1
|
Tocci P, Caprara V, Roman C, Sestito R, Rosanò L, Bagnato A. YAP signaling orchestrates the endothelin-1-guided invadopodia formation in high-grade serous ovarian cancer. Biosci Rep 2024; 44:BSR20241320. [PMID: 39495612 DOI: 10.1042/bsr20241320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/06/2024] Open
Abstract
The high-grade serous ovarian cancer (HG-SOC) is a notoriously challenging disease, characterized by a rapid peritoneal dissemination. HG-SOC cells leverage actin-rich membrane protrusions, known as invadopodia, to degrade the surrounding extracellular matrix (ECM) and invade, initiating the metastatic cascade. In HG-SOC, the endothelin-1 (ET-1)/endothelin A receptor (ETAR)-driven signaling coordinates invadopodia activity, however how this axis integrates pro-oncogenic signaling routes, as YAP-driven one, impacting on the invadopodia-mediated ECM degradation and metastatic progression, deserves a deeper investigation. Herein, we observed that downstream of the ET-1/ET-1R axis, the RhoC and Rac1 GTPases, acting as signaling intermediaries, promote the de-phosphorylation and nuclear accumulation of YAP. Conversely, the treatment with the dual ETA/ETB receptor antagonist, macitentan, inhibits the ET-1-driven YAP activity. Similarly, RhoC silencing, or cell transfection with a dominant inactive form of Rac1, restores YAP phosphorylation. Mechanistically, the ET-1R/YAP signal alliance coordinates invadopodia maturation into ECM-degrading structures, indicating how such ET-1R-guided protein network represents a route able to enhance the HG-SOC invasive potential. At functional level, we found that the interconnection between the ET-1R/RhoC and YAP signals is required for MMP-2 and MMP-9 proteolytic functions, cell invasion, and cytoskeleton architecture changes, supporting the HG-SOC metastatic strength. In HG-SOC patient-derived xenografts (PDX) macitentan, turning-off the invadopodia regulators RhoC/YAP, halts the metastatic colonization. ET-1R targeting, hindering the YAP activity, weakens the invadopodia machinery, embodying a promising therapeutic avenue to prevent peritoneal dissemination in HG-SOC.
Collapse
Affiliation(s)
- Piera Tocci
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Celia Roman
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Rosanò
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), Rome 00185, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
2
|
Li S, Lei N, Chen M, Guo R, Han L, Qiu L, Wu F, Jiang S, Tong N, Wang K, Li Y, Chang L. Exploration of organoids in ovarian cancer: From basic research to clinical translation. Transl Oncol 2024; 50:102130. [PMID: 39303357 PMCID: PMC11437877 DOI: 10.1016/j.tranon.2024.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Ovarian cancer is a highly heterogeneous tumor with a poor prognosis. The lack of reliable and efficient research models that can accurately mimic heterogeneity has impeded in-depth investigations and hindered the clinical translation of research findings in ovarian cancer. Organoid models have emerged as a promising in vitro approach, demonstrating remarkable fidelity to the histological, molecular, genomic, and transcriptomic features of their tissues of origin. In recent years, organoids have contributed to advancing our understanding of ovarian cancer initiation, metastasis, and drug resistance mechanisms, as well as facilitating clinical screening of effective therapeutic agents. The establishment of high-throughput organoid culture systems, coupled with cutting-edge technologies such as organ-on-a-chip, genetic engineering, and 3D printing, has tremendous potential for accelerating ovarian cancer research translation. In this review, we present a comprehensive overview of the latest exploration of organoids in basic ovarian cancer research and clinical translation. Furthermore, we discuss the prospects and challenges associated with the use of organoids and related novel technologies in the context of ovarian cancer. This review provides insights into the application of organoids in ovarian cancer.
Collapse
Affiliation(s)
- Siyu Li
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyu Chen
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Liping Han
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Luojie Qiu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Fengling Wu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Shan Jiang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ningyao Tong
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Kunmei Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Yong Li
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia; Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia.
| | - Lei Chang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China.
| |
Collapse
|
3
|
Liu Y, Xiao H, Zeng H, Xiang Y. Beyond tumor‑associated macrophages involved in spheroid formation and dissemination: Novel insights for ovarian cancer therapy (Review). Int J Oncol 2024; 65:117. [PMID: 39513610 PMCID: PMC11575928 DOI: 10.3892/ijo.2024.5705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Ovarian cancer (OC) is the most common and deadly malignant tumor of the female reproductive system. When OC cells detach from the primary tumor and enter the ascitic microenvironment, they are present as individual cells or multicellular spheroids in ascites. These spheroids, composed of cancer and non‑malignant cells, are metastatic units and play a crucial role in the progression of OC. However, little is known about the mechanism of spheroid formation and dissemination. Tumor‑associated macrophages (TAMs) in the center of spheroids are key in spheroid formation and metastasis and provide a potential target for OC therapy. The present review summarizes the key biological features of spheroids, focusing on the role of TAMs in spheroid formation, survival and peritoneal metastasis, and the strategies targeting TAMs to provide new insights in treating OC.
Collapse
Affiliation(s)
- Yuchen Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Haoyue Xiao
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Hai Zeng
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
4
|
Galvagno F, Leuci V, Massa A, Donini C, Rotolo R, Capellero S, Proment A, Vitali L, Lombardi AM, Tuninetti V, D'Ambrosio L, Merlini A, Vigna E, Valabrega G, Primo L, Puliafito A, Sangiolo D. Three-dimensional dynamics of mesothelin-targeted CAR.CIK lymphocytes against ovarian cancer peritoneal carcinomatosis. Cancer Immunol Immunother 2024; 74:6. [PMID: 39487859 PMCID: PMC11531451 DOI: 10.1007/s00262-024-03860-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/09/2024] [Indexed: 11/04/2024]
Abstract
Intraperitoneal cellular immunotherapy with CAR-redirected lymphocytes is an intriguing approach to target peritoneal carcinomatosis (PC) from ovarian cancer (OC), which is currently evaluated in clinical trials. PC displays a composite structure with floating tumor cells within ascites and solid-like masses invading the peritoneum. Therefore, a comprehensive experimental model is crucial to optimize CAR-cell therapies in such a peculiar environment. Here, we explored the activity of cytokine-induced killer lymphocytes (CIK), redirected by CAR against mesothelin (MSLN-CAR.CIK), within reductionistic 3D models resembling the structural complexity of both liquid and solid components of PC. MSLN-CAR.CIK effectively killed and were functionally efficient against OC targets. In a "floating-like" 3D context with floating OC spheroids, both tumor localization and killing by MSLN-CAR.CIK were significantly boosted by fluid flow. In a "solid-like" context, MSLN-CAR.CIK were recruited through the extracellular matrix on embedded tumor aggregates, with variable kinetics depending on the effector-target distance. Furthermore, MSLN-CAR.CIK penetrated the inner levels of OC spheroids exerting effective tumor killing. Our findings provide currently unknown therapeutically relevant information on intraperitoneal approaches with CAR.CIK, supporting further developments and improvements for clinical studies in the context of locoregional cell therapy approaches for patients with PC from OC.
Collapse
Grants
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 2023-2024 Italian Ministry of Health, Ricerca Corrente
- 23211 Fondazione AIRC per la ricerca sul cancro ETS
- 25040 Fondazione AIRC per la ricerca sul cancro ETS
- 20259 Fondazione AIRC per la ricerca sul cancro ETS
- 2019 Ricerca Locale, Università degli Studi di Torino
- 2022 Ricerca Locale, Università degli Studi di Torino
- 2022 Fondazione CRT
- RCR-2019-23669115 CAR-T Grant
- PON 2014-2020, DM 1062/2021 PNR 2021-2027
Collapse
Affiliation(s)
- Federica Galvagno
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - Valeria Leuci
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - Annamaria Massa
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - Chiara Donini
- Department of Oncology, University of Torino, Turin, Italy
| | - Ramona Rotolo
- Department of Oncology, University of Torino, Turin, Italy
| | | | | | - Letizia Vitali
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Valentina Tuninetti
- Department of Oncology, University of Torino, Turin, Italy
- Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| | - Lorenzo D'Ambrosio
- Department of Oncology, University of Torino, Turin, Italy
- Medical Oncology, AOU San Luigi Gonzaga, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Torino, Turin, Italy
- Medical Oncology, AOU San Luigi Gonzaga, Orbassano, TO, Italy
| | - Elisa Vigna
- Department of Oncology, University of Torino, Turin, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, Turin, Italy
- Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| | - Luca Primo
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy
| | - Alberto Puliafito
- Department of Oncology, University of Torino, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, TO, Italy.
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy.
| |
Collapse
|
5
|
Maas RJA, Hoogstad-van Evert JS, Hagemans IM, Brummelman J, van Ens D, de Jonge PKJD, Hooijmaijers L, Mahajan S, van der Waart AB, Hermans CKJC, de Klein J, Woestenenk R, van Herwaarden AE, Schaap NPM, Rezaeifard S, Tauriello DVF, Zusterzeel PLM, Ottevanger N, Jansen JH, Hobo W, Dolstra H. Increased peritoneal TGF-β1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer. Front Immunol 2024; 15:1448041. [PMID: 39376560 PMCID: PMC11456434 DOI: 10.3389/fimmu.2024.1448041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cell-mediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-β1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-β1 exposure. Moreover, inhibition of TGF-β1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-β1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-β1 signaling could increase NK cell immune responses in high-grade EOC patients.
Collapse
Affiliation(s)
- Ralph J. A. Maas
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janneke S. Hoogstad-van Evert
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology Amphia, Breda, Netherlands
| | - Iris M. Hagemans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolanda Brummelman
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Diede van Ens
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul K. J. D. de Jonge
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Laura Hooijmaijers
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Shweta Mahajan
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anniek B. van der Waart
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Charlotte K. J. C. Hermans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janne de Klein
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Woestenenk
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonius E. van Herwaarden
- Diagnostic Laboratory – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Somayeh Rezaeifard
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Petra L. M. Zusterzeel
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nelleke Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joop H. Jansen
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Willemijn Hobo
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Harry Dolstra
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
6
|
Bischof K, Cremaschi A, Eroukhmanoff L, Landskron J, Flage-Larsen LL, Gade A, Bjørge L, Urbanucci A, Taskén K. Patient-derived acellular ascites fluid affects drug responses in ovarian cancer cell lines through the activation of key signalling pathways. Mol Oncol 2024. [PMID: 39245677 DOI: 10.1002/1878-0261.13726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/03/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Malignant ascites is commonly produced in advanced epithelial ovarian cancer (EOC) and serves as unique microenvironment for tumour cells. Acellular ascites fluid (AAF) is rich in signalling molecules and has been proposed to play a role in the induction of chemoresistance. Through in vitro testing of drug sensitivity and by assessing intracellular phosphorylation status in response to mono- and combination treatment of five EOC cell lines after incubation with AAFs derived from 20 different patients, we investigated the chemoresistance-inducing potential of ascites. We show that the addition of AAFs to the culture media of EOC cell lines has the potential to induce resistance to standard-of-care drugs (SCDs). We also show that AAFs induce time- and concentration-dependent activation of downstream signalling to signal transducer and activator of transcription 3 (STAT3), and concomitantly altered phosphorylation of mitogen-activated protein kinase kinase (MEK), phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) and nuclear factor NF-kappa-B (NFκB). Antibodies targeting the interleukin-6 receptor (IL6R) effectively blocked phosphorylation of STAT3 and STAT1. Treatments with SCDs were effective in reducing cell viability in only a third of 30 clinically relevant conditions examined, defined as combinations of drugs, different cell lines and AAFs. Combinations of SCDs and novel therapeutics such as trametinib, fludarabine or rapamycin were superior in another third. Notably, we could nominate effective treatment combinations in almost all conditions except in 4 out of 30 conditions, in which trametinib or fludarabine showed higher efficacy alone. Taken together, our study underscores the importance of the molecular characterisation of individual patients' AAFs and the impact on treatment resistance as providing clinically meaningful information for future precision treatment approaches in EOC.
