospholipases. To overcome this, metabolically stable LPC analogs were synthesized by replacing the acyl group at the sn-1 position with an alkyl group with 12–20 carbons. The sn-2 position was either an OH, H, or a methoxy (OCH3) group [56,57,58][56][57][58]. These ether LPC analogs, the ALPs (Figure 1), demonstrated increased half-life and significantly higher ability to boost antibody production in response to various antigens compared to LPC [59]. Thus, ALPs were initially developed for use as potential immunomodulators. Subsequently, a number of other ether LPC analogs were observed to have cytotoxic activity against a variety of cancer cells in vitro with 1-O-octadecyl-2-O-methyl-glycerophosphocholine (ET-18-OCH3; aka edelfosine) being the most active [60,61,62][60][61][62]. Further testing of ALPs using in vivo mouse and rat tumor models demonstrated antitumor activity by reducing tumor growth and metastasis [63,64][63][64]. While several different pathways contributing to cell death in vitro have been examined, including inhibition of protein kinase C [65[65][66],66], decreasing arachidonate release [66], abrogating phosphatidylcholine biosynthesis [67[67][68],68], and altering cell membranes [69[69][70],70], edelfosine exhibited antitumor activity against a wide range of cancer cells by inducing apoptosis [71,72][71][72]. ALPs have been tested in clinical trials and exhibit high tolerance to reversible toxicities and produce tumor stasis or a decrease in the tumor growth rate [63,64,71,73,74,75,76,77][63][64][71][73][74][75][76][77]. Despite having promising clinical efficacy in vivo, the compounds are not indicated for the treatment of human cancers.
Studies to identify the minimum structure of ALPs required for anticancer activity led to the development of the alkylphospholipids (APLs; Figure 1). These compounds lack the glycerol backbone and consist of an alkylchain esterified to a phosphobase. The prototype of this group is hexadecylphosphocholine (miltefosine) [71,78][71][78]. While miltefosine was initially investigated as an anticancer agent, tests were stopped due to toxic side effects. It is currently used for leishmaniasis and amoeboid infections. Several analogs of APLs, including erucylphosphocholine [79] and perifosine [80,81][80][81], have been synthesized and their antitumor activities characterized [82]. Perifosine was under development to treat human cancers as it blocks PI3K/AKT/mTOR signaling [83]. Perifosine showed considerable promise in early phase clinical trials but failed in Phase III trials for colorectal cancer and refractory multiple myeloma [84]. The mode of action of these compounds appears to be similar to that of the ALPs in killing cells by inducing apoptosis [79,82][79][82].
GAELs were developed by replacing the phosphocholine of edelfosine with a monosaccharide moiety via an O-glycosidic bond [85]. The cancer cell killing activity of the GAEL compounds that were initially synthesized was weak compared to that of edelfosine. Substitution of the O-glycosidic to an S-glycosidic bond to improve lipophilicity did not significantly change the cancer cell-killing characteristics of the GAEL compounds [86,87][86][87]. Subsequent structure–activity relationship (SAR) studies determined that replacing the OH at position 2 of the glucose with an NH2 yielded cancer cell-killing activities that were similar or superior to edelfosine [88]. This discovery opened up the field and led to the development of potent analogs and the further identification and characterization that GAELs were distinct from ALPs and APLs by inducing a non-apoptotic (caspase-independent) form of cell death.

