1
|
Wang L, Hu Z, Bai H, Chang L, Chen C, Li W. miRNA-105-5p regulates the histone deacetylase HDAC2 through FOXG1 to affect the malignant biological behavior of triple-negative breast cancer cells. Am J Med Sci 2025; 369:513-523. [PMID: 39313115 DOI: 10.1016/j.amjms.2024.09.009] [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: 09/07/2023] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer (BC). Some potential molecular targets have been identified, and miR-105-5p was found to be abnormally expressed in TNBC tissues. OBJECTIVE The objective of this study was to probe the effect of miR-105-5p on TNBC via FOXG1/HDAC2-mediated acetylation. METHODS An animal model of TNBC was established by injecting BC cells into the axillary area of nude mice. The levels of miR-105-5p, FOXG1, HDAC2, Bcl-2, Bax, and Ki67 were detected via RT‒qPCR, Western blotting and immunohistochemistry. Flow cytometry, CCK-8, Transwell and colony formation assays were used to measure apoptosis, proliferation and migration, respectively. Total histone acetylation levels were measured by ELISA. The binding of FOXG1 to HDAC2 was detected by co-immunoprecipitation. The binding relationship between miR-105-5p and FOXG1 was verified using a dual-luciferase reporter gene assay. RESULTS In this study, miR-105-5p and HDAC2 were highly expressed in the MDA-MB-231 and BT-549 BC cell lines, whereas FOXG1 was expressed at low levels. The inhibition of miR-105-5p inhibited the proliferation and migration of MDA-MB-231 and BT-549 cells and promoted their apoptosis. Bioinformatics analysis revealed that miR-105-5p and FOXG1 had a negative targeting regulatory relationship. FOXG1 overexpression had a similar effect on cancer cells as the inhibition of miR-105-5p. Moreover, experiments revealed that FOXG1 and HDAC2 could bind to each other and that HDAC2 overexpression or treatment with the histone acetyltransferase inhibitor Garcinol weakened the effect of FOXG1 overexpression. In addition, FOXG1 knockdown inhibited the effect of the miR-105-5p inhibitor, while Garcinol treatment further enhanced the effect of FOXG1 knockdown, inhibited histone acetylation, promoted the proliferation and migration of cancer cells, and inhibited apoptosis. Moreover, the in vivo results confirmed the in vitro results. CONCLUSION miR-105-5p promotes HDAC2 expression by reducing FOXG1, inhibits histone acetylation, and aggravates the malignant biological behavior of TNBC cells.
Collapse
Affiliation(s)
- Li Wang
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Zaoxiu Hu
- Department of Pathology, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Han Bai
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Li Chang
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China
| | - Ceshi Chen
- Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China.
| | - Wenhui Li
- Department of Radiotherapy, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650000, Yunnan, China.
| |
Collapse
|
2
|
Wang C, Yuan F, Yan Z, Zhang T, Fu C, Li Y, Dai G, Kim HS, Xia S, Yu L, Debnath S, Ren WX, Shu J, Qiu M, Kim JS. High Entropy 2D Layered Double Hydroxide Nanosheet Toward Cascaded Nanozyme-Initiated Chemodynamic and Immune Synergistic Therapy. J Am Chem Soc 2025; 147:136-148. [PMID: 39477803 DOI: 10.1021/jacs.4c04523] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
High-entropy nanomaterials (HEMs) are a hot topic in the fields of energy and catalysis. However, in terms of promising biomedical applications, potential therapeutic studies involving HEMs are unprecedented. Herein, we demonstrated high entropy two-dimensional layered double hydroxide (HE-LDH) nanoplatforms with versatile physicochemical advantages that reprogram the tumor microenvironment (TME) and provide antitumor treatment via cascaded nanoenzyme-initiated chemodynamic and immune synergistic therapy. In response to the TME, the multifunctional HE-LDHs sequentially release metal ions, such as Co2+, Fe3+, and Cu2+, exhibiting exquisite superoxide dismutase (SOD), peroxidase (POD), and glutathione peroxidase (GPX) activities. The multiple enzymatic activities convert specific tumor metabolites into a continuous supply of cytotoxic reactive oxygen species (ROS) to relieve hypoxia under a TME. Thus, HE-LDHs facilitate robust nanozyme-initiated chemodynamic therapy (NCDT), achieving high therapeutic efficacy without obvious side effects. In addition, the release of Zn2+ from the HE-LDH matrix triggers the cyclic GMP-AMP synthase/stimulator of interferon gene (cGAS/STING) signaling pathway, boosting the innate immunotherapeutic efficacy. The intratumoral applications of the nanocomposite in tumor-bearing mice models indicate that HE-LDH-mediated NCDT and immune synergistic therapy effectively upregulated the expression of relevant antitumor cytokines and induced cytotoxic T lymphocyte infiltration, showing superior efficacy in inhibiting tumor growth. Therefore, this work opens a new research direction toward synchronized NCDT and immunotherapy of tumors using HEMs for advanced healthcare.
Collapse
Affiliation(s)
- Chen Wang
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Fengying Yuan
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Zichao Yan
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Tianqi Zhang
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Chenchen Fu
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Ya Li
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Guidong Dai
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Hyeong Seok Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Shuwei Xia
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Liangmin Yu
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | | | - Wen Xiu Ren
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Jian Shu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P. R. China
| | - Meng Qiu
- College of Chemistry and Chemical Engineering, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, P. R. China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
3
|
Pandey P, Lakhanpal S, Mahmood D, Kang HN, Kim B, Kang S, Choi J, Choi M, Pandey S, Bhat M, Sharma S, Khan F, Park MN, Kim B. An updated review summarizing the anticancer potential of flavonoids via targeting NF-kB pathway. Front Pharmacol 2025; 15:1513422. [PMID: 39834817 PMCID: PMC11743680 DOI: 10.3389/fphar.2024.1513422] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Nuclear factor-κB (NF-κB) cell signaling pathway is essential for the progression and development of numerous human disorders, including cancer. NF-κB signaling pathway regulates a wide range of physiological processes, such as cell survival, growth, and migration. Deregulated NF-kB signaling resulted in unregulated cell proliferation, viability, movement, and invasion, thus promoting tumor development. Recent findings have increasingly shown that plant derived phytochemicals that inhibit NF-κB signaling have the potential to be employed in cancer therapeutics. Flavonoids are a group of polyphenolic natural compounds present in various plants and their fruits, vegetables, and leaves. These compounds have numerous medicinal properties owing to their antioxidant, anti-inflammatory, antiviral, and antitumor characteristics. The main mechanism by which these flavonoids exhibit their anticancer potential is via potent antioxidative and immunomodulatory actions. Current research reports have demonstrated that these flavonoids exhibited their anticancer effects via suppressing the NF-κB signaling. Based on these facts, we have comprehensively outlined the cancer promoting role of NF-κB pathway in various processes including tumor progression, drug resistance, angiogenesis and metastasis. In addition to these, we also summarize the anticancer potential of flavonoids by specifically targeting the NF-κB pathway in various types of cancers.
