1
|
Ebrahim NAA, Soliman SMA. Advanced Biomaterials and Biomedical Devices for Studying Tumor-Associated Fibroblasts: Current Trends, Innovations, and Future Prospects. BIOMEDICAL MATERIALS & DEVICES 2025. [DOI: 10.1007/s44174-025-00287-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 04/23/2025]
|
2
|
Cheng F, Ma X, Cheng Z, Wang Y, Zhang X, Ma C. Predict value of tumor markers combined with interleukins for therapeutic efficacy and prognosis in ovarian cancer patients. Am J Cancer Res 2024; 14:4868-4879. [PMID: 39553206 PMCID: PMC11560821 DOI: 10.62347/gsrd2580] [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: 06/25/2024] [Accepted: 09/29/2024] [Indexed: 11/19/2024] Open
Abstract
Ovarian cancer (OC) is the most prevalent and fatal malignancy of the female reproductive system, with the majority of patients diagnosed at an advanced stage due to the lack of early screening. Despite surgery and chemotherapy being the standard treatments, overall survival rates have not improved significantly, highlighting the need for new biomarkers for therapeutic efficacy and prognostic evaluation. This study aimed to clarify the application value of tumor markers (TMs), including carbohydrate antigen 125 (CA125), alpha-fetoprotein (AFP), and carcinoembryonic antigen (CEA), combined with interleukins (ILs), such as IL-1β, IL-2, IL-6, IL-8, and IL-10, in the evaluation of therapeutic efficacy and prognosis of OC, and to establish a prediction model. A retrospective analysis was conducted on 184 OC patients treated at the Affiliated Hospital of Henan University of Traditional Chinese Medicine from February 2020 to February 2023. Serum levels of CA125, AFP, and CEA were quantified by chemiluminescence immunoassay, and ILs by enzyme-linked immunosorbent assays. Significant decreases in CA125, AFP, CEA, IL-1β, IL-2, IL-6, and IL-10 levels were observed after treatment (all P<0.001), while IL-8 levels showed no significant change (P=0.597). The death group exhibited notably higher levels of CA125, IL-6, and IL-8 than the survival group (all P<0.001). Cox regression analysis identified CA125, IL-8, histological grading, ascites, intravascular tumor thrombus, and International Federation of Gynecology and Obstetrics (FIGO) staging as independent prognostic factors. The Nomogram model based on these factors showed strong predictive ability in predicting patient mortality with an area under the curve (AUC) of 0.756. In conclusion, the combination of TMs and ILs is valuable in evaluating therapeutic efficacy and prognosis in OC. Dynamic monitoring of CA125, IL-6, and IL-8 can guide clinical treatment adjustments, improving diagnostic accuracy and prognosis reliability.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Gynecology, The Third Affiliated Hospital of Henan University of Chinese MedicineZhengzhou 450000, Henan, China
| | - Xijia Ma
- AAB 904, Level 9, Academic and Administrative Building, Baptist University Road Campus, Hong Kong Baptist UniversityKowloon Tong, Hong Kong, China
| | - Zhenyang Cheng
- Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai 20000, China
| | - Yami Wang
- Department of Research, Third Affiliated Hospital of Henan University of Traditional Chinese MedicineZhengzhou 450000, Henan, China
| | - Xuelin Zhang
- Research and Experiment Center, Third Affiliated Hospital of Henan University of Traditional Chinese MedicineZhengzhou 450000, Henan, China
| | - Chunzheng Ma
- Department of Oncology, Henan Province Hospital of TCMZhengzhou 450000, Henan, China
| |
Collapse
|
3
|
