1
|
Kim G, Zhu R, Yu S, Fan B, Jeon H, Leon J, Webber MJ, Wang Y. Enhancing Gene Delivery to Breast Cancer with Highly Efficient siRNA Loading and pH-Responsive Small Extracellular Vesicles. ACS Biomater Sci Eng 2025; 11:213-227. [PMID: 39713992 DOI: 10.1021/acsbiomaterials.4c01595] [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: 12/24/2024]
Abstract
Small extracellular vesicles (sEVs) are promising nanocarriers for drug delivery to treat a wide range of diseases due to their natural origin and innate homing properties. However, suboptimal therapeutic effects, attributed to ineffective targeting, limited lysosomal escape, and insufficient delivery, remain challenges in effectively delivering therapeutic cargo. Despite advances in sEV-based drug delivery systems, conventional approaches need improvement to address low drug-loading efficiency and to develop surface functionalization techniques for precise targeting of cells of interest, all while preserving the membrane integrity of sEVs. We report an enhanced gene delivery system using multifunctional sEVs for highly efficient siRNA loading and delivery. The integration of chiral graphene quantum dots enhanced the loading capacity while preserving the structural integrity of the sEVs. Additionally, lysosomal escape is facilitated by functionalizing sEVs with pH-responsive peptides, fully harnessing the inherent homing effect of sEVs for targeted and precise delivery. These sEVs achieved a 1.74-fold increase in cytosolic cargo delivery compared to unmodified sEVs, resulting in substantial gene silencing of around 73%. Our approach has significant potential to advance sEV-based gene delivery in order to accelerate clinical progress.
Collapse
Affiliation(s)
- Gaeun Kim
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Runyao Zhu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Sihan Yu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Bowen Fan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Hyunsu Jeon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Jennifer Leon
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Matthew J Webber
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| | - Yichun Wang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana46556, United States
| |
Collapse
|
2
|
Khashei Varnamkhasti K, Moghanibashi M, Naeimi S. Implications of ZNF334 gene in lymph node metastasis of lung SCC: potential bypassing of cellular senescence. J Transl Med 2024; 22:372. [PMID: 38637790 PMCID: PMC11025273 DOI: 10.1186/s12967-024-05115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The primary goal of this work is to identify biomarkers associated with lung squamous cell carcinoma and assess their potential for early detection of lymph node metastasis. METHODS This study investigated gene expression in lymph node metastasis of lung squamous cell carcinoma using data from the Cancer Genome Atlas and R software. Protein-protein interaction networks, hub genes, and enriched pathways were analyzed. ZNF334 and TINAGL1, two less explored genes, were further examined through in vitro, ex vivo, and in vivo experiments to validate the findings from bioinformatics analyses. The role of ZNF334 and TINAGL1 in senescence induction was assessed after H2O2 and UV induced senescence phenotype determined using β-galactosidase activity and cell cycle status assay. RESULTS We identified a total of 611 up- and 339 down-regulated lung squamous cell carcinoma lymph node metastasis-associated genes (FDR < 0.05). Pathway enrichment analysis highlighted the central respiratory pathway within mitochondria for the subnet genes and the nuclear DNA-directed RNA polymerases for the hub genes. Significantly down regulation of ZNF334 gene was associated with malignancy lymph node progression and senescence induction has significantly altered ZNF334 expression (with consistency in bioinformatics, in vitro, ex vivo, and in vivo results). Deregulation of TINAGL1 expression with inconsistency in bioinformatics, in vitro (different types of lung squamous cancer cell lines), ex vivo, and in vivo results, was also associated with malignancy lymph node progression and altered in senescence phenotype. CONCLUSIONS ZNF334 is a highly generalizable gene to lymph node metastasis of lung squamous cell carcinoma and its expression alter certainly under senescence conditions.
