The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities
Abstract
:1. Introduction
1.1. OC Biology and Subtypes
1.2. Classification and Histopathology
1.3. Diagnosis and Standard of Care
2. The TME
3. Tumor Vasculature
Tumor Metabolism
4. Stromal Compartment
5. Immune Environment in Ovarian Tumors
Immune Cells in the TME
6. Barriers in the TME That Inhibit Therapeutic Success
7. Remodeling the TME to Enhance Therapeutic Efficacy
8. Conclusions and Future Directions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Peres, L.C.; Cushing-Haugen, K.L.; Kobel, M.; Harris, H.R.; Berchuck, A.; Rossing, M.A.; Schildkraut, J.M.; A Doherty, J. Invasive Epithelial Ovarian Cancer Survival by Histotype and Disease Stage. J. Natl. Cancer Inst. 2019, 111, 60–68. [Google Scholar] [CrossRef] [PubMed]
- Jayson, G.C.; Kohn, E.C.; Kitchener, H.C.; Ledermann, J.A. Ovarian cancer. Lancet 2014, 384, 1376–1388. [Google Scholar] [CrossRef] [PubMed]
- Arora, T.; Mullangi, S.; Lekkala, M.R. Ovarian Cancer. In StatPearls; Ineligible Companies: Treasure Island, FL, USA, 2024. [Google Scholar]
- Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef] [PubMed]
- Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2019, 6, 160. [Google Scholar] [CrossRef] [PubMed]
- Hynes, R.O.; Naba, A. Overview of the matrisome—An inventory of extracellular matrix constituents and functions. Cold Spring Harb. Perspect. Biol. 2012, 4, a004903. [Google Scholar] [CrossRef] [PubMed]
- Walker, C.; Mojares, E.; del Río Hernández, A. Role of Extracellular Matrix in Development and Cancer Progression. Int. J. Mol. Sci. 2018, 19, 3028. [Google Scholar] [CrossRef] [PubMed]
- Radisky, E.S. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J. Biol. Chem. 2024, 300, 107347. [Google Scholar] [CrossRef] [PubMed]
- Amos, S.E.; Choi, Y.S. The Cancer Microenvironment: Mechanical Challenges of the Metastatic Cascade. Front. Bioeng. Biotechnol. 2021, 9, 625859. [Google Scholar] [CrossRef] [PubMed]
- Sleeboom, J.J.F.; van Tienderen, G.S.; Schenke-Layland, K.; van der Laan, L.J.W.; Khalil, A.A.; Verstegen, M.M.A. The extracellular matrix as hallmark of cancer and metastasis: From biomechanics to therapeutic targets. Sci. Transl. Med. 2024, 16, eadg3840. [Google Scholar] [CrossRef]
- Lampi, M.C.; Reinhart-King, C.A. Targeting extracellular matrix stiffness to attenuate disease: From molecular mechanisms to clinical trials. Sci. Transl. Med. 2018, 10, eaao0475. [Google Scholar] [CrossRef]
- Labiche, A.; Heutte, N.; Herlin, P.; Chasle, J.; Gauduchon, P.; Elie, N. Stromal compartment as a survival prognostic factor in advanced ovarian carcinoma. Int. J. Gynecol. Cancer 2010, 20, 28–33. [Google Scholar] [CrossRef] [PubMed]
- Joshi, R.S.; Kanugula, S.S.; Sudhir, S.; Pereira, M.P.; Jain, S.; Aghi, M.K. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers 2021, 13, 1399. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, G.M.; Galpin, K.J.C.; McCloskey, C.W.; Vanderhyden, B.C. The Tumor Microenvironment of Epithelial Ovarian Cancer and Its Influence on Response to Immunotherapy. Cancers 2018, 10, 242. [Google Scholar] [CrossRef] [PubMed]
- Xing, F.; Saidou, J.; Watabe, K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. Landmark Ed. 2010, 15, 166–179. [Google Scholar] [CrossRef] [PubMed]
- Desmoulière, A.; Redard, M.; Darby, I.; Gabbiani, G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. Am. J. Pathol. 1995, 146, 56–66. [Google Scholar] [PubMed]
- Bremnes, R.M.; Dønnem, T.; Al-Saad, S.; Al-Shibli, K.; Andersen, S.; Sirera, R.; Camps, C.; Marinez, I.; Busund, L.-T. The role of tumor stroma in cancer progression and prognosis: Emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J. Thorac. Oncol. 2011, 6, 209–217. [Google Scholar] [CrossRef] [PubMed]
- Helms, E.; Onate, M.K.; Sherman, M.H. Fibroblast Heterogeneity in the Pancreatic Tumor Microenvironment. Cancer Discov. 2020, 10, 648–656. [Google Scholar] [CrossRef] [PubMed]
- Davidson, B.; Trope, C.G.; Reich, R. The role of the tumor stroma in ovarian cancer. Front. Oncol. 2014, 4, 104. [Google Scholar] [CrossRef] [PubMed]
- Kamat, A.A.; Fletcher, M.; Gruman, L.M.; Mueller, P.; Lopez, A.; Landen, C.N., Jr.; Han, L.; Gershenson, D.M.; Sood, A.K. The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin. Cancer Res. 2006, 12, 1707–1714. [Google Scholar] [CrossRef]
- Luo, Z.; Wang, Q.; Lau, W.B.; Lau, B.; Xu, L.; Zhao, L.; Yang, H.; Feng, M.; Xuan, Y.; Yang, Y.; et al. Tumor microenvironment: The culprit for ovarian cancer metastasis? Cancer Lett. 2016, 377, 174–182. [Google Scholar] [CrossRef]
- Bamberger, E.S.; Perrett, C.W. Angiogenesis in epithelian ovarian cancer. Mol. Pathol. 2002, 55, 348–359. [Google Scholar] [CrossRef]
- Teleanu, R.I.; Chircov, C.; Grumezescu, A.M.; Teleanu, D.M. Tumor Angiogenesis and Anti-Angiogenic Strategies for Cancer Treatment. J. Clin. Med. 2019, 9, 84. [Google Scholar] [CrossRef] [PubMed]