Liu X, Ping G, Ji D, Wen Z, Chen Y. Reclassify High-Grade Serous Ovarian Cancer Patients Into Different Molecular Subtypes With Discrepancy Prognoses and Therapeutic Responses Based on Cancer-Associated Fibroblast-Enriched Prognostic Genes. Biomed Eng Comput Biol 2024; 15:11795972241274024. [PMID: 39221174 PMCID: PMC11365035 DOI: 10.1177/11795972241274024] [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: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in the metastasis and therapeutic response of high-grade serous ovarian cancer (HGSC). Our study intended to select HGSC patients with unfavorable prognoses and therapeutic responses based on CAF-enriched prognostic genes. The bulk RNA and single-cell RNA sequencing (scRNA-seq) data of tumor tissues were collected from the TCGA and GEO databases. The infiltrated levels of immune and stromal cells were estimated by multiple immune deconvolution algorithms and verified through immunohistochemical analysis. The univariate Cox regression analyses were used to identify prognostic genes. Gene Set Enrichment Analysis (GSEA) was conducted to annotate enriched gene sets. The Genomics of Drug Sensitivity in Cancer (GDSC) database was used to explore potential alternative drugs. We found the infiltered levels of CAFs were remarkedly elevated in advanced and metastatic HGSC tissues and identified hundreds of genes specifically enriched in CAFs. Then we selected 6 CAF-enriched prognostic genes based on which HGSC patients were reclassified into 2 subclusters with discrepancy prognoses. Further analysis revealed that the HGSC patients in cluster-2 tended to undergo poor responses to traditional chemotherapy and immunotherapy. Subsequently, we selected 24 novel potential therapeutic drugs for cluster-2 HGSC patients. Moreover, we discovered a positive correlation of infiltrated levels between CAFs and monocytes/macrophages in HGSC tissues. Collectively, our study successfully reclassified HGSC patients into 2 different subgroups that have discrepancy prognoses and responses to current therapeutic methods.
Collapse
Affiliation(s)
- Xiangxiang Liu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Guoqiang Ping
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongze Ji
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhifa Wen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Coursier D, Calvo F. CAFs vs. TECs: when blood feuds fuel cancer progression, dissemination and therapeutic resistance. Cell Oncol (Dordr) 2024; 47:1091-1112. [PMID: 38453816 PMCID: PMC11322395 DOI: 10.1007/s13402-024-00931-z] [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] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
Neoplastic progression involves complex interactions between cancer cells and the surrounding stromal milieu, fostering microenvironments that crucially drive tumor progression and dissemination. Of these stromal constituents, cancer-associated fibroblasts (CAFs) emerge as predominant inhabitants within the tumor microenvironment (TME), actively shaping multiple facets of tumorigenesis, including cancer cell proliferation, invasiveness, and immune evasion. Notably, CAFs also orchestrate the production of pro-angiogenic factors, fueling neovascularization to sustain the metabolic demands of proliferating cancer cells. Moreover, CAFs may also directly or indirectly affect endothelial cell behavior and vascular architecture, which may impact in tumor progression and responses to anti-cancer interventions. Conversely, tumor endothelial cells (TECs) exhibit a corrupted state that has been shown to affect cancer cell growth and inflammation. Both CAFs and TECs are emerging as pivotal regulators of the TME, engaging in multifaceted biological processes that significantly impact cancer progression, dissemination, and therapeutic responses. Yet, the intricate interplay between these stromal components and the orchestrated functions of each cell type remains incompletely elucidated. In this review, we summarize the current understanding of the dynamic interrelationships between CAFs and TECs, discussing the challenges and prospects for leveraging their interactions towards therapeutic advancements in cancer.
