1
|
Zhou R, Yin L, Zhang X, Zhu K. SFRP1 reduces neutrophil infiltration and inhibits the Wnt/β-catenin pathway to alleviate oral submucous fibrosis. In Vitro Cell Dev Biol Anim 2024; 60:1034-1045. [PMID: 39017751 DOI: 10.1007/s11626-024-00945-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/24/2024] [Indexed: 07/18/2024]
Abstract
Oral submucous fibrosis (OSF) is a precancerous condition characterized by oral mucosal atrophy with fibrosis of the submucosal tissue. OSF has a high prevalence, and treatment requires improvement. Our study aims to investigate the role and mechanism of secreted frizzled-related protein 1 (SFRP1) in OSF. We constructed an arecoline-induced OSF mice model. Through Pearson's correlation analysis, we investigated the association between SFRP1 levels and expressions of proteins related to the Wnt/β-catenin signaling pathway, as well as the correlation between SFRP1 levels and the degree of neutrophil infiltration. Moreover, neutrophil infiltration intensity, tissue fibrosis degree, and levels of β-catenin, Cyclin D1, and c-myc were evaluated after SFRP1 overexpression treatment through immunohistochemical and biochemical assays. A Wnt/β-catenin pathway activator was used to investigate the molecular mechanism of SFRP1 in the arecoline-induced OSF cell model. Compared with the control group, mice in the OSF group exhibited increased collagen deposition and more severe fibrosis in the oral mucosal tissue (OMT). In the OMT of OSF mice, the levels of SFRP1 were decreased. The levels of SFRP1 exhibited a negative correlation with the levels of Wnt/β-catenin proteins and neutrophil infiltration in the OMT. Upon SFRP1 overexpression, there was a reduction in neutrophil infiltration and fibrosis in the OMT, as well as inhibition of Wnt/β-catenin-related proteins. In vitro, the Wnt/β-catenin pathway activator further reversed the effect of SFRP1 overexpression on OSF. SFRP1 attenuates OSF by reducing neutrophil infiltration and inhibiting the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Stomatology, Changsha Hospital for Maternal and Child Health Care, Changsha, China
| | - Lin Yin
- Department of Stomatology, Changsha Hospital for Maternal and Child Health Care, Changsha, China
| | - Xin Zhang
- Department of Stomatology, Changsha Hospital for Maternal and Child Health Care, Changsha, China
| | - Keke Zhu
- Department of Stomatology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
2
|
Patra A, Ghosh SS, Saini GK. Exploring potential molecular targets and therapeutic efficacy of beauvericin in triple-negative breast cancer cells. Comput Biol Chem 2024; 112:108154. [PMID: 39029290 DOI: 10.1016/j.compbiolchem.2024.108154] [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: 04/01/2024] [Revised: 06/02/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Triple negative breast cancer (TNBC) presents a significant global health concern due to its aggressive nature, high mortality rate and limited treatment options, highlighting the urgent need for targeted therapies. Beauvericin, a bioactive fungal secondary metabolite, possess significant anticancer potential, although its molecular targets in cancer cells remain unexplored. This study has investigated possible molecular targets of beauvericin and its therapeutic insights in TNBC cells. In silico studies using molecular docking and MD simulation predicted the molecular targets of beauvericin. The identified targets included MRP-1 (ABCC1), HDAC-1, HDAC-2, LCK and SYK with average binding energy of -90.1, -44.3, -72.1, -105 and -60.8 KJ/mol, respectively, implying its multifaceted roles in reversing drug resistance, inhibiting epigenetic modulators and oncogenic tyrosine kinases. Beauvericin has significantly reduced the viability of MDA-MB-231 and MDA-MB-468 cells, with IC50 concentrations of 4.4 and 3.9 µM, while concurrently elevating the intracellular ROS by 9.0 and 7.9 folds, respectively. Subsequent reduction of mitochondrial transmembrane potential in TNBC cells, has confirmed the induction of oxidative stress, leading to apoptotic cell death, as observed by flow cytometric analyses. Beauvericin has also arrested cell cycle at G1-phase and impaired the spheroid formation and clonal expansion abilities of TNBC cells. The viability of spheroids was reduced upon beauvericin treatment, exhibiting IC50 concentrations of 10.3 and 6.2 µM in MDA-MB-468 and MDA-MB-231 cells, respectively. In conclusion, beauvericin has demonstrated promising therapeutic potential against TNBC cells through possible inhibition of MRP-1 (ABCC1), HDAC-1, HDAC-2, LCK and SYK.
