1
|
Liu C, Yin Q, Wu Z, Li W, Huang J, Chen B, Yang Y, Zheng X, Zeng L, Wang J. Inflammation and Immune Escape in Ovarian Cancer: Pathways and Therapeutic Opportunities. J Inflamm Res 2025; 18:895-909. [PMID: 39867950 PMCID: PMC11762012 DOI: 10.2147/jir.s503479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies, largely due to its late-stage diagnosis and high recurrence rates. Chronic inflammation is a critical driver of OC progression, contributing to immune evasion, tumor growth, and metastasis. Inflammatory cytokines, including IL-6, TNF-α, and IL-8, as well as key signaling pathways such as nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3), are upregulated in OC, promoting a tumor-promoting environment. The tumor microenvironment (TME) is characterized by immune cells like tumor-associated macrophages (TAMs) and regulatory T cells (Tregs), which suppress anti-tumor immune responses, facilitating immune evasion. Furthermore, OC cells utilize immune checkpoint pathways, including PD-1/PD-L1, to inhibit cytotoxic T cell activity. Targeting these inflammatory and immune evasion mechanisms offers promising therapeutic strategies. COX-2 inhibitors, Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway blockers, and NF-kB inhibitors have shown potential in preclinical studies, while immune checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 have been explored with mixed results in OC. Additionally, emerging research on the microbiome and inflammation-related biomarkers, such as microRNAs (miRNAs) and exosomes, points to new opportunities for early detection and precision medicine. Future approaches to OC treatment must focus on personalized strategies that target the inflammatory TME, integrating anti-inflammatory therapies with immunotherapy to enhance patient outcomes. Continued research into the interplay between inflammation and immune evasion in OC is essential for developing effective, long-lasting treatments.
Collapse
Affiliation(s)
- Chunyan Liu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Qinan Yin
- Department of Radiation Oncology, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, People’s Republic of China
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Zhaoying Wu
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Wenhui Li
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Jun Huang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Bo Chen
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yanjun Yang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Xuewei Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Li Zeng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Jingjing Wang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
2
|
Cerda‐Troncoso C, Grünenwald F, Arias‐Muñoz E, Cavieres VA, Caceres‐Verschae A, Hernández S, Gaete‐Ramírez B, Álvarez‐Astudillo F, Acuña RA, Ostrowski M, Burgos PV, Varas‐Godoy M. Chemo-small extracellular vesicles released in cisplatin-resistance ovarian cancer cells are regulated by the lysosomal function. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e157. [PMID: 38947172 PMCID: PMC11212338 DOI: 10.1002/jex2.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 04/03/2024] [Accepted: 04/29/2024] [Indexed: 07/02/2024]
Abstract
Chemoresistance is a common problem in ovarian cancer (OvCa) treatment, where resistant cells, in response to chemotherapy, secrete small extracellular vesicles (sEVs), known as chemo-sEVs, that transfer resistance to recipient cells. sEVs are formed as intraluminal vesicles (ILVs) within multivesicular endosomes (MVEs), whose trafficking is regulated by Ras-associated binding (RAB) GTPases that mediate sEVs secretion or lysosomal degradation. A decrease in lysosomal function can promote sEVs secretion, but the relationship between MVEs trafficking pathways and sEVs secretion in OvCa chemoresistance is unclear. Here, we show that A2780cis cisplatin (CCDP) resistant OvCa cells had an increased number of MVEs and ILVs structures, higher levels of Endosomal Sorting Complex Required for Transport (ESCRTs) machinery components, and RAB27A compared to A2780 CDDP-sensitive OvCa cells. CDDP promoted the secretion of chemo-sEVs in A2780cis cells, enriched in DNA damage response proteins. A2780cis cells exhibited poor lysosomal function with reduced levels of RAB7, essential in MVEs-Lysosomal trafficking. The silencing of RAB27A in A2780cis cells prevents the Chemo-EVs secretion, reduces its chemoresistance and restores lysosomal function and levels of RAB7, switching them into an A2780-like cellular phenotype. Enhancing lysosomal function with rapamycin reduced chemo-sEVs secretion. Our results suggest that adjusting the balance between secretory MVEs and lysosomal MVEs trafficking could be a promising strategy for overcoming CDDP chemoresistance in OvCa.