Collapse
Affiliation(s)
- Pratibha Pandey
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Danish Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Byunggyu Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Min Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, India
| | - Mahakshit Bhat
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Fahad Khan
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Shi X, Cheng X, Jiang A, Shi W, Zhu L, Mou W, Glaviano A, Liu Z, Cheng Q, Lin A, Wang L, Luo P. Immune Checkpoints in B Cells: Unlocking New Potentials in Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403423. [PMID: 39509319 PMCID: PMC11653663 DOI: 10.1002/advs.202403423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/26/2024] [Indexed: 11/15/2024]
Abstract
B cells are crucial component of humoral immunity, and their role in the tumor immune microenvironment (TME) has garnered significant attention in recent years. These cells hold great potential and application prospects in the field of tumor immunotherapy. Research has demonstrated that the TME can remodel various B cell functions, including proliferation, differentiation, antigen presentation, and antibody production, thereby invalidating the anti-tumor effects of B cells. Concurrently, numerous immune checkpoints (ICs) on the surface of B cells are upregulated. Aberrant B-cell IC signals not only impair the function of B cells themselves, but also modulate the tumor-killing effects of other immune cells, ultimately fostering an immunosuppressive TME and facilitating tumor immune escape. Blocking ICs on B cells is beneficial for reversing the immunosuppressive TME and restoring anti-tumor immune responses. In this paper, the intricate connection between B-cell ICs and the TME is delved into, emphasizing the critical role of targeting B-cell ICs in anti-tumor immunity, which may provide valuable insights for the future development of tumor immunotherapy based on B cells.
Collapse
Affiliation(s)
- Xiaoye Shi
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiangshu Cheng
- College of Bioinformatics Science and TechnologyHarbin Medical University157 Baojian Road. Nangang District, HarbinHeilongiiang150076China
| | - Aimin Jiang
- Department of UrologyChanghai HospitalNaval Medical University (Second Military Medical University)Shanghai200433China
| | - Wenjie Shi
- Molecular and Experimental SurgeryUniversity Clinic for General‐Visceral‐Vascular‐ and Trans‐Plantation SurgeryMedical Faculty University Hospital MagdeburgOtto‐von Guericke University39120MagdeburgGermany
| | - Lingxuan Zhu
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
| | - Weiming Mou
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- Department of UrologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Antonino Glaviano
- Department of BiologicalChemical and Pharmaceutical Sciences and TechnologiesUniversity of PalermoPalermo90123Italy
| | - Zaoqu Liu
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Anqi Lin
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
| | - Linhui Wang
- Department of UrologyChanghai HospitalNaval Medical University (Second Military Medical University)Shanghai200433China
| | - Peng Luo
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdong510282China
- Cancer Centre and Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacau SAR999078China
| |
Collapse
|
5
|
Yu M, Li D, Zhang L, Wang K. Identification and validation of a prognostic model based on immune-related genes in ovarian carcinoma. PeerJ 2024; 12:e18235. [PMID: 39494284 PMCID: PMC11531744 DOI: 10.7717/peerj.18235] [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: 06/11/2024] [Accepted: 09/15/2024] [Indexed: 11/05/2024] Open
Abstract
Background A novel valuable prognostic model has been developed on the basis of immune-related genes (IRGs), which could be used to estimate overall survival (OS) in ovarian cancer (OC) patients in The Cancer Genome Atlas (TCGA) dataset and the International Cancer Genome Consortium (ICGC) dataset. Methods This prognostic model was engineered by employing LASSO regression in training cohort (TCGA dataset). The corresponding growth predictive values of this model for individualized survival was evaluated using survival analysis, receiver operating characteristic curve (ROC curve), and risk curve analysis. Combined with clinical characteristics, a model risk score nomogram for OS was well built. Thereafter, depended on the model risk score, patients were divided into high and low risk subgroups. The survival difference between these subgroups was measured using Kaplan-Meier survival method. In addition, correlations containing pathway enrichment, treatment, immune cell infiltration and the prognostic model were also analyzed. We established the ovarian cancer cell line W038 for this study and identified the performances of GBP1P1 knockdown on a series of activities including cellular proliferation, apoptosis, migration, and invasion of W038 cells in vitro. Results We constructed a 25-genes prognostic model (TNFAIP8L3, PI3, TMEM181, GBP1P1 (LOC400759), STX18, KIF26B, MRPS11, CACNA1C, PACSIN3, GMPR, MANF, PYGB, SNRPA1, ST7L, ZBP1, BMPR1B-DT, STAC2, LINC02585, LYPD6, NSG1, ACOT13, FAM120B, LEFTY1, SULT1A2, FZD3). The areas under the curves (AUC) of 1, 2 and 3 years were 0.806, 0.773 and 0.762, in the TCGA cohort, respectively. Besides, the effectiveness of the model was verified using ICGC testing data. Univariate and multivariate Cox regression analysis exposes the risk score as an independent prognosis predictor for OS both in the TCGA and ICGC cohort. In summary, we utilized comprehensive bioinformatics analysis to build an effective prognostic gene model for OC patients. These bioinformatic results suggested that GBP1P1 could act as a novel biomarker for OC. GBP1P1 knockdown substantially inhibited the proliferation, migration, and invasion of W038 cells in vitro, and increased the percentage of apoptotic W038 cells. Conclusions The analyses of genetic status of patients with 25-genes model might improve the ability to predict the prognosis of patients with OC and help to select patients suit able to therapies. Immune-related gene GBP1P1 might serve as prognostic biomarker for OC.