- Joyce, J.A.; Baruch, A.; Chehade, K.; Meyer-Morse, N.; Giraudo, E.; Tsai, F.Y.; Greenbaum, D.C.; Hager, J.H.; Bogyo, M.; Hanahan, D. Cathepsin cysteine proteases are effectors of invasive growth and angiogenesis during multistage tumorigenesis. Cancer Cell 2004, 5, 443–453. [Google Scholar] [CrossRef] [PubMed]
- Deng, T.; Lu, X.; Jia, X.; Du, J.; Wang, L.; Cao, B.; Yang, M.; Yin, Y.; Liu, F. Cathepsins and cancer risk: A Mendelian randomization study. Front. Endocrinol. 2024, 15, 1428433. [Google Scholar] [CrossRef] [PubMed]
- Pranjol, M.Z.I.; Gutowski, N.J.; Hannemann, M.; Whatmore, J.L. Cathepsin L Induces Proangiogenic Changes in Human Omental Microvascular Endothelial Cells via Activation of the ERK1/2 Pathway. Curr. Cancer Drug Targets 2019, 19, 231–242. [Google Scholar] [CrossRef] [PubMed]
- Pranjol, Z.I.; Zinovkin, D.A.; Maskell, A.R.T.; Stephens, L.J.; Achinovich, S.L.; Los’, D.M.; Nadyrov, E.A.; Hannemann, M.; Gutowski, N.J.; Whatmore, J.L. Cathepsin L-induced galectin-1 may act as a proangiogenic factor in the metastasis of high-grade serous carcinoma. J. Transl. Med. 2019, 17, 216. [Google Scholar] [CrossRef] [PubMed]
- Hu, L.; Roth, J.M.; Brooks, P.; Luty, J.; Karpatkin, S. Thrombin up-regulates cathepsin D which enhances angio-genesis, growth, and metastasis. Cancer Res. 2008, 68, 4666–4673. [Google Scholar] [CrossRef] [PubMed]
- Briozzo, P.; Badet, J.; Capony, F.; Pieri, I.; Montcourrier, P.; Barritault, D.; Rochefort, H. MCF7 mammary cancer cells respond to bFGF and internalize it following its release from extracellular matrix: A permissive role of cathepsin D. Exp. Cell Res. 1991, 194, 252–259. [Google Scholar] [CrossRef] [PubMed]
- Mustafa, A.; Elkhamisy, F.; Arghiani, N.; Pranjol, M.Z.I. Potential crosstalk between pericytes and cathepsins in the tumour microenvironment. Biomed. Pharmacother. 2023, 164, 114932. [Google Scholar] [CrossRef]
- Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef]
- Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
- Attane, C.; Muller, C. Drilling for Oil: Tumor-Surrounding Adipocytes Fueling Cancer. Trends Cancer 2020, 6, 593–604. [Google Scholar] [CrossRef] [PubMed]
- Kimura, H.; Braun, R.D.; Ong, E.T.; Hsu, R.; Secomb, T.W.; Papahadjopoulos, D.; Hong, K.; Dewhirst, M.W. Fluctuations in red cell flux in tumor microvessels can lead to transient hypoxia and reoxygenation in tumor parenchyma. Cancer Res. 1996, 56, 5522–5528. [Google Scholar] [PubMed]
- Hashizume, H.; Baluk, P.; Morikawa, S.; McLean, J.W.; Thurston, G.; Roberge, S.; Jain, R.K.; McDonald, D.M. Openings between defective endothelial cells explain tumor vessel leakiness. Am. J. Pathol. 2000, 156, 1363–1380. [Google Scholar] [CrossRef] [PubMed]
- Yu, P.; Wang, Y.; Yuan, D.; Sun, Y.; Qin, S.; Li, T. Vascular normalization: Reshaping the tumor microenvironment and augmenting antitumor immunity for ovarian cancer. Front. Immunol. 2023, 14, 1276694. [Google Scholar] [CrossRef] [PubMed]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic require-ments of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Liu, N. Noncoding RNAs in the Glycolysis of Ovarian Cancer. Front. Pharmacol. 2022, 13, 855488. [Google Scholar] [CrossRef]
- Bogdanov, A.; Bogdanov, A.; Chubenko, V.; Volkov, N.; Moiseenko, F.; Moiseyenko, V. Tumor acidity: From hallmark of cancer to target of treatment. Front. Oncol. 2022, 12, 979154. [Google Scholar] [CrossRef]
- Andreucci, E.; Peppicelli, S.; Ruzzolini, J.; Bianchini, F.; Biagioni, A.; Papucci, L.; Magnelli, L.; Mazzanti, B.; Stecca, B.; Calorini, L. The acidic tumor microenvironment drives a stem-like phenotype in melanoma cells. J. Mol. Med. 2020, 98, 1431–1446. [Google Scholar] [CrossRef]
- Da Silva, A.C.; Jammal, M.P.; Crispim, P.C.A.; Murta, E.F.C.; Nomelini, R.S. The Role of Stroma in Ovarian Cancer. Immunol. Investig. 2020, 49, 406–424. [Google Scholar] [CrossRef]
- Nowak, M.; Klink, M. The Role of Tumor-Associated Macrophages in the Progression and Chemoresistance of Ovarian Cancer. Cells 2020, 9, 1299. [Google Scholar] [CrossRef] [PubMed]
- Solinas, G.; Germano, G.; Mantovani, A.; Allavena, P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J. Leukoc. Biol. 2009, 86, 1065–1073. [Google Scholar] [CrossRef] [PubMed]
- Larionova, I.; Cherdyntseva, N.; Liu, T.; Patysheva, M.; Rakina, M.; Kzhyshkowska, J. Interaction of tu-mor-associated macrophages and cancer chemotherapy. Oncoimmunology 2019, 8, 1596004. [Google Scholar] [CrossRef] [PubMed]
- Plikus, M.V.; Wang, X.; Sinha, S.; Forte, E.; Thompson, S.M.; Herzog, E.L.; Driskell, R.R.; Rosenthal, N.; Biernaskie, J.; Horsley, V. Fibroblasts: Origins, definitions, and functions in health and disease. Cell 2021, 184, 3852–3872. [Google Scholar] [CrossRef] [PubMed]
- Cirri, P.; Chiarugi, P. Cancer associated fibroblasts: The dark side of the coin. Am. J. Cancer Res. 2011, 1, 482–497. [Google Scholar] [PubMed]
- Wright, K.; Ly, T.; Kriet, M.; Czirok, A.; Thomas, S.M. Cancer-Associated Fibroblasts: Master Tumor Microenvironment Modifiers. Cancers 2023, 15, 1899. [Google Scholar] [CrossRef] [PubMed]