Collapse
Affiliation(s)
- Diane Coursier
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain.
| |
Collapse
|
5
|
Fan FM, Fleishman JS, Chen J, Chen ZS, Dong HH. New insights into the mechanism of resistance to lenvatinib and strategies for lenvatinib sensitization in hepatocellular carcinoma. Drug Discov Today 2024; 29:104069. [PMID: 38936692 DOI: 10.1016/j.drudis.2024.104069] [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: 02/28/2024] [Revised: 05/04/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Lenvatinib is a multikinase inhibitor that suppresses vascular endothelial growth factor receptor (VEGFR), fibroblast growth factor receptor (FGFR), platelet-derived growth factor receptor α (PDGFRα), as well as the proto-oncogenes RET and KIT. Lenvatinib has been approved by the US Food and Drug Administration (FDA) for the first-line treatment of hepatocellular carcinoma (HCC) due to its superior efficacy when compared to sorafenib. Unfortunately, the development of drug resistance to lenvatinib is becoming increasingly common. Thus, there is an urgent need to identify the factors that lead to drug resistance and ways to mitigate it. We summarize the molecular mechanisms that lead to lenvatinib resistance (LR) in HCC, which involve programmed cell death (PCD), translocation processes, and changes in the tumor microenvironment (TME), and provide strategies to reverse resistance.
Collapse
Affiliation(s)
- Fei-Mu Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St John's University, Queens, NY 11439, USA
| | - Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China.
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St John's University, Queens, NY 11439, USA.
| | - Han-Hua Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan 430000, China.
| |
Collapse
|
6
|
Sulaiman R, Koirala N, Aske JC, Lin X, Rojas-Espaillat L, Starks D, Dale A, Gaster K, De P, Dey N. A landscape of patient-derived cancer-associated fibroblast signals in endometrial cancers. Am J Cancer Res 2024; 14:467-489. [PMID: 38455423 PMCID: PMC10915338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/02/2023] [Indexed: 03/09/2024] Open
Abstract
In conversation with endometrial tumor cells, the endometrial cancer-associated fibroblasts (CAFs) are the "partners in crime" of uterine neoplasm's highly heterogeneous tumor microenvironment (TME). We designed a laboratory-friendly method to culture endometrial CAFs on a patient-to-patient basis for studying the CAF-TME and CAF-tumor cell interaction(s). Here, we present a comprehensive characterization of endometrial CAFs derived from patients' tumor tissues (T) and tumor-adjacent normal tissues (N). We used more than 80 T and N from 53 consecutive consented patients with endometrial cancers at the Avera Cancer Institute. We derived TCAF and NCAF in a non-enzymatic feeder-layer culture and characterized their expression of markers by qRT-PCR, flow cytometry, immunocytochemistry, immunofluorescence, and Western blot. Although similar in the expression pattern of EpCAM-/CK18-/vimentin+ as in ovarian CAFs, endometrial NCAFs, and TCAFs characteristically presented dual morphology in culture. Endometrial CAFs were EpCAM-/CK18-/CD45-/CD31-/SMA+/TE-7+/PDGFRA+/CXCL12+/Meflin+/CD155+/CD90+ with patient-specific positivity for S100A4/FAP/PD-L1/CD44. Endometrial CAFs expressed mRNAs for signaling proteins of several pathways and receptor-ligands, including (1) cell cycle pathway, (2) TGF pathway, (3) FGF pathway, (4) Wnt-beta-catenin pathway, (5) HER pathway, (6) tyrosine kinase receptor ligands, and (7) steroid receptors. We tested the hypoxic response of CAFs to show that endometrial CAFs upregulate MMP1 in a HIF-1a-independent manner. In trying to delineate the relationship between expressions of CAF markers and T-cells in the tumor tissue, we observed that FAP-positive CAFs that are derived from CD4/CD8 positive tumor tissue expressed CXCL12 mRNA. The data indicate the role of the CXCL12-CXCR4 pathway of the CAF-rich stroma in the lymphocytic infiltration of the tumor. We demonstrate that endometrial CAFs can be cultured in an enzymatic-digestion-independent manner, and their signaling landscape can be mapped toward understanding CAF-TME dialogue. Our data will help unearth the functional relevance of endometrial CAFs in the context of clinical outcomes and designing CAF-inclusive therapy in the future.