Collapse
Affiliation(s)
- Min Yu
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Dan Li
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Li Zhang
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ke Wang
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
6
|
Li Y, Ma J, Ma X, Chen C, Ruan M, Yang W, Shui R. PD-L1 expression and tumor-infiltrating lymphocytes: Correlations and prognostic values in Chinese triple-negative breast cancer patients with different molecular subtyping. Pathol Res Pract 2024; 262:155556. [PMID: 39216323 DOI: 10.1016/j.prp.2024.155556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To investigate the correlation between programmed death ligand-1 (PD-L1) expression and tumor-infiltrating lymphocytes (TILs) and evaluate the prognostic value of PD-L1 and TILs in Chinese triple-negative breast cancer (TNBC) patients with different molecular subtype METHODS: This retrospective study was conducted at 2020. Specifically, the pre-chemotherapy clinical data and non-stained tissue blocks of 465 TNBC patients visited the Fudan University Shanghai Cancer Center (FUSCC) between 2008 and 2014 were collected, with their blocks sliced and stained using PD-L1(SP142), and the outcome of subsequent chemotherapy obtained in 2020. The relapse-free survival (RFS) of the study population was calculated. The baseline PD-L1 expression status correlations with TILs and molecular subtypes were assessed using Spearman's rank correlation analysis and the Kruskal-Wallis test. Kaplan-Meier survival analyses were undertaken to evaluate the prognosis value of TILs and PD-L1 expression. RESULTS PD-L1 expression status on IC was moderately and positively correlated with stromal tumor-infiltrating lymphocytes (sTILs) (rs = 0.502, P <0.001) and iTILs (rs = 0.410, P < 0.001), respectively. PD-L1 expression status and TILs showed significant differences among molecular subtypes (P < 0.001), with the highest proportion of PD-L1+ and high TILs patients observed in the immunomodulatory (IM) subtype. TILs were significantly associated with RFS. Moreover, sTILs could act as an independent predictor of RFS (RR 0.953, 95 % CI 0.920 ∼ 0.987, P = 0.007), while PD-L1 expression status did not show the same prognostic significance. CONCLUSIONS The incorporation of pre-treatment TILs and PD-L1 expression status as valuable tools for optimizing patient selection for immunotherapy and managing the risks associated with chemotherapy in Chinese TNBC patients. DATA AVAILABILITY The data sets generated and analyzed during the current study are available from the corresponding author.
Collapse
Affiliation(s)
- Yanping Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jing Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoxi Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Chen
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Miao Ruan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wentao Yang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Ruohong Shui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Chaudhry Z, Boyadzhyan A, Sasaninia K, Rai V. Targeting Neoantigens in Cancer: Possibilities and Opportunities in Breast Cancer. Antibodies (Basel) 2024; 13:46. [PMID: 38920970 PMCID: PMC11200483 DOI: 10.3390/antib13020046] [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: 05/13/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
As one of the most prevalent forms of cancer worldwide, breast cancer has garnered significant attention within the clinical research setting. While traditional treatment employs a multidisciplinary approach including a variety of therapies such as chemotherapy, hormone therapy, and even surgery, researchers have since directed their attention to the budding role of neoantigens. Neoantigens are defined as tumor-specific antigens that result from a multitude of genetic alterations, the most prevalent of which is the single nucleotide variant. As a result of their foreign nature, neoantigens elicit immune responses upon presentation by Major Histocompatibility Complexes I and II followed by recognition by T cell receptors. Previously, researchers have been able to utilize these immunogenic properties and manufacture neoantigen-specific T-cells and neoantigen vaccines. Within the context of breast cancer, biomarkers such as tumor protein 53 (TP53), Survivin, Partner and Localizer of BRCA2 (PALB2), and protein tyrosine phosphatase receptor T (PTPRT) display exceeding potential to serve as neoantigens. However, despite their seemingly limitless potential, neoantigens must overcome various obstacles if they are to be fairly distributed to patients. For instance, a prolonged period between the identification of a neoantigen and the dispersal of treatment poses a serious risk within the context of breast cancer. Regardless of these current obstacles, it appears highly promising that future research into neoantigens will make an everlasting impact on the health outcomes within the realm of breast cancer. The purpose of this literature review is to comprehensively discuss the etiology of various forms of breast cancer and current treatment modalities followed by the significance of neoantigens in cancer therapeutics and their application to breast cancer. Further, we have discussed the limitations, future directions, and the role of transcriptomics in neoantigen identification and personalized medicine. The concepts discussed in the original and review articles were included in this review article.
Collapse
Affiliation(s)
| | | | | | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (Z.C.); (A.B.); (K.S.)
| |
Collapse
|
8
|
Wang Y, Bergman DR, Trujillo E, Pearson AT, Sweis RF, Jackson TL. Mathematical model predicts tumor control patterns induced by fast and slow cytotoxic T lymphocyte killing mechanisms. Sci Rep 2023; 13:22541. [PMID: 38110479 PMCID: PMC10728095 DOI: 10.1038/s41598-023-49467-6] [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: 09/08/2023] [Accepted: 12/08/2023] [Indexed: 12/20/2023] Open
Abstract
Immunotherapy has dramatically transformed the cancer treatment landscape largely due to the efficacy of immune checkpoint inhibitors (ICIs). Although ICIs have shown promising results for many patients, the low response rates in many cancers highlight the ongoing challenges in cancer treatment. Cytotoxic T lymphocytes (CTLs) execute their cell-killing function via two distinct mechanisms: a fast-acting, perforin-mediated process and a slower, Fas ligand (FasL)-driven pathway. Evidence also suggests that the preferred killing mechanism of CTLs depends on the antigenicity of tumor cells. To determine the critical factors affecting responses to ICIs, we construct an ordinary differential equation model describing in vivo tumor-immune dynamics in the presence of active or blocked PD-1/PD-L1 immune checkpoint. Specifically, we identify important aspects of the tumor-immune landscape that affect tumor size and composition in the short and long term. We also generate a virtual cohort of mice with diverse tumor and immune attributes to simulate the outcomes of immune checkpoint blockade in a heterogeneous population. By identifying key tumor and immune characteristics associated with tumor elimination, dormancy, and escape, we predict which fraction of a population potentially responds well to ICIs and ways to enhance therapeutic outcomes with combination therapy.
Collapse
Affiliation(s)
- Yixuan Wang
- Department of Mathematics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daniel R Bergman
- Department of Mathematics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Erica Trujillo
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, 60637, USA
| | - Alexander T Pearson
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, 60637, USA
| | - Randy F Sweis
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, 60637, USA.
| | - Trachette L Jackson
- Department of Mathematics, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
9
|
Wang Y, Lu L, Ling C, Zhang P, Han R. Potential of Dietary HDAC2i in Breast Cancer Patients Receiving PD-1/PD-L1 Inhibitors. Nutrients 2023; 15:3984. [PMID: 37764768 PMCID: PMC10537481 DOI: 10.3390/nu15183984] [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: 08/02/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is a lethal malignancy with high morbidity and mortality but lacks effective treatments thus far. Despite the introduction of immune checkpoint inhibitors (ICIs) (including PD-1/PD-L1 inhibitors), durable and optimal clinical benefits still remain elusive for a considerable number of BC patients. To break through such a dilemma, novel ICI-based combination therapy has been explored for enhancing the therapeutic effect. Recent evidence has just pointed out that the HDAC2 inhibitor (HDAC2i), which has been proven to exhibit an anti-cancer effect, can act as a sensitizer for ICIs therapy. Simultaneously, dietary intervention, as a crucial supportive therapy, has been reported to provide ingredients containing HDAC2 inhibitory activity. Thus, the novel integration of dietary intervention with ICIs therapy may offer promising possibilities for improving treatment outcomes. In this study, we first conducted the differential expression and prognostic analyses of HDAC2 and BC patients using the GENT2 and Kaplan-Meier plotter platform. Then, we summarized the potential diet candidates for such an integrated therapeutic strategy. This article not only provides a whole new therapeutic strategy for an HDAC2i-containing diet combined with PD-1/PD-L1 inhibitors for BC treatment, but also aims to ignite enthusiasm for exploring this field.