3. GAEL Structure–Activity Relationship

Several GAEL prototypes have been described [50,52,88,89,90,91,92,93][50][52][88][89][90][91][92][93] (Figure 2). The key event to the development of these compounds was the discovery by the Bittman group that the replacement of neutral thio-glucose by a basic 2-amino-β-D-glucose scaffold produced a potent GAEL [β-GLN (1-O-hexadecyl-2-O-methyl-3-O-(2′-amino-2′-deoxy-β-D-glucopyranosyl)-sn-glycerol)] with antiproliferative effects (Figure 2, compound 2). Cell killing activity was tested in various human epithelial cancer cell lines, including A549, MCF-7, A427, and T84, with the greatest sensitivity in HGSOC OVCAR-3 cells [88]. Indeed, compared to edelfosine, β-GLN was five times more effective at killing OVCAR-3 cells [88]. Further experiments showed that acylation of the basic 2-amino group in β-GLN resulted in a 2–3-fold reduction in anticancer activity, indicating that the basic primary 2-amino group is critical for the antitumor effect. Further modification of β-GLN to contain an α-glucosidic linkage of 2-amino-D-glucose (α-GLN; Figure 2, compound 1) displayed two-fold enhanced cell-killing activity compared to β-GLN [91]. By contrast, replacement of the O-glycosidic linkage with S-glycosidic linkage and modifications on the glycero-ether moiety, in addition to guanidinylation of the amino group, resulted in reduced cell-killing effects [91]. In order to increase the metabolic stability of β-GLN, C-glycosidic analogs were generated (Figure 2, compounds 3 and 4) that retained antiproliferative effects [92,93][92][93]. Studies to replace the α-O-glucosidic linkage with an α-O-galactosidic or α-O-mannosidic linkage revealed that both α-O-galactosidic and α-O-glucosidic linkages displayed comparable cell-killing effects while a >4-fold reduction in cell killing activity was observed with the α-O-mannosylated analog [52]. Introducing a second amino group at the 6-position in α-GLN or β-GLN (Figure 2, compounds 5 and 6) enhanced the cell-killing effect. Interestingly, both dibasic analogs displayed potent anticancer stem cell (CSC) activity against BT-474 and DU-145 stem cells with superior activity compared to salinomycin [50]. Dibasic analogs of α-GLN or β-GLN were produced in which D-glucose (D-Glc) was replaced by glycosidase-resistant L-glucose (L-Glc) (Figure 2, compounds 7 and 8) [90]. Moreover, decoration of the dibasic L-Glc scaffold by attachment of a phenyl group further enhanced the cell-killing effect [90]. The incorporation of a third amino group and amphiphilic modulation of the D-Glc scaffold also enhanced the cell-killing effects of GAELs [94]. While first-generation GAELs were synthesized with D-linked-sugars, their potential susceptibility to circulating and cellular glycosidases in vivo led to the development of L-sugar-linked GAEL analogs that demonstrated equivalent or enhanced activity [90]. Humans do not metabolize L-sugars; thus, L-GAELs are predicted to have longer half-lives in vivo. A structure–activity map that summarizes the structural modifications of the antitumor effect is shown in Figure 3.
Figure 2. Structural evolution of GAEL prototypes. 1 = α-GLN, 2 = β-GLN; 3 and 4 are C-glycosidic analogs; 5 and 6 incorporate a second amino group at the 6 position; dibasic analogs 7 and 8 with L-glucose; 9 = 3-amino-1-O-hexadecyloxy-2R-(O–α-L-Rhamnopyranosyl)-sn-glycerol (L-Rham).
Figure 3. GAEL structure–activity relationship map. Data compiled from references [50,52,88,90,91,92,94].
 GAEL structure–activity relationship map. Data compiled from references [50][52][88][90][91][92][94].
One of the challenges to exploiting GAELs for therapeutic applications and animal studies is the requirement for multi-step synthesis resulting in low isolation yields. Synthetic processes required 12 or more synthetic steps. To reduce the number of synthetic steps, a more easily accessible L-rhamnose-based cationic lipid was generated (Figure 2, compound 9; 3-amino-1-O-hexadecyloxy-2R-(O–α-L-rhamnopyranosyl)-sn-glycerol (L-Rham)) [95]. This L-Rham-based GAEL differs from previous GAELs by the glycosidic linkage and glycero backbone; this compound retained cell-killing effects and was found to possess antitumor effects and therefore was used for further in vitro and in vivo studies [49,95][49][95]. GAELs possess many desirable anticancer characteristics that include: (1) killing chemoresistant cell lines and primary HGSOC cells grown as 2D or 3D cultures and causing the disaggregation of 3D spheroids [83,84,92,93][83][84][92][93]; (2) killing human cancer stem cell-enriched fractions from breast and prostate cell lines [83,84,94][83][84][94]; and (3) killing cells via an apoptosis-independent mechanism of action [47,48,50,51,52][47][48][50][51][52]. This latter characteristic distinguishes GAELs from most of the currently used chemotherapeutic drugs that rely on apoptosis to kill HGSOC cells.

References

  1. CCS. Canadian Cancer Statistics 2021. 2021. Available online: https://cdn.cancer.ca/-/media/files/research/cancer-statistics/2021-statistics/2021-pdf-en-final.pdf?rev=2b9d2be7a2d34c1dab6a01c6b0a6a32d&hash=01DE85401DBF0217F8B64F2B7DF43986 (accessed on 14 June 2022).