- Sun, W.; Fu, S. Role of cancer-associated fibroblasts in tumor structure, composition and the microenvironment in ovarian cancer. Oncol. Lett. 2019, 18, 2173–2178. [Google Scholar] [CrossRef] [PubMed]
- Fujisawa, M.; Moh-Moh-Aung, A.; Zeng, Z.; Yoshimura, T.; Wani, Y.; Matsukawa, A. Ovarian stromal cells as a source of cancer-associated fibroblasts in human epithelial ovarian cancer: A histopathological study. PLoS ONE 2018, 13, e0205494. [Google Scholar] [CrossRef]
- Barrett, R.L.; Pure, E. Cancer-associated fibroblasts an their influence on tumor immunity and immunotherapy. Elife 2020, 9, e57243. [Google Scholar] [CrossRef]
- Elyada, E.; Bolisetty, M.; Laise, P.; Flynn, W.F.; Courtois, E.T.; Burkhart, R.A.; Teinor, J.A.; Belleau, P.; Biffi, G.; Lucito, M.S.; et al. Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov. 2019, 9, 1102–1123. [Google Scholar] [CrossRef]
- Zheng, X.; Turkowski, K.; Mora, J.; Brüne, B.; Seeger, W.; Weigert, A.; Savai, R. Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2017, 8, 48436–48452. [Google Scholar] [CrossRef] [PubMed]
- Comito, G.; Giannoni, E.; Segura, C.P.; Barcellos-De-Souza, P.; Raspollini, M.R.; Baroni, G.; Lanciotti, M.; Serni, S.; Chiarugi, P. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene 2014, 33, 2423–2431. [Google Scholar] [CrossRef] [PubMed]
- Monteran, L.; Erez, N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front. Immunol. 2019, 10, 1835. [Google Scholar] [CrossRef] [PubMed]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef] [PubMed]
- Knutson, K.L.; Lu, H.; Stone, B.; Reiman, J.M.; Behrens, M.D.; Prosperi, C.M.; Gad, E.A.; Smorlesi, A.; Disis, M.L. Immunoediting of Cancers May Lead to Epithelial to Mesenchymal Transition. Front. Immunol. 2006, 177, 1526–1533. [Google Scholar] [CrossRef]
- Garnier, L.; Gkountidi, A.-O.; Hugues, S. Tumor-Associated Lymphatic Vessel Features and Immunomodulatory Functions. Front. Immunol. 2019, 10, 720. [Google Scholar] [CrossRef]
- Koukourakis, M.I.; Giatromanolaki, A. Tumor draining lymph nodes, immune response, and radiotherapy: Towards a revisal of therapeutic principles. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188704. [Google Scholar] [CrossRef]
- Stylianopoulos, T.; Martin, J.D.; Snuderl, M.; Mpekris, F.; Jain, S.R.; Jain, R.K. Coevolution of solid stress and in-terstitial fluid pressure in tumors during progression: Implications for vascular collapse. Cancer Res. 2013, 73, 3833–3841. [Google Scholar] [CrossRef]
- Fucikova, J.; Hensler, M.; Kasikova, L.; Lanickova, T.; Pasulka, J.; Rakova, J.; Drozenova, J.; Fredriksen, T.; Hraska, M.; Hrnciarova, T.; et al. An Autologous Dendritic Cell Vaccine Promotes Anticancer Immunity in Patients with Ovarian Cancer with Low Mutational Burden and Cold Tumors. Clin. Cancer Res. 2022, 28, 3053–3065. [Google Scholar] [CrossRef]
- Fanale, D.; Dimino, A.; Pedone, E.; Brando, C.; Corsini, L.R.; Filorizzo, C.; Fiorino, A.; Lisanti, M.C.; Magrin, L.; Randazzo, U.; et al. Prognostic and Predictive Role of Tumor-Infiltrating Lymphocytes (TILs) in Ovarian Cancer. Cancers 2022, 14, 4344. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.W.Y.; Dand, S.; Doig, L.; Papenfuss, A.T.; Scott, C.L.; Ho, G.; Ooi, J.D. T-Cell Receptor Therapy in the Treatment of Ovarian Cancer: A Mini Review. Front. Immunol. 2021, 12, 672502. [Google Scholar] [CrossRef] [PubMed]
- Tang, S.; Ning, Q.; Yang, L.; Mo, Z.; Tang, S. Mechanisms of immune escape in the cancer immune cycle. Int. Immunopharmacol. 2020, 86, 106700. [Google Scholar] [CrossRef] [PubMed]
- Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2015; Volume 35. [Google Scholar]
- Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef] [PubMed]
- Wei, C.; Liu, X.; Wang, Q.; Li, Q.; Xie, M. Identification of Hypoxia Signature to Assess the Tumor Immune Microenvironment and Predict Prognosis in Patients with Ovarian Cancer. Int. J. Endocrinol. 2021, 2021, 4156187. [Google Scholar] [CrossRef] [PubMed]
- Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [PubMed]
- Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef]
- Yu, J.; Zhuang, A.; Gu, X.; Hua, Y.; Yang, L.; Ge, S.; Ruan, J.; Chai, P.; Jia, R.; Fan, X. Nuclear PD-L1 promotes EGR1-mediated angiogenesis and accelerates tumorigenesis. Cell Discov. 2023, 9, 33. [Google Scholar] [CrossRef] [PubMed]
- Yu, W.; Hua, Y.; Qiu, H.; Hao, J.; Zou, K.; Li, Z.; Hu, S.; Guo, P.; Chen, M.; Sui, S.; et al. PD-L1 promotes tumor growth and progression by activating WIP and beta-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis. 2020, 11, 506. [Google Scholar] [CrossRef]
- Barsoum, I.B.; Smallwood, C.A.; Siemens, D.R.; Graham, C.H. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014, 74, 665–674. [Google Scholar] [CrossRef]
- Koh, Y.W.; Lee, S.J.; Han, J.H.; Haam, S.; Jung, J.; Lee, H.W. PD-L1 protein expression in non-small-cell lung cancer and its relationship with the hypoxia-related signaling pathways: A study based on immunohistochemistry and RNA sequencing data. Lung Cancer 2019, 129, 41–47. [Google Scholar] [CrossRef]
- Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef] [PubMed]