Collapse
Affiliation(s)
- Katharina Bischof
- Department of Cancer Immunology, Institute for Cancer Research, University of Oslo, Norway
- Division of Cancer Medicine, Department of Gynecological Oncology, Oslo University Hospital, Norway
| | - Andrea Cremaschi
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Norway
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lena Eroukhmanoff
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Norway
| | - Johannes Landskron
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Norway
| | - Lise-Lotte Flage-Larsen
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Norway
| | - Alexandra Gade
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo, Norway
| | - Line Bjørge
- Department of Obstetrics and Gynaecology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Alfonso Urbanucci
- Faculty of Medicine and Health Technology, TAYS Cancer Centre and FICAN Mid, Tampere University, Finland
- Department of Tumor Biology, Institute for Cancer Research, University of Oslo, Norway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, University of Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
7
|
Almeida-Nunes DL, Nunes M, Osório H, Ferreira V, Lobo C, Monteiro P, Abreu MH, Bartosch C, Silvestre R, Dinis-Oliveira RJ, Ricardo S. Ovarian cancer ascites proteomic profile reflects metabolic changes during disease progression. Biochem Biophys Rep 2024; 39:101755. [PMID: 38974022 PMCID: PMC11225207 DOI: 10.1016/j.bbrep.2024.101755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 07/09/2024] Open
Abstract
Ovarian cancer (OC) patients develop ascites, an accumulation of ascitic fluid in the peritoneal cavity anda sign of tumour dissemination within the peritoneal cavity. This body fluid is under-researched, mainly regarding the ascites formed during tumour progression that have no diagnostic value and, therefore, are discarded. We performed a discovery proteomics study to identify new biomarkers in the ascites supernatant of OC patients. In this preliminary study, we analyzed a small amount of OC ascites to highlight the importance of not discarding such biological material during treatment, which could be valuable for OC management. Our findings reveal that OC malignant ascitic fluid (MAF) displays a proliferative environment that promotes the growth of OC cells that shift the metabolic pathway using alternative sources of nutrients, such as the cholesterol pathway. Also, OC ascites drained from patients during treatment showed an immunosuppressive environment, with up-regulation of proteins from the signaling pathways of IL-4 and IL-13 and down-regulation from the MHC-II. This preliminary study pinpointed a new protein (Transmembrane Protein 132A) in the OC context that deserves to be better explored in a more extensive cohort of patients' samples. The proteomic profile of MAF from OC patients provides a unique insight into the metabolic kinetics of cancer cells during disease progression, and this information can be used to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Diana Luísa Almeida-Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
| | - Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - Hugo Osório
- Proteomics Scientific Platform, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Department of Pathology, Faculty of Medicine from University of Porto (FMUP), 4200-319, Porto, Portugal
| | - Verónica Ferreira
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Cláudia Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Paula Monteiro
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Miguel Henriques Abreu
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Carla Bartosch
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (CI-IPO-Porto) / Health Research Network (RISE@CI-IPO-Porto), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072, Porto, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine from University of Minho, 4710-057, Braga, Portugal
- ICVS/3B's – PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- UCIBIO - Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
- FOREN – Forensic Science Experts, Dr. Mário Moutinho Avenue, No. 33-A, 1400-136, Lisbon, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135, Porto, Portugal
- Associate Laboratory I4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116, Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116, Gandra, Portugal
| |
Collapse
|
8
|
Zhang W, Li J, Chen Q, Jin H, Zhou L, Liu L. Prediction of postoperative residual primary ovarian neoplasm or metastatic lesion close to rectum of serous ovarian carcinoma based on clinical and MR T1-DEI features. Acta Radiol 2024; 65:1153-1163. [PMID: 39043176 DOI: 10.1177/02841851241262520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
BACKGROUND The optimal primary debulking surgery outcome of serous ovarian carcinoma (SOC) is greatly affected by primary ovarian neoplasm or metastatic lesion close to the rectum. PURPOSE To study the risk factors affecting postoperative residual primary ovarian neoplasm or metastatic lesion close to the rectum of SOC. MATERIAL AND METHODS The clinical and MRI data of 164 patients with SOC eligible from institution A (training and test groups) and 36 patients with SOC eligible from institution B (external validation group) were collected and retrospectively analyzed. The clinical data included age, serum carbohydrate antigen 125 (CA-125), human epididymis protein 4, and neutrophil-to-lymphocyte ratio (NLR). Magnetic resonance imaging (MRI) data included ovarian mass distribution, maximum diameter of ovarian mass, ovarian mass features, degree of rectal invasion of the primary ovarian neoplasm or metastatic lesion, and amount of ascites. A model was established using multivariate logistic regression. RESULTS By univariate and multivariate logistic regressions, CA-125 (P = 0.024, odds ratio [OR] = 3.798, 95% confidence interval [CI] = 1.24-13.32), NLR (P = 0.037, OR = 3.543, 95% CI = 1.13-12.72), and degree of rectal invasion of the primary ovarian neoplasm or metastatic lesion (P < 0.001, OR = 37.723, 95% CI = 7.46-266.88) were screened as independent predictors. The area under the curve values of the model in the training, test, and external validation groups were 0.860, 0.764, and 0.778, respectively. CONCLUSION The clinical-radiological model based on T1-weighted dual-echo MRI can be used non-invasively to predict postoperative residual ovarian neoplasm or metastasis close to SOC in the rectum.
Collapse
Affiliation(s)
- Wenfei Zhang
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, Chongqing, PR China
| | - Juncai Li
- Department of Surgery, The People's Hospital of Yubei District of Chongqing City, Chongqing, PR China
| | - Qiao Chen
- School of Public Health, Chongqing Medical University, Chongqing, PR China
| | - Hongliang Jin
- Department of Osteology, The People's Hospital of Yubei District of Chongqing City, Chongqing, PR China
| | - Linyi Zhou
- Department of Radiology, Daping Hospital, Army Medical Center, Army Medical University, Chongqing, PR China
| | - Li Liu
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, Chongqing, PR China
| |
Collapse
|
9
|
Wang H, Yung MM, Xuan Y, Chen F, Chan W, Siu MK, Long R, Jia S, Liang Y, Xu D, Song Z, Tsui SK, Ngan HY, Chan KK, Chan DW. Polyunsaturated fatty acids promote M2-like TAM deposition via dampening RhoA-YAP1 signaling in the ovarian cancer microenvironment. Exp Hematol Oncol 2024; 13:90. [PMID: 39198883 PMCID: PMC11360340 DOI: 10.1186/s40164-024-00558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Peritoneal metastases frequently occur in epithelial ovarian cancer (EOC), resulting in poor prognosis and survival rates. Tumor-associated-macrophages (TAMs) massively infiltrate into ascites spheroids and are multi-polarized as protumoral M2-like phenotype, orchestrating the immunosuppression and promoting tumor progression. However, the impact of omental conditioned medium/ascites (OCM/AS) on TAM polarization and its function in tumor progression remains elusive. METHODS The distribution and polarization of TAMs in primary and omental metastatic EOC patients' tumors and ascites were examined by m-IHC, FACS analysis, and immunofluorescence. QPCR, immunofluorescence, FACS analysis, lipid staining assay, ROS assay, and Seahorse real-time cell metabolic assay characterized TAMs as being polarized in the ascites microenvironment. The oncogenic role of TAMs in tumor cells was demonstrated by co-cultured migration/invasion, proliferation, and spheroid formation assays. Mechanistic studies of the regulations of TAM polarization were performed by using RNA-Seq, GTPase pull-down, G-LISA activation assays, and other biochemical assays. A Yap1 macrophages (MФs) conditional knockout (cKO) mouse model demonstrated the roles of YAP1 in TAM polarization status and its pro-metastatic function. Finally, the anti-metastatic potential of targeting TAMs through restoring YAP1 by pharmacological agonist XMU MP1 was demonstrated in vitro and in vivo. RESULTS Abundant polyunsaturated fatty acids (PUFAs) in OCM/AS suppressed RhoA-GTPase activities, which, in turn, downregulated nuclear YAP1 in MФs, leading to increased protumoral TAM polarization accompanied by elevated OXPHOS metabolism. Abolishment of YAP1 in MФs further confirmed that a higher M2/M1 ratio of TAM polarization could alleviate CD8+ T cell infiltration and cytotoxicity in vivo. Consistently, the loss of YAP1 has been observed in EOC metastatic tissues, suggesting its clinical relevance. On the contrary, restoration of YAP1 expression by pharmaceutical inhibition of MST1/2 induced conversion of M2-to-M1-like polarized MФs, elevating the infiltration of CD8+ T cells and attenuating tumor growth. CONCLUSION This study revealed that PUFAs-enriched OCM/AS of EOC promotes M2-like TAM polarization through RhoA-YAP1 inhibition, where YAP1 downregulation is required for accelerating protumoral M2-like TAM polarization, thereby causing immunosuppression and enhancing tumor progression. Conversion of M2-to-M1-like polarized MФs through Yap1 activation inhibits tumor progression and contributes to developing potential TAMs-targeted immunotherapies in combating EOC peritoneal metastases.
Collapse
Affiliation(s)
- Huogang Wang
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Mingo Mh Yung
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Yang Xuan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Fushun Chen
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Waisun Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Michelle Ky Siu
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Runying Long
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Shuo Jia
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P.R. China
| | - Yonghao Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P.R. China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Stephen Kw Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Hextan Ys Ngan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Karen Kl Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
| | - David W Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China.
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, P.R. China.
| |
Collapse
|
10
|
Tocci P, Roman C, Sestito R, Caprara V, Sacconi A, Molineris I, Tonon G, Blandino G, Bagnato A. The endothelin-1-driven tumor-stroma feed-forward loops in high-grade serous ovarian cancer. Clin Sci (Lond) 2024; 138:851-862. [PMID: 38884602 PMCID: PMC11230866 DOI: 10.1042/cs20240346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/18/2024]
Abstract
The high-grade serous ovarian cancer (HG-SOC) tumor microenvironment (TME) is constellated by cellular elements and a network of soluble constituents that contribute to tumor progression. In the multitude of the secreted molecules, the endothelin-1 (ET-1) has emerged to be implicated in the tumor/TME interplay; however, the molecular mechanisms induced by the ET-1-driven feed-forward loops (FFL) and associated with the HG-SOC metastatic potential need to be further investigated. The tracking of the patient-derived (PD) HG-SOC cell transcriptome by RNA-seq identified the vascular endothelial growth factor (VEGF) gene and its associated signature among those mostly up-regulated by ET-1 and down-modulated by the dual ET-1R antagonist macitentan. Within the ligand-receptor pairs concurrently expressed in PD-HG-SOC cells, endothelial cells and activated fibroblasts, we discovered two intertwined FFL, the ET-1/ET-1R and VEGF/VEGF receptors, concurrently activated by ET-1 and shutting-down by macitentan, or by the anti-VEGF antibody bevacizumab. In parallel, we observed that ET-1 fine-tuned the tumoral and stromal secretome toward a pro-invasive pattern. Into the fray of the HG-SOC/TME double and triple co-cultures, the secretion of ET-1 and VEGF, that share a common co-regulation, was inhibited upon the administration of macitentan. Functionally, macitentan, mimicking the effect of bevacizumab, interfered with the HG-SOC/TME FFL-driven communication that fuels the HG-SOC invasive behavior. The identification of ET-1 and VEGF FFL as tumor and TME actionable vulnerabilities, reveals how ET-1R blockade, targeting the HG-SOC cells and the TME simultaneously, may represent an effective therapeutic option for HG-SOC patients.
Collapse
Affiliation(s)
- Piera Tocci
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Celia Roman
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Rosanna Sestito
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Valentina Caprara
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Sacconi
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Ivan Molineris
- Department of Life Science and System Biology, University of Turin, Turin, Italy
| | - Giovanni Tonon
- Center for Omics Sciences (COSR) and Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
11
|
Villagomez FR, Lang J, Rosario FJ, Nunez-Avellaneda D, Webb P, Neville M, Woodruff ER, Bitler BG. Claudin-4 Modulates Autophagy via SLC1A5/LAT1 as a Mechanism to Regulate Micronuclei. CANCER RESEARCH COMMUNICATIONS 2024; 4:1625-1642. [PMID: 38867360 PMCID: PMC11218812 DOI: 10.1158/2767-9764.crc-24-0240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
Genome instability is a hallmark of cancer crucial for tumor heterogeneity and is often a result of defects in cell division and DNA damage repair. Tumors tolerate genomic instability, but the accumulation of genetic aberrations is regulated to avoid catastrophic chromosomal alterations and cell death. In ovarian cancer tumors, claudin-4 is frequently upregulated and closely associated with genome instability and worse patient outcomes. However, its biological association with regulating genomic instability is poorly understood. Here, we used CRISPR interference and a claudin mimic peptide to modulate the claudin-4 expression and its function in vitro and in vivo. We found that claudin-4 promotes a tolerance mechanism for genomic instability through micronuclei generation in tumor cells. Disruption of claudin-4 increased autophagy and was associated with the engulfment of cytoplasm-localized DNA. Mechanistically, we observed that claudin-4 establishes a biological axis with the amino acid transporters SLC1A5 and LAT1, which regulate autophagy upstream of mTOR. Furthermore, the claudin-4/SLC1A5/LAT1 axis was linked to the transport of amino acids across the plasma membrane as one of the potential cellular processes that significantly decreased survival in ovarian cancer patients. Together, our results show that the upregulation of claudin-4 contributes to increasing the threshold of tolerance for genomic instability in ovarian tumor cells by limiting its accumulation through autophagy. SIGNIFICANCE Autophagy regulation via claudin-4/SLC1A5/LAT1 has the potential to be a targetable mechanism to interfere with genomic instability in ovarian tumor cells.