Collapse
Affiliation(s)
- Yuqian Wang
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
| | - Changquan Ling
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - Ping Zhang
- Center for Integrative Conservation, Yunnan Key Laboratory for the Conservation of Tropical Rainforests and Asian Elephants, Xishuangbanna Tropical Botanical Garden, Xishuangbanna 666303, China
| | - Rui Han
- Department of Chinese Medicine Oncology, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
- Department of Chinese Medicine, Naval Medical University, Shanghai 200433, China
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA
- School of Medicine, Center for Biomedical Data Science, Yale University, 60 College Street, New Haven, CT 06520, USA
- Yale Cancer Center, Yale University, 60 College Street, New Haven, CT 06520, USA
- Department of Oncology, The First Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
10
|
Lin J, Fang W, Xiang Z, Wang Q, Cheng H, Chen S, Fang J, Liu J, Wang Q, Lu Z, Ma L. Glycolytic enzyme HK2 promotes PD-L1 expression and breast cancer cell immune evasion. Front Immunol 2023; 14:1189953. [PMID: 37377974 PMCID: PMC10291184 DOI: 10.3389/fimmu.2023.1189953] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Immune therapies targeting the PD-1/PD-L1 pathway have been employed in the treatment of breast cancer, which requires aerobic glycolysis to sustain breast cancer cells growth. However, whether PD-L1 expression is regulated by glycolysis in breast cancer cells remains to be further elucidated. Here, we demonstrate that glycolytic enzyme hexokinase 2 (HK2) plays a crucial role in upregulating PD-L1 expression. Under high glucose conditions, HK2 acts as a protein kinase and phosphorylates IκBα at T291 in breast cancer cells, leading to the rapid degradation of IκBα and activation of NF-κB, which enters the nucleus and promotes PD-L1 expression. Immunohistochemistry staining of human breast cancer specimens and bioinformatics analyses reveals a positive correlation between HK2 and PD-L1 expression levels, which are inversely correlated with immune cell infiltration and survival time of breast cancer patients. These findings uncover the intrinsic and instrumental connection between aerobic glycolysis and PD-L1 expression-mediated tumor cell immune evasion and underscore the potential to target the protein kinase activity of HK2 for breast cancer treatment.
Collapse
Affiliation(s)
- Jichun Lin
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenshuo Fang
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhuo Xiang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qingqing Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Huapeng Cheng
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Shimin Chen
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Fang
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Leina Ma
- Department of Oncology, the Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
11
|
Li H, Wang L, Zhang W, Dong Y, Cai Y, Huang X, Dong X. Overexpression of PKMYT1 associated with poor prognosis and immune infiltration may serve as a target in triple-negative breast cancer. Front Oncol 2023; 12:1002186. [PMID: 36793346 PMCID: PMC9922894 DOI: 10.3389/fonc.2022.1002186] [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: 07/24/2022] [Accepted: 09/14/2022] [Indexed: 01/31/2023] Open
Abstract
Breast cancer (BC) is one of the most common malignancies among women worldwide. It is necessary to search for improvement in diagnosis and treatment methods to improve the prognosis. Protein kinase, membrane associated tyrosine/threonine 1 (PKMYT1), a member of the Wee family of protein kinases, has been studied in some tumors except BC. This study has explored that PKMYT1 functional role by bioinformatics methods combined with local clinical samples and experiments. Comprehensive analysis showed that PKMYT1 expression was higher in BC tissues, especially in advanced patients than that in normal breast tissues. The expression of PKMYT1 was an independent determinant for BC patients' prognosis when combined with the clinical features. In addition, based on multi-omics analysis, we found that the PKMYT1 expression was closely relevant to several oncogenic or tumor suppressor gene variants. The analysis of single-cell sequencing indicated that PKMYT1 expression was upregulated in triple-negative breast cancer (TNBC), consistent with the results of bulk RNA-sequencing. High PKMYT1 expression was correlated with a poor prognosis. Functional enrichment analysis revealed that PKMYT1 expression was associated with cell cycle-related, DNA replication-related, and cancer-related pathways. Further research revealed that PKMYT1 expression was linked to immune cell infiltration in the tumor microenvironment. Additionally, loss-of-function experiments in vitro were performed to investigate the role of PKMYT1. TNBC cell lines' proliferation, migration, and invasion were inhibited when PKMYT1 expression was knock-down. Besides, the down-regulation of PKMYT1 induced apoptosis in vitro. As a result, PKMYT1 might be a biomarker for prognosis and a therapeutic target for TNBC.