Collapse
Affiliation(s)
- Raed Sulaiman
- Department of Pathology, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Nischal Koirala
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
- Comprehensive Cancer Center, The Ohio State University Wexner Medical CenterColumbus, OH 43210, USA
| | - Jennifer C Aske
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Xiaoqian Lin
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Luis Rojas-Espaillat
- Department of Gynecologic Oncology, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - David Starks
- Department of Gynecologic Oncology, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Adam Dale
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Kris Gaster
- Assistant VP Outpatient Cancer Clinics, Avera Cancer InstituteSioux Falls, SD 57108, USA
| | - Pradip De
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
- Department of Internal Medicine, University of South Dakota SSOMSioux Falls, SD 57108, USA
- VieCureGreenwood Village, CO 80111, USA
| | - Nandini Dey
- Translational Oncology Laboratory, Avera Cancer InstituteSioux Falls, SD 57108, USA
| |
Collapse
|
7
|
Peng X, Zheng J, Liu T, Zhou Z, Song C, Geng Y, Wang Z, Huang Y. Tumor Microenvironment Heterogeneity, Potential Therapeutic Avenues, and Emerging Therapies. Curr Cancer Drug Targets 2024; 24:288-307. [PMID: 37537777 DOI: 10.2174/1568009623666230712095021] [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: 04/17/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVE This review describes the comprehensive portrait of tumor microenvironment (TME). Additionally, we provided a panoramic perspective on the transformation and functions of the diverse constituents in TME, and the underlying mechanisms of drug resistance, beginning with the immune cells and metabolic dynamics within TME. Lastly, we summarized the most auspicious potential therapeutic strategies. RESULTS TME is a unique realm crafted by malignant cells to withstand the onslaught of endogenous and exogenous therapies. Recent research has revealed many small-molecule immunotherapies exhibiting auspicious outcomes in preclinical investigations. Furthermore, some pro-immune mechanisms have emerged as a potential avenue. With the advent of nanosystems and precision targeting, targeted therapy has now transcended the "comfort zone" erected by cancer cells within TME. CONCLUSION The ceaseless metamorphosis of TME fosters the intransigent resilience and proliferation of tumors. However, existing therapies have yet to surmount the formidable obstacles posed by TME. Therefore, scientists should investigate potential avenues for therapeutic intervention and design innovative pharmacological and clinical technologies.
Collapse
Affiliation(s)
- Xintong Peng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jingfan Zheng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Tianzi Liu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ziwen Zhou
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Chen Song
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Geng
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zichuan Wang
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Huang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
8
|
Chen T, Wang Y, Zhu L, Wu J, Lin J, Huang W, Yan D. Hybrid Membrane Camouflaged Chemodrug-Gene Nanoparticles for Enhanced Combination Therapy of Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58067-58078. [PMID: 38056905 DOI: 10.1021/acsami.3c10586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Recently, cell membrane camouflaged nanoparticles (NPs) endowed with natural cellular functions have been extensively studied in various biomedical fields. However, there are few reports about such biomimetic NPs used to codeliver chemodrug and genes for synergistic cancer treatment up to now. Herein, we first prepare chemodrug-gene nanoparticles (Mito-Her2 NPs) by the electrostatic interaction coself-assembly of mitoxantrone hydrochloride (Mito) and human epidermal growth factor receptor-2 antisense oligonucleotide (Her2 ASO). Then, Mito-Her2 NPs are coated by a hybrid membrane (RSHM), consisting of the red blood cell membrane (RBCM) and the SKOV3 ovarian cancer cell membrane (SCM), to produce biomimetic chemodrug-gene nanoparticles (Mito-Her2@RSHM NPs) for combination therapy of ovarian cancer. Mito-Her2@RSHM NPs integrate the advantages of RBCM (e.g., good immune evasion capability and long circulation lifetime in the blood) and SCM (e.g., highly specific cognate recognition) together and improve the anticancer efficacy of Mito-Her2 NPs. The results show that Mito-Her2@RSHM NPs can be devoured by SKOV3 ovarian cancer cells and effectively degraded to release Her2 ASOs and Mito simultaneously. Her2 ASOs can inhibit the expression of endogenous Her2 genes and recover cancer cells' sensitivity to Mito, which ultimately led to a high apoptosis rate of 75.7% in vitro. Mito-Her2@RSHM NPs also show a high tumor suppression rate of 83.33 ± 4.16% in vivo without significant damage to normal tissues. In summary, Mito-Her2@RSHM NPs would be expected as a versatile and safe nanodrug delivery platform with high efficiency for chemo-gene combined cancer treatment.