Collapse
Affiliation(s)
- Arupam Patra
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, North Guwahati, Assam 781039, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, North Guwahati, Assam 781039, India.
| | - Gurvinder Kaur Saini
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, North Guwahati, Assam 781039, India.
| |
Collapse
|
3
|
Lapenta C, Santini SM, Antonacci C, Donati S, Cecchetti S, Frittelli P, Catalano P, Urbani F, Macchia I, Spada M, Vitale S, Michelini Z, Corsi DC, Zeuner A, Dattilo R, Tamburo De Bella M. Anti-Tumor Immunity to Patient-Derived Breast Cancer Cells by Vaccination with Interferon-Alpha-Conditioned Dendritic Cells (IFN-DC). Vaccines (Basel) 2024; 12:1058. [PMID: 39340087 PMCID: PMC11435915 DOI: 10.3390/vaccines12091058] [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: 08/05/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Breast cancer represents one of the leading causes of death among women. Surgery can be effective, but once breast cancer has metastasized, it becomes extremely difficult to treat. Conventional therapies are associated with substantial toxicity and poor efficacy due to tumor heterogeneity, treatment resistance and disease relapse. Moreover, immune checkpoint blockade appears to offer limited benefit in breast cancer. The poor tumor immunogenicity and the immunosuppressive tumor microenvironment result in scarce T-cell infiltration, leading to a low response rate. Thus, there is considerable interest in the development of improved active immunotherapies capable of sensitizing a patient's immune system against tumor cells. METHODS We evaluated the in vitro anti-tumor activity of a personalized vaccine based on dendritic cells generated in the presence of interferon (IFN)-α and granulocyte-macrophage colony-stimulating factor (IFN-DC) and loaded with an oxidized lysate from autologous tumor cells expanded as 3D organoid culture maintaining faithful tumor antigenic profiles. RESULTS Our findings demonstrate that stimulation of breast cancer patients' lymphocytes with autologous IFN-DC led to efficient Th1-biased response and the generation in vitro of potent cytotoxic activity toward the patients' own tumor cells. CONCLUSIONS This approach can be potentially applied in association with checkpoint blockade and chemotherapy in the design of new combinatorial therapies for breast cancer.
Collapse
Affiliation(s)
- Caterina Lapenta
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Stefano Maria Santini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Celeste Antonacci
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Serena Cecchetti
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Patrizia Frittelli
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Piera Catalano
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Sara Vitale
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Zuleika Michelini
- National Center for Global Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Domenico Cristiano Corsi
- Medical Oncology Unit, Fatebenefratelli-Isola Tiberina Hospital, 00186 Rome, Italy; (P.F.); (P.C.); (D.C.C.)
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Rosanna Dattilo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (S.D.); (F.U.); (I.M.); (S.V.); (A.Z.); (R.D.)
| | - Manuela Tamburo De Bella
- Hospital Clinical Networks Governance e DM70/15 Monitoring—AGENAS—National Agency for Regional Health Services, 00187 Rome, Italy;
| |
Collapse
|
4
|
Alqathama A. Natural products as promising modulators of breast cancer immunotherapy. Front Immunol 2024; 15:1410300. [PMID: 39050852 PMCID: PMC11266008 DOI: 10.3389/fimmu.2024.1410300] [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: 03/31/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024] Open
Abstract
Breast cancer (BC) is the most common malignancy among women and is considered a major global health challenge worldwide due to its high incidence and mortality rates. Treatment strategies for BC is wide-ranging and include surgery, radiotherapy, chemotherapy, targeted hormonal therapy and immunotherapy. Immunotherapy has gained popularity recently and is often integrated as a component of personalized cancer care because it aims to strengthen the immune system and enable it to recognize and eradicate transformed cells. It has fewer side-effects and lower toxicity than other treatment strategies, such as chemotherapy. Many natural products are being investigated for a wide range of therapeutic pharmacological properties, such as immune system modulation and activity against infection, auto-immune disease, and cancer. This review presents an overview of the major immune response-related pathways in BC, followed by detailed explanation of how natural compounds can act as immunomodulatory agents against biomolecular targets. Research has been carried out on many forms of natural products, including extracts, isolated entities, synthetic derivatives, nanoparticles, and combinations of natural compounds. Findings have shown significant regulatory effects on immune cells and immune cytokines that lead to immunogenic cancer cell death, as well as upregulation of macrophages and CD+8 T cells, and increased natural killer cell and dendritic cell activity. Natural products have also been found to inhibit some immuno-suppressive cells such as Treg and myeloid-derived suppressor cells, and to decrease immunosuppressive factors such as TGF-β and IL-10. Also, some natural compounds have been found to target and hinder immune checkpoints such as PD-L1.