Collapse
Affiliation(s)
- Huihui Li
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Wang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| | - Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youting Dong
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Yefeng Cai
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Department of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoli Huang
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Department of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,*Correspondence: Xiaoli Huang, ; Xubin Dong,
| | - Xubin Dong
- Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,*Correspondence: Xiaoli Huang, ; Xubin Dong,
| |
Collapse
|
12
|
Cui H, Ren X, Dai L, Chang L, Liu D, Zhai Z, Kang H, Ma X. Comprehensive analysis of nicotinamide metabolism-related signature for predicting prognosis and immunotherapy response in breast cancer. Front Immunol 2023; 14:1145552. [PMID: 36969219 PMCID: PMC10031006 DOI: 10.3389/fimmu.2023.1145552] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Background Breast cancer (BC) is the most common malignancy among women. Nicotinamide (NAM) metabolism regulates the development of multiple tumors. Herein, we sought to develop a NAM metabolism-related signature (NMRS) to make predictions of survival, tumor microenvironment (TME) and treatment efficacy in BC patients. Methods Transcriptional profiles and clinical data from The Cancer Genome Atlas (TCGA) were analyzed. NAM metabolism-related genes (NMRGs) were retrieved from the Molecular Signatures Database. Consensus clustering was performed on the NMRGs and the differentially expressed genes between different clusters were identified. Univariate Cox, Lasso, and multivariate Cox regression analyses were sequentially conducted to develop the NAM metabolism-related signature (NMRS), which was then validated in the International Cancer Genome Consortium (ICGC) database and Gene Expression Omnibus (GEO) single-cell RNA-seq data. Further studies, such as gene set enrichment analysis (GSEA), ESTIMATE, CIBERSORT, SubMap, and Immunophenoscore (IPS) algorithm, cancer-immunity cycle (CIC), tumor mutation burden (TMB), and drug sensitivity were performed to assess the TME and treatment response. Results We identified a 6-gene NMRS that was significantly associated with BC prognosis as an independent indicator. We performed risk stratification according to the NMRS and the low-risk group showed preferable clinical outcomes (P < 0.001). A comprehensive nomogram was developed and showed excellent predictive value for prognosis. GSEA demonstrated that the low-risk group was predominantly enriched in immune-associated pathways, whereas the high-risk group was enriched in cancer-related pathways. The ESTIMATE and CIBERSORT algorithms revealed that the low-risk group had a higher abundance of anti-tumor immunocyte infiltration (P < 0.05). Results of Submap, IPS, CIC, TMB, and external immunotherapy cohort (iMvigor210) analyses showed that the low-risk group were indicative of better immunotherapy response (P < 0.05). Conclusions The novel signature offers a promising way to evaluate the prognosis and treatment efficacy in BC patients, which may facilitate clinical practice and management.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaobin Ma
- *Correspondence: Xiaobin Ma, ; Huafeng Kang,
| |
Collapse
|
13
|
Yaman I, Ağaç Çobanoğlu D, Xie T, Ye Y, Amit M. Advances in understanding cancer-associated neurogenesis and its implications on the neuroimmune axis in cancer. Pharmacol Ther 2022; 239:108199. [PMID: 35490859 PMCID: PMC9991830 DOI: 10.1016/j.pharmthera.2022.108199] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/07/2023]
Abstract
Nerves and immunologic mediators play pivotal roles in body homeostasis by interacting with each other through diverse mechanisms. The spread of nerves in the tumor microenvironment increases tumor cell proliferation and disease progression, and this correlates with poor patient outcomes. The effects of sympathetic and parasympathetic nerves on cancer regulation are being investigated. Recent findings demonstrate the possibility of developing therapeutic strategies that target the tumor microenvironment and its components such as immune cells, neurotransmitters, and extracellular vesicles. Therefore, examining and understanding the mechanisms and pathways associated with the sympathetic and parasympathetic nervous systems, neurotransmitters, cancer-derived mediators and their interactions with the immune system in the tumor microenvironment may lead to the development of new cancer treatments. This review discusses the effects of nerve cells, immune cells, and cancer cells have on each other that regulate neurogenesis, cancer progression, and dissemination.
Collapse
Affiliation(s)
- Ismail Yaman
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Didem Ağaç Çobanoğlu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tongxin Xie
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yi Ye
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Head and Neck Surgery, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
14
|
Temsirolimus Enhances Anti-Cancer Immunity by Inducing Autophagy-Mediated Degradation of the Secretion of Small Extracellular Vesicle PD-L1. Cancers (Basel) 2022; 14:cancers14174081. [PMID: 36077620 PMCID: PMC9454510 DOI: 10.3390/cancers14174081] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Immune checkpoint blockade therapies (ICBT) have increasing importance in patient survival and prognosis because it enhances immune cell activation by inhibiting the binding of programmed death-ligand 1 (PD-L1) of tumor and programmed death-1 (PD-1) of T cells. However, tumor-derived small extracellular vesicle (sEV) PD-L1 trigger low reactivity in immunotherapy because it promotes tumor growth and metastasis and inhibits activation of immune cell. In this study, temsirolimus (TEM) which the Food and Drug Administration (FDA) approved as a targeted anti-cancer drug, inhibited tumor-derived sEV PD-L1 secretion by activating autophagy. In addition, TEM induced systemic anti-cancer immunity by increasing the number and activation of CD4+ and CD8+ T cells. Therefore, TEM showed that the anti-cancer effect was better in the breast cancer-bearing-immunocompetent mice than in the nude mice. In summary, we suggest that TEM can overcome sEV PD-L1-mediated immunosuppression in patients with cancer through activation of the immune system in the body by inhibiting tumor-derived sEV PD-L1. Abstract Tumor-derived small extracellular vesicle (sEV) programmed death-ligand 1 (PD-L1) contributes to the low reactivity of cells to immune checkpoint blockade therapy (ICBT), because sEV PD-L1 binds to programmed death 1 (PD-1) in immune cells. However, there are no commercially available anti-cancer drugs that activate immune cells by inhibiting tumor-derived sEV PD-L1 secretion and cellular PD-L1. Here, we aimed to investigate if temsirolimus (TEM) inhibits both sEV PD-L1 and cellular PD-L1 levels in MDA-MB-231 cells. In cancer cell autophagy activated by TEM, multivesicular bodies (MVBs) associated with the secretion of sEV are degraded through colocalization with autophagosomes or lysosomes. TEM promotes CD8+ T cell-mediated anti-cancer immunity in co-cultures of CD8+ T cells and tumor cells. Furthermore, the combination therapy of TEM and anti-PD-L1 antibodies enhanced anti-cancer immunity by increasing both the number and activity of CD4+ and CD8+ T cells in the tumor and draining lymph nodes (DLNs) of breast cancer-bearing immunocompetent mice. In contrast, the anti-cancer effect of the combination therapy with TEM and anti-PD-L1 antibodies was reversed by the injection of exogenous sEV PD-L1. These findings suggest that TEM, previously known as a targeted anti-cancer drug, can overcome the low reactivity of ICBT by inhibiting sEV PD-L1 and cellular PD-L1 levels.