  2. Global Burden of Disease Cancer Collaboration; Fitzmaurice, C.; Akinyemiju, T.F.; Al Lami, F.H.; Alam, T.; Alizadeh-Navaei, R. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2016: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2018, 4, 1553–1568.
  3. Buys, S.S.; Partridge, E.; Black, A.; Johnson, C.C.; Lamerato, L.; Isaacs, C. Effect of screening on ovarian cancer mortality: The Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Randomized Controlled Trial. JAMA 2011, 305, 2295–2303.
  4. Gilbert, L.; Basso, O.; Sampalis, J.; Karp, I.; Martins, C.; Feng, J. Assessment of symptomatic women for early diagnosis of ovarian cancer: Results from the prospective DOvE pilot project. Lancet Oncol. 2012, 13, 285–291.
  5. Goff, B.A.; Mandel, L.S.; Drescher, C.W.; Urban, N.; Gough, S.; Schurman, K.M. Development of an ovarian cancer symptom index: Possibilities for earlier detection. Cancer 2007, 109, 221–227.
  6. Jacobs, I.J.; Menon, U.; Ryan, A.; Gentry-Maharaj, A.; Burnell, M.; Kalsi, J.K. Ovarian cancer screening and mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A randomised controlled trial. Lancet 2016, 387, 945–956.
  7. Lu, K.H.; Skates, S.; Hernandez, M.A.; Bedi, D.; Bevers, T.; Leeds, L. A 2-stage ovarian cancer screening strategy using the Risk of Ovarian Cancer Algorithm (ROCA) identifies early-stage incident cancers and demonstrates high positive predictive value. Cancer 2013, 119, 3454–3461.
  8. Altman, A.D.; Lambert, P.; Love, A.J.; Turner, D.; Lotocki, R.; Dean, E. Examining the Effects of Time to Diagnosis, Income, Symptoms, and Incidental Detection on Overall Survival in Epithelial Ovarian Cancer: Manitoba Ovarian Cancer Outcomes (MOCO) Study Group. Int. J. Gynecol. Cancer 2017, 27, 1637–1644.
  9. Nagle, C.M.; Francis, J.E.; Nelson, A.E.; Zorbas, H.; Luxford, K.; de Fazio, A. Reducing time to diagnosis does not improve outcomes for women with symptomatic ovarian cancer: A report from the Australian Ovarian Cancer Study Group. J. Clin. Oncol. 2011, 29, 2253–2258.
  10. McGuire, W.P.; Hoskins, W.J.; Brady, M.F.; Kucera, P.R.; Partridge, E.E.; Look, K.Y. 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.
  11. Piccart, M.J.; Bertelsen, K.; James, K.; Cassidy, J.; Mangioni, C.; Simonsen, E. 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.
  12. International Collaborative Ovarian Neoplasm Group. 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.
  13. Ozols, R.F.; Bundy, B.N.; Greer, B.E.; Fowler, J.M.; Clarke-Pearson, D.; Burger, R.A. 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.
  14. Sherman-Baust, C.A.; Becker, K.G.; Wood, W.H., III; Zhang, Y.; Morin, P.J. Gene expression and pathway analysis of ovarian cancer cells selected for resistance to cisplatin, paclitaxel, or doxorubicin. J. Ovarian Res. 2011, 4, 21.
  15. Suh, D.H.; Kim, M.K.; No, J.H.; Chung, H.H.; Song, Y.S. Metabolic approaches to overcoming chemoresistance in ovarian cancer. Ann. N. Y. Acad. Sci. 2011, 1229, 53–60.
  16. Kehoe, S.; Hook, J.; Nankivell, M.; Jayson, G.C.; Kitchener, H.; Lopes, T. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): An open-label, randomised, controlled, non-inferiority trial. Lancet 2015, 386, 249–257.
  17. May, T.; Altman, A.; McGee, J.; Lu, L.; Xu, W.; Lane, K. Examining Survival Outcomes of 852 Women with Advanced Ovarian Cancer: A Multi-institutional Cohort Study. Int. J. Gynecol. Cancer 2018, 28, 925–931.
  18. Vergote, I.; Trope, C.G.; Amant, F.; Kristensen, G.B.; Ehlen, T.; Johnson, N. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 2010, 363, 943–953.