Collapse
Affiliation(s)
- Fabian R. Villagomez
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Julie Lang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Fredrick J. Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Daniel Nunez-Avellaneda
- Deputy Directorate of Technological Development, Linkage, and Innovation, National Council of Humanities, Sciences, and Technologies, Mexico City, Mexico.
| | - Patricia Webb
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Margaret Neville
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Elizabeth R. Woodruff
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| | - Benjamin G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
12
|
Kaur K, Sanghu J, Memarzadeh S, Jewett A. Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas. Vaccines (Basel) 2024; 12:677. [PMID: 38932405 PMCID: PMC11209217 DOI: 10.3390/vaccines12060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
High-grade serous ovarian cancers (HGSOCs) likely consist of poorly differentiated stem-like cells (PDSLCs) and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs, contributing to disease progression and tumor relapse. Primary NK cells are known to effectively lyse PDSLCs, but they exhibit low or minimal cytotoxic potential against well-differentiated tumors. We have introduced and discussed the characteristics of super-charged NK (sNK) cells in this review. sNK cells, in comparison to primary NK cells, exhibit a significantly higher capability for the direct killing of both PDSLCs and well-differentiated tumors. In addition, sNK cells secrete significantly higher levels of cytokines, especially those known to induce the differentiation of tumors. In addition, we propose that a combination of sNK and chemotherapy could be one of the most effective strategies to eliminate the heterogeneous population of ovarian tumors; sNK cells can lyse both PDSLCs and well-differentiated tumors, induce the differentiation of PDSLCs, and could be used in combination with chemotherapy to target both well-differentiated and NK-induced differentiated tumors.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| | - Jashan Sanghu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
13
|
Balan D, Kampan NC, Plebanski M, Abd Aziz NH. Unlocking ovarian cancer heterogeneity: advancing immunotherapy through single-cell transcriptomics. Front Oncol 2024; 14:1388663. [PMID: 38873253 PMCID: PMC11169633 DOI: 10.3389/fonc.2024.1388663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Ovarian cancer, a highly fatal gynecological cancer, warrants the need for understanding its heterogeneity. The disease's prevalence and impact are underscored with statistics on mortality rates. Ovarian cancer is categorized into distinct morphological groups, each with its characteristics and prognosis. Despite standard treatments, survival rates remain low due to relapses and chemoresistance. Immune system involvement is evident in ovarian cancer's progression, although the tumor employs immune evasion mechanisms. Immunotherapy, particularly immune checkpoint blockade therapy, is promising, but ovarian cancer's heterogeneity limits its efficacy. Single-cell sequencing technology could be explored as a solution to dissect the heterogeneity within tumor-associated immune cell populations and tumor microenvironments. This cutting-edge technology has the potential to enhance diagnosis, prognosis, and personalized immunotherapy in ovarian cancer, reflecting its broader application in cancer research. The present review focuses on recent advancements and the challenges in applying single-cell transcriptomics to ovarian cancer.
Collapse
Affiliation(s)
- Dharvind Balan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Nor Haslinda Abd Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Li R, Xiong Z, Ma Y, Li Y, Yang Y, Ma S, Ha C. Enhancing precision medicine: a nomogram for predicting platinum resistance in epithelial ovarian cancer. World J Surg Oncol 2024; 22:81. [PMID: 38509620 PMCID: PMC10956367 DOI: 10.1186/s12957-024-03359-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND This study aimed to develop a novel nomogram that can accurately estimate platinum resistance to enhance precision medicine in epithelial ovarian cancer(EOC). METHODS EOC patients who received primary therapy at the General Hospital of Ningxia Medical University between January 31, 2019, and June 30, 2021 were included. The LASSO analysis was utilized to screen the variables which contained clinical features and platinum-resistance gene immunohistochemistry scores. A nomogram was created after the logistic regression analysis to develop the prediction model. The consistency index (C-index), calibration curve, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA) were used to assess the nomogram's performance. RESULTS The logistic regression analysis created a prediction model based on 11 factors filtered down by LASSO regression. As predictors, the immunohistochemical scores of CXLC1, CXCL2, IL6, ABCC1, LRP, BCL2, vascular tumor thrombus, ascites cancer cells, maximum tumor diameter, neoadjuvant chemotherapy, and HE4 were employed. The C-index of the nomogram was found to be 0.975. The nomogram's specificity is 95.35% and its sensitivity, with a cut-off value of 165.6, is 92.59%, as seen by the ROC curve. After the nomogram was externally validated in the test cohort, the coincidence rate was determined to be 84%, and the ROC curve indicated that the nomogram's AUC was 0.949. CONCLUSION A nomogram containing clinical characteristics and platinum gene IHC scores was developed and validated to predict the risk of EOC platinum resistance.
Collapse
Affiliation(s)
- Ruyue Li
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Zhuo Xiong
- Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Department of Gynecologic Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Yuan Ma
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
- Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Yongmei Li
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Yu'e Yang
- Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Shaohan Ma
- Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Chunfang Ha
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
- Department of Gynecologic Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
- Key Laboratory of Reproduction and Genetic of Ningxia Hui Autonomous Region, Key Laboratory of Fertility Preservation and Maintenance of Ningxia Medical University and Ministry of Education of China, Department of Histology and Embryology in, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China.
| |
Collapse
|
15
|
Constantinescu DR, Sorop A, Ghionescu AV, Lixandru D, Herlea V, Bacalbasa N, Dima SO. EM-transcriptomic signature predicts drug response in advanced stages of high-grade serous ovarian carcinoma based on ascites-derived primary cultures. Front Pharmacol 2024; 15:1363142. [PMID: 38510654 PMCID: PMC10953505 DOI: 10.3389/fphar.2024.1363142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction: High-grade serous ovarian carcinoma (HGSOC) remains a medical challenge despite considerable improvements in the treatment. Unfortunately, over 75% of patients have already metastasized at the time of diagnosis. Advances in understanding the mechanisms underlying how ascites cause chemoresistance are urgently needed to derive novel therapeutic strategies. This study aimed to identify the molecular markers involved in drug sensitivity and highlight the use of ascites as a potential model to investigate HGSOC treatment options. Methods: After conducting an in silico analysis, eight epithelial-mesenchymal (EM)-associated genes related to chemoresistance were identified. To evaluate differences in EM-associated genes in HGSOC samples, we analyzed ascites-derived HGSOC primary cell culture (AS), tumor (T), and peritoneal nodule (NP) samples. Moreover, in vitro experiments were employed to measure tumor cell proliferation and cell migration in AS, following treatment with doxorubicin (DOX) and cisplatin (CIS) and expression of these markers. Results: Our results showed that AS exhibits a mesenchymal phenotype compared to tumor and peritoneal nodule samples. Moreover, DOX and CIS treatment leads to an invasive-intermediate epithelial-to-mesenchymal transition (EMT) state of the AS by different EM-associated marker expression. For instance, the treatment of AS showed that CDH1 and GATA6 decreased after CIS exposure and increased after DOX treatment. On the contrary, the expression of KRT18 has an opposite pattern. Conclusion: Taken together, our study reports a comprehensive investigation of the EM-associated genes after drug exposure of AS. Exploring ascites and their associated cellular and soluble components is promising for understanding the HGSOC progression and treatment response at a personalized level.
Collapse
Affiliation(s)
| | - Andrei Sorop
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Daniela Lixandru
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Vlad Herlea
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Department of Pathology-Fundeni Clinical Institute, Bucharest, Romania
| | - Nicolae Bacalbasa
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Simona Olimpia Dima
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
- Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
16
|
Grunt TW, Wagner R, Ries A, Berghoff AS, Preusser M, Grusch M, Valent P. Targeting endogenous fatty acid synthesis stimulates the migration of ovarian cancer cells to adipocytes and promotes the transport of fatty acids from adipocytes to cancer cells. Int J Oncol 2024; 64:24. [PMID: 38214315 PMCID: PMC10807641 DOI: 10.3892/ijo.2024.5612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
Despite significant advances in oncology, 1 of 108 female patients succumb to ovarian cancer (OC) each year. Improved novel treatments against this aggressive disease would be a major improvement. The growth of OC cells has been demonstrated to be highly dependent on lipids. OC cells are abundantly present in the abdominal cavity and omentum, the main sites of OC expansion. Accordingly, it has been attempted not only to block the hyperactive synthesis of fatty acids (FAs) in cancer cells, but also to disrupt lipid supply. While either strategy has yielded promising results as monotherapy, the induction of resistance pathways diminishing the anticancer effects is yet conceivable. The endogenous regulation of lipid biosynthesis in OC has been extensively studied. However, the role of stromal cells in the modulation of the effects of anti‑lipogenic drugs has not yet been well documented. The present study thus examined the interaction between OC cells and associated stromal cells, when de novo FA synthesis was blocked. It has recently been revealed by the authors that when FA are provided to OC cells in monoculture, the lipid deficiency induced by pharmacological inhibition of FA synthase (FASN), the key enzyme of endogenous FA synthesis, cannot be compensated through an increased FA uptake by OC cells. In the present study, OC cells were co‑cultured with adipocytes preloaded with fluorescent FA and the effects of FASN‑inhibition on OC homing to adipocytes and the transcellular delivery of fluorescent FA from adipocytes to OC cells were examined. The FASN inhibitors, G28UCM and Fasnall, stimulated the spontaneous migration of A2780 OC cells in a concentration‑dependent manner and stimulated the transfer of FA from adipocytes to OC cells. Similar effects were observed with all types of adipocytes tested. The models applied in the present study demonstrated that co‑cultured cancer‑associated adipocytes may attenuate the anticancer effects of FASN inhibitors by attracting tumor cells and by supplying the cells with FA. This lipid‑mediated dependency may provide a rationale for the design of new treatment approaches for the treatment of OC.
Collapse
Affiliation(s)
- Thomas W. Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Renate Wagner
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Alexander Ries
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Anna S. Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Grusch
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Peter Valent
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
- Division of Hematology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
17
|
Śliwa A, Szczerba A, Pięta PP, Białas P, Lorek J, Nowak-Markwitz E, Jankowska A. A Recipe for Successful Metastasis: Transition and Migratory Modes of Ovarian Cancer Cells. Cancers (Basel) 2024; 16:783. [PMID: 38398174 PMCID: PMC10886816 DOI: 10.3390/cancers16040783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
One of the characteristic features of ovarian cancer is its early dissemination. Metastasis and the invasiveness of ovarian cancer are strongly dependent on the phenotypical and molecular determinants of cancer cells. Invasive cancer cells, circulating tumor cells, and cancer stem cells, which are responsible for the metastatic process, may all undergo different modes of transition, giving rise to mesenchymal, amoeboid, and redifferentiated epithelial cells. Such variability is the result of the changing needs of cancer cells, which strive to survive and colonize new organs. This would not be possible if not for the variety of migration modes adopted by the transformed cells. The most common type of metastasis in ovarian cancer is dissemination through the transcoelomic route, but transitions in ovarian cancer cells contribute greatly to hematogenous and lymphatic dissemination. This review aims to outline the transition modes of ovarian cancer cells and discuss the migratory capabilities of those cells in light of the known ovarian cancer metastasis routes.
Collapse
Affiliation(s)
- Aleksandra Śliwa
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Anna Szczerba
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Paweł Piotr Pięta
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Piotr Białas
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Jakub Lorek
- Gynecologic Oncology Department, Poznan University of Medical Sciences, 33 Polna Street, 60-101 Poznan, Poland
| | - Ewa Nowak-Markwitz
- Gynecologic Oncology Department, Poznan University of Medical Sciences, 33 Polna Street, 60-101 Poznan, Poland
| | - Anna Jankowska
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| |
Collapse
|
18
|
Cao L, Zhang S, Peng H, Lin Y, Xi Z, Lin W, Guo J, Wu G, Yu F, Zhang H, Ye H. Identification and validation of anoikis-related lncRNAs for prognostic significance and immune microenvironment characterization in ovarian cancer. Aging (Albany NY) 2024; 16:1463-1483. [PMID: 38226979 PMCID: PMC10866438 DOI: 10.18632/aging.205439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024]
Abstract
Anoikis, a form of apoptotic cell death resulting from inadequate cell-matrix interactions, has been implicated in tumor progression by regulating tumor angiogenesis and metastasis. However, the potential roles of anoikis-related long non-coding RNAs (arlncRNAs) in the tumor microenvironment are not well understood. In this study, five candidate lncRNAs were screened through least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analysis based on differentially expressed lncRNAs associated with anoikis-related genes (ARGs) from TCGA and GSE40595 datasets. The prognostic accuracy of the risk model was evaluated using Kaplan-Meier survival analysis and receiver operating characteristic (ROC) curves. Furthermore, Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis (GSEA) analyses revealed significant differences in immune-related hallmarks and signal transduction pathways between the high-risk and low-risk groups. Additionally, immune infiltrate analysis showed significant differences in the distribution of macrophages M2, follicular T helper cells, plasma cells, and neutrophils between the two risk groups. Lastly, silencing the expression of PRR34_AS1 and SPAG5_AS1 significantly increased anoikis-induced cell death in ovarian cancer cells. In conclusion, our study constructed a risk model that can predict clinicopathological features, tumor microenvironment characteristics, and prognosis of ovarian cancer patients. The immune-related pathways identified in this study may offer new treatment strategies for ovarian cancer.