Collapse
Affiliation(s)
| | - Mehdi Moghanibashi
- Department of Genetics, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran.
| | - Sirous Naeimi
- Department of Genetics, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
3
|
Romo A, Rodríguez TM, Yu G, Dewey RA. Chimeric TβRII-SE/Fc overexpression by a lentiviral vector exerts strong antitumoral activity on colorectal cancer-derived cell lines in vitro and on xenografts. Cancer Gene Ther 2024; 31:174-185. [PMID: 37993543 DOI: 10.1038/s41417-023-00694-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
The TGF signaling pathway is a key regulator of cancer progression. In this work, we report for the first time the antitumor activity of TβRII-SE/Fc, a novel peptibody whose targeting domain is comprised of the soluble endogenous isoform of the human TGF-β type II receptor (TβRII-SE). Overexpression of TβRIISE/Fc reduces in vitro cell proliferation and migration while inducing cell cycle arrest and apoptosis in human colorectal cancer-derived cell lines. Moreover, TβRII-SE/Fc overexpression reduces tumorigenicity in BALB/c nude athymic mice. Our results revealed that TRII-SE/Fc-expressing tumors were significantly reduced in size or were even incapable of developing. We also demonstrated that the novel peptibody has the ability to inhibit the canonical TGF-β and BMP signaling pathways while identifying SMAD-dependent and independent proteins involved in tumor progression that are modulated by TβRII-SE/Fc. These findings provide insights into the underlying mechanism responsible for the antitumor activity of TβRII-SE/Fc. Although more studies are required to demonstrate the effectiveness and safety of the novel peptibody as a new therapeutic for the treatment of cancer, our initial in vitro and in vivo results in human colorectal tumor-derived cell lines are highly encouraging. Our results may serve as the foundation for further research and development of a novel biopharmaceutical for oncology.
Collapse
Affiliation(s)
- Ana Romo
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina
- RADBIO S.A.S., Sunchales, Argentina
| | - Tania Melina Rodríguez
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina
| | - Guo Yu
- Bio X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ricardo Alfredo Dewey
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina.
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina.
- RADBIO S.A.S., Sunchales, Argentina.
- Centro de Medicina Traslacional (CEMET), Hospital de Alta Complejidad en Red "El Cruce" Nestor Carlos Kirchner, Florencio Varela, Argentina.
| |
Collapse
|
4
|
Dong S, Chen C, Di C, Wang S, Dong Q, Lin W, Liu D. The Association between NADPH Oxidase 2 (NOX2) and Drug Resistance in Cancer. Curr Cancer Drug Targets 2024; 24:1195-1212. [PMID: 38362697 DOI: 10.2174/0115680096277328240110062433] [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: 08/16/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 02/17/2024]
Abstract
NADPH oxidase, as a major source of intracellular reactive oxygen species (ROS), assumes an important role in the immune response and oxidative stress response of the body. NADPH oxidase 2 (NOX2) is the first and most representative member of the NADPH oxidase family, and its effects on the development of tumor cells are gaining more and more attention. Our previous study suggested that NCF4 polymorphism in p40phox, a key subunit of NOX2, affected the outcome of diffuse large B-cell lymphoma patients treated with rituximab. It hypothesized that NOX2-mediated ROS could enhance the cytotoxic effects of some anti-tumor drugs in favor of patients with tumors. Several reviews have summarized the role of NOX2 and its congeners-mediated ROS in anti-tumor therapy, but few studies focused on the relationship between the expression of NOX2 and anti-tumor drug resistance. In this article, we systematically introduced the NOX family, represented by NOX2, and a classification of the latest inhibitors and agonists of NOX2. It will help researchers to have a more rational and objective understanding of the dual role of NOX2 in tumor drug resistance and is expected to provide new ideas for oncology treatment and overcoming drug resistance in cancer.