  19. Katsumata, N.; Yasuda, M.; Takahashi, F.; Isonishi, S.; Jobo, T.; Aoki, D. Dose-dense paclitaxel once a week in combination with carboplatin every 3 weeks for advanced ovarian cancer: A phase 3, open-label, randomised controlled trial. Lancet 2009, 374, 1331–1338.
  20. Pignata, S.; Scambia, G.; Katsaros, D.; Gallo, C.; Pujade-Lauraine, E.; De Placido, S. Carboplatin plus paclitaxel once a week versus every 3 weeks in patients with advanced ovarian cancer (MITO-7): A randomised, multicentre, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 396–405.
  21. Armstrong, D.K.; Bundy, B.; Wenzel, L.; Huang, H.Q.; Baergen, R.; Lele, S. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N. Engl. J. Med. 2006, 354, 34–43.
  22. Van Driel, W.J.; Koole, S.N.; Sikorska, K.; Schagen van Leeuwen, J.H.; Schreuder, H.W.R.; Hermans, R.H.M. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. N. Engl. J. Med. 2018, 378, 230–240.
  23. Burger, R.A.; Brady, M.F.; Bookman, M.A.; Fleming, G.F.; Monk, B.J.; Huang, H. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N. Engl. J. Med. 2011, 365, 2473–2483.
  24. Chan, J.K.; Brady, M.F.; Penson, R.T.; Huang, H.; Birrer, M.J.; Walker, J.L. Weekly vs. Every-3-Week Paclitaxel and Carboplatin for Ovarian Cancer. N. Engl. J. Med. 2016, 374, 738–748.
  25. Oza, A.M.; Cook, A.D.; Pfisterer, J.; Embleton, A.; Ledermann, J.A.; Pujade-Lauraine, E. Standard chemotherapy with or without bevacizumab for women with newly diagnosed ovarian cancer (ICON7): Overall survival results of a phase 3 randomised trial. Lancet Oncol. 2015, 16, 928–936.
  26. Walker, J.L.; Brady, M.F.; Wenzel, L.; Fleming, G.F.; Huang, H.Q.; DiSilvestro, P.A. Randomized Trial of Intravenous Versus Intraperitoneal Chemotherapy Plus Bevacizumab in Advanced Ovarian Carcinoma: An NRG Oncology/Gynecologic Oncology Group Study. J. Clin. Oncol. 2019, 37, 1380–1390.
  27. DiSilvestro, P.; Colombo, N.; Harter, P.; Gonzalez-Martin, A.; Ray-Coquard, I.; Coleman, R.L. Maintenance Treatment of Newly Diagnosed Advanced Ovarian Cancer: Time for a Paradigm Shift? Cancers 2021, 13, 5756.
  28. Markman, M.; Liu, P.Y.; Wilczynski, S.; Monk, B.; Copeland, L.J.; Alvarez, R.D. Phase III randomized trial of 12 versus 3 months of maintenance paclitaxel in patients with advanced ovarian cancer after complete response to platinum and paclitaxel-based chemotherapy: A Southwest Oncology Group and Gynecologic Oncology Group trial. J. Clin. Oncol. 2003, 21, 2460–2465.
  29. Banerjee, S.; Moore, K.N.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A. Maintenance olaparib for patients with newly diagnosed advanced ovarian cancer and a BRCA mutation (SOLO1/GOG 3004): 5-year follow-up of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 1721–1731.
  30. Gonzalez-Martin, A.; Pothuri, B.; Vergote, I.; DePont Christensen, R.; Graybill, W.; Mirza, M.R. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2391–2402.
  31. Konstantinopoulos, P.A.; Lheureux, S.; Moore, K.N. PARP Inhibitors for Ovarian Cancer: Current Indications, Future Combinations, and Novel Assets in Development to Target DNA Damage Repair. Am. Soc. Clin. Oncol. Educ. Book 2020, 40, e116–e131.
  32. Ledermann, J.; Harter, P.; Gourley, C.; Friedlander, M.; Vergote, I.; Rustin, G. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 2012, 366, 1382–1392.
  33. Ledermann, J.; Harter, P.; Gourley, C.; Friedlander, M.; Vergote, I.; Rustin, G. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: A preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol. 2014, 15, 852–861.
  34. Ray-Coquard, I.; Pautier, P.; Pignata, S.; Perol, D.; Gonzalez-Martin, A.; Berger, R. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2416–2428.