- Kryczek, I.; Wei, S.; Zhu, G.; Myers, L.; Mottram, P.; Cheng, P.; Chen, L.; Coukos, G.; Zou, W. Relationship between B7-H4, regulatory T cells, and patient outcome in human ovarian carcinoma. Cancer Res. 2007, 67, 8900–8905. [Google Scholar] [CrossRef]
- Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
- Dufait, I.; Pardo, J.; Escors, D.; De Vlaeminck, Y.; Jiang, H.; Keyaerts, M.; De Ridder, M.; Breckpot, K. Perforin and Granzyme B Expressed by Murine Myeloid-Derived Suppressor Cells: A Study on Their Role in Outgrowth of Cancer Cells. Cancers 2019, 11, 808. [Google Scholar] [CrossRef] [PubMed]
- Saleh, R.; Elkord, E. FoxP3+ T regulatory cells in cancer: Prognostic biomarkers and therapeutic targets. Cancer Lett. 2020, 490, 174–185. [Google Scholar] [CrossRef]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Y.; Li, B.; Liang, Y.; Reeves, P.M.; Qu, X.; Ran, C.; Liu, Q.; Callahan, M.V.; Sluder, A.E.; Gelfand, J.A.; et al. Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. 2019, 33, 6596–6608. [Google Scholar] [CrossRef]
- Chou, F.-C.; Chen, H.-Y.; Kuo, C.-C.; Sytwu, H.-K. Role of Galectins in Tumors and in Clinical Immunotherapy. Int. J. Mol. Sci. 2018, 19, 430. [Google Scholar] [CrossRef]
- Astorgues-Xerri, L.; Riveiro, M.E.; Tijeras-Raballand, A.; Serova, M.; Neuzillet, C.; Albert, S.; Raymond, E.; Faivre, S. Unraveling galectin-1 as a novel therapeutic target for cancer. Cancer Treat. Rev. 2014, 40, 307–319. [Google Scholar] [CrossRef]
- Cagnoni, A.J.; Giribaldi, M.L.; Blidner, A.G.; Cutine, A.M.; Gatto, S.G.; Morales, R.M.; Salatino, M.; Abba, M.C.; Croci, D.O.; Mariño, K.V.; et al. Galectin-1 fosters an immunosuppressive microenvironment in colorectal cancer by reprogramming CD8+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2021, 118, e2102950118. [Google Scholar] [CrossRef] [PubMed]
- Yakushina, V.D.; Vasil’eva, O.A.; Ryazantseva, N.V.; Novitsky, V.V.; Tashireva, L.A. The effects of galectin-1 on the gene expression of the transcription factors TBX21, GATA-3, FOXP3 and RORC. Mol. Cell. Biochem. 2015, 398, 245–249. [Google Scholar] [CrossRef] [PubMed]
- Schweer, D.; McAtee, A.; Neupane, K.; Richards, C.; Ueland, F.; Kolesar, J. Tumor-Associated Macrophages and Ovarian Cancer: Implications for Therapy. Cancers 2022, 14, 2220. [Google Scholar] [CrossRef] [PubMed]
- Cheng, H.; Wang, Z.; Fu, L.; Xu, T. Macrophage Polarization in the Development and Progression of Ovarian Cancers: An Overview. Front. Oncol. 2019, 9, 421. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Cai, D.-J.; Li, B. Ovarian cancer stem-like cells elicit the polarization of M2 macrophages. Mol. Med. Rep. 2015, 11, 4685–4693. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; He, Y.; Sun, X.; Li, Q.; Wang, W.; Zhao, A.; Di, W. A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients. J. Ovarian Res. 2014, 7, 19. [Google Scholar] [CrossRef]
- Lan, C.; Huang, X.; Lin, S.; Huang, H.; Cai, Q.; Wan, T.; Lu, J.; Liu, J. Expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer. Technol. Cancer Res. Treat. 2013, 12, 259–267. [Google Scholar] [CrossRef]
- Yuan, X.; Zhang, J.; Li, D.; Mao, Y.; Mo, F.; Du, W.; Ma, X. Prognostic significance of tumor-associated macrophages in ovarian cancer: A meta-analysis. Gynecol. Oncol. 2017, 147, 181–187. [Google Scholar]
- Tiwari, A.; Trivedi, R.; Lin, S.-Y. Tumor microenvironment: Barrier or opportunity towards effective cancer therapy. J. Biomed. Sci. 2022, 29, 1–27. [Google Scholar] [CrossRef]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef]
- Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [PubMed]
- Khalaf, K.; Hana, D.; Chou, J.T.; Singh, C.; Mackiewicz, A.; Kaczmarek, M. Aspects of the Tumor Microenviron-ment Involved in Immune Resistance and Drug Resistance. Front. Immunol. 2021, 12, 656364. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Mangala, L.S.; Mooberry, L.; Bayraktar, E.; Dasari, S.K.; Ma, S.; Ivan, C.; Court, K.A.; Rodriguez-Aguayo, C.; Bayraktar, R.; et al. Identifying and targeting angiogenesis-Related microRNAs in ovarian cancer. Oncogene 2019, 38, 6095–6108. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Zhang, Z.; Wei, X.; Dai, R. Small-molecule inhibitor of Bcl-2 (TW-37) suppresses growth and enhances cisplatin-induced apoptosis in ovarian cancer cells. J. Ovarian Res. 2015, 8, 3. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Grande, F.; Garofalo, A.; Neamati, N.; Gu, D.; Liu, H.; Su, G.H.; Zhang, X.; Chin-Sinex, H.; Hanenberg, H.; et al. Discovery of a novel orally active small-molecule gp130 inhibitor for the treatment of ovarian cancer. Mol. Cancer Ther. 2013, 12, 937–949. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Butkevich, A.N.; Yamada, R.; Zhou, Y.; Debnath, B.; Duncan, R.; Zandi, E.; Petasis, N.A.; Neamati, N. Discovery of an orally active small-molecule irreversible inhibitor of protein disulfide isomerase for ovarian cancer treatment. Proc. Natl. Acad. Sci. USA 2012, 109, 16348–16353. [Google Scholar] [CrossRef] [PubMed]