Collapse
Affiliation(s)
- Shiqi Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Chao Chen
- Department of laboratory, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Chang Di
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Shufan Wang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Quan Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Wenxin Lin
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| | - Duo Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, 150086, China
| |
Collapse
|
5
|
Anderson R, Rapoport BL, Steel HC, Theron AJ. Pro-Tumorigenic and Thrombotic Activities of Platelets in Lung Cancer. Int J Mol Sci 2023; 24:11927. [PMID: 37569299 PMCID: PMC10418868 DOI: 10.3390/ijms241511927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Aside from their key protective roles in hemostasis and innate immunity, platelets are now recognized as having multifaceted, adverse roles in the pathogenesis, progression and outcome of many types of human malignancy. The most consistent and compelling evidence in this context has been derived from the notable association of elevated circulating platelet counts with the onset and prognosis of various human malignancies, particularly lung cancer, which represents the primary focus of the current review. Key topics include an overview of the association of lung cancer with the circulating platelet count, as well as the mechanisms of platelet-mediated, pro-tumorigenic immunosuppression, particularly the role of transforming growth factor beta 1. These issues are followed by a discussion regarding the pro-tumorigenic role of platelet-derived microparticles (PMPs), the most abundant type of microparticles (MPs) in human blood. In this context, the presence of increased levels of PMPs in the blood of lung cancer patients has been associated with tumor growth, invasion, angiogenesis and metastasis, which correlate with disease progression and decreased survival times. The final section of the review addresses, firstly, the role of cancer-related platelet activation and thrombosis in the pathogenesis of secondary cardiovascular disorders and the associated mortality, particularly in lung cancer, which is second only to disease progression; secondly, the review addresses the potential role of antiplatelet agents in the adjunctive therapy of cancer.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| | - Bernardo L. Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
- The Medical Oncology Centre of Rosebank, Johannesburg 2196, South Africa
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (B.L.R.); (H.C.S.); (A.J.T.)
| |
Collapse
|
6
|
Attaallah A, Elmrazeky AR, El-Beltagy AEFBM, Abdelaziz KK, Soliman MF. Modulatory role of Coriandrum sativum (coriander) extract against diabetic complications on the gonads of female rats and their offspring. Tissue Cell 2023; 83:102127. [PMID: 37331322 DOI: 10.1016/j.tice.2023.102127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/10/2023] [Accepted: 06/06/2023] [Indexed: 06/20/2023]
Abstract
It is well known that diabetes is associated with impairment of ovarian and testicular structure and function. Coriander (Coriandrum sativum L.) is identified as one of the oldest herbal plants valued for its nutritional and medicinal properties. This work is mainly designed to evaluate the possible modulatory role of dry coriander fruit extract against gonadal impairments associated with diabetes in female rats and their pups. Twenty-four pregnant rats were divided into four groups (n = 6): group I served as control, group II was administered daily with coriander fruit extract (250 mg/kg b.wt), group III was injected interaperitoneally with a single dose of streptozotocin (STZ) (80 mg/kg b.wt), and group IV was injected with single dose of STZ and post administered coriander extract. The experiment was conducted from the 4th day of gestation till the end of weaning. At the end of the experiment, the mothers' rats and their offspring were weighed, sacrificed, the ovaries from mothers as well as ovaries and testes from offspring were immediately excised, and processed for histological, immunohistochemical and evaluation of apoptosis and transforming growth factorβ (TGF-β). Also, blood samples were collected and analyzed to estimate the levels of sex hormones as well as antioxidants.In STZ induced diabetes in mother's rats and their offspring, the ovarian sections revealed severe histopathological signs included several atretic follicles, dilated and congested blood capillaries. Additionally, the testicular sections of offspring appeared with destructive seminiferous tubules. Immunohistochemically, the ovarian sections displayed weak to negative expression for calretinin marker however the testicular sections showed strong expression for Bax protein (apoptotic marker) and weak to negative expression for Ki67 protein (proliferative marker). Also, the mean % values of positively expressed cells for TGF-β and annexin-v markers (late and early apoptosis indicator) were significantly elevated in the ovarian and testicular tissues of STZ-induced group of mother's rats and their pups if compared with control. Further results revealed that the levels of insulin, FSH, LH, estrogen, SOD and CAT were significantly decreased if compared with control however the levels of MDA and NO were significantly elevated. Administration of coriander fruit extract to diabetic rats successfully alleviated most of the altered histological, immunohistochemical, biochemical, and apoptotic changes induced by diabetes. Coriandrum sativum fruit extract has a powerful ameliorative role against STZ-induced diabetic gonadal dysfunctions in female rats and their offspring.