Collapse
Affiliation(s)
- Lixue Cao
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shaofen Zhang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Haojie Peng
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yongqing Lin
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihui Xi
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wumei Lin
- Department of Gynecology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jialing Guo
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Geyan Wu
- Biomedicine Research Centre, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haiyan Ye
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Rebordão-Pires M, Estrada MF, Gomes A, Silva F, Baptista C, Ramos MJ, Fortuna A, Simões P, Sousa G, Marreiros A, Fior R. Relapsed Ovarian Cancer Patients with Ascites and/or Pleural Effusion Still Benefit from Treatment: A Real-Life Study. Cancers (Basel) 2023; 16:162. [PMID: 38201589 PMCID: PMC10778384 DOI: 10.3390/cancers16010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
(1) Background: Relapsed HGSOC with ascites and/or pleural effusion is a poor-prognostic population and poorly represented in clinical studies. We questioned if these patients are worth treating. In other words, if these patients received the most effective treatment, would it change the course of this disease? To our knowledge this is the first real-life study to evaluate this question in this low-survival population. (2) Methods: To tackle this question we performed a retrospective, multi-centric, real-life study, that reviewed relapsed HGSOC patients with ascites and/or pleural effusion. Our rationale was to compare the OS of two groups of patients: responders, i.e., patients who had an imagological response to treatment (complete/partial response/stable disease, RECIST criteria) versus non-responders (no response/progression upon treatment). We evaluated the predictive value of clinical variables that are available in a real-life setting (e.g., staging, chemotherapy, surgery, platinum-sensitivity). Multivariate logistic regression and survival analysis was conducted. A two-step cluster analysis SPSS tool was used for subgroup analysis. Platinum sensitivity/resistance was also analyzed, as well as multivariate and cluster analysis. (3) Results: We included 57 patients, 41.4% first line responders and 59.6% non-responders. The median OS of responders was 23 months versus 8 months in non-responders (p < 0.001). This difference was verified in platinum-sensitive (mOS 28 months vs. 8 months, p < 0.001) and platinum-resistant populations (mOS 16 months vs. 7 months, p < 0.001). Thirty-one patients reached the second line, of which only 10.3% responded to treatment. Three patients out of thirty-one who did not respond in the first line of relapse, responded in the second line. In the second line, the mOS for the responders' group vs. non-responders was 31 months versus 13 months (p = 0.02). The two step cluster analysis tool found two different subgroups with different prognoses based on overall response rate, according to consolidation chemotherapy, neoadjuvant chemotherapy, FIGO staging and surgical treatment. Cluster analysis showed that even patients with standard clinical and treatment variables associated with poor prognosis might achieve treatment response (the opposite being also true). (4) Conclusions: Our data clearly show that relapsed HGSOC patients benefit from treatment. If given an effective treatment upfront, this can lead to a ~3 times increase in mOS for these patients. Moreover, this was irrespective of patient disease and treatment characteristics. Our results highlight the urgent need for a sensitivity test to tailor treatments and improve efficacy rates in a personalized manner.
Collapse
Affiliation(s)
- Mariana Rebordão-Pires
- Medical Oncology Unit, Instituto Português De Oncologia De Coimbra Francisco Gentil, 3000-075 Coimbra, Portugal; (M.R.-P.); (G.S.)
| | - Marta F. Estrada
- Cancer Development and Innate Immune Evasion Laboratory, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | - António Gomes
- Surgery Unit, Hospital Vila Franca de Xira, 2600-153 Vila Franca de Xira, Portugal;
| | - Filipa Silva
- Gynecology Unit, Champalimaud Foundation, 1400-038 Lisboa, Portugal;
| | - Carlota Baptista
- Medical Oncology Unit, Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (C.B.); (P.S.)
| | - Maria João Ramos
- Medical Oncology Unit, Centro Hospitalar Universitário de Santo António, 4099-001 Porto, Portugal;
| | - Ana Fortuna
- Medical Oncology Unit, Centro Hospitalar do Algarve, 8500-338 Portimão, Portugal;
| | - Pedro Simões
- Medical Oncology Unit, Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (C.B.); (P.S.)
| | - Gabriela Sousa
- Medical Oncology Unit, Instituto Português De Oncologia De Coimbra Francisco Gentil, 3000-075 Coimbra, Portugal; (M.R.-P.); (G.S.)
| | - Ana Marreiros
- Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal;
- Algarve Biomedical Center Research Institute, University of Algarve, 8005-139 Faro, Portugal
| | - Rita Fior
- Cancer Development and Innate Immune Evasion Laboratory, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| |
Collapse
|
20
|
Blanc-Durand F, Clemence Wei Xian L, Tan DSP. Targeting the immune microenvironment for ovarian cancer therapy. Front Immunol 2023; 14:1328651. [PMID: 38164130 PMCID: PMC10757966 DOI: 10.3389/fimmu.2023.1328651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
Ovarian cancer (OC) is an aggressive malignancy characterized by a complex immunosuppressive tumor microenvironment (TME). Immune checkpoint inhibitors have emerged as a breakthrough in cancer therapy by reactivating the antitumor immune response suppressed by tumor cells. However, in the case of OC, these inhibitors have failed to demonstrate significant improvements in patient outcomes, and existing biomarkers have not yet identified promising subgroups. Consequently, there remains a pressing need to understand the interplay between OC tumor cells and their surrounding microenvironment to develop effective immunotherapeutic approaches. This review aims to provide an overview of the OC TME and explore its potential as a therapeutic strategy. Tumor-infiltrating lymphocytes (TILs) are major actors in OC TME. Evidence has been accumulating regarding the spontaneous TILS response against OC antigens. Activated T-helpers secrete a wide range of inflammatory cytokines with a supportive action on cytotoxic T-cells. Simultaneously, mature B-cells are recruited and play a significant antitumor role through opsonization of target antigens and T-cell recruitment. Macrophages also form an important subset of innate immunity (M1-macrophages) while participating in the immune-stimulation context. Finally, OC has shown to engage a significant natural-killer-cells immune response, exerting direct cytotoxicity without prior sensitization. Despite this initial cytotoxicity, OC cells develop various strategies to induce an immune-tolerant state. To this end, multiple immunosuppressive molecules are secreted to impair cytotoxic cells, recruit regulatory cells, alter antigen presentation, and effectively evade immune response. Consequently, OC TME is predominantly infiltrated by immunosuppressive cells such as FOXP3+ regulatory T-cells, M2-polarized macrophages and myeloid-derived suppressor cells. Despite this strong immunosuppressive state, PD-1/PD-L1 inhibitors have failed to improve outcomes. Beyond PD-1/PD-L1, OC expresses multiple other immune checkpoints that contribute to immune evasion, and each representing potential immune targets. Novel immunotherapies are attempting to overcome the immunosuppressive state and induce specific immune responses using antibodies adoptive cell therapy or vaccines. Overall, the OC TME presents both opportunities and obstacles. Immunotherapeutic approaches continue to show promise, and next-generation inhibitors offer exciting opportunities. However, tailoring therapies to individual immune characteristics will be critical for the success of these treatments.
Collapse
Affiliation(s)
- Felix Blanc-Durand
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - Lai Clemence Wei Xian
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine and Cancer Science Institute (CSI), National University of Singapore (NUS), Singapore, Singapore
| | - David S. P. Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University Centre for Cancer Research (N2CR) and Cancer Science Institute (CSI), National University of Singapore, Singapore, Singapore
| |
Collapse
|
21
|
Monavarian M, Page EF, Rajkarnikar R, Kumari A, Macias LQ, Massicano F, Lee NY, Sahoo S, Hempel N, Jolly MK, Ianov L, Worthey E, Singh A, Broude EV, Mythreye K. Development of adaptive anoikis resistance promotes metastasis that can be overcome by CDK8/19 Mediator kinase inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.04.569970. [PMID: 38106208 PMCID: PMC10723298 DOI: 10.1101/2023.12.04.569970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Anoikis resistance or evasion of cell death triggered by cell detachment into suspension is a hallmark of cancer that is concurrent with cell survival and metastasis. The effects of frequent matrix detachment encounters on the development of anoikis resistance in cancer remains poorly defined. Here we show using a panel of ovarian cancer models, that repeated exposure to suspension stress in vitro followed by attached recovery growth leads to the development of anoikis resistance paralleling in vivo development of anoikis resistance in ovarian cancer ascites. This resistance is concurrent with enhanced invasion, chemoresistance and the ability of anoikis adapted cells to metastasize to distant sites. Adapted anoikis resistant cells show a heightened dependency on oxidative phosphorylation and can also evade immune surveillance. We find that such acquired anoikis resistance is not genetic, as acquired resistance persists for a finite duration in the absence of suspension stress. Transcriptional reprogramming is however essential to this process, as acquisition of adaptive anoikis resistance in vitro and in vivo is exquisitely sensitive to inhibition of CDK8/19 Mediator kinase, a pleiotropic regulator of transcriptional reprogramming. Our data demonstrate that growth after recovery from repeated exposure to suspension stress is a direct contributor to metastasis and that inhibition of CDK8/19 Mediator kinase during such adaptation provides a therapeutic opportunity to prevent both local and distant metastasis in cancer.
Collapse
Affiliation(s)
- Mehri Monavarian
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| | - Emily Faith Page
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| | - Resha Rajkarnikar
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| | - Asha Kumari
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| | - Liz Quintero Macias
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| | - Felipe Massicano
- UAB Biological Data Science Core, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nam Y Lee
- Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, 85721, USA
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Nadine Hempel
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh School of Medicine Pittsburgh PA 15213
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, 560012, India
| | - Lara Ianov
- UAB Biological Data Science Core, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elizabeth Worthey
- UAB Biological Data Science Core, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716, USA
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Karthikeyan Mythreye
- Division of Molecular Cellular Pathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama, Heersink School of Medicine, Birmingham, AL, USA
| |
Collapse
|
22
|
Choi JA, Kim H, Kwon H, Lee EH, Cho H, Chung JY, Kim JH. Ascitic autotaxin as a potential prognostic, diagnostic, and therapeutic target for epithelial ovarian cancer. Br J Cancer 2023; 129:1184-1194. [PMID: 37596406 PMCID: PMC10539369 DOI: 10.1038/s41416-023-02355-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Malignant ascites contributes to the metastatic process by facilitating the multifocal dissemination of ovarian tumour cells onto the peritoneal surface. However, the prognostic and diagnostic relevance of ascitic fluid remains largely unknown. Herein, we investigated the potential clinical value and therapeutic utility of ascitic autotaxin (ATX) in epithelial ovarian cancer (EOC). METHODS ATX expression was assessed in clinical samples. Spheroid-forming assay, real-time PCR, western blot analysis, invadopodia assay, and adhesion assays were performed. RESULTS Ascitic ATX expression was highly elevated in patients with ovarian cancer compared to those with benign ascites and was associated with advanced stage, high grade, and a short disease-free period in patients with EOC. Combining the diagnostic ability of ascitic ATX and serum CA-125 levels significantly improved the area under the curve (AUC) value for EOC compared to serum CA125 level alone. This marker combination showed a large odds ratio for short disease-free period in high-risk EOC groups. Functional studies revealed that ascitic ATX was required for maintaining cancer stem cell-like characteristics and invadopodia formation. CONCLUSION Ascitic ATX levels may serve as a useful prognostic indicator for predicting aggressive behaviour in EOC. ATX-linked invadopodia are a potential target to prevent peritoneal dissemination in ovarian cancer.
Collapse
Affiliation(s)
- Jung-A Choi
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyosun Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hyunja Kwon
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Elizabeth Hyeji Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
23
|
Jiao Y, Peng X, Wang Y, Hao Z, Chen L, Wu M, Zhang Y, Li J, Li W, Zhan X. Malignant ascites supernatant enhances the proliferation of gastric cancer cells partially via the upregulation of asparagine synthetase. Oncol Lett 2023; 26:418. [PMID: 37664666 PMCID: PMC10472050 DOI: 10.3892/ol.2023.14005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/09/2023] [Indexed: 09/05/2023] Open
Abstract
Malignant ascites (MA) is a common manifestation of advanced gastric cancer (GC) with peritoneal metastasis (PM), which usually indicates a poor prognosis. The present study aimed to explore the effects of MA, a unique microenvironment of PM, on the proliferation of cancer cells and investigate the underlying mechanisms. Ex vivo experiments demonstrated that GC cells treated with MA exhibited enhanced proliferation. RNA sequencing indicated that asparagine synthetase (ASNS) was one of the differentially expressed genes in GC cells following incubation with MAs. Furthermore, the present study suggested that MA induced an upregulation of ASNS expression and the stimulatory effect of MA on cancer cell proliferation was alleviated upon ASNS downregulation. Activating transcription factor 4 (ATF4), a pivotal transcription factor regulating ASNS, was upregulated when cells were treated with MA supernatant. After ATF4 knockdown, the proliferation of MA-treated GC cells and the expression of ASNS decreased. In addition, the decline in the proliferation of the ATF4-downregulated AGS GC cell line was rescued by ASNS upregulation. The findings indicated that MA could promote the proliferation of GC cells via activation of the ATF4-ASNS axis. Hence, it may be a potential target for treating GC with PM and MA.