Collapse
Affiliation(s)
- Tianbao Chen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yuling Wang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Lijuan Zhu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200217, China
| | - Jingchun Wu
- Zhejiang Haobang Chemical Co., LTD, 26 Luyin Road, Quzhou Hi-Tech Industrial Park, Zhejiang 324100, China
| | - Jintang Lin
- Zhejiang Haobang Chemical Co., LTD, 26 Luyin Road, Quzhou Hi-Tech Industrial Park, Zhejiang 324100, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
9
|
Sulaiman R, De P, Aske JC, Lin X, Dale A, Koirala N, Gaster K, Espaillat LR, Starks D, Dey N. Tumor-TME Bipartite Landscape of PD-1/PD-L1 in Endometrial Cancers. Int J Mol Sci 2023; 24:11079. [PMID: 37446260 DOI: 10.3390/ijms241311079] [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: 04/15/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
The bipartite landscape of tumor cells and stromal cells determines a tumor's response to treatment during disease management. In endometrial cancers (ECs), the mechanistic contribution of PD-L1/L2 and PD-1 signaling of the host's tumor microenvironment (TME) (CAF and immune cells) in the context of the tumor cells is elusive. To understand the tumor-stroma-immune crosstalk, we studied the compartmental pattern of PD-L1/L2 and PD-1 expression in EC tissues and their matched CAFs. Over 116 surgically resected tumors (T) and the tumor-adjacent normal tissues (N) were obtained from consented unselected consecutive patients. IHC was performed in T, N-epi-thelium, and the stromal mesenchymal environment (SME; mesenchyme) in the T and N tissues. The staining intensity and distribution patterns of PD-L1/L2 and PD-1 in the FFPE sections of T and N were evaluated by a pathologist using a standard scoring system of TPS and CPS. We tested the PD-L1/L2 and PD-1 immune landscape of tumor-TME pair and normal epithelial-stromal mesenchyme pairs from patients with different grades of disease vis-à-vis their CAF PD-L1 levels. We used qRT-PCR to determine the expressions of mRNAs, while the flow cytometry and ICC determined the level of expression of proteins. We observed higher levels of PD-L1 mRNA and protein expression in primary CAFs from the resected tumor tissue compared to the tumor-adjacent normal tissues. We also determined the expression of patients' soluble PD-L1/L2 as peripheral readouts of PD-L1/L2 and PD-1. As we evaluated the results in the context of their pathological parameters, such as grades, stages, lymphovascular invasion, percentage of myometrial invasion, and dMMR in patients, the dominance of PD-L1 expression in TME was positively correlated to the higher pathological grades of tumors, and its relationship with the dMMR. Since the neutralization of CD8-positive cytotoxic T-cells is PD-L1-dependent, our data indicate that irrespective of the PD-L1 positivity of tumor cells, the PD-L1-positive CAFs can play a critical role in bringing out an additional load of PD-L1 for an effective engagement of PD-1 within a tumor mass.