- Kashani, B.; Zandi, Z.; Bashash, D.; Zaghal, A.; Momeny, M.; Poursani, E.M.; Pourbagheri-Sigaroodi, A.; Mousavi, S.A.; Ghaffari, S.H. Small molecule inhibitor of TLR4 inhibits ovarian cancer cell proliferation: New insight into the anticancer effect of TAK-242 (Resatorvid). Cancer Chemother. Pharmacol. 2020, 85, 47–59. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.; Zhou, F. Efficacy and safety of anti-angiogenic drug monotherapy and combination therapy for ovarian cancer: A meta-analysis and trial sequential analysis of randomized controlled trials. Front. Pharmacol. 2024, 15, 1423891. [Google Scholar] [CrossRef] [PubMed]
- Rendell, A.; Thomas-Bland, I.; McCuish, L.; Taylor, C.; Binju, M.; Yu, Y. Targeting Tyrosine Kinases in Ovarian Cancer: Small Molecule Inhibitor and Monoclonal Antibody, Where Are We Now? Biomedicines 2022, 10, 2113. [Google Scholar] [CrossRef]
- Hainsworth, J.D.; Thompson, D.S.; Bismayer, J.A.; Gian, V.G.; Merritt, W.M.; Whorf, R.C.; Finney, L.H.; Dudley, B.S. Paclitaxel/carboplatin with or without sorafenib in the first-line treatment of patients with stage III/IV epithelial ovarian cancer: A randomized phase II study of the Sarah Cannon Research Institute. Cancer Med. 2015, 4, 673–681. [Google Scholar] [CrossRef]
- Despierre, E.; Vergote, I.; Anderson, R.; Coens, C.; Katsaros, D.; Hirsch, F.R.; Boeckx, B.; Varella-Garcia, M.; Ferrero, A.; Ray-Coquard, I.; et al. Epidermal Growth Factor Receptor (EGFR) Pathway Biomarkers in the Randomized Phase III Trial of Erlotinib Versus Observation in Ovarian Cancer Patients with No Evidence of Disease Progression after First-Line Platinum-Based Chemotherapy. Target. Oncol. 2015, 10, 583–596. [Google Scholar] [CrossRef] [PubMed]
- Lheureux, S.; Prokopec, S.D.; Oldfield, L.E.; Gonzalez-Ochoa, E.; Bruce, J.P.; Wong, D.; Danesh, A.; Torti, D.; Torchia, J.; Fortuna, A.; et al. Identifying Mechanisms of Resistance by Circulating Tumor DNA in EVOLVE, a Phase II Trial of Cediranib Plus Olaparib for Ovarian Cancer at Time of PARP Inhibitor Progression. Clin. Cancer Res. 2023, 29, 3706–3716. [Google Scholar] [CrossRef]
- Gonzalez-Martin, A.; Pothuri, B.; Vergote, I.; DePont Christensen, R.; Graybill, W.; Mirza, M.R.; McCormick, C.; Lorusso, D.; Hoskins, P.; Freyer, G.; et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N. Engl. J. Med. 2019, 381, 2391–2402. [Google Scholar] [CrossRef]
- Matulonis, U.A.; Shapira-Frommer, R.; Santin, A.D.; Lisyanskaya, A.S.; Pignata, S.; Vergote, I.; Raspagliesi, F.; Sonke, G.S.; Birrer, M.; Provencher, D.M.; et al. Antitumor activity and safety of pembrolizumab in patients with advanced re-current ovarian cancer: Results from the phase II KEYNOTE-100 study. Ann. Oncol. 2019, 30, 1080–1087. [Google Scholar] [CrossRef] [PubMed]
- Kverneland, A.H.; Pedersen, M.; Westergaard, M.C.W.; Nielsen, M.; Borch, T.H.; Olsen, L.R.; Aasbjerg, G.; Santegoets, S.J.; van der Burg, S.H.; Milne, K.; et al. Adoptive cell therapy in combination with checkpoint inhibitors in ovarian cancer. Oncotarget 2020, 11, 2092–2105. [Google Scholar] [CrossRef] [PubMed]
- Farokhi Boroujeni, S.; Rodriguez, G.; Galpin, K.; Yakubovich, E.; Murshed, H.; Ibrahim, D.; Asif, S.; Vanderhyden, B.C. BRCA1 and BRCA2 deficient tumour models generate distinct ovarian tumour microenvironments and differential responses to therapy. J. Ovarian Res. 2023, 16, 231. [Google Scholar] [CrossRef]
- Feng, B.; Zhou, F.; Hou, B.; Wang, D.; Wang, T.; Fu, Y.; Ma, Y.; Yu, H.; Li, Y. Binary Cooperative Prodrug Nano-particles Improve Immunotherapy by Synergistically Modulating Immune Tumor Microenvironment. Adv. Mater. 2018, 30, e1803001. [Google Scholar] [CrossRef] [PubMed]
- Raja, F.A.; Chopra, N.; Ledermann, J.A. Optimal first-line treatment in ovarian cancer. Ann. Oncol. 2012, 23 (Suppl. S10), x118–x127. [Google Scholar] [CrossRef] [PubMed]
- Ventola, C.L. Cancer Immunotherapy, Part 3: Challenges and Future Trends. Pharm. Ther. 2017, 42, 514–521. [Google Scholar]
- Yang, C.; Xia, B.-R.; Zhang, Z.-C.; Zhang, Y.-J.; Lou, G.; Jin, W.-L. Immunotherapy for Ovarian Cancer: Adjuvant, Combination, and Neoadjuvant. Front. Immunol. 2020, 11, 577869. [Google Scholar] [CrossRef]
- Johnson, R.L.; Cummings, M.; Thangavelu, A.; Theophilou, G.; de Jong, D.; Orsi, N.M. Barriers to Immunotherapy in Ovarian Cancer: Metabolic, Genomic, and Immune Perturbations in the Tumour Microenvironment. Cancers 2021, 13, 6231. [Google Scholar] [CrossRef] [PubMed]
- McFarland, C.D.; Yaglom, J.A.; Wojtkowiak, J.W.; Scott, J.G.; Morse, D.L.; Sherman, M.Y.; Mirny, L.A. The Dam-aging Effect of Passenger Mutations on Cancer Progression. Cancer Res. 2017, 77, 4763–4772. [Google Scholar] [CrossRef] [PubMed]
- Rad, H.S.; Monkman, J.; Warkiani, M.E.; Ladwa, R.; O’Byrne, K.; Rezaei, N.; Kulasinghe, A. Understanding the tumor microenvironment for effective immunotherapy. Med. Res. Rev. 2021, 41, 1474–1498. [Google Scholar]
- Morand, S.; Devanaboyina, M.; Staats, H.; Stanbery, L.; Nemunaitis, J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int. J. Mol. Sci. 2021, 22, 6532. [Google Scholar] [CrossRef] [PubMed]