Collapse
Affiliation(s)
- Yuan Jiao
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yujie Wang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Ling Chen
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Meihong Wu
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Jie Li
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, Shanghai 200433, P.R. China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
24
|
Hrvat A, Schmidt M, Wagner B, Zwanziger D, Kimmig R, Volbracht L, Brandau S, Mallmann-Gottschalk N. Electrolyte imbalance causes suppression of NK and T cell effector function in malignant ascites. J Exp Clin Cancer Res 2023; 42:235. [PMID: 37684704 PMCID: PMC10485936 DOI: 10.1186/s13046-023-02798-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Malignant ascites commonly occurs in advanced or recurrent stages of epithelial ovarian cancer during peritoneal carcinomatosis and is correlated with poor prognosis. Due to its complex composition of cellular and acellular components malignant ascites creates a unique tumor microenvironment, which mediates immunosuppression and promotes progression of disease. However, the immunosuppressive mechanisms remain poorly understood. METHODS In the present study, we explored the antitumor activity of healthy donor NK and T cells directed against ovarian cancer cells in presence of malignant ascites derived from patients with advanced or recurrent peritoneal carcinomatosis. A wide range of methods was used to study the effect of ascites on NK and T cells (FACS, ELISA, EliSpot, qPCR, Live-cell and confocal microscopy, Western blot and electrolyte flux assays). The ascites components were assessed using quantitative analysis (nephelometry, potentiometry and clinical chemistry) and separation methods (dialysis, ultracentrifugal filtration and lipid depletion). RESULTS Ascites rapidly inhibited NK cell degranulation, tumor lysis, cytokine secretion and calcium signaling. Similarly, target independent NK and T cell activation was impaired in ascites environment. We identified imbalanced electrolytes in ascites as crucial factors causing extensive immunosuppression of NK and T cells. Specifically, high sodium, low chloride and low potassium content significantly suppressed NK-mediated cytotoxicity. Electrolyte imbalance led to changes in transcription and protein expression of electrolyte channels and impaired NK and T cell activation. Selected inhibitors of sodium electrolyte channels restored intracellular calcium flux, conjugation, degranulation and transcript expression of signaling molecules. The levels of ascites-mediated immunosuppression and sodium/chloride/potassium imbalance correlated with poor patient outcome and selected molecular alterations were confirmed in immune cells from ovarian cancer patients. CONCLUSION Our data suggest a novel electrolyte-based mechanism of immunosuppression in malignant ascites of patients with peritoneal carcinomatosis. We show for the first time that the immunosuppression of NK cytotoxicity in coculture assays is correlated to patient poor survival. Therapeutic application of sodium channel inhibitors may provide new means for restoring immune cell activity in ascites or similar electrolyte imbalanced environments.
Collapse
Affiliation(s)
- Antonio Hrvat
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Mathias Schmidt
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Bernd Wagner
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
| | - Denise Zwanziger
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, 45147, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147, Essen, Germany
| | - Lothar Volbracht
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
| | - Sven Brandau
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
- partner site Essen-Düsseldorf, German Cancer Consortium (DKTK), 45147, Essen, Germany.
| | - Nina Mallmann-Gottschalk
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Department of Gynecology and Obstetrics, University Hospital of Cologne, 50931, Cologne, Germany
| |
Collapse
|
25
|
Wilczyński JR, Wilczyński M, Paradowska E. "DEPHENCE" system-a novel regimen of therapy that is urgently needed in the high-grade serous ovarian cancer-a focus on anti-cancer stem cell and anti-tumor microenvironment targeted therapies. Front Oncol 2023; 13:1201497. [PMID: 37448521 PMCID: PMC10338102 DOI: 10.3389/fonc.2023.1201497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer, especially high-grade serous type, is the most lethal gynecological malignancy. The lack of screening programs and the scarcity of symptomatology result in the late diagnosis in about 75% of affected women. Despite very demanding and aggressive surgical treatment, multiple-line chemotherapy regimens and both approved and clinically tested targeted therapies, the overall survival of patients is still unsatisfactory and disappointing. Research studies have recently brought some more understanding of the molecular diversity of the ovarian cancer, its unique intraperitoneal biology, the role of cancer stem cells, and the complexity of tumor microenvironment. There is a growing body of evidence that individualization of the treatment adjusted to the molecular and biochemical signature of the tumor as well as to the medical status of the patient should replace or supplement the foregoing therapy. In this review, we have proposed the principles of the novel regimen of the therapy that we called the "DEPHENCE" system, and we have extensively discussed the results of the studies focused on the ovarian cancer stem cells, other components of cancer metastatic niche, and, finally, clinical trials targeting these two environments. Through this, we have tried to present the evolving landscape of treatment options and put flesh on the experimental approach to attack the high-grade serous ovarian cancer multidirectionally, corresponding to the "DEPHENCE" system postulates.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother's Health Center-Research Institute, Lodz, Poland
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| |
Collapse
|
26
|
Malgundkar SH, Tamimi Y. Exosomes as crucial emerging tools for intercellular communication with therapeutic potential in ovarian cancer. Future Sci OA 2023; 9:FSO833. [PMID: 37006229 PMCID: PMC10051132 DOI: 10.2144/fsoa-2022-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than two-thirds of epithelial ovarian cancer (EOC) patients are diagnosed at advanced stages due to the lack of sensitive biomarkers. Currently, exosomes are intensively investigated as non-invasive cancer diagnostic markers. Exosomes are nanovesicles released in the extracellular milieu with the potential to modulate recipient cells' behavior. EOC cells release many altered exosomal cargoes that exhibit clinical relevance to tumor progression. Exosomes represent powerful therapeutic tools (drug carriers or vaccines), posing a promising option in clinical practice for curing EOC in the near future. In this review, we highlight the importance of exosomes in cell–cell communication, epithelial–mesenchymal transition (EMT), and their potential to serve as diagnostic and prognostic factors, particularly in EOC.
Collapse
Affiliation(s)
- Shika Hanif Malgundkar
- Department of Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, PO Box 35, PC 123, Muscat, Sultanate of Oman
| | - Yahya Tamimi
- Department of Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, PO Box 35, PC 123, Muscat, Sultanate of Oman
| |
Collapse
|
27
|
Rickard BP, Overchuk M, Obaid G, Ruhi MK, Demirci U, Fenton SE, Santos JH, Kessel D, Rizvi I. Photochemical Targeting of Mitochondria to Overcome Chemoresistance in Ovarian Cancer †. Photochem Photobiol 2023; 99:448-468. [PMID: 36117466 PMCID: PMC10043796 DOI: 10.1111/php.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy with a stubborn mortality rate of ~65%. The persistent failure of multiline chemotherapy, and significant tumor heterogeneity, has made it challenging to improve outcomes. A target of increasing interest is the mitochondrion because of its essential role in critical cellular functions, and the significance of metabolic adaptation in chemoresistance. This review describes mitochondrial processes, including metabolic reprogramming, mitochondrial transfer and mitochondrial dynamics in ovarian cancer progression and chemoresistance. The effect of malignant ascites, or excess peritoneal fluid, on mitochondrial function is discussed. The role of photodynamic therapy (PDT) in overcoming mitochondria-mediated resistance is presented. PDT, a photochemistry-based modality, involves the light-based activation of a photosensitizer leading to the production of short-lived reactive molecular species and spatiotemporally confined photodamage to nearby organelles and biological targets. The consequential effects range from subcytotoxic priming of target cells for increased sensitivity to subsequent treatments, such as chemotherapy, to direct cell killing. This review discusses how PDT-based approaches can address key limitations of current treatments. Specifically, an overview of the mechanisms by which PDT alters mitochondrial function, and a summary of preclinical advancements and clinical PDT experience in ovarian cancer are provided.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson TX 95080, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Suzanne E. Fenton
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David Kessel
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Imran Rizvi
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; North Carolina State University, Raleigh, NC 27606, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Kampan NC, Kartikasari AER, Deceneux C, Madondo MT, McNally OM, Flanagan KL, Aziz NA, Stephens AN, Reynolds J, Quinn MA, Plebanski M. Combining TNFR2-Expressing Tregs and IL-6 as Superior Diagnostic Biomarkers for High-Grade Serous Ovarian Cancer Masses. Cancers (Basel) 2023; 15:667. [PMID: 36765633 PMCID: PMC9913655 DOI: 10.3390/cancers15030667] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
We hypothesised that the inclusion of immunosuppressive and inflammatory biomarkers in HGSOC patients would improve the sensitivity and specificity of the preoperative marker prediction of malignancy in patients with ovarian masses. We tested a panel of 29 soluble immune factors by multiplex bead immunoassay and 16 phenotypic T cell markers by flow cytometry in pre-treatment blood samples from 66 patients undergoing surgery for suspected ovarian cancer or ovarian cancer risk reduction. The potential diagnostic utility of all parameters was explored using Volcano plots, principal component analysis (PCA) and receiver operator characteristic (ROC) analysis. We also assessed the effect of culturing PBMCs from 20 healthy donors in the presence of malignant ascites fluid. The combination of TNFR2+ Tregs and IL-6 in the pre-treatment blood of patients with advanced HGSOC effectively discriminated patients with benign or malignant ovarian masses. In vitro culturing of the PBMCs of healthy donors in malignant ascites promoted an increase in TNFR2-expressing Tregs, which were decreased following blockade with IL-6 or STAT3 activity. Pre-treatment serum IL-6 and peripheral blood TNFR2+ Tregs may be potential clinical biomarkers that can discriminate patients with malignant compared to benign ovarian cancer masses, and the relationship between IL-6 and TNFR2+ Treg is likely to be mediated via the STAT3 signalling pathway.
Collapse
Affiliation(s)
- Nirmala Chandralega Kampan
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | | | - Cyril Deceneux
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| | - Mutsa Tatenda Madondo
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Orla M. McNally
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
- Department of Obstetrics and Gynaecology, Melbourne University, Parkville, VIC 3052, Australia
| | - Katie Louise Flanagan
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
- School of Health Sciences and School of Medicine, University of Tasmania, Hobart, TAS 7005, Australia
| | - Norhaslinda A. Aziz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Andrew N. Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3800, Australia
- Epworth Research Institute, Epworth Healthcare, Richmond, VIC 3121, Australia
| | - John Reynolds
- Biostatistics Consulting Platform, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
| | - Michael A. Quinn
- Oncology Unit, Royal Women’s Hospital, 20 Flemington Road, Parkville, VIC 3052, Australia
| | - Magdalena Plebanski
- Department of Immunology & Pathology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Level 6, The Alfred, Commercial Road, Melbourne, VIC 3181, Australia
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia
| |
Collapse
|
29
|
Al-Marzouki L, Stavrakos VS, Pal S, Giannias B, Bourdeau F, Rayes R, Bertos N, Najmeh S, Spicer JD, Cools-Lartigue J, Bailey SD, Ferri L, Sangwan V. Soluble factors in malignant ascites promote the metastatic adhesion of gastric adenocarcinoma cells. Gastric Cancer 2023; 26:55-68. [PMID: 36059037 DOI: 10.1007/s10120-022-01338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/25/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Adenocarcinoma of the proximal stomach is the fastest rising malignancy in North America. It is commonly associated with peritoneal accumulation of malignant ascites (MA), a fluid containing cancer and inflammatory cells and soluble proteins. Peritoneal metastasis (PM) is the most common site of gastric cancer (GC) progression after curative-intent surgery and is the leading cause of death among GC patients. METHODS/RESULTS Using a panel of gastric adenocarcinoma cell lines (human: MKN 45, SNU-5; murine: NCC-S1M), we demonstrate that prior incubation of GC cells with MA results in a significant (> 1.7-fold) increase in the number of cells capable of adhering to human peritoneal mesothelial cells (HPMC) (p < 0.05). We then corroborate these findings using an ex vivo PM model and show that MA also significantly enhances the ability of GC cells to adhere to strips of human peritoneum (p < 0.05). Using a multiplex ELISA, we identify MIF and VEGF as consistently elevated across MA samples from GC patients (p < 0.05). We demonstrate that agents that block the effects of MIF or VEGF abrogate the ability of MA to stimulate the adhesion of GC cells to adhere to human peritoneum and promote both ex vivo and in vivo metastases. CONCLUSION Agents targeting MIF or VEGF may be relevant to the treatment or prevention of PM in GC patients.