Collapse
Affiliation(s)
- Aljawharah Alqathama
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
5
|
Zhao Z, Ma X, Cai Z. The potential role of CD8+ cytotoxic T lymphocytes and one branch connected with tissue-resident memory in non-luminal breast cancer. PeerJ 2024; 12:e17667. [PMID: 39006029 PMCID: PMC11246025 DOI: 10.7717/peerj.17667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Advances in understanding the pathological mechanisms of breast cancer have resulted in the emergence of novel therapeutic strategies. However, triple-negative breast cancer (TNBC), a molecular subtype of breast cancer with a poor prognosis, lacks classical and general therapeutic targets, hindering the clinical application of several therapies to breast cancer. As insights into the unique immunity and molecular mechanisms of TNBC have become more extensive, immunotherapy has gradually become a valuable complementary approach to classical radiotherapy and chemotherapy. CD8+ cells are significant actors in the tumor immunity cycle; thus, research on TNBC immunotherapy is increasingly focused in this direction. Recently, CD8+ tissue-resident memory (TRM) cells, a subpopulation of CD8+ cells, have been explored in relation to breast cancer and found to seemingly play an undeniably important role in tumor surveillance and lymphocytic infiltration. In this review, we summarize the recent advances in the mechanisms and relative targets of CD8+ T cells, and discuss the features and potential applications of CD8+ TRM cells in non-luminal breast cancer immunotherapy.
Collapse
Affiliation(s)
- Ziqi Zhao
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Xinyu Ma
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhengang Cai
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
6
|
Sivaganesh V, Ta TM, Peethambaran B. Pentagalloyl Glucose (PGG) Exhibits Anti-Cancer Activity against Aggressive Prostate Cancer by Modulating the ROR1 Mediated AKT-GSK3β Pathway. Int J Mol Sci 2024; 25:7003. [PMID: 39000112 PMCID: PMC11241829 DOI: 10.3390/ijms25137003] [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: 05/09/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Androgen-receptor-negative, androgen-independent (ARneg-AI) prostate cancer aggressively proliferates and metastasizes, which makes treatment difficult. Hence, it is necessary to continue exploring cancer-associated markers, such as oncofetal Receptor Tyrosine Kinase like Orphan Receptor 1 (ROR1), which may serve as a form of targeted prostate cancer therapy. In this study, we identify that Penta-O-galloyl-β-D-glucose (PGG), a plant-derived gallotannin small molecule inhibitor, modulates ROR1-mediated oncogenic signaling and mitigates prostate cancer phenotypes. Results indicate that ROR1 protein levels were elevated in the highly aggressive ARneg-AI PC3 cancer cell line. PGG was selectively cytotoxic to PC3 cells and induced apoptosis of PC3 (IC50 of 31.64 µM) in comparison to normal prostate epithelial RWPE-1 cells (IC50 of 74.55 µM). PGG was found to suppress ROR1 and downstream oncogenic pathways in PC3 cells. These molecular phenomena were corroborated by reduced migration, invasion, and cell cycle progression of PC3 cells. PGG minimally and moderately affected RWPE-1 and ARneg-AI DU145, respectively, which may be due to these cells having lower levels of ROR1 expression in comparison to PC3 cells. Additionally, PGG acted synergistically with the standard chemotherapeutic agent docetaxel to lower the IC50 of both compounds about five-fold (combination index = 0.402) in PC3 cells. These results suggest that ROR1 is a key oncogenic driver and a promising target in aggressive prostate cancers that lack a targetable androgen receptor. Furthermore, PGG may be a selective and potent anti-cancer agent capable of treating ROR1-expressing prostate cancers.