Collapse
|
15
|
Fang Y, Liu J, Zhang Q, She C, Zheng R, Zhang R, Chen Z, Chen C, Wu J. Overexpressed VDAC1 in breast cancer as a novel prognostic biomarker and correlates with immune infiltrates. World J Surg Oncol 2022; 20:211. [PMID: 35729567 PMCID: PMC9215028 DOI: 10.1186/s12957-022-02667-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/28/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND More and more evidence suggests that cancer is a mitochondrial metabolic disease recently and mitochondria dysfunction is critical to tumorigenesis. As a gatekeeper of mitochondria, the voltage-dependent anion channel 1 (VDAC1) is associated with the development of breast cancer (BC). However, its potential mechanism and clinical significance remain unclear; thus, in this research, we aimed to explore it. METHODS VDAC1 expression in BC tissues and normal tissues was obtained from The Cancer Genome Atlas (TCGA) and validated by datasets from the gene expression omnibus (GEO) database. Then, the relationships between VDAC1 expression and clinicopathological features were analyzed. Receiver operating characteristics (ROC) curves were used to identify the diagnostic value of VDAC1. The prognostic value was evaluated by Kaplan-Meier survival curves and Cox regression analysis. VDAC1 with its co-expression genes were subjected to enrichment analysis to explore potential mechanisms in BC and the protein-protein interaction (PPI) network was constructed. At last, the association between VDAC1 expression and infiltration levels of immune cell infiltration by various methods, as well as their corresponding markers, was analyzed. We also analyzed the correction between VDAC1 expression and eight immune checkpoint genes and the tumor immune dysfunction and exclusion (TIDE) scores of each BC sample in TCGA were calculated and the differences between high and low VDAC1 expression groups were analyzed. RESULTS VDAC1 expression was remarkably elevated in BC (p < 0.001), and high expression of VDAC1 was associated with the positive expression of ER (p = 0.004), PR (p = 0.033), and HER2 (p = 0.001). ROC analysis suggested that VDAC1 had diagnosed value in BC. The Kaplan-Meier analysis suggested that higher expression of VDAC1 was associated with shorter overall survival (OS), and further Cox regression analysis revealed that VDAC1 was an independent factor of unfavorable prognosis in BC patients. Enrichment analysis of VDAC1 and its co-expression suggested that VDAC1 was related to the regulation of mitochondrial energy metabolism and protein modification, and the HIF-1 singing pathway might be the potential mechanism in BC. Notably, we found that VDAC1 expression was infiltration levels of most types of immune cells, as well as the expression of marker genes of immune cells. The ICGs PDCD1, CTLA4, LAG3, SIGLEC15, and TIGIT were negatively corrected with VDAC1 expression in BC. TIDE scores between the low and high expression groups showed no difference. CONCLUSION Overexpressed VDAC1 in BC could be severed as a novel biomarker for diagnosis and VDAC1 was an independent factor for adverse prognosis prediction. Our study revealed that VDAC1 might inhibit tumor immunity and might be a novel therapeutic target in BC.
Collapse
Affiliation(s)
- Yutong Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Junpeng Liu
- Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Qunchen Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Chuanghong She
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Rongji Zheng
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Rendong Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Zexiao Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Chunfa Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| | - Jundong Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Rd, Shantou, 515041 Guangdong China
- The Department of Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, 515041 Guangdong China
| |
Collapse
|
16
|
Huang L, Liu X, Li L, Wang L, Wu N, Liu Z. Novel immune subtypes identification of HER2-positive breast cancer based on immunogenomic landscape. Med Oncol 2022; 39:92. [PMID: 35568771 DOI: 10.1007/s12032-022-01690-3] [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: 11/17/2021] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
HER2 positive BC is heterogeneous. But few studies discussed the classification of HER2-positive BC based on immune-related signatures. Using three publicly BC genomics datasets, we classified HER2 positive BC based on 33 immune-related signatures and used unsupervised machine learning methods to predict and perform the classification. We grouped three HER2-positive BC subtypes that we called Immune-High (IM-H), Immune-Medium (IM-M), and Immune-Low (IM-L), and manifested this categorization was predictable, duplicable and reliable by analyzing another dataset. Compared to other subtypes, IM-H had a higher immune cell infiltration level and stronger anti-tumor immune activities, as well as better clinical survival outcome. Besides these signatures, there were some cancer-related pathways which were hyperactivated in IM-H, including cytokine-cytokine receptor interactions, antigen processing and presentation pathways, natural killer cell-mediated cytotoxicity, Th1 and Th2 cell differentiation, chemokine signaling pathway, Th17 cell differentiation, B and T cell receptor signaling, NF-kappa B signaling, PD-L1 expression and PD-1 checkpoint pathway in cancer, TNF signaling, IL-17 signaling, NOD-like receptor signaling and Toll-like receptor signaling. By contrast, IM-L showed depressed immune-related signatures and enhanced activation of lycosylphosphatidylinositol-anchor biosynthesis and mismatch repair. Moreover, we discovered a gene co-expression network focused on eight transcription factor genes (EOMES, TBX21, GFI1, IRF4, POU2AF1, CIITA, FOXP3 and TOX) and one tumor suppress gene (PRF1), which were closely related with tumor immune. We identified three HER2-positive BC subtypes based on immune-related signatures, which had potential clinical implications and promoted the optimal stratification of HER2-positive BC responsive to immunotherapy.
Collapse
Affiliation(s)
- Lingli Huang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Xin Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Li Li
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Lei Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Nan Wu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Zhixian Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
17
|
Le TMD, Yoon AR, Thambi T, Yun CO. Polymeric Systems for Cancer Immunotherapy: A Review. Front Immunol 2022; 13:826876. [PMID: 35273607 PMCID: PMC8902250 DOI: 10.3389/fimmu.2022.826876] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy holds enormous promise to create a new outlook of cancer therapy by eliminating tumors via activation of the immune system. In immunotherapy, polymeric systems play a significant role in improving antitumor efficacy and safety profile. Polymeric systems possess many favorable properties, including magnificent biocompatibility and biodegradability, structural and component diversity, easy and controllable fabrication, and high loading capacity for immune-related substances. These properties allow polymeric systems to perform multiple functions in immunotherapy, such as immune stimulants, modifying and activating T cells, delivery system for immune cargos, or as an artificial antigen-presenting cell. Among diverse immunotherapies, immune checkpoint inhibitors, chimeric antigen receptor (CAR) T cell, and oncolytic virus recently have been dramatically investigated for their remarkable success in clinical trials. In this report, we review the monotherapy status of immune checkpoint inhibitors, CAR-T cell, and oncolytic virus, and their current combination strategies with diverse polymeric systems.
Collapse
Affiliation(s)
- Thai Minh Duy Le
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| | - Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea.,GeneMedicine CO., Ltd., Seoul, South Korea
| |
Collapse
|
18
|
Necroptosis-Associated lncRNA Prognostic Model and Clustering Analysis: Prognosis Prediction and Tumor-Infiltrating Lymphocytes in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:7099930. [PMID: 35528236 PMCID: PMC9068297 DOI: 10.1155/2022/7099930] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 12/19/2022]
Abstract
Necroptosis plays an important role in tumor genesis and progression. This study aims to identify necroptosis-related lncRNAs (NR-lncRNAs) in breast cancer (BC), and their prognostic value and relationship with the tumor immune environment (TIE) through bioinformatics. Methods. A total of 67 necroptosis-related genes (NRGs) are retrieved, and 13 prognostically relevant NR-lncRNAs are identified by co-expression and Univariate Cox regression analyses. After unsupervised clustering analysis, the patients are classified into three clusters, and their survival and immune infiltration are compared. Lasso regression analysis is conducted to construct a prognostic model using eight lncRNAs (USP30-AS1, AC097662.1, AC007686.3, AL133467.1, AP006284.1, NDUFA6-DT, LINC01871, AL135818.1). The model is validated by Kaplan-Meier survival analysis, Multivariate Cox regression analysis, and receiver-operating characteristic (ROC) curves. Correlation analysis is useful to identify associations between risk scores and clinicopathological features. GSEA, drug prediction, and immune checkpoints analysis are further used to differentiate between the risk groups. Results. The C3 cluster has longer overall survival (OS) and the highest immune score, indicative of an immunologically hot tumor that may be sensitive to immunotherapy. Furthermore, the OS is significantly higher in the low-risk group, even after dividing the patients into subgroups with different clinical characteristics. The area under the ROC curve (AUC) for 1-, 3-, and 5-year survival in the training set are 0.761, 0.734, and 0.664, respectively, which indicate the moderate predictive performance of the model. Conclusion. NR-lncRNAs can predict the prognosis of BC, distinguish between hot and cold tumors, and are potential predictive markers of the immunotherapy response.