- Palaia, I.; Tomao, F.; Sassu, C.M.; Musacchio, L.; Benedetti Panici, P. Immunotherapy For Ovarian Cancer: Recent Advances And Combination Therapeutic Approaches. OncoTargets Ther. 2020, 13, 6109–6129. [Google Scholar] [CrossRef] [PubMed]
- Muthukutty, P.; Woo, H.Y.; Ragothaman, M.; Yoo, S.Y. Recent Advances in Cancer Immunotherapy Delivery Modalities. Pharmaceutics 2023, 15, 504. [Google Scholar] [CrossRef] [PubMed]
- Cunningham, N.; Lapointe, R.; Lerouge, S. Biomaterials for enhanced immunotherapy. APL Bioeng. 2022, 6, 041502. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Hong, Z.; Zhang, C.; Wang, L.; Han, Z.; Ma, D. Immune checkpoint therapy for solid tumours: Clinical dilemmas and future trends. Signal Transduct. Target. Ther. 2023, 8, 320. [Google Scholar] [CrossRef] [PubMed]
- Blanc-Durand, F.; Genestie, C.; Galende, E.Y.; Gouy, S.; Morice, P.; Pautier, P.; Maulard, A.; Mesnage, S.; Le Formal, A.; Brizais, C.; et al. Distribution of novel immune-checkpoint targets in ovarian cancer tumor microenvironment: A dynamic landscape. Gynecol. Oncol. 2021, 160, 279–284. [Google Scholar] [CrossRef]
- Lampert, E.J.; Zimmer, A.S.; Padget, M.R.; Cimino-Mathews, A.; Nair, J.R.; Liu, Y.; Swisher, E.M.; Hodge, J.W.; Nixon, A.B.; Nichols, E.; et al. Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: A Proof-of-Concept Phase II Study. Clin. Cancer Res. 2020, 26, 4268–4279. [Google Scholar] [CrossRef]
- Schoutrop, E.; El-Serafi, I.; Poiret, T.; Zhao, Y.; Gultekin, O.; He, R.; Moyano-Galceran, L.; Carlson, J.W.; Lehti, K.; Hassan, M.; et al. Mesothelin-Specific CAR T Cells Target Ovarian Cancer. Cancer Res. 2021, 81, 3022–3035. [Google Scholar] [CrossRef] [PubMed]
- Xia, A.-L.; Wang, X.-C.; Lu, Y.-J.; Lu, X.-J.; Sun, B. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: Challenges and opportunities. Oncotarget 2017, 8, 90521–90531. [Google Scholar] [CrossRef] [PubMed]
- Dong, X.; Ren, J.; Amoozgar, Z.; Lee, S.; Datta, M.; Roberge, S.; Duquette, M.; Fukumura, D.; Jain, R.K. Anti-VEGF therapy improves EGFR-vIII-CAR-T cell delivery and efficacy in syngeneic glioblastoma models in mice. J. ImmunoTherapy Cancer 2023, 11, e005583. [Google Scholar] [CrossRef]
- Kang, C.; Jeong, S.-Y.; Song, S.Y.; Choi, E.K. The emerging role of myeloid-derived suppressor cells in radiotherapy. Radiat. Oncol. J. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Pegram, H.J.; Lee, J.C.; Hayman, E.G.; Imperato, G.H.; Tedder, T.F.; Sadelain, M.; Brentjens, R.J. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood 2012, 119, 4133–4141. [Google Scholar] [CrossRef] [PubMed]
- Krenciute, G.; Prinzing, B.L.; Yi, Z.; Wu, M.F.; Liu, H.; Dotti, G.; Balyasnikova, I.V.; Gottschalk, S. Transgenic Ex-pression of IL15 Improves Antiglioma Activity of IL13Ralpha2-CAR T Cells but Results in Antigen Loss Variants. Cancer Immunol. Res. 2017, 5, 571–581. [Google Scholar] [CrossRef] [PubMed]
- Spolski, R.; Leonard, W.J. Interleukin-21: A double-edged sword with therapeutic potential. Nat. Rev. Drug Discov. 2014, 13, 379–395. [Google Scholar] [CrossRef] [PubMed]
- Shimabukuro-Vornhagen, A.; Godel, P.; Subklewe, M.; Stemmler, H.J.; Schlosser, H.A.; Schlaak, M.; Kochanek, M.; Boll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. ImmunoTherapy Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [PubMed]
- Chow, S.; Berek, J.S.; Dorigo, O. Development of Therapeutic Vaccines for Ovarian Cancer. Vaccines 2020, 8, 657. [Google Scholar] [CrossRef]
- Block, M.S.; Dietz, A.B.; Gustafson, M.P.; Kalli, K.R.; Erskine, C.L.; Youssef, B.; Vijay, G.V.; Allred, J.B.; Pavelko, K.D.; Strausbauch, M.A.; et al. Th17-inducing autologous dendritic cell vaccination promotes antigen-specific cellular and humoral immunity in ovarian cancer patients. Nat. Commun. 2020, 11, 5173. [Google Scholar] [CrossRef]
- Kooti, W.; Esmaeili Gouvarchin Ghaleh, H.; Farzanehpour, M.; Dorostkar, R.; Jalali Kondori, B.; Bolandian, M. Oncolytic Viruses and Cancer, Do You Know the Main Mechanism? Front. Oncol. 2021, 11, 761015. [Google Scholar] [CrossRef] [PubMed]
- Yun, C.-O.; Hong, J.; Yoon, A.-R. Current clinical landscape of oncolytic viruses as novel cancer immunotherapeutic and recent preclinical advancements. Front. Immunol. 2022, 13, 953410. [Google Scholar] [CrossRef] [PubMed]
- Manyam, M.; Stephens, A.J.; Kennard, J.A.; LeBlanc, J.; Ahmad, S.; Kendrick, J.E.; Holloway, R.W. A phase 1b study of intraperitoneal oncolytic viral immunotherapy in platinum-resistant or refractory ovarian cancer. Gynecol. Oncol. 2021, 163, 481–489. [Google Scholar] [CrossRef] [PubMed]
- Santry, L.A.; van Vloten, J.P.; Knapp, J.P.; Matuszewska, K.; McAusland, T.M.; Minott, J.A.; Mould, R.C.; Stegelmeier, A.A.; Major, P.P.; Wootton, S.K.; et al. Tumour vasculature: Friend or foe of oncolytic viruses? Cytokine Growth Factor Rev. 2020, 56, 69–82. [Google Scholar] [CrossRef] [PubMed]