Collapse
Affiliation(s)
- Vignesh Sivaganesh
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Ave, Philadelphia, PA 19131, USA
| | - Tram M. Ta
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
| | - Bela Peethambaran
- Department of Biology, Saint Joseph’s University, 600 S 43rd St, Philadelphia, PA 19104, USA; (V.S.); (T.M.T.)
| |
Collapse
|
7
|
Zhang X, Li R, Wang G. PDL1-Based Nomogram May Be of Potential Clinical Utility for Predicting Survival Outcome in Stage III Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:731-746. [PMID: 37905205 PMCID: PMC10613449 DOI: 10.2147/bctt.s435980] [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: 08/18/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Purpose Programmed cell death ligand 1 (PDL1) has the predictive and prognostic value in a great deal of cancers. This study aims to explore the expression of PDL1 in stage III breast cancer (BC) and its correlation with clinical outcome. Methods The protein expression of PDL1 in tumor tissues was determined by immunohistochemistry (IHC). The correlations between PDL1 and clinicopathological variables were performed by χ²-tests or Fisher's exact tests. The Cox proportional hazards model was used for univariate and multivariate analysis of the potential prognostic factors. Survival curves were estimated based on Kaplan-Meier analyses, and Log Rank test was used to contrast factors influencing the survival outcome. Results On the basis of the semiquantitative scoring method for PDL1 expression, the patients were divided into low PDL1 expression group (109 cases) and high PDL1 expression group (107 cases). PDL1 expression was correlated with positive lymph nodes, positive axillary lymph nodes, postoperative radiotherapy, and CK5/6 expression (P < 0.05). The PDL1 expression in tumor tissues was discovered to be a potential prognostic risk factor with the disease-free survival (DFS) and overall survival (OS) for stage III BC. Moreover, patients with high PDL1 expression showed longer lifetime (DFS and OS) compared to those with low PDL1 expression in total patient population (P < 0.05). Moreover, the nomogram showed that the prediction line is in good agreement with the reference line for postoperative 1-, 3-, and 5-year lifetime. The DCA curve showed that the 3- and 5-year lifetime by nomogram had so much better divination of the clinical application than only by PDL1. Conclusion PDL1 is a latent prognostic factor in stage III BC and is closely related to some clinicopathological features. PDL1 expression in tumor tissues is significantly associated with better lifetime rate in stage III BC.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Ruzhe Li
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| | - Guonian Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
8
|
The New Frontier of Immunotherapy: Chimeric Antigen Receptor T (CAR-T) Cell and Macrophage (CAR-M) Therapy against Breast Cancer. Cancers (Basel) 2023; 15:cancers15051597. [PMID: 36900394 PMCID: PMC10000829 DOI: 10.3390/cancers15051597] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Breast cancer represents one of the most common tumor histologies. To date, based on the specific histotype, different therapeutic strategies, including immunotherapies, capable of prolonging survival are used. More recently, the astonishing results that were obtained from CAR-T cell therapy in haematological neoplasms led to the application of this new therapeutic strategy in solid tumors as well. Our article will deal with chimeric antigen receptor-based immunotherapy (CAR-T cell and CAR-M therapy) in breast cancer.
Collapse
|
9
|
Lu B, Natarajan E, Balaji Raghavendran HR, Markandan UD. Molecular Classification, Treatment, and Genetic Biomarkers in Triple-Negative Breast Cancer: A Review. Technol Cancer Res Treat 2023; 22:15330338221145246. [PMID: 36601658 PMCID: PMC9829998 DOI: 10.1177/15330338221145246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Breast cancer is the most common malignancy and the second most common cause of cancer-related mortality in women. Triple-negative breast cancers do not express estrogen receptors, progesterone receptors, or human epidermal growth factor receptor 2 and have a higher recurrence rate, greater metastatic potential, and lower overall survival rate than those of other breast cancers. Treatment of triple-negative breast cancer is challenging; molecular-targeted therapies are largely ineffective and there is no standard treatment. In this review, we evaluate current attempts to classify triple-negative breast cancers based on their molecular features. We also describe promising treatment methods with different advantages and discuss genetic biomarkers and other prediction tools. Accurate molecular classification of triple-negative breast cancers is critical for patient risk categorization, treatment decisions, and surveillance. This review offers new ideas for more effective treatment of triple-negative breast cancer and identifies novel targets for drug development.