Collapse
Affiliation(s)
- Luai Al-Marzouki
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Vivian S Stavrakos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Sanjima Pal
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Betty Giannias
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - France Bourdeau
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Roni Rayes
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Sara Najmeh
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan D Spicer
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Jonathan Cools-Lartigue
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada
| | - Swneke D Bailey
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Lorenzo Ferri
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
- Division of Thoracic and Upper GI Surgery, Montreal General Hospital, 1650 Cedar Avenue, Room L8-325, Montreal, QC, H3G 1A4, Canada.
| | - Veena Sangwan
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Surgery, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
30
|
Chung YH, Volckaert BA, Steinmetz NF. Metastatic Colon Cancer Treatment Using S100A9-Targeted Cowpea Mosaic Virus Nanoparticles. Biomacromolecules 2022; 23:5127-5136. [PMID: 36375170 PMCID: PMC9772157 DOI: 10.1021/acs.biomac.2c00873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peritoneal metastases (PMs) occur due to the metastasis of gynecological and gastrointestinal cancers such as ovarian, colon, pancreatic, or gastric tumors. PM outgrowth is often fatal, and patients with PMs have a median survival of 6 months. Cowpea mosaic virus (CPMV) has been shown, when injected intratumorally, to act as an immunomodulator reversing the immunosuppressive tumor microenvironment, therefore turning cold tumors hot and priming systemic antitumor immunity. However, not all tumors are injectable, and PMs especially will require targeted treatments to direct CPMV toward the disseminated tumor nodules. Toward this goal, we designed and tested a CPMV nanoparticle targeted to S100A9, a key immune mediator for many cancer types indicated in cancer growth, invasiveness, and metastasis. Here, we chose to use an intraperitoneal (IP) colon cancer model, and analysis of IP gavage fluid demonstrates that S100A9 is upregulated following IP challenge. S100A9-targeted CPMV particles displaying peptide ligands specific for S100A9 homed to IP-disseminated tumors, and treatment led to improved survival and decreased tumor burden. Targeting CPMV to S100A9 improves preclinical outcomes and harbors the potential of utilizing CPMV for the treatment of IP-disseminated diseases.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Britney A. Volckaert
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093-0021, United States
| | - Nicole F. Steinmetz
- Corresponding Author: Nicole F. Steinmetz – Department of Bioengineering, Moores Cancer Center, Department of NanoEngineering, Department of Radiology, Institute for Materials Discovery and Design, Center for Nano-Immuno Engineering, and Center for Engineering in Cancer, University of California, San Diego, La Jolla, California 92093-0021, United States;
| |
Collapse
|
31
|
Pharmacological Inhibition of Lipid Import and Transport Proteins in Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14236004. [PMID: 36497485 PMCID: PMC9737127 DOI: 10.3390/cancers14236004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy with a 5-year survival rate of 49%. This is caused by late diagnosis when cells have already metastasized into the peritoneal cavity and to the omentum. OC progression is dependent on the availability of high-energy lipids/fatty acids (FA) provided by endogenous de novo biosynthesis and/or through import from the microenvironment. The blockade of these processes may thus represent powerful strategies against OC. While this has already been shown for inhibition of FA/lipid biosynthesis, evidence of the role of FA/lipid import/transport is still sparse. Therefore, we treated A2780 and SKOV3 OC cells with inhibitors of the lipid uptake proteins fatty acid translocase/cluster of differentiation 36 (FAT/CD36) and low-density lipoprotein (LDL) receptor (LDLR), as well as intracellular lipid transporters of the fatty acid-binding protein (FABP) family, fatty acid transport protein-2 (FATP2/SLC27A2), and ADP-ribosylation factor 6 (ARF6), which are overexpressed in OC. Proliferation was determined by formazan dye labeling/photometry and cell counting. Cell cycle analysis was performed by propidium iodide (PI) staining, and apoptosis was examined by annexin V/PI and active caspase 3 labeling and flow cytometry. RNA-seq data revealed altered stress and metabolism pathways. Overall, the small molecule inhibitors of lipid handling proteins BMS309403, HTS01037, NAV2729, SB-FI-26, and sulfosuccinimidyl oleate (SSO) caused a drug-specific, dose-/time-dependent inhibition of FA/LDL uptake, associated with reduced proliferation, cell cycle arrest, and apoptosis. Our findings indicate that OC cells are very sensitive to lipid deficiency. This dependency should be exploited for development of novel strategies against OC.
Collapse
|
32
|
Zhan S, Yung MMH, Siu MKY, Jiao P, Ngan HYS, Chan DW, Chan KKL. New Insights into Ferroptosis Initiating Therapies (FIT) by Targeting the Rewired Lipid Metabolism in Ovarian Cancer Peritoneal Metastases. Int J Mol Sci 2022; 23:ijms232315263. [PMID: 36499591 PMCID: PMC9737695 DOI: 10.3390/ijms232315263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/04/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers worldwide. The poor prognosis of this malignancy is substantially attributed to the inadequate symptomatic biomarkers for early diagnosis and effective remedies to cure the disease against chemoresistance and metastasis. Ovarian cancer metastasis is often relatively passive, and the single clusters of ovarian cancer cells detached from the primary ovarian tumor are transcoelomic spread by the peritoneal fluid throughout the peritoneum cavity and omentum. Our earlier studies revealed that lipid-enriched ascitic/omental microenvironment enforced metastatic ovarian cancer cells to undertake metabolic reprogramming and utilize free fatty acids as the main energy source for tumor progression and aggression. Intriguingly, cell susceptibility to ferroptosis has been tightly correlated with the dysregulated fatty acid metabolism (FAM), and enhanced iron uptake as the prominent features of ferroptosis are attributed to the strengthened lipid peroxidation and aberrant iron accumulation, suggesting that ferroptosis induction is a targetable vulnerability to prevent cancer metastasis. Therefore, the standpoints about tackling altered FAM in combination with ferroptosis initiation as a dual-targeted therapy against advanced ovarian cancer were highlighted herein. Furthermore, a discussion on the prospect and challenge of inducing ferroptosis as an innovative therapeutic approach for reversing remedial resistance in cancer interventions was included. It is hoped this proof-of-concept review will indicate appropriate directions for speeding up the translational application of ferroptosis-inducing compounds (FINs) to improve the efficacy of ovarian cancer treatment.
Collapse
Affiliation(s)
- Shijie Zhan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mingo M. H. Yung
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michelle K. Y. Siu
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Peili Jiao
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hextan Y. S. Ngan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David W. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- School of Medicine, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China
- Correspondence: (D.W.C.); (K.K.L.C.); Tel.: +86-755-2351-6153 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2255-0947 (K.K.L.C.)
| | - Karen K. L. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Correspondence: (D.W.C.); (K.K.L.C.); Tel.: +86-755-2351-6153 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2255-0947 (K.K.L.C.)
| |
Collapse
|
33
|
Hoarau-Véchot J, Blot-Dupin M, Pauly L, Touboul C, Rafii S, Rafii A, Pasquier J. Akt-Activated Endothelium Increases Cancer Cell Proliferation and Resistance to Treatment in Ovarian Cancer Cell Organoids. Int J Mol Sci 2022; 23:ijms232214173. [PMID: 36430649 PMCID: PMC9694384 DOI: 10.3390/ijms232214173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Ovarian cancer (OC) is a heterogeneous disease characterized by its late diagnosis (FIGO stages III and IV) and the importance of abdominal metastases often observed at diagnosis. Detached ovarian cancer cells (OCCs) float in ascites and form multicellular spheroids. Here, we developed endothelial cell (EC)-based 3D spheroids to better represent in vivo conditions. When co-cultured in 3D conditions, ECs and OCCs formed organized tumor angiospheres with a core of ECs surrounded by proliferating OCCs. We established that Akt and Notch3/Jagged1 pathways played a role in angiosphere formation and peritoneum invasion. In patients' ascites we found angiosphere-like structures and demonstrated in patients' specimens that tumoral EC displayed Akt activation, which supports the importance of Akt activation in ECs in OC. Additionally, we demonstrated the importance of FGF2, Pentraxin 3 (PTX3), PD-ECGF and TIMP-1 in angiosphere organization. Finally, we confirmed the role of Notch3/Jagged1 in OCC-EC crosstalk relating to OCC proliferation and during peritoneal invasion. Our results support the use of multicellular spheroids to better model tumoral and stromal interaction. Such models could help decipher the complex pathways playing critical roles in metastasis spread and predict tumor response to chemotherapy or anti-angiogenic treatment.
Collapse
Affiliation(s)
- Jessica Hoarau-Véchot
- Department of Genetic Medicine and Obstetrics and Gynecology, Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
| | - Morgane Blot-Dupin
- Faculté de Médecine de Créteil UPEC—Paris XII, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France
| | - Léa Pauly
- Faculté de Médecine de Créteil UPEC—Paris XII, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Centre Hospitalier Intercommunal de Créteil, 40 Avenue de Verdun, 94000 Créteil, France
| | - Cyril Touboul
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), UMR_S 938, Centre de Recherche Saint-Antoine, Team Cancer Biology and Therapeutics, Institut Universitaire de Cancérologie, Sorbonne Université, 75012 Paris, France
- Department of Obstetrics and Gynecology, Hôpital Tenon, Assistance Publique Des Hôpitaux de Paris, GRC-6 UPMC, Université Pierre et Marie Curie, 75005 Paris, France
| | - Shahin Rafii
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Arash Rafii
- Department of Genetic Medicine and Obstetrics and Gynecology, Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jennifer Pasquier
- Department of Genetic Medicine and Obstetrics and Gynecology, Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar
- Correspondence:
| |
Collapse
|
34
|
Schoutrop E, Moyano-Galceran L, Lheureux S, Mattsson J, Lehti K, Dahlstrand H, Magalhaes I. Molecular, cellular and systemic aspects of epithelial ovarian cancer and its tumor microenvironment. Semin Cancer Biol 2022; 86:207-223. [PMID: 35395389 DOI: 10.1016/j.semcancer.2022.03.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/11/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Ovarian cancer encompasses a heterogeneous group of malignancies that involve the ovaries, fallopian tubes and the peritoneal cavity. Despite major advances made within the field of cancer, the majority of patients with ovarian cancer are still being diagnosed at an advanced stage of the disease due to lack of effective screening tools. The overall survival of these patients has, therefore, not substantially improved over the past decades. Most patients undergo debulking surgery and treatment with chemotherapy, but often micrometastases remain and acquire resistance to the therapy, eventually leading to disease recurrence. Here, we summarize the current knowledge in epithelial ovarian cancer development and metastatic progression. For the most common subtypes, we focus further on the properties and functions of the immunosuppressive tumor microenvironment, including the extracellular matrix. Current and future treatment modalities are discussed and finally we provide an overview of the different experimental models used to develop novel therapies.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Stephanie Lheureux
- University of Toronto, Toronto, Ontario, Canada; Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Toronto, Ontario, Canada
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hanna Dahlstrand
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical unit Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
35
|
Abd-Elghany AA, Ahmed SM, Masoud MA, Atia T, Waggiallah HA, El-Sakhawy MA, Mohamad EA. Annona squamosa L. Extract-Loaded Niosome and Its Anti-Ehrlich Ascites’ Carcinoma Activity. ACS OMEGA 2022; 7:38436-38447. [DOI: 10.1021/acsomega.2c03649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/11/2022] [Indexed: 12/26/2022]
Affiliation(s)
- Amr A. Abd-Elghany
- Radiology and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam Bin Abdul-Aziz University, Al-Kharj11942, Saudi Arabia
- Biophysics Department, Faculty of Science, Cairo University, Giza12613, Egypt
| | - Samya Mahmoud Ahmed
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR), Egyptian Drug Authority (EDA), Giza12654, Egypt
| | - Marwa A. Masoud
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Egyptian Drug Authority (EDA), Giza12654, Egypt
| | - Tarek Atia
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdul-Aziz University, Al-Kharj11942, Saudi Arabia
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdul-Aziz University, Al-Kharj11942, Saudi Arabia
| | - Mohamed A. El-Sakhawy
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdul-Aziz University, Al-Kharj11942, Saudi Arabia
- Department of Medicinal and Aromatic Plants, Desert Research Center, Cairo11753, Egypt
| | - Ebtesam A. Mohamad
- Radiology and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam Bin Abdul-Aziz University, Al-Kharj11942, Saudi Arabia
| |
Collapse
|
36
|
Ogasawara F, Higuchi T, Nishimori T, Hashida Y, Kojima K, Daibata M. Targeting
VEGF
with bevacizumab inhibits malignant effusion formation of primary human herpesvirus 8‐unrelated effusion large B‐cell lymphoma in vivo. J Cell Mol Med 2022; 26:5580-5589. [PMID: 36209502 PMCID: PMC9667516 DOI: 10.1111/jcmm.17570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Primary human herpesvirus 8 (HHV8)‐unrelated effusion large B‐cell lymphoma (ELBCL) is recognized as a new clinical entity, but its pathogenesis and therapeutic strategies remain largely unknown. We have generated two mouse models with profuse lymphomatous effusions that resemble HHV8‐unrelated ELBCL occurring in humans, by grafting the cell lines designated as Pell‐1 and Pell‐2. Using these in vivo models, we evaluated the potential role of vascular endothelial growth factor (VEGF) in the pathogenesis of HHV8‐unrelated ELBCL. Both Pell‐1 and Pell‐2 cells consistently produced very high levels of VEGF in mice, in contrast to in vitro findings of relatively low VEGF production in culture medium by HHV8‐unrelated ELBCL cells, especially Pell‐1 cells. Conversely, returning Pell‐1 cells grown in mice to culture medium markedly suppressed VEGF production to the original in vitro level. These findings suggest that the tumour microenvironment plays a role in the steady production of VEGF. We also found that the interaction between HHV8‐unrelated ELBCL cells and peritoneal mesothelial cells increased the production of VEGF in vitro. Finally, we found that bevacizumab significantly suppressed effusion formation and lymphoma cell growth in both mouse models. These results suggest that bevacizumab is a rational approach to the treatment of HHV8‐unrelated ELBCL.