  35. Leary, A.; Tan, D.; Ledermann, J. Immune checkpoint inhibitors in ovarian cancer: Where do we stand? Ther. Adv. Med. Oncol. 2021, 13, 17588359211039899.
  36. Arnold, M.; Rutherford, M.J.; Bardot, A.; Ferlay, J.; Andersson, T.M.; Myklebust, T.A. Progress in cancer survival, mortality, and incidence in seven high-income countries 1995-2014 (ICBP SURVMARK-2): A population-based study. Lancet Oncol. 2019, 20, 1493–1505.
  37. Cabasag, C.J.; Butler, J.; Arnold, M.; Rutherford, M.; Bardot, A.; Ferlay, J. Exploring variations in ovarian cancer survival by age and stage (ICBP SurvMark-2): A population-based study. Gynecol. Oncol. 2020, 157, 234–244.
  38. Coleman, M.P.; Forman, D.; Bryant, H.; Butler, J.; Rachet, B.; Maringe, C. Cancer survival in Australia, Canada, Denmark, Norway, Sweden, and the UK, 1995–2007 (the International Cancer Benchmarking Partnership): An analysis of population-based cancer registry data. Lancet 2011, 377, 127–138.
  39. Lee, J.Y.; Kim, S.; Kim, Y.T.; Lim, M.C.; Lee, B.; Jung, K.W. Changes in ovarian cancer survival during the 20 years before the era of targeted therapy. BMC Cancer 2018, 18, 601.
  40. Lin, J.J.; Egorova, N.; Franco, R.; Prasad-Hayes, M.; Bickell, N.A. Ovarian Cancer Treatment and Survival Trends Among Women Older Than 65 Years of Age in the United States, 1995–2008. Obstet. Gynecol. 2016, 127, 81–89.
  41. Norell, C.H.; Butler, J.; Farrell, R.; Altman, A.; Bentley, J.; Cabasag, C.J. Exploring international differences in ovarian cancer treatment: A comparison of clinical practice guidelines and patterns of care. Int. J. Gynecol. Cancer 2020, 30, 1748–1756.
  42. Vogler, W.R.; Liu, J.; Volpert, O.; Ades, E.W.; Bouck, N. The anticancer drug edelfosine is a potent inhibitor of neovascularization in vivo. Cancer Investig. 1998, 16, 549–553.
  43. Croft, S.L.; Engel, J. Miltefosine—Discovery of the antileishmanial activity of phospholipid derivatives. Trans. R. Soc. Trop. Med. Hyg. 2006, 100, S4–S8.
  44. Erdlenbruch, B.; Jendrossek, V.; Marx, M.; Hunold, A.; Eibl, H.; Lakomek, M. Antitumor effects of erucylphosphocholine on brain tumor cells in vitro and in vivo. Anticancer Res. 1998, 18, 2551–2557.
  45. Gills, J.J.; Dennis, P.A. Perifosine: Update on a novel Akt inhibitor. Curr. Oncol. Rep. 2009, 11, 102–110.
  46. Arthur, G.; Schweizer, F.; Ogunsina, M. Synthetic glycosylated ether glycerolipids as anticancer agents. In Carbohydrates in Drug Design and Discovery; Jiménez-Barbero, J., Cañada, F.J., Martin-Santamaria, S., Eds.; Royal Society of Chemistry: Cambridge, UK, 2015.
  47. Jahreiss, L.; Renna, M.; Bittman, R.; Arthur, G.; Rubinsztein, D.C. 1-O-Hexadecyl-2-O-methyl-3-O-(2′-acetamido-2′-deoxy-beta-D-glucopyranosyl)-sn-gly cerol (Gln) induces cell death with more autophagosomes which is autophagy-independent. Autophagy 2009, 5, 835–846.
  48. Moraya, A.I.; Ali, J.L.; Samadder, P.; Liang, L.; Morrison, L.C.; Werbowetski-Ogilvie, T.E. Novel glycolipid agents for killing cisplatin-resistant human epithelial ovarian cancer cells. J. Exp. Clin. Cancer Res. 2017, 36, 67.
  49. Nachtigal, M.W.; Musaphir, P.; Dhiman, S.; Altman, A.D.; Schweizer, F.; Arthur, G. Cytotoxic capacity of a novel glycosylated antitumor ether lipid in chemotherapy-resistant high grade serous ovarian cancer in vitro and in vivo. Transl. Oncol. 2021, 14, 101203.