Collapse
Affiliation(s)
- Boya Lu
- Department of Mechanical Engineering, Faculty of Engineering,
Technology and Built Environment, UCSI University,
Kuala Lumpur, Malaysia,Boya Lu, MD, Department of Mechanical
Engineering, Faculty of Engineering, Technology and Built Environment, UCSI
University, No 1, Jalan Menara Gading, UCSI Heights (Taman Connaught), Cheras,
56000, Kuala Lumpur, Malaysia.
| | - Elango Natarajan
- Department of Mechanical Engineering, Faculty of Engineering,
Technology and Built Environment, UCSI University,
Kuala Lumpur, Malaysia
| | - Hanumantha Rao Balaji Raghavendran
- Faculty of Clinical Research, Central Research Facility, Sri
Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu,
India
| | | |
Collapse
|
10
|
The Structure, Function and Regulation of Protein Tyrosine Phosphatase Receptor Type J and Its Role in Diseases. Cells 2022; 12:cells12010008. [PMID: 36611803 PMCID: PMC9818648 DOI: 10.3390/cells12010008] [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: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Protein tyrosine phosphatase receptor type J (PTPRJ), also known as DEP-1, HPTPη, or CD148, belongs to the R3 subfamily of receptor protein tyrosine phosphatases (RPTPs). It was first identified as an antioncogene due to its protein level being significantly downregulated in most epithelial tumors and cancer cell lines (e.g., colon, lung, thyroid, breast, and pancreas). PTPRJ regulates mouse optic nerve projection by inhibiting the phosphorylation of the erythropoietin-producing hepatocellular carcinoma (Eph) receptor and abelson murine leukemia viral oncogene homolog 1 (c-Abl). PTPRJ is crucial for metabolism. Recent studies have demonstrated that PTPRJ dephosphorylates JAK2 at positions Y813 and Y868 to inhibit leptin signaling. Akt is more phosphorylated at the Ser473 and Thr308 sites in Ptprj-/- mice, suggesting that PTPRJ may be a novel negative regulator of insulin signaling. PTPRJ also plays an important role in balancing the pro- and anti-osteoclastogenic activity of the M-CSF receptor (M-CSFR), and in maintaining NFATc1 expression during the late stages of osteoclastogenesis to promote bone-resorbing osteoclast (OCL) maturation. Furthermore, multiple receptor tyrosine kinases (RTKs) as substrates of PTPRJ are probably a potential therapeutic target for many types of diseases, such as cancer, neurodegenerative diseases, and metabolic diseases, by inhibiting their phosphorylation activity. In light of the important roles that PTPRJ plays in many diseases, this review summarizes the structural features of the protein, its expression pattern, and the physiological and pathological functions of PTPRJ, to provide new ideas for treating PTPRJ as a potential therapeutic target for related metabolic diseases and cancer.