Collapse
Affiliation(s)
- Fumiya Ogasawara
- Department of Microbiology and Infection Kochi Medical School, Kochi University Nankoku Japan
- Department of Hematology Kochi Medical School, Kochi University Nankoku Japan
| | - Tomonori Higuchi
- Department of Microbiology and Infection Kochi Medical School, Kochi University Nankoku Japan
| | - Tomohiro Nishimori
- Department of Microbiology and Infection Kochi Medical School, Kochi University Nankoku Japan
| | - Yumiko Hashida
- Department of Microbiology and Infection Kochi Medical School, Kochi University Nankoku Japan
| | - Kensuke Kojima
- Department of Hematology Kochi Medical School, Kochi University Nankoku Japan
| | - Masanori Daibata
- Department of Microbiology and Infection Kochi Medical School, Kochi University Nankoku Japan
| |
Collapse
|
37
|
Immune Tumor Microenvironment in Ovarian Cancer Ascites. Int J Mol Sci 2022; 23:ijms231810692. [PMID: 36142615 PMCID: PMC9504085 DOI: 10.3390/ijms231810692] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) has a specific type of metastasis, via transcoelomic, and most of the patients are diagnosed at advanced stages with multiple tumors spread within the peritoneal cavity. The role of Malignant Ascites (MA) is to serve as a transporter of tumor cells from the primary location to the peritoneal wall or to the surface of the peritoneal organs. MA comprise cellular components with tumor and non-tumor cells and acellular components, creating a unique microenvironment capable of modifying the tumor behavior. These microenvironment factors influence tumor cell proliferation, progression, chemoresistance, and immune evasion, suggesting that MA play an active role in OC progression. Tumor cells induce a complex immune suppression that neutralizes antitumor immunity, leading to disease progression and treatment failure, provoking a tumor-promoting environment. In this review, we will focus on the High-Grade Serous Carcinoma (HGSC) microenvironment with special attention to the tumor microenvironment immunology.
Collapse
|
38
|
Semibulk RNA-seq analysis as a convenient method for measuring gene expression statuses in a local cellular environment. Sci Rep 2022; 12:15309. [PMID: 36097044 PMCID: PMC9468030 DOI: 10.1038/s41598-022-19391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
When biologically interpretation of the data obtained from the single-cell RNA sequencing (scRNA-seq) analysis is attempted, additional information on the location of the single cells, behavior of the surrounding cells, and the microenvironment they generate, would be very important. We developed an inexpensive, high throughput application while preserving spatial organization, named “semibulk RNA-seq” (sbRNA-seq). We utilized a microfluidic device specifically designed for the experiments to encapsulate both a barcoded bead and a cell aggregate (a semibulk) into a single droplet. Using sbRNA-seq, we firstly analyzed mouse kidney specimens. In the mouse model, we could associate the pathological information with the gene expression information. We validated the results using spatial transcriptome analysis and found them highly consistent. When we applied the sbRNA-seq analysis to the human breast cancer specimens, we identified spatial interactions between a particular population of immune cells and that of cancer-associated fibroblast cells, which were not precisely represented solely by the single-cell analysis. Semibulk analysis may provide a convenient and versatile method, compared to a standard spatial transcriptome sequencing platform, to associate spatial information with transcriptome information.
Collapse
|
39
|
Generating patient-derived ascites-dependent xenograft mouse models of peritoneal carcinomatosis. STAR Protoc 2022; 3:101548. [PMID: 35842864 PMCID: PMC9294250 DOI: 10.1016/j.xpro.2022.101548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/26/2022] [Accepted: 06/17/2022] [Indexed: 11/24/2022] Open
Abstract
Clinically relevant animal models are crucial for effective development of therapeutics for peritoneal carcinomatosis (PC). This protocol describes the generation of patient-derived ascites-dependent xenograft (PDADX) models from the cellular component of ascites. The use of routine intraperitoneal injection of the fluid component of ascites is analogous to the biological events occurring intra-abdominally in patients with PC. By serving as a proxy, PDADX models represent a valuable tool for preclinical testing of new therapeutics for PC. For complete details on the use and execution of this protocol, please refer to Hendrikson et al. (2022). Generation of patient-derived ascites-dependent xenograft (PDADX) models Serial passages of PDADX tumors in mice Evaluation of patient-specific drug efficacy utilizing PDADX models
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
|
40
|
Platelet Activation in Ovarian Cancer Ascites: Assessment of GPIIb/IIIa and PF4 in Small Extracellular Vesicles by Nano-Flow Cytometry Analysis. Cancers (Basel) 2022; 14:cancers14174100. [PMID: 36077635 PMCID: PMC9454670 DOI: 10.3390/cancers14174100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Platelets play a critical role in coagulation and fibrinolysis processes, but recent literature also indicates their central involvement in immune response, cancer progression and metastasis. During platelet activation, small extracellular vesicles (EVs) are released. The ascites are a fluid developing in the peritoneum of ovarian cancer patients in an advanced stage. This study analysed the expression of platelet markers GPIIb/IIIa and PF4 in small-EVs populations isolated from the ascitic fluid of patients with advanced ovarian cancer. The percentage of platelet-derived small-EVs was positively correlated with platelet distribution width to platelet count in sera (PDW/PLT), a surrogate indicator of platelet activation. Overall, we presented a method that can be helpful in future studies to determine the correlation between the extent of platelet activation in ascites and disease status. Abstract In ovarian cancer, ascites represent the microenvironment in which the platelets extravasate to play their role in the disease progression. We aimed to develop an assay to measure ascites’ platelet activation. We enriched small extracellular vesicles (EVs) (40–200 nm) from ascites of high-grade epithelial ovarian cancer patients (n = 12) using precipitation with polyethylene glycol, and we conducted single-particle phenotyping analysis by nano-flow cytometry after labelling and ultra-centrifugation. Atomic force microscopy single-particle nanomechanical analysis showed heterogeneous distributions in the size of the precipitated particles and their mechanical stiffness. Samples were fluorescently labelled with antibodies specific to the platelet markers GPIIb/IIIa and PF4, showing 2.6 to 18.16% of all particles stained positive for the biomarkers and, simultaneously, the EV membrane labelling. Single-particle phenotyping analysis allowed us to quantify the total number of non-EV particles, the number of small-EVs and the number of platelet-derived small-EVs, providing a platelet activation assessment independent of the ascites volume. The percentage of platelet-derived small-EVs was positively correlated with platelet distribution width to platelet count in sera (PDW/PLT). Overall, we presented a high-throughput method that can be helpful in future studies to determine the correlation between the extent of platelet activation in ascites and disease status.
Collapse
|
41
|
Kono Y, Fushimi A, Yoshizawa Y, Higaki K, Ogawara KI. Effects of particle size and release property of paclitaxel-loaded nanoparticles on their peritoneal retention and therapeutic efficacy against mouse malignant ascites. Int J Pharm 2022; 623:121904. [PMID: 35716981 DOI: 10.1016/j.ijpharm.2022.121904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/11/2022] [Accepted: 06/07/2022] [Indexed: 11/25/2022]
Abstract
Malignant ascites accounts for abdominal pain, dyspnea and anorexia, all of which decrease quality of life in cancer patients. Intraperitoneal chemotherapy is a useful method for managing malignant ascites and nanoparticulate drug delivery system makes it more effective by increasing peritoneal retention of anti-cancer drugs. In this study, we prepared paclitaxel-loaded emulsions and liposomes with different particle sizes and drug release properties, and evaluated their peritoneal retention and therapeutic efficacy in Ehrlich's ascites carcinoma (EAC)-bearing mice. Liposomes with the size of 100 nm were rapidly absorbed from peritoneal cavity into blood after intraperitoneal injection into EAC-bearing mice, whereas 1000-nm liposomes were highly retained in peritoneal cavity. Accordingly, 1000 nm liposomes significantly prolonged survival time of EAC-bearing mice but did not inhibit the ascites accumulation because of too poor paclitaxel release. On the other hand, although peritoneal retention of emulsions themselves was similar irrespective of their sizes, 270-nm emulsions showed the higher PTX retention in ascites than other emulsions, and resulted in significantly prolonged survival time and lower accumulation of ascites in EAC-bearing mice. These results indicate that not only particle size but drug release property is one of key determinants of the biodisposition and therapeutic efficacy of intraperitoneally injected nanoparticulate PTX against malignant ascites.
Collapse
Affiliation(s)
- Yusuke Kono
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Aya Fushimi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Yuta Yoshizawa
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Ken-Ichi Ogawara
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
42
|
Hassan AA, Artemenko M, Tang MKS, Shi Z, Chen LY, Lai HC, Yang Z, Shum HC, Wong AST. Ascitic fluid shear stress in concert with hepatocyte growth factor drive stemness and chemoresistance of ovarian cancer cells via the c-Met-PI3K/Akt-miR-199a-3p signaling pathway. Cell Death Dis 2022; 13:537. [PMID: 35676254 PMCID: PMC9177676 DOI: 10.1038/s41419-022-04976-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 01/21/2023]
Abstract
Overcoming drug resistance is an inevitable challenge to the success of cancer treatment. Recently, in ovarian cancer, a highly chemoresistant tumor, we demonstrated an important role of shear stress in stem-like phenotype and chemoresistance using a three-dimensional microfluidic device, which most closely mimics tumor behavior. Here, we examined a new mechanosensitive microRNA-miR-199a-3p. Unlike most key microRNA biogenesis in static conditions, we found that Dicer, Drosha, and Exportin 5 were not involved in regulating miR-199a-3p under ascitic fluid shear stress (0.02 dynes/cm2). We further showed that hepatocyte growth factor (HGF), but not other ascitic cytokines/growth factors such as epidermal growth factor and tumor necrosis factor α or hypoxia, could transcriptionally downregulate miR-199a-3p through its primary transcript miR-199a-1 and not miR-199a-2. Shear stress in the presence of HGF resulted in a concerted effect via a specific c-Met/PI3K/Akt signaling axis through a positive feedback loop, thereby driving cancer stemness and drug resistance. We also showed that miR-199a-3p expression was inversely correlated with enhanced drug resistance properties in chemoresistant ovarian cancer lines. Patients with low miR-199a-3p expression were more resistant to platinum with a significantly poor prognosis. miR-199a-3p mimic significantly suppressed ovarian tumor metastasis and its co-targeting in combination with cisplatin or paclitaxel further decreased the peritoneal dissemination of ovarian cancer in mice. These findings unravel how biophysical and biochemical cues regulate miR-199a-3p and is important in chemoresistance. miR-199a-3p mimics may serve as a novel targeted therapy for effective chemosensitization.
Collapse
Affiliation(s)
- Ayon A. Hassan
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Margarita Artemenko
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Maggie K. S. Tang
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong ,grid.493736.cLaboratory for Synthetic Chemistry and Chemical Biology Limited, 17 W, Hong Kong Science and Technology Parks, Shatin, New Territories Hong Kong
| | - Zeyu Shi
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Lin-Yu Chen
- grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei, 23561 Taiwan
| | - Hung-Cheng Lai
- grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, Taipei, 23561 Taiwan ,grid.412896.00000 0000 9337 0481Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Zhenyu Yang
- grid.194645.b0000000121742757Department of Mechanical Engineering, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| | - Ho-Cheung Shum
- grid.194645.b0000000121742757Department of Mechanical Engineering, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong ,Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories Hong Kong
| | - Alice S. T. Wong
- grid.194645.b0000000121742757School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Pokfulam Road, Hong Kong
| |
Collapse
|
43
|
Spheroid Formation and Peritoneal Metastasis in Ovarian Cancer: The Role of Stromal and Immune Components. Int J Mol Sci 2022; 23:ijms23116215. [PMID: 35682890 PMCID: PMC9181487 DOI: 10.3390/ijms23116215] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological cancers, with the worst prognosis and the highest mortality rate. Peritoneal dissemination (or carcinomatosis) accompanied by ascites formation is the most unfavorable factor in the progression and recurrence of OC. Tumor cells in ascites are present as either separate cells or, more often, as cell aggregates, i.e., spheroids which promote implantation on the surface of nearby organs and, at later stages, metastases to distant organs. Malignant ascites comprises a unique tumor microenvironment; this fact may be of relevance in the search for new prognostic and predictive factors that would make it possible to personalize the treatment of patients with OC. However, the precise mechanisms of spheroid formation and carcinomatosis are still under investigation. Here, we summarize data on ascites composition as well as the activity of fibroblasts and macrophages, the key stromal and immune components, in OC ascites. We describe current knowledge about the role of fibroblasts and macrophages in tumor spheroid formation, and discuss the specific functions of fibroblasts, macrophages and T cells in tumor peritoneal dissemination and implantation.