  50. Ogunsina, M.; Samadder, P.; Idowu, T.; Arthur, G.; Schweizer, F. Design, synthesis and evaluation of cytotoxic properties of bisamino glucosylated antitumor ether lipids against cancer cells and cancer stem cells. Med. Chem. Comm. 2016, 7, 2100–2110.
  51. Samadder, P.; Bittman, R.; Byun, H.S.; Arthur, G. A glycosylated antitumor ether lipid kills cells via paraptosis-like cell death. Biochem. Cell Biol. 2009, 87, 401–414.
  52. Samadder, P.; Xu, Y.Z.; Schweizer, F.; Arthur, G. Cytotoxic properties of D-gluco-, D-galacto- and D-manno-configured 2-amino-2-deoxy-glycerolipids against epithelial cancer cell lines and BT-474 breast cancer stem cells. Eur. J. Med. Chem. 2014, 78, 225–235.
  53. Burdzy, K.; Munder, P.G.; Fischer, H.; Westphal, O. Increase in the Phagocytosis of Peritoneal Macrophages by Lysolecithin. Z. Naturforsch. B. 1964, 19, 1118–1120.
  54. Fischer, H. Lysolecithin and the Action of Complement. Ann. N. Y. Acad. Sci. 1964, 116, 1063–1070.
  55. Munder, P.G.; Modolell, M.; Ferber, E.; Fischer, H. Phospholipids in quartz-damaged macrophages. Biochem. Z. 1966, 344, 310–313.
  56. Arnold, D.; Weltzien, H.U.; Westphal, O. Concerning the synthesis of lysolecithin and its ether analogs. Justus Liebigs Ann. Chem. 1967, 709, 234–239.
  57. Eibl, H.; Westphal, O. Palmitoyl-propandiol-(1.3)-phosphorylcholine (2-desoxylysolecithin) and omega.omega’-alkanediol-analogs. Justus Liebigs Ann. Chem. 1967, 709, 244–245.
  58. Weltzien, H.U.; Westphal, O. O-methylated and O-acetylated lysolecithin. Justus Liebigs Ann. Chem. 1967, 709, 240–243.
  59. Munder, P.G.; Modolell, M.; Andreesen, R.; Weltzien, H.U.; Westphal, O. Lysophosphatidylcholine (Lysolecithin) and its Synthetic Analogues. Immunomodulating and Other Biologic Effects. In Immunostimulation; Chedid, L., Miescher, P.A., Mueller-Eberhard, H.J., Eds.; Springer: Berlin/Heidelberg, Germany, 1980.
  60. Andreesen, R.; Modolell, M.; Weltzien, H.U.; Eibl, H.; Common, H.H.; Lohr, G.W. Selective destruction of human leukemic cells by alkyl-lysophospholipids. Cancer Res. 1978, 38, 3894–3899.
  61. Modolell, M.; Andreesen, R.; Pahlke, W.; Brugger, U.; Munder, P.G. Disturbance of phospholipid metabolism during the selective destruction of tumor cells induced by alkyl-lysophospholipids. Cancer Res. 1979, 39, 4681–4686.
  62. Tarnowski, G.S.; Mountain, I.M.; Stock, C.C.; Munder, P.G.; Weltzien, H.U.; Westphal, O. Effect of lysolecithin and analogs on mouse ascites tumors. Cancer Res. 1978, 38, 339–344.
  63. Berdel, W.E.; Bausert, W.R.; Weltzien, H.U.; Modolell, M.L.; Widmann, K.H.; Munder, P.G. The influence of alkyl-lysophospholipids and lysophospholipid-activated macrophages on the development of metastasis of 3-Lewis lung carcinoma. Eur. J. Cancer 1980, 16, 1199–1204.
  64. Berger, M.R.; Munder, P.G.; Schmahl, D.; Westphal, O. Influence of the alkyllysophospholipid ET-18-OCH3 on methylnitrosourea-induced rat mammary carcinomas. Oncology 1984, 41, 109–113.
  65. Helfman, D.M.; Barnes, K.C.; Kinkade, J.M.; Vogler, W.R.; Shoji, M.; Kuo, J.F. Phospholipid-Sensitive Ca-2+-Dependent Protein-Phosphorylation System in Various Types of Leukemic-Cells from Human Patients and in Human-Leukemic Cell Line-Hl60 and Line-K562, and Its Inhibition by Alkyl-Lysophospholipid. Cancer Res. 1983, 43, 2955–2961.