Collapse
Affiliation(s)
- Amany Attaallah
- Zoology Department, Faculty of Science, Damanhur University, Egypt.
| | | | | | | | - Mona Fm Soliman
- Histology Department, Faculty of Medicine, Mansoura University, Egypt
| |
Collapse
|
7
|
Augustyniak A, Mc Mahon H. Dietary marine-derived ingredients for stimulating hair cell cycle. Biomed Pharmacother 2023; 163:114838. [PMID: 37156114 DOI: 10.1016/j.biopha.2023.114838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023] Open
Abstract
In normal condition human hair growth occurs through three phases, anagen (growth phase included about 85 % of hairs, last from 2 to 6 years), catagen (transitional phase lasting up to 2 weeks) and telogen (resting phase which last from 1 to 4 months). Natural dynamics of the hair growth process can be impaired by several factors, such as genetic predisposition, hormonal disorders, aging, poor nutrition or stress, which can lead to the slowdown in the growth of hair or even hair loss. The aim of the study was to assess the hair growth promotion effect of marine-derived ingredients, hair supplement Viviscal® and its raw components (marine protein complex AminoMarC®, shark and oyster extract). Cytotoxicity, production of alkaline phosphatase and glycosaminoglycans, as well as expression of genes involved in hair cycle-related pathways were investigated using dermal papilla cells, both immortalized and primary cell lines. Tested marine compounds showed no evidence of cytotoxicity under in vitro conditions. Viviscal® significantly increased proliferation of dermal papilla cells. Moreover, tested samples stimulated cells to produce alkaline phosphatase and glycosaminoglycans. Increased expression of hair cell cycle-related genes was also observed. The obtained results indicate that marine-derived ingredients stimulate hair growth through anagen activation.
Collapse
Affiliation(s)
- Aleksandra Augustyniak
- Circular Bioeconomy Research Group, Shannon Applied Biotechnology Centre, Munster Technological University, V92CX88 Tralee, Ireland.
| | - Helena Mc Mahon
- Circular Bioeconomy Research Group, Shannon Applied Biotechnology Centre, Munster Technological University, V92CX88 Tralee, Ireland
| |
Collapse
|
8
|
Qin J, Cao M, Hu X, Tan W, Ma B, Cao Y, Chen Z, Li Q, Hu G. Dual inhibitors of ASK1 and PDK1 kinases: Design, synthesis, molecular docking and mechanism studies of N-benzyl pyridine-2-one containing derivatives as anti-fibrotic agents. Eur J Med Chem 2023; 247:115057. [PMID: 36603508 DOI: 10.1016/j.ejmech.2022.115057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Utilizing fragment-based hybrid designing strategies, 24 N-benzyl pyridine-2-one containing derivatives were synthesized by successfully incorporating 6-(4H-1,2,4-triazol-3-yl) pyridin-2-amine of scaffold of ASK1 inhibitor (GS-444217). These newly synthesized compounds were screened in cell-free ASK1 and PDK1 kinase and cellular vitality assays. Among all compounds tested, both 21c and 21d displayed single digit potency of 9.13, 1.73 nM in inhibiting ASK1, and exhibited excellent enzyme inhibitory activity against PDK1 (the inhibition rates at 10 μM were 13.63% and 23.80%, respectively). Specifically, both compounds inhibited the TGF-β1 induced fibrotic response and blocked the up-regulated protein expression levels of ASK1-p38/JNK signaling pathways and possessed the potency in reducing PDK1/Akt phosphorylation. The results herein showed the potential lead characteristics of 21c or 21d as dual inhibitors ASK1/PDK1 kinases.
Collapse
Affiliation(s)
- Jia Qin
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Meng Cao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Xinlan Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Wenhua Tan
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Binghao Ma
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanyuan Cao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
9
|
3-Bromopyruvate inhibits pancreatic tumor growth by stalling glycolysis, and dismantling mitochondria in a syngeneic mouse model. Am J Cancer