Collapse
|
11
|
Nicolini A, Ferrari P, Carpi A. Immune Checkpoint Inhibitors and Other Immune Therapies in Breast Cancer: A New Paradigm for Prolonged Adjuvant Immunotherapy. Biomedicines 2022; 10:biomedicines10102511. [PMID: 36289773 PMCID: PMC9599105 DOI: 10.3390/biomedicines10102511] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/25/2022] [Accepted: 10/02/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Breast cancer is the most common form of cancer in women worldwide. Advances in the early diagnosis and treatment of cancer in the last decade have progressively decreased the cancer mortality rate, and in recent years, immunotherapy has emerged as a relevant tool against cancer. HER2+ and triple-negative breast cancers (TNBCs) are considered more immunogenic and suitable for this kind of treatment due to the higher rate of tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) expression. In TNBC, genetic aberrations further favor immunogenicity due to more neo-antigens in cancer cells. Methods: This review summarizes the principal ongoing conventional and investigational immunotherapies in breast cancer. Particularly, immune checkpoint inhibitors (ICIs) and their use alone or combined with DNA damage repair inhibitors (DDRis) are described. Then, the issue on immunotherapy with monoclonal antibodies against HER-2 family receptors is updated. Other investigational immunotherapies include a new schedule based on the interferon beta-interleukin-2 sequence that was given in ER+ metastatic breast cancer patients concomitant with anti-estrogen therapy, which surprisingly showed promising results. Results: Based on the scientific literature and our own findings, the current evaluation of tumor immunogenicity and the conventional model of adjuvant chemotherapy (CT) are questioned. Conclusions: A novel strategy based on additional prolonged adjuvant immunotherapy combined with hormone therapy or alternated with CT is proposed.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy
- Correspondence:
| | - Paola Ferrari
- Unit of Oncology, Department of Medical and Oncological Area, Azienda Ospedaliera-Universitaria Pisana, 56125 Pisa, Italy
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
12
|
Yang Y, Liu HL, Liu YJ. A Novel Five-Gene Signature Related to Clinical Outcome and Immune Microenvironment in Breast Cancer. Front Genet 2022; 13:912125. [PMID: 35646102 PMCID: PMC9136328 DOI: 10.3389/fgene.2022.912125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 11/30/2022] Open
Abstract
Breast cancer (BC) is the most frequent cancer in women and the main cause of cancer-related deaths in the globe, according to the World Health Organization. The need for biomarkers that can help predict survival or guide treatment decisions in BC patients is critical in order to provide each patient with an individualized treatment plan due to the wide range of prognoses and therapeutic responses. A reliable prognostic model is essential for determining the best course of treatment for patients. Patients’ clinical and pathological data, as well as their mRNA expression levels at level 3, were gleaned from the TCGA databases. Differentially expressed genes (DEGs) between BC and non-tumor specimens were identified. Tumor immunity analyses have been utilized in order to decipher molecular pathways and their relationship to the immune system. The expressions of KIF4A in BC cells were determined by RT-PCR. To evaluate the involvement of KIF4A in BC cell proliferation, CCK-8 tests were used. In this study, utilizing FC > 4 and p < 0.05, we identified 140 upregulated genes and 513 down-regulated genes. A five-gene signature comprising SFRP1, SAA1, RBP4, KIF4A and COL11A1 was developed for the prediction of overall survivals of BC. Overall survival was distinctly worse for patients in the high-risk group than those in the low-risk group. Cancerous and aggressiveness-related pathways and decreased B cell, T cell CD4+, T cell CD8+, Neutrophil and Myeloid dendritic cells levels were seen in the high-risk group. In addition, we found that KIF4A was highly expressed in BC and its silence resulted in the suppression of the proliferation of BC cells. Taken together, as a possible prognostic factor for BC, the five-gene profile created and verified in this investigation could guide the immunotherapy selection.
Collapse
|
13
|
Sawasdee N, Wattanapanitch M, Thongsin N, Phanthaphol N, Chiawpanit C, Thuwajit C, Yenchitsomanus PT, Panya A. Doxorubicin sensitizes breast cancer cells to natural killer cells in connection with increased Fas receptors. Int J Mol Med 2022; 49:40. [PMID: 35119077 PMCID: PMC8815410 DOI: 10.3892/ijmm.2022.5095] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/12/2022] [Indexed: 11/15/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women. Although standard treatments are successful in patients with BC diagnosed at an early stage, an alternative treatment is required for patients with advanced-stage disease who do not respond to these treatments. The concept of using chemotherapy to sensitize cancer cells to become susceptible to immunotherapy was recently introduced and may be used as an alternative treatment for BC. The chemotherapeutic drug doxorubicin has been reported to sensitize cancer cells; however, the efficacy to sensitize the solid spheroids, in addition to its underlying mechanism regarding how doxorubicin sensitizes BC, has not previously been explored. In the present study, the effectiveness of a combined treatment of doxorubicin and natural killer-92 (NK-92) cells against BC in either 2D or 3D spheroid models, and its association with Fas receptor (FasR) expression, was demonstrated. The BC (MCF7) cell line expressing a higher level of FasR was more sensitive to NK-92 cell killing than the MDA-MB-231 cell line, which expressed a lower level of FasR. A sublethal dose of doxorubicin caused a significant improvement in NK cytotoxicity. Concordantly, a significant reduction in cell viability was observed in the doxorubicin-treated MCF7 spheroids. Notably, flow cytometric analysis revealed significantly increased FasR expression in the MCF7 cells, suggesting the underlying sensitization mechanism of doxorubicin in BC was related to the FasR upregulation. The present findings supported the use of combined doxorubicin and NK immunotherapy in BC treatment.