  66. Parker, J.; Daniel, L.W.; Waite, M. Evidence of Protein-Kinase-C Involvement in Phorbol Diester-Stimulated Arachidonic-Acid Release and Prostaglandin Synthesis. J. Biol. Chem. 1987, 262, 5385–5393.
  67. Daniel, L.W.; Etkin, L.A.; Morrison, B.T.; Parker, J.; Morris-Natschke, S.; Surles, J.R. Ether lipids inhibit the effects of phorbol diester tumor promoters. Lipids 1987, 22, 851–855.
  68. Vogler, W.R.; Whigham, E.; Bennett, W.D.; Olson, A.C. Effect of alkyl-lysophospholipids on phosphatidylcholine biosynthesis in leukemic cell lines. Exp. Hematol. 1985, 13, 629–633.
  69. Berdel, W.E.; Fromm, M.; Fink, U.; Pahlke, W.; Bicker, U.; Reichert, A. Cytotoxicity of thioether-lysophospholipids in leukemias and tumors of human origin. Cancer Res. 1983, 43, 5538–5543.
  70. Storme, G.A.; Berdel, W.E.; van Blitterswijk, W.J.; Bruyneel, E.A.; De Bruyne, G.K.; Mareel, M.M. Antiinvasive effect of racemic 1-O-octadecyl-2-O-methylglycero-3-phosphocholine on MO4 mouse fibrosarcoma cells in vitro. Cancer Res. 1985, 45, 351–357.
  71. Houlihan, W.J.; Lohmeyer, M.; Workman, P.; Cheon, S.H. Phospholipid antitumor agents. Med. Res. Rev. 1995, 15, 157–223.
  72. Smets, L.A.; Van Rooij, H.; Salomons, G.S. Signalling steps in apoptosis by ether lipids. Apoptosis 1999, 4, 419–427.
  73. Berdel, W.E.; Fink, U.; Rastetter, J. Clinical phase I pilot study of the alkyl lysophospholipid derivative ET-18-OCH3. Lipids 1987, 22, 967–969.
  74. Drings, P.; Günther, I.; Gatzemeier, U.; Ulbrich, F.; Khanavkar, B.; Schreml, W. Final Evaluation of a Phase II Study on the Effect of Edelfosine (an Ether Lipid) in Advanced Non-Small-Cell Bronchogenic Carcinoma. Onkologie 1992, 15, 375–382.
  75. Gajate, C.; Mollinedo, F. Biological activities, mechanisms of action and biomedical prospect of the antitumor ether phospholipid ET-18-OCH(3) (edelfosine), a proapoptotic agent in tumor cells. Curr. Drug Metab. 2002, 3, 491–525.
  76. Herrmann, D.B.; Neumann, H.A.; Berdel, W.E.; Heim, M.E.; Fromm, M.; Boerner, D. Phase I trial of the thioether phospholipid analogue BM 41.440 in cancer patients. Lipids 1987, 22, 962–966.
  77. Hilgard, P.; Stekar, J.; Voegeli, R.; Harleman, J.H. Experimental therapeutic studies with miltefosine in rats and mice. Prog. Exp. Tumor Res. 1992, 34, 116–130.
  78. Unger, C.; Eibl, H. Hexadecylphosphocholine: Preclinical and the first clinical results of a new antitumor drug. Lipids 1991, 26, 1412–1417.
  79. Jendrossek, V.; Erdlenbruch, B.; Hunold, A.; Kugler, W.; Eibl, H.; Lakomek, M. Erucylphosphocholine, a novel antineoplastic ether lipid, blocks growth and induces apoptosis in brain tumor cell lines in vitro. Int. J. Oncol. 1999, 14, 15–22.
  80. Kondapaka, S.B.; Singh, S.S.; Dasmahapatra, G.P.; Sausville, E.A.; Roy, K.K. Perifosine, a novel alkylphospholipid, inhibits protein kinase B activation. Mol. Cancer Ther. 2003, 2, 1093–1103.
  81. Richardson, P.G.; Eng, C.; Kolesar, J.; Hideshima, T.; Anderson, K.C. Perifosine, an oral, anti-cancer agent and inhibitor of the Akt pathway: Mechanistic actions, pharmacodynamics, pharmacokinetics, and clinical activity. Expert Opin. Drug Metab. Toxicol. 2012, 8, 623–633.