- Beug, S.T.; Pichette, S.J.; St-Jean, M.; Holbrook, J.; Walker, D.E.; LaCasse, E.C.; Korneluk, R.G. Combination of IAP Antagonists and TNF-α-Armed Oncolytic Viruses Induce Tumor Vascular Shutdown and Tumor Regression. Mol. Ther. Oncol. 2018, 10, 28–39. [Google Scholar] [CrossRef] [PubMed]
- Fu, R.; Qi, R.; Xiong, H.; Lei, X.; Jiang, Y.; He, J.; Chen, F.; Zhang, L.; Qiu, D.; Chen, Y.; et al. Combination therapy with oncolytic virus and T cells or mRNA vaccine amplifies antitumor effects. Signal Transduct. Target. Ther. 2024, 9, 118. [Google Scholar] [CrossRef] [PubMed]
- Santry, L.A.; van Vloten, J.P.; AuYeung, A.W.K.; Mould, R.C.; Yates, J.G.E.; McAusland, T.M.; Petrik, J.J.; Major, P.P.; Bridle, B.W.; Wootton, S.K. Recombinant Newcastle disease viruses expressing immunological checkpoint inhibitors induce a pro-inflammatory state and enhance tumor-specific immune responses in two murine models of cancer. Front. Microbiol. 2024, 15, 1325558. [Google Scholar] [CrossRef]
- Matuszewska, K.; Santry, L.A.; van Vloten, J.P.; Auyeung, A.W.K.; Major, P.P.; Lawler, J.; Wootton, S.K.; Bridle, B.W.; Petrik, J. Combining Vascular Normalization with an Oncolytic Virus Enhances Immunotherapy in a Preclinical Model of Advanced-Stage Ovarian Cancer. Clin. Cancer Res. 2019, 25, 1624–1638. [Google Scholar] [CrossRef]
- Lynam, S.; Lugade, A.A.; Odunsi, K. Immunotherapy for Gynecologic Cancer: Current Applications and Future Directions. Clin. Obstet. Gynecol. 2020, 63, 48–63. [Google Scholar] [CrossRef]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
- Shuel, S.L. Targeted cancer therapies: Clinical pearls for primary care. Can. Fam. Physician 2022, 68, 515–518. [Google Scholar] [CrossRef] [PubMed]
- Turk, A.A.; Wisinski, K.B. PARP inhibitors in breast cancer: Bringing synthetic lethality to the bedside. Cancer 2018, 124, 2498–2506. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Xu, S.; Cheng, S.; Yang, J.; Wang, Y. Clinical application of PARP inhibitors in ovarian cancer: From mo-lecular mechanisms to the current status. J. Ovarian Res. 2023, 16, 6. [Google Scholar] [CrossRef] [PubMed]
- Boussios, S.; Rassy, E.; Moschetta, M.; Ghose, A.; Adeleke, S.; Sanchez, E.; Sheriff, M.; Chargari, C.; Pavlidis, N. BRCA Mutations in Ovarian and Prostate Cancer: Bench to Bedside. Cancers 2022, 14, 3888. [Google Scholar] [CrossRef] [PubMed]
- Rose, P.G. Ovarian cancer recurrence: Is the definition of platinum sensitivity modified by PARPi, bevacizumab or other intervening treatments?: A clinical perspective. Cancer Drug Resist. 2022, 5, 415–423. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Pujade-Lauraine, E.; Ledermann, J.A.; Selle, F.; Gebski, V.; Penson, R.T.; Oza, A.M.; Korach, J.; Huzarski, T.; Poveda, A.; Pignata, S.; et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1274–1284. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.M.; Ledermann, J.A.; Kohn, E.C. PARP Inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann. Oncol. 2014, 25, 32–40. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.J.; Huang, R.; Shen, Y.W.; Liu, J.; Wu, Y.; Jin, J.M.; Zhang, H.; Sun, Y.; Chen, H.Z.; Luan, X. Enhanced anti-tumor efficacy by inhibiting HIF-1alpha to reprogram TAMs via core-satellite upconverting nanoparticles with curcumin mediated photodynamic therapy. Biomater. Sci. 2021, 9, 6403–6415. [Google Scholar] [CrossRef]
- Ning, F.; Cole, C.B.; Annunziata, C.M. Driving Immune Responses in the Ovarian Tumor Microenvironment. Front. Oncol. 2020, 10, 604084. [Google Scholar] [CrossRef]
- Ding, L.; Kim, H.-J.; Wang, Q.; Kearns, M.; Jiang, T.; Ohlson, C.E.; Li, B.B.; Xie, S.; Liu, J.F.; Stover, E.H.; et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep. 2018, 25, 2972–2980.e5. [Google Scholar] [CrossRef] [PubMed]
- Martincuks, A.; Song, J.; Kohut, A.; Zhang, C.; Li, Y.J.; Zhao, Q.; Mak, E.; Rodriguez-Rodriguez, L.; Yu, H.; Cristea, M. PARP Inhibition Activates STAT3 in Both Tumor and Immune Cells Underlying Therapy Resistance and Immunosuppression In Ovarian Cancer. Front. Oncol. 2021, 11, 724104. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.-B.; Tang, Y.-L.; Liang, X.-H. Targeting VEGF pathway to normalize the vasculature: An emerging insight in cancer therapy. OncoTargets Ther. 2018, 11, 6901–6909. [Google Scholar] [CrossRef] [PubMed]
- Seebacher, N.A.; Stacy, A.E.; Porter, G.M.; Merlot, A.M. Clinical development of targeted and immune based an-ti-cancer therapies. J. Exp. Clin. Cancer Res. 2019, 38, 156. [Google Scholar] [CrossRef]
- Marchetti, C.; Muzii, L.; Romito, A.; Benedetti Panici, P. First-line treatment of women with advanced ovarian cancer: Focus on bevacizumab. OncoTargets Ther. 2019, 12, 1095–1103. [Google Scholar] [CrossRef] [PubMed]
- Oza, A.M.; Cook, A.D.; Pfisterer, J.; Embleton, A.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
- Niu, G.; Chen, X. Vascular Endothelial Growth Factor as an Anti-Angiogenic Target for Cancer Therapy. Curr. Drug Targets 2010, 11, 1000–1017. [Google Scholar] [CrossRef] [PubMed]
- Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microen-vironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Song, S.; Duan, N.; Wang, F.; Wang, Y.; Yang, Y.; Peng, C.; Li, J.; Nie, D.; Zhang, X.; et al. MT1-MMP-Activated Liposomes to Improve Tumor Blood Perfusion and Drug Delivery for Enhanced Pancreatic Cancer Therapy. Adv. Sci. 2020, 7, 1902746. [Google Scholar] [CrossRef]
- Heil, F.; Babitzki, G.; Julien-Laferriere, A.; Ooi, C.-H.; Hidalgo, M.; Massard, C.; Martinez-Garcia, M.; Le Tourneau, C.; Kockx, M.; Gerber, P.; et al. Vanucizumab mode of action: Serial biomarkers in plasma, tumor, and skin-wound-healing biopsies. Transl. Oncol. 2021, 14, 100984. [Google Scholar] [CrossRef]
- Hidalgo, M.; Martinez-Garcia, M.; Le Tourneau, C.; Massard, C.; Garralda, E.; Boni, V.; Taus, A.; Albanell, J.; Sablin, M.P.; Alt, M.; et al. First-in-Human Phase I Study of Single-agent Vanucizumab, A First-in-Class Bispecific An-ti-Angiopoietin-2/Anti-VEGF-A Antibody, in Adult Patients with Advanced Solid Tumors. Clin. Cancer Res. 2018, 24, 1536–1545. [Google Scholar] [CrossRef] [PubMed]
- Russell, S.; Duquette, M.; Liu, J.; Drapkin, R.; Lawler, J.; Petrik, J. Combined therapy with thrombospondin-1 type I repeats (3TSR) and chemotherapy induces regression and significantly improves survival in a preclinical model of advanced stage epithelial ovarian cancer. FASEB J. 2015, 29, 576–588. [Google Scholar] [CrossRef] [PubMed]
- Scharovsky, O.G.; Mainetti, L.E.; Rozados, V.R. Metronomic Chemotherapy: Changing the Paradigm That More Is Better. Curr. Oncol. 2009, 16, 7–15. [Google Scholar] [CrossRef] [PubMed]
- Fotopoulou, C. Limitations to the use of carboplatin-based therapy in advanced ovarian cancer. Eur. J. Cancer Suppl. 2014, 12, 13–16. [Google Scholar] [CrossRef] [PubMed]
- Aston, W.J.; Hope, D.E.; Nowak, A.K.; Robinson, B.W.; Lake, R.A.; Lesterhuis, W.J. A systematic investigation of the maximum tolerated dose of cytotoxic chemotherapy with and without supportive care in mice. BMC Cancer 2017, 17, 684. [Google Scholar] [CrossRef]
- Matuszewska, K.; Ten Kortenaar, S.; Pereira, M.; Santry, L.A.; Petrik, D.; Lo, K.M.; Bridle, B.W.; Wootton, S.K.; Lawler, J.; Petrik, J. Addition of an Fc-IgG induces receptor clustering and increases the in vitro efficacy and in vivo anti-tumor properties of the thrombospondin-1 type I repeats (3TSR) in a mouse model of advanced stage ovarian cancer. Gynecol. Oncol. 2022, 164, 154–169. [Google Scholar] [CrossRef]
Inhibitor | Target | Effect | Reference |
---|---|---|---|
miR-204-5p-inh | miR-204-5p | Inhibits angiogenesis and ovarian tumor growth | [95] |
TW-37 | Bcl-2 | Apoptosis of ovarian cancer cells; resensitization to cisplatin | [96] |
SC144 | Glycoprotein 130 (gp130) | Delays ovarian tumor progression | [97] |
PACMA 31 | Protein Disulfide Isomerase (PDI) | Reduces survival and proliferation of ovarian cancer cells; delays tumor growth | [98] |
TAK242 | Toll-like receptor (TLR) 4 | Suppression of ovarian cancer cell inflammation; induction of cell cycle arrest and apoptosis | [99] |
Bevacizumab | VEGF | Has shown clinical efficacy, particularly when used in combination | [100] |
Cetuximab | EGFR | Reduces cell proliferation and inhibits cell cycle | [101] |
Pertuzumab | HER2 | Targets HER2 dimerization domain—inhibits HER signaling and reduces cell proliferation | [101] |
Sorafenib | Multi-receptor tyrosine kinases | Cytotoxic to tumor cells; anti-angiogenic | [102] |
Erlotinib | EGFR | Can reduce ovarian cancer progression in EGFR-positive tumors | [103] |
Olaparib | PARP | Synthetic lethality in ovarian tumors; enhanced efficacy in patients with BRCA mutations or homologous recombination-deficient tumors | [104] |
Niraparib | PARP | Synthetic lethality; efficacy appears not to be reliant on BRCA status | [105] |
Pembrolizumab | PD-1 | Reduces tumor immunosuppression; reduced tumor progression in clinical trials | [106] |
Ipilimumab | CTLA-4 | Enhances expansion of tumor-infiltrating lymphocytes in the ovarian TME | [107] |
Durvalumab | PD-L1 | Reduces immunosuppression in the ovarian TME | [108] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Garlisi, B.; Lauks, S.; Aitken, C.; Ogilvie, L.M.; Lockington, C.; Petrik, D.; Eichhorn, J.S.; Petrik, J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Curr. Oncol. 2024, 31, 3826-3844. https://doi.org/10.3390/curroncol31070283
Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS, Petrik J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Current Oncology. 2024; 31(7):3826-3844. https://doi.org/10.3390/curroncol31070283
Chicago/Turabian StyleGarlisi, Bianca, Sylvia Lauks, Caroline Aitken, Leslie M. Ogilvie, Cielle Lockington, Duncan Petrik, Jan Soeren Eichhorn, and Jim Petrik. 2024. "The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities" Current Oncology 31, no. 7: 3826-3844. https://doi.org/10.3390/curroncol31070283
APA StyleGarlisi, B., Lauks, S., Aitken, C., Ogilvie, L. M., Lockington, C., Petrik, D., Eichhorn, J. S., & Petrik, J. (2024). The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Current Oncology, 31(7), 3826-3844. https://doi.org/10.3390/curroncol31070283