Collapse
Affiliation(s)
- Raed Sulaiman
- Department of Pathology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Pradip De
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
- Department of Internal Medicine, University of South Dakota SSOM, Sioux Falls, SD 57108, USA
- Viecure, Greenwood Village, CO 80111, USA
| | - Jennifer C Aske
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Xiaoqian Lin
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Adam Dale
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Nischal Koirala
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Kris Gaster
- Assistant VP Outpatient Cancer Clinics, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Luis Rojas Espaillat
- Department of Gynecologic Oncology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - David Starks
- Department of Gynecologic Oncology, Avera Cancer Institute, Sioux Falls, SD 57108, USA
| | - Nandini Dey
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57108, USA
- Department of Internal Medicine, University of South Dakota SSOM, Sioux Falls, SD 57108, USA
| |
Collapse
|
10
|
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
|
11
|
Sulaiman R, De P, Aske JC, Lin X, Dale A, Gaster K, Espaillat LR, Starks D, Dey N. A CAF-Based Two-Cell Hybrid Co-Culture Model to Test Drug Resistance in Endometrial Cancers. Biomedicines 2023; 11:biomedicines11051326. [PMID: 37238998 DOI: 10.3390/biomedicines11051326] [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: 04/07/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The management of advanced or recurrent endometrial cancers presents a challenge due to the development of resistance to treatments. The knowledge regarding the role of the tumor microenvironment (TME) in determining the disease's progression and treatment outcome has evolved in recent years. As a TME component, cancer-associated fibroblasts (CAFs) are essential in developing drug-induced resistance in various solid tumors, including endometrial cancers. Hence, an unmet need exists to test the role of endometrial CAF in overcoming the roadblock of resistance in endometrial cancers. We present a novel tumor-TME two-cell ex vivo model to test CAF's role in resisting the anti-tumor drug, paclitaxel. Endometrial CAFs, both NCAFs (tumor-adjacent normal-tissue-derived CAFs) and TCAFs (tumor-tissue-derived CAFs) were validated by their expression markers. Both TCAFs and NCAFs expressed positive markers of CAF, including SMA, FAP, and S100A4, in varying degrees depending on the patients, while they consistently lacked the negative marker of CAF, EpCAM, as tested via flow cytometry and ICC. CAFs expressed TE-7 and immune marker, PD-L1, via ICC. CAFs better resisted the growth inhibitory effect of paclitaxel on endometrial tumor cells in 2D and 3D formats compared to the resistance of the tumoricidal effect of paclitaxel in the absence of CAFs. TCAF resisted the growth inhibitory effect of paclitaxel on endometrial AN3CA and RL-95-2 cells in an HyCC 3D format. Since NCAF similarly resisted the growth inhibitor action of paclitaxel, we tested NCAF and TCAF from the same patient to demonstrate the protective action of NCAF and TCAF in resisting the tumoricidal effect of paclitaxel in AN3CA in both 2D and 3D matrigel formats. Using this hybrid co-culture CAF and tumor cells, we established a patient-specific, laboratory-friendly, cost-effective, and time-sensitive model system to test drug resistance. The model will help test the role of CAFs in developing drug resistance and contribute to understanding tumor cell-CAF dialogue in gynecological cancers and beyond.
Collapse
Affiliation(s)
- Raed Sulaiman
- Department of Pathology, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - Pradip De
- Translational Oncology Laboratory, Avera Research Institute, Sioux Falls, SD 57105, USA
- Department of Internal Medicine, University of South Dakota SSOM, USD, Sioux Falls, SD 57105, USA
- Viecure, Greenwood Village, CO 80111, USA
| | - Jennifer C Aske
- Translational Oncology Laboratory, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - Xiaoqian Lin
- Translational Oncology Laboratory, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - Adam Dale
- Translational Oncology Laboratory, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - Kris Gaster
- Assistant VP Outpatient Cancer Clinics, Avera Cancer Institute, Sioux Falls, SD 57105, USA
| | - Luis Rojas Espaillat
- Department of Gynecologic Oncology, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - David Starks
- Department of Gynecologic Oncology, Avera Research Institute, Sioux Falls, SD 57105, USA
| | - Nandini Dey
- Translational Oncology Laboratory, Avera Research Institute, Sioux Falls, SD 57105, USA
- Department of Internal Medicine, University of South Dakota SSOM, USD, Sioux Falls, SD 57105, USA
| |
Collapse
|