  82. Van Blitterswijk, W.J.; Verheij, M. Anticancer mechanisms and clinical application of alkylphospholipids. Biochim. Biophys. Acta 2013, 1831, 663–674.
  83. Keane, N.A.; Glavey, S.V.; Krawczyk, J.; O’Dwyer, M. AKT as a therapeutic target in multiple myeloma. Expert Opin. Ther. Targets 2014, 18, 897–915.
  84. Zentaris, A. Assessment of Efficacy and Safety of Perifosine, Bortezomib and Dexamethasone in Multiple Myeloma Patients. 2013. Available online: https://www.clinicaltrials.gov/ct2/show/NCT01002248 (accessed on 14 June 2022).
  85. Weber, N.; Benning, H. Synthesis of Ether Glyceroglycolipids. Chem. Phys. Lipids 1986, 41, 93–100.
  86. Guivisdalsky, P.N.; Bittman, R.; Smith, Z.; Blank, M.L.; Snyder, F.; Howard, S. Synthesis and antineoplastic properties of ether-linked thioglycolipids. J. Med. Chem. 1990, 33, 2614–2621.
  87. Lu, X.L.; Rengan, K.; Bittman, R.; Arthur, G. The Alpha-Anomers and Beta-Anomers of 1-O-Hexadecyl-2-O-Methyl-3-S-Thioglucosyl-Sn-Glycerol Inhibit the Proliferation of Epithelial Cancer Cell-Lines. Oncol. Rep. 1994, 1, 933–936.
  88. Erukulla, R.K.; Zhou, X.; Samadder, P.; Arthur, G.; Bittman, R. Synthesis and evaluation of the antiproliferative effects of 1-O-hexadecyl-2-O-methyl-3-O-(2′-acetamido-2′-deoxy-beta-D-glucopyranosyl)-sn-glycerol and 1-O-hexadecyl-2-O-methyl-3-0-(2′-amino-2′-deoxy-beta-D-glucopyranosyl)-sn-glycerol on epithelial cancer cell growth. J. Med. Chem. 1996, 39, 1545–1548.
  89. Idowu, T.; Samadder, P.; Arthur, G.; Schweizer, F. Design, synthesis and antitumor properties of glycosylated antitumor ether lipid (GAEL)- chlorambucil-hybrids. Chem. Phys. Lipids 2016, 194, 139–148.
  90. Ogunsina, M.; Samadder, P.; Idowu, T.; Arthur, G.; Schweizer, F. Replacing D-Glucosamine with Its L-Enantiomer in Glycosylated Antitumor Ether Lipids (GAELs) Retains Cytotoxic Effects against Epithelial Cancer Cells and Cancer Stem Cells. J. Med. Chem. 2017, 60, 2142–2147.
  91. Xu, Y.Z.; Ogunsina, M.; Samadder, P.; Arthur, G.; Schweizer, F. StructureActivity Relationships of Glucosamine-Derived Glycerolipids: The Role of the Anomeric Linkage, the Cationic Charge and the Glycero Moiety on the Antitumor Activity. ChemMedChem 2013, 8, 511–520.
  92. Yang, G.L.; Franck, R.W.; Bittman, R.; Samadder, P.; Arthur, G. Synthesis and growth inhibitory properties of glucosamine-derived glycerolipids. Org. Lett. 2001, 3, 197–200.
  93. Yang, G.L.; Franck, R.W.; Byun, H.S.; Bittman, R.; Samadder, P.; Arthur, G. Convergent C-glycolipid synthesis via the Ramberg-Backlund reaction: Active antiproliferative glycolipids. Org. Lett. 1999, 1, 2149–2151.
  94. Idowu, T.; Samadder, P.; Arthur, G.; Schweizer, F. Amphiphilic Modulation of Glycosylated Antitumor Ether Lipids Results in a Potent Triamino Scaffold against Epithelial Cancer Cell Lines and BT474 Cancer Stem Cells. J. Med. Chem. 2017, 60, 9724–9738.
  95. Ogunsina, M.; Samadder, P.; Idowu, T.; Nachtigal, M.W.; Schweizer, F.; Arthur, G. Syntheses of L-Rhamnose linked amino glycerolipids and their cytotoxic activities against human cancer cells. Molecules 2020, 25, 566.
More