Collapse
Affiliation(s)
- Cristóbal Cerda‐Troncoso
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
| | - Felipe Grünenwald
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Eloísa Arias‐Muñoz
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Viviana A. Cavieres
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Albano Caceres‐Verschae
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Sergio Hernández
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Belén Gaete‐Ramírez
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | | | - Rodrigo A. Acuña
- Centro de Medicina Regenerativa, Facultad de MedicinaClínica Alemana Universidad del DesarrolloSantiagoChile
| | - Matias Ostrowski
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS)Universidad de Buenos Aires (UBA)Buenos AiresArgentina
| | - Patricia V. Burgos
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
| | - Manuel Varas‐Godoy
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
- Advanced Center for Chronic DiseasesSantiagoChile
| |
Collapse
|
3
|
Khan MW, Zou C, Hassan S, Din FU, Abdoul Razak MY, Nawaz A, Alam Zeb, Wahab A, Bangash SA. Cisplatin and oleanolic acid Co-loaded pH-sensitive CaCO 3 nanoparticles for synergistic chemotherapy. RSC Adv 2022; 12:14808-14818. [PMID: 35702211 PMCID: PMC9109477 DOI: 10.1039/d2ra00742h] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/01/2022] [Indexed: 01/06/2023] Open
Abstract
Despite being one of the most potent anticancer agents, cisplatin (CDDP) clinical usage is limited owing to the acquired resistance and severe adverse effects including nephrotoxicity. The current work has offered a unique approach by designing a pH-sensitive calcium carbonate drug delivery system for CDDP and oleanolic acid (OA) co-delivery, with an enhanced tumor efficacy and reduced unwanted effects. Micro emulsion method was employed to generate calcium carbonate cores (CDDP encapsulated) followed by lipid coating along with the OA loading resulting in the generation of lipid-coated cisplatin/oleanolic acid calcium carbonate nanoparticles (CDDP/OA-LCC NPs). In vitro biological assays confirmed the synergistic apoptotic effect of CDDP and OA against HepG2 cells. It was further verified in vivo through the tumor-bearing nude mice model where NPs exhibited enhanced satisfactory antitumor efficacy in contrast to free drug solutions. In vivo pharmacokinetic study demonstrated that a remarkable long circulation time with a constant therapeutic concentration for both drugs could be achieved via this drug delivery system. In addition, the in vivo imaging study revealed that DiR-loaded NPs were concentrated more in tumors for a longer period of time as compared to other peritoneal tissues in tumor bearing mice, demonstrating the site specificity of the delivery system. On the other hand, hematoxylin and eosin (H&E) staining of Kunming mice kidney tissue sections revealed that OA greatly reduced CDDP induced nephrotoxicity in the formulation. Overall, these results confirmed that our pH-sensitive dual loaded drug delivery system offers a handy direction for effective and safer combination chemotherapy.
Collapse
Affiliation(s)
- Muhammad Waseem Khan
- Institute of Pharmaceutical Sciences, Khyber Medical University Peshawar Pakistan +92-3459146065
| | - Chenming Zou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei 430030 China
| | - Said Hassan
- Institute of Biotechnology and Microbiology, Bacha Khan University Charsadda Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Quaid-I-Azam University Islamabad 45320 Pakistan
| | - Mahaman Yacoubou Abdoul Razak
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
| | - Asif Nawaz
- Faculty of Pharmacy, Gomal University Dera Ismail Khan Pakistan
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology Kohat Pakistan
| | - Sudhair Abbas Bangash
- Faculty of Life Science, Department of Pharmacy, Sarhad University of Science and Information Technology Peshawar Pakistan
| |
Collapse
|
4
|
Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022; 232:114205. [DOI: 10.1016/j.ejmech.2022.114205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
5
|
Zhou M, Zhao G, Zhou X, Xuan R, Chen Y, Wang Y. Clinicopathological analysis and prognostic significance of NF-κB p65 and IKKβ protein and mRNA expression in nasopharyngeal carcinoma. J Int Med Res 2022; 50:3000605211069195. [PMID: 34994235 PMCID: PMC8743962 DOI: 10.1177/03000605211069195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective To investigate the clinicopathological significance of NF-κB p65 and IKKβ protein and mRNA expression in nasopharyngeal carcinoma (NPC) patients from Guangdong Province, China. Methods Data and tissues from patients with NPC were retrospectively studied. Immunohistochemical staining and quantitative reverse transcription polymerase chain reaction were used to evaluate and compare NF-κB p65 and IKKβ protein and mRNA levels, respectively, in 60 NPC and 30 nasopharyngitis tissue samples. Statistical analysis was conducted to determine correlations between NF-κB p65 and IKKβ protein and mRNA levels with clinicopathological characteristics and prognoses of NPC patients. Results NF-κB p65 and IKKβ protein and mRNA expression in NPC were significantly correlated with tumor size, lymph node metastasis, and TNM stage. NF-κB p65 and IKKβ protein and mRNA levels were significantly increased in NPC patients with deep tumor invasion (T3–4), lymph node metastasis, and stage III/IV disease; high NF-κB p65 and IKKβ mRNA expression were associated with significantly shorter disease-free survival rates compared with cases showing low NF-κB p65 and IKKβ mRNA expression. Conclusions NF-κB p65 and IKKβ may affect the prognosis of NPC patients and could be potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Mei Zhou