Collapse
|
19
|
Liu B, Ji Q, Cheng Y, Liu M, Zhang B, Mei Q, Liu D, Zhou S. Biomimetic GBM-targeted drug delivery system boosting ferroptosis for immunotherapy of orthotopic drug-resistant GBM. J Nanobiotechnology 2022; 20:161. [PMID: 35351131 PMCID: PMC8962245 DOI: 10.1186/s12951-022-01360-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/08/2022] [Indexed: 01/13/2023] Open
Abstract
Background Clinical studies have shown that the efficacy of programmed cell death receptor-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors on glioblastoma (GBM) is much lower than what is expected because of the low immunogenicity of GBM. Ferroptosis of cancer cells can induce the maturation of dendritic cells (DC cells) and increase the activity of T cell. The activated T cells release IFN-γ, which subsequently induces the ferroptosis of cancer cells. Thus, the aim of this paper is to set up a new GBM-targeted drug delivery system (Fe3O4-siPD-L1@M-BV2) to boost ferroptosis for immunotherapy of drug-resistant GBM. Results Fe3O4-siPD-L1@M-BV2 significantly increased the accumulation of siPD-L1 and Fe2+ in orthotopic drug-resistant GBM tissue in mice. Fe3O4-siPD-L1@M-BV2 markedly decreased the protein expression of PD-L1 and increased the ratio between effector T cells and regulatory T cells in orthotopic drug-resistant GBM tissue. Moreover, Fe3O4-siPD-L1@M-BV2 induced ferroptosis of GBM cells and maturation of DC cell, and it also increased the ratio between M1-type microglia and M2-type microglia in orthotopic drug-resistant GBM tissue. Finally, the growth of orthotopic drug-resistant GBM in mice was significantly inhibited by Fe3O4-siPD-L1@M-BV2. Conclusion The mutual cascade amplification effect between ferroptosis and immune reactivation induced by Fe3O4-siPD-L1@M-BV2 significantly inhibited the growth of orthotopic drug-resistant GBM and prolonged the survival time of orthotopic drug-resistant GBM mice. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01360-6.
Collapse
Affiliation(s)
- Bao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Bangle Zhang
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Qibing Mei
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China
| | - Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China. .,Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
20
|
Gershtein ES, Korotkova EA, Vorotnikov IK, Sokolov NY, Ermilova VD, Mochalova AS, Kushlinskii NE. Soluble forms of PD-1/PD-L immune checkpoint receptor and ligand in blood serum of breast cancer patients: association with clinical pathologic factors and molecular type of the tumor. Klin Lab Diagn 2022; 67:76-80. [PMID: 35192751 DOI: 10.51620/0869-2084-2022-67-2-76-80] [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: 06/14/2023]
Abstract
Results of enzyme-linked immunosorbent assay of the soluble forms of PD-1/PD-L immune checkpoint receptor and ligand (sPD-1 and sPD-L1) in pretreatment blood serum of 88 breast cancer patients at various disease stages aged 30-83 years are presented. The control group included 55 practically healthy women aged 19-82 years. Serum sPD-1 and sPD-L1 levels in breast cancer patients highly significantly (p<0.0001) differ from control and these changes are opposite: soluble receptor level is more than 6-fold decreased, while soluble ligand concentration - 5.5 fold increased. Both markers separately, as well as their ratio demonstrate very high sensitivity (94-100%) and specificity (95-100%) in relation to healthy control. No statistically significant associations of sPD-1 and sPD-L1 levels with clinical stage, individual TNM system criteria, tumor histological structure, grade, receptor status, and molecular type were established. In particular, no significant peculiarities of the markers' levels in triple negative breast cancer successfully treated with anti-PD-1/PD-L1 preparations were revealed. Long-term follow-up and dynamic studies of sPD-1 and sPD-L1serum levels in the course of treatment are required for evaluation of their independent from clinical and morphological factors prognostic significance and the possibility of application as low invasive tests for prediction and monitoring of corresponding targeted therapy efficiency.
Collapse
Affiliation(s)
| | - E A Korotkova
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| | - I K Vorotnikov
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| | - N Yu Sokolov
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| | - V D Ermilova
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| | - A S Mochalova
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| | - N E Kushlinskii
- N.N. Blokhin National Medical Research Center of Oncology Ministry of Health of the Russian Federation
| |
Collapse
|
21
|
Zhang J, Lin Y, Lin Z, Wei Q, Qian J, Ruan R, Jiang X, Hou L, Song J, Ding J, Yang H. Stimuli-Responsive Nanoparticles for Controlled Drug Delivery in Synergistic Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103444. [PMID: 34927373 PMCID: PMC8844476 DOI: 10.1002/advs.202103444] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/28/2021] [Indexed: 05/10/2023]
Abstract
Cancer immunotherapy has achieved promising clinical progress over the recent years for its potential to treat metastatic tumors and inhibit their recurrences effectively. However, low patient response rates and dose-limiting toxicity remain as major dilemmas for immunotherapy. Stimuli-responsive nanoparticles (srNPs) combined with immunotherapy offer the possibility to amplify anti-tumor immune responses, where the weak acidity, high concentration of glutathione, overexpressions of enzymes, and reactive oxygen species, and external stimuli in tumors act as triggers for controlled drug release. This review highlights the design of srNPs based on tumor microenvironment and/or external stimuli to combine with different anti-tumor drugs, especially the immunoregulatory agents, which eventually realize synergistic immunotherapy of malignant primary or metastatic tumors and acquire a long-term immune memory to prevent tumor recurrence. The authors hope that this review can provide theoretical guidance for the construction and clinical transformation of smart srNPs for controlled drug delivery in synergistic cancer immunotherapy.