Collapse
|
44
|
Luo N, Sulaiman Z, Wang C, Ding J, Chen Y, Liu B, Cheng Z, Liu S. Hsa_circ_0000497 and hsa_circ_0000918 contributed to peritoneal metastasis of ovarian cancer via ascites. J Transl Med 2022; 20:201. [PMID: 35538537 PMCID: PMC9092689 DOI: 10.1186/s12967-022-03404-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/24/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE As a common complication of epithelial ovarian cancer (EOC), malignant ascites contributes to the peritoneal metastasis of EOC. CircRNAs play essential roles in tumor metastasis. However, no circRNAs have been reported to be involved in EOC peritoneal metastasis via ascites. METHODS Total of 22 samples from 9 EOC patients containing primary lesions (T), tumor cells from ascites (ASC), and metastatic lesions (M) were included for RNA sequencing to identify differentially expressed circRNAs and mRNAs among different tumors. Bioinformatic analyses, including single-sample Gene Set Enrichment Analysis and soft cluster analysis, were performed to find circRNAs potentially correlated with ascitic metastasis. Wound healing and transwell analysis were performed to evaluate tumor cells metastasis in vitro. Quantitative real-time PCR and western-blot were used for gene expression evaluation. RESULTS According to transcriptomic analysis, ASC showed mesenchymal phenotype while T and M showed epithelial phenotype. 10 circRNAs were differentially expressed among ASC, T, and M. Among them, hsa_circ_0000497 and hsa_circ_0000918 were significantly up-regulated in ASC. Functional analysis showed that both hsa_circ_0000497 and hsa_circ_0000918 promoted metastasis of EOC via epithelial-mesenchymal transition (EMT) in vitro. The regulatory network construction identified 8 miRNAs and 19 mRNAs, and 7 miRNAs and 17 mRNAs as potential downstream target genes of hsa_circ_0000497 and hsa_circ_0000918, respectively, which may play pivotal roles in EOC ascitic metastasis. CONCLUSIONS circRNAs (hsa_circ_0000497 and hsa_circ_0000918) contribute to metastasis of EOC via ascites by regulating EMT. These circRNAs may serve as novel potential therapeutic targets or prognostic biomarkers for EOC peritoneal metastasis.
Collapse
Affiliation(s)
- Ning Luo
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zubaidan Sulaiman
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Chunyan Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jinye Ding
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yingying Chen
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Biting Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Shupeng Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
- Institute of Gynecological Minimally Invasive Medicine, School of Medicine, Tongji University, Shanghai, 200072, China.
- Department of Obstetrics and Gynecology, Putuo People's Hospital, Tongji University, Shanghai, 200060, China.
| |
Collapse
|
45
|
Wang W, Jo H, Park S, Kim H, Kim SI, Han Y, Lee J, Seol A, Kim J, Lee M, Lee C, Dhanasekaran DN, Ahn T, Song YS. Integrated analysis of ascites and plasma extracellular vesicles identifies a miRNA-based diagnostic signature in ovarian cancer. Cancer Lett 2022; 542:215735. [DOI: 10.1016/j.canlet.2022.215735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 01/03/2023]
|
46
|
Uno K, Iyoshi S, Yoshihara M, Kitami K, Mogi K, Fujimoto H, Sugiyama M, Koya Y, Yamakita Y, Nawa A, Kanayama T, Tomita H, Enomoto A, Kajiyama H. Metastatic Voyage of Ovarian Cancer Cells in Ascites with the Assistance of Various Cellular Components. Int J Mol Sci 2022; 23:4383. [PMID: 35457198 PMCID: PMC9031612 DOI: 10.3390/ijms23084383] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and has a unique metastatic route using ascites, known as the transcoelomic root. However, studies on ascites and contained cellular components have not yet been sufficiently clarified. In this review, we focus on the significance of accumulating ascites, contained EOC cells in the form of spheroids, and interaction with non-malignant host cells. To become resistant against anoikis, EOC cells form spheroids in ascites, where epithelial-to-mesenchymal transition stimulated by transforming growth factor-β can be a key pathway. As spheroids form, EOC cells are also gaining the ability to attach and invade the peritoneum to induce intraperitoneal metastasis, as well as resistance to conventional chemotherapy. Recently, accumulating evidence suggests that EOC spheroids in ascites are composed of not only cancer cells, but also non-malignant cells existing with higher abundance than EOC cells in ascites, including macrophages, mesothelial cells, and lymphocytes. Moreover, hetero-cellular spheroids are demonstrated to form more aggregated spheroids and have higher adhesion ability for the mesothelial layer. To improve the poor prognosis, we need to elucidate the mechanisms of spheroid formation and interactions with non-malignant cells in ascites that are a unique tumor microenvironment for EOC.
Collapse
Affiliation(s)
- Kaname Uno
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 223-62 Lund, Sweden
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Discipline of Obstetrics and Gynecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiko Yamakita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan;
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| |
Collapse
|
47
|
Ayyagari V, Li M, Pasman Z, Wang X, Louis S, Diaz-Sylvester P, Groesch K, Wilson T, Brard L. Assessment of the diagnostic and prognostic relevance of ACAT1 and CE levels in plasma, peritoneal fluid and tumor tissue of epithelial ovarian cancer patients - a pilot study. BMC Cancer 2022; 22:387. [PMID: 35399074 PMCID: PMC8994887 DOI: 10.1186/s12885-022-09476-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 03/14/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Abnormal accumulation of acyl-CoA cholesterol acyltransferase-1 (ACAT1) and ACAT1-mediated cholesterol esterified with fatty acids (CE) contribute to cancer progression in various cancers. Our findings of increased CE and ACAT1 levels in epithelial ovarian cancer (EOC) cell lines prompted us to investigate whether such an increase occurs in primary clinical samples obtained from human subjects diagnosed with EOC. We evaluated the diagnostic/prognostic potential of ACAT1 and CE in EOC by: 1) assessing ACAT1 and CE levels in plasma, peritoneal fluid, and ovarian/tumor tissues; 2) assessing diagnostic performance by Receiver Operating Characteristic (ROC) analysis; and 3) comparing expression of ACAT1 and CE with that of tumor proliferation marker, Ki67.
Methods
ACAT1 protein levels in plasma, peritoneal fluid and tissue were measured via enzyme-linked immunosorbent assay. Tissue expression of ACAT1 and Ki67 proteins were confirmed by immunohistochemistry and mRNA transcript levels were evaluated using quantitative real-time polymerase chain reaction (qRT-PCR). CE levels were assessed in plasma, peritoneal fluid (colorimetric assay) and in tissue (thin layer chromatography).
Results
Preoperative levels of ACAT1 and CE on the day of surgery were significantly higher in tissue and peritoneal fluid from EOC patients vs. the non-malignant group, which included subjects with benign tumors and normal ovaries; however, no significant differences were observed in plasma. In tissue and peritoneal fluid, positive correlations were observed between CE and ACAT1 levels, as well as between ACAT1/CE and Ki67.
Conclusions
ACAT1 and CE accumulation may be linked to the aggressive potential of EOC; therefore, these mediators may be useful biomarkers for EOC prognosis and target-specific treatments.
Collapse
|
48
|
Dunn ZS, Li YR, Yu Y, Lee D, Gibbons A, Kim JJ, Zhou TY, Li M, Nguyen M, Cen X, Zhou Y, Wang P, Yang L. Minimally Invasive Preclinical Monitoring of the Peritoneal Cavity Tumor Microenvironment. Cancers (Basel) 2022; 14:1775. [PMID: 35406547 PMCID: PMC8997523 DOI: 10.3390/cancers14071775] [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] [Received: 03/07/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022] Open
Abstract
Intraperitoneal (i.p.) experimental models in mice can recapitulate the process of i.p. dissemination in abdominal cancers and may help uncover critical information about future successful clinical treatments. i.p. cellular composition is studied in preclinical models addressing a wide spectrum of other pathophysiological states such as liver cirrhosis, infectious disease, autoimmunity, and aging. The peritoneal cavity is a multifaceted microenvironment that contains various immune cell populations, including T, B, NK, and various myeloid cells, such as macrophages. Analysis of the peritoneal cavity is often obtained by euthanizing mice and performing terminal peritoneal lavage. This procedure inhibits continuous monitoring of the peritoneal cavity in a single mouse and necessitates the usage of more mice to assess the cavity at multiple timepoints, increasing the cost, time, and variability of i.p. studies. Here, we present a simple, novel method termed in vivo intraperitoneal lavage (IVIPL) for the minimally invasive monitoring of cells in the peritoneal cavity of mice. In this proof-of-concept, IVIPL provided real-time insights into the i.p. tumor microenvironment for the development and study of ovarian cancer therapies. Specifically, we studied CAR-T cell therapy in a human high-grade serous ovarian cancer (HGSOC) xenograft mouse model, and we studied the immune composition of the i.p. tumor microenvironment (TME) in a mouse HGSOC syngeneic model.
Collapse
Affiliation(s)
- Zachary Spencer Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; (Z.S.D.); (P.W.)
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Derek Lee
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Alicia Gibbons
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - James Joon Kim
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Tian Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Mulin Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Mya Nguyen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Xinjian Cen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; (Z.S.D.); (P.W.)
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Y.); (D.L.); (A.G.); (J.J.K.); (T.Y.Z.); (M.L.); (M.N.); (X.C.); (Y.Z.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
49
|
MacPherson S, Keyes S, Kilgour MK, Smazynski J, Chan V, Sudderth J, Turcotte T, Devlieger A, Yu J, Huggler KS, Cantor JR, DeBerardinis RJ, Siatskas C, Lum JJ. Clinically relevant T cell expansion media activate distinct metabolic programs uncoupled from cellular function. Mol Ther Methods Clin Dev 2022; 24:380-393. [PMID: 35284590 PMCID: PMC8897702 DOI: 10.1016/j.omtm.2022.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/11/2022] [Indexed: 12/17/2022]
Abstract
Ex vivo expansion conditions used to generate T cells for immunotherapy are thought to adopt metabolic phenotypes that impede therapeutic efficacy in vivo. The comparison of five different culture media used for clinical T cell expansion revealed unique optima based on different output variables, including proliferation, differentiation, function, activation, and mitochondrial phenotypes. The extent of proliferation and function depended on the culture media rather than stimulation conditions. Moreover, the expanded T cell end products adapted their metabolism when switched to a different media formulation, as shown by glucose and glutamine uptake and patterns of glucose isotope labeling. However, adoption of these metabolic phenotypes was uncoupled to T cell function. Expanded T cell products cultured in ascites from ovarian cancer patients displayed suppressed mitochondrial activity and function irrespective of the ex vivo expansion media. Thus, ex vivo T cell expansion media have profound impacts on metabolism and function.
Collapse
Affiliation(s)
- Sarah MacPherson
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada
| | - Sarah Keyes
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada
| | - Marisa K Kilgour
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Julian Smazynski
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Vanessa Chan
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Jessica Sudderth
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Jessie Yu
- Stemcell Technologies Canada Inc., Vancouver, BC, Canada
| | - Kimberly S Huggler
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jason R Cantor
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R6V5, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
50
|
Liu C, Zhang Y, Li X, Wang D. Ovarian cancer-specific dysregulated genes with prognostic significance: scRNA-Seq with bulk RNA-Seq data and experimental validation. Ann N Y Acad Sci 2022; 1512:154-173. [PMID: 35247207 DOI: 10.1111/nyas.14748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022]
Abstract
A major cause of gynecological cancer -related deaths worldwide, ovarian cancer is characterized by heterogeneity in both tumor cells and the tumor microenvironment (TME). Our study aimed to characterize tumor cell heterogeneity and the infiltration of M2 tumor-associated macrophages (TAMs) in the ovarian cancer TME by single-cell RNA-Seq (scRNA-Seq) analysis combined with bulk RNA sequencing (bulk RNA-Seq). Several highly variable genes were identified in ovarian cancer tissues, and tumor cell heterogeneity and infiltrating immune tumor cell heterogeneity were characterized in ovarian cancer cells. M2 TAMs in the TME were the predominant phenotype of TAM. Further, M2 TAM infiltration in the TME was negatively correlated with poor prognosis of ovarian cancer patients. Four M2 TAM-associated genes (SLAMF7, GNAS, TBX2-AS1, and LYPD6) correlated with the prognostic survival of ovarian cancer patients. Knockdown of SLAMF7 or GNAS mRNA repressed malignancy and cisplatin resistance of ovarian cancer cells. ScRNA-Seq combined with bulk RNA-Seq identified the same four genes associated with M2 TAMs. The prognostic risk score model based on these four genes may hold favorable predictive value for the prognosis of ovarian cancer patients.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohan Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|