- Department of Pathology, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| | - Gang Zhao
- Department of Ultrasonography, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| | - Xiaojun Zhou
- Department of Otorhinolaryngology, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| | - Ruixia Xuan
- Department of Pathology, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| | - Yuwei Chen
- Department of Pathology, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| | - Yun Wang
- Department of Pathology, Traditional Chinese Medical Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
6
|
Zhao P, Yun Q, Li A, Li R, Yan Y, Wang Y, Sun H, Damirin A. LPA3 is a precise therapeutic target and potential biomarker for ovarian cancer. Med Oncol 2022; 39:17. [PMID: 34982278 DOI: 10.1007/s12032-021-01616-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
Current studies have demonstrated that significant increased LPA levels to be observed in ascites in patients with ovarian cancer. Although several studies have shown that Lysophosphatidic acid (LPA) related to the progression of ovarian cancer, which LPA receptors (LPARs) and G-coupled protein subtypes mediated in LPA actions have not been clearly elucidated. This study aimed to clarify the roles of LPA and it is subtype-specific LPARs mediating mechanisms in ovarian cancer integrated using bioinformatic analysis and biological experimental approaches. The big data analysis shown that LPA3 was the only differentially expressed LPA receptor among the six LPARs in ovarian cancer and further verified in immunohistochemistry of tissue microarrays. Also found that LPA3 was also highly expressed in ovarian cancer tissue and ovarian cancer cells. Importantly, LPA significantly promoted the proliferation and migration of LPA3-overexpressing ovarian cancer cells, while the LPA-induced actions blocked by Ki16425, a LPAR1/3 antagonist treated, and LPA3-shRNA transfected. In vivo study indicated that the LPA3-overexpressing cell-derived tumors metastasis, tumors volume, and tumors mass were apparently increased in xenografted nude mice. In addition, we also observed that LPA3 was differential high expression in ovarian cancer tissue of the patients. Our studies further confirmed the LPA3/Gi/MAPKs/NF-κB signals were involved in LPA-induced oncogenic actions in ovarian cancer cells. Our findings indicated that the LPA3 might be a novel precise therapeutic target and potential biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Pengfei Zhao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Qingru Yun
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Aodungerile Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Rong Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yali Yan
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Hongju Sun
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| | - Alatangaole Damirin
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
7
|
Utilizing Patient-Derived Epithelial Ovarian Cancer Tumor Organoids to Predict Carboplatin Resistance. Biomedicines 2021; 9:biomedicines9081021. [PMID: 34440225 PMCID: PMC8394135 DOI: 10.3390/biomedicines9081021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
The development of patient-derived tumor organoids (TOs) from an epithelial ovarian cancer tumor obtained at the time of primary or interval debulking surgery has the potential to play an important role in precision medicine. Here, we utilized TOs to test front-line chemotherapy sensitivity and to investigate genomic drivers of carboplatin resistance. We developed six high-grade, serous epithelial ovarian cancer tumor organoid lines from tissue obtained during debulking surgery (two neoadjuvant-carboplatin-exposed and four chemo-naïve). Each organoid line was screened for sensitivity to carboplatin at four different doses (100, 10, 1, and 0.1 µM). Cell viability curves and resultant EC50 values were determined. One organoid line, UK1254, was predicted to be resistant to carboplatin based on its EC50 value (50.2 µM) being above clinically achievable Cmax. UK1254 had a significantly shorter PFS than the rest of the subjects (p = 0.0253) and was treated as a platinum-resistant recurrence. Subsequent gene expression analysis revealed extensively interconnected, differentially expressed pathways related to NF-kB, cellular differentiation (PRDM6 activation), and the linkage of B-cell receptor signaling to the PI3K-Akt signaling pathway (PI3KAP1 activation). This study demonstrates that patient-derived tumor organoids can be developed from patients at the time of primary or interval debulking surgery and may be used to predict clinical platinum sensitivity status or to investigate drivers of carboplatin resistance.
Collapse
|