Collapse
Affiliation(s)
- Jin Zhang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Yandai Lin
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Zhe Lin
- Ruisi (Fujian) Biomedical Engineering Research Center Co LtdFuzhou350100P. R. China
| | - Qi Wei
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Jiaqi Qian
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Renjie Ruan
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Xiancai Jiang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Linxi Hou
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| |
Collapse
|
22
|
Synergistic antitumor efficacy of PD-1-conjugated PTX- and ZSQ-loaded nanoliposomes against multidrug-resistant liver cancers. Drug Deliv Transl Res 2022; 12:2550-2560. [PMID: 35031972 DOI: 10.1007/s13346-021-01106-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide with poor chemotherapeutic efficiency due to multidrug resistance (MDR); it is very important to develop a targeted nanocarrier for the treatment of HCC. In this study, a programmed death ligand 1 (PD-L1)-conjugated nanoliposome was constructed for co-delivery of paclitaxel (PTX) and P-glycoprotein (P-gp) inhibitor zosuquidar (ZSQ) to overcome MDR in human HCC cells and tumors in vivo. Transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) were used to examine the nanoparticles morphology and size; PD-1-conjugated PTX and ZSQ-loaded nanoliposomes (PD-PZLP) revealed a spherical shape with a size of 139.5 ± 10.7 nm. Then, the physicochemical properties, as well as the drug loading capacity, release profile, cellular uptake, and cytotoxicity of the dual drug-encapsulated nanoliposomes were characterized. PD-PZLP displayed a high drug loading capacity of 20 ~ 30% for both PTX and ZSQ; the drug release of PTX and ZSQ in pH 5.0 was significantly faster than in pH 7.4. Cellular uptake study demonstrated PD-PZLP had higher internalization efficiency than non-targeted PZLP. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and reactive oxygen species (ROS) analysis demonstrated that PD-PZLP triggered an excessive ROS reaction and cell apoptosis compared with that of free PTX or ZSQ, which was also consistent with the cell antiproliferative effects in MTT assay. Furthermore, PD-PZLP could enhance synergistic antitumor effects on 7721/ADM xenograft tumor model, which also significantly alleviated hepatotoxicity as evident from the decreased aspartate transaminase (AST) and alanine transaminase (ALT) levels. Overall, PD-PZLP exhibited high loading capacity, significant synergistic effects, promising antitumor efficacy, and the lowest toxicity, which provide a promising strategy to overcome MDR in HCC.
Collapse
|
23
|
Ayoub NM, Fares M, Marji R, Al Bashir SM, Yaghan RJ. Programmed Death-Ligand 1 Expression in Breast Cancer Patients: Clinicopathological Associations from a Single-Institution Study. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:603-615. [PMID: 34803400 PMCID: PMC8597920 DOI: 10.2147/bctt.s333123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/28/2021] [Indexed: 11/23/2022]
Abstract
PURPOSE Tumor expression of programmed death-ligand 1 (PD-L1) is associated with evasion of immune response in several types of malignancies and such expression may render patients eligible for PD-L1 inhibitors. The use of immune checkpoint blockade therapy has been recently approved for the treatment of breast cancer. However, PD-L1 expression data are lacking among Jordanian breast cancer patients. In this study, the tumor PD-L1 expression was characterized in breast cancer patients to assess their eligibility for immune checkpoint blockade therapy. The study also aimed to explore the association between tumoral PD-L1 expression and the clinicopathologic characteristics and the prognostic factors in patients with breast cancer. PATIENTS AND METHODS Tissue samples were available from 153 female patients with primary invasive breast cancer. Immunohistochemistry was performed on paraffin-embedded tumor sections that were stained with a PD-L1 antibody. Expression of tumor PD-L1 was correlated with demographics, clinicopathologic characteristics, and prognosis. RESULTS The mean age at diagnosis was 54.2±12.8 years (median 52, interquartile range 45-65). The percentage of PD-L1-positive tumors was 26.1%. PD-L1 expression on tumor cells significantly and positively correlated with tumor size (rho=0.174, p=0.032). PD-L1 positivity was significantly associated with the grade of carcinoma (p=0.001), HER2-positivity (p=0.015), and lymphovascular invasion (p=0.036). PD-L1 intensity was significantly associated with tumor stage (p=0.046). No significant associations were observed for the PD-L1 expression status or intensity with patient menopausal status, hormone receptor expression, and molecular subtypes. PD-L1 expression significantly correlated with a worse prognosis of breast cancer patients at the time of diagnosis (rho=0.230, p=0.005). CONCLUSION Tumor PD-L1 expression was associated with advanced clinicopathologic features and worse prognosis in this cohort of Jordanian breast cancer patients. Future studies are needed to better understand the impact of PD-L1 blockade therapy on treatment outcomes in eligible breast cancer patients in Jordan.
Collapse
Affiliation(s)
- Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, 22110, Jordan
| | - Mona Fares
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, 22110, Jordan
| | - Raya Marji
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Samir M Al Bashir
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Rami J Yaghan
- Department of Surgery, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
24
|
Xu P, Xiong W, Lin Y, Fan L, Pan H, Li Y. Histone deacetylase 2 knockout suppresses immune escape of triple-negative breast cancer cells via downregulating PD-L1 expression. Cell Death Dis 2021; 12:779. [PMID: 34365463 PMCID: PMC8349356 DOI: 10.1038/s41419-021-04047-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023]
Abstract
The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacetylase 2 (HDAC2) is required for IFN signaling. Here, we investigated the regulation of HDAC2 on the IFNγ-induced PD-L1 expression in TNBC cells. We found the HDAC2 and PD-L1 expression in TNBC was significantly higher than that in non-TNBC, and HDAC2 was positively correlated with PD-L1 expression. HDAC2 promoted PD-L1 induction by upregulating the phosphorylation of JAK1, JAK2, and STAT1, as well as the translocation of STAT1 to the nucleus and the recruitment of STAT1 to the PD-L1 promoter. Meanwhile, HDAC2 was recruited to the PD-L1 promoter by STAT1, and HDAC2 knockout compromised IFNγ-induced upregulation of H3K27, H3K9 acetylation, and the BRD4 recruitment in PD-L1 promoter. In addition, significant inhibition of proliferation, colony formation, migration, and cell cycle of TNBC cells were observed following knockout of HDAC2 in vitro. Furthermore, HDAC2 knockout reduced IFNγ-induced PD-L1 expression, lymphocyte infiltration, and retarded tumor growth and metastasis in the breast cancer mouse models. This study may provide evidence that HDAC2 promotes IFNγ-induced PD-L1 expression, suggesting a way for enhanced antitumor immunity when targeting the HDAC2 in TNBC.
Collapse
Affiliation(s)
- Pengfei Xu
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, 7 Raoping Road, Shantou, 515041, China
| | - Wei Xiong
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China
| | - Yun Lin
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China
| | - Liping Fan
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China
| | - Hongchao Pan
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, 7 Raoping Road, Shantou, 515041, China
| | - Yaochen Li
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, 515041, China.
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, 7 Raoping Road, Shantou, 515041, China.
| |
Collapse
|