Collapse
Affiliation(s)
- Nunghathai Sawasdee
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Methichit Wattanapanitch
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nontaphat Thongsin
- Siriraj Center for Regenerative Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nattaporn Phanthaphol
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chutipa Chiawpanit
- Department of Biology, Industry and Medicine, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Aussara Panya
- Department of Biology, Industry and Medicine, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
14
|
Protein Tyrosine Phosphatases: Mechanisms in Cancer. Int J Mol Sci 2021; 22:ijms222312865. [PMID: 34884670 PMCID: PMC8657787 DOI: 10.3390/ijms222312865] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Protein tyrosine kinases, especially receptor tyrosine kinases, have dominated the cancer therapeutics sphere as proteins that can be inhibited to selectively target cancer. However, protein tyrosine phosphatases (PTPs) are also an emerging target. Though historically known as negative regulators of the oncogenic tyrosine kinases, PTPs are now known to be both tumor-suppressive and oncogenic. This review will highlight key protein tyrosine phosphatases that have been thoroughly investigated in various cancers. Furthermore, the different mechanisms underlying pro-cancerous and anti-cancerous PTPs will also be explored.
Collapse
|
15
|
Henriques B, Mendes F, Martins D. Immunotherapy in Breast Cancer: When, How, and What Challenges? Biomedicines 2021; 9:1687. [PMID: 34829916 PMCID: PMC8616011 DOI: 10.3390/biomedicines9111687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/27/2022] Open
Abstract
Breast Cancer (BC) is the second most frequent cause of cancer death among women worldwide and, although there have been significant advances in BC therapies, a significant percentage of patients develop metastasis and disease recurrence. Since BC was demonstrated to be an immunogenic tumor, immunotherapy has broken through as a significant therapy strategy against BC. Over the years, immunotherapy has improved the survival rate of HER2+ BC patients due to the approval of some monoclonal antibodies (mAbs) such as Trastuzumab, Pertuzumab and, recently, Margetuximab, along with the antibody-drug conjugates (ADC) Trastuzumab-Emtansine (T-DM1) and Trastuzumab Deruxtecan. Immune checkpoint inhibitors (ICI) showed promising efficacy in triple-negative breast cancer (TNBC) treatment, namely Atezolizumab and Pembrolizumab. Despite the success of immunotherapy, some patients do not respond to immunotherapy or those who respond to the treatment relapse or progress. The main causes of these adverse events are the complex, intrinsic or extrinsic resistance mechanisms. In this review, we address the different immunotherapy approaches approved for BC and some of the mechanisms responsible for resistance to immunotherapy.
Collapse
Affiliation(s)
- Beatriz Henriques
- Politécnico de Coimbra, ESTeSC, UCPCBL, 3046-854 Coimbra, Portugal; (B.H.); (F.M.)
| | - Fernando Mendes
- Politécnico de Coimbra, ESTeSC, UCPCBL, 3046-854 Coimbra, Portugal; (B.H.); (F.M.)
- Laboratório de Investigação em Ciências Aplicadas à Saúde (LabinSaúde), Politécnico de Coimbra, ESTESC, 3046-854 Coimbra, Portugal
- Biophysics Institute of Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
- European Association for Professions in Biomedical Sciences, B-1000 Brussels, Belgium
| | - Diana Martins
- Politécnico de Coimbra, ESTeSC, UCPCBL, 3046-854 Coimbra, Portugal; (B.H.); (F.M.)
- Laboratório de Investigação em Ciências Aplicadas à Saúde (LabinSaúde), Politécnico de Coimbra, ESTESC, 3046-854 Coimbra, Portugal
- Biophysics Institute of Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), University of Coimbra, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-504 Coimbra, Portugal
| |
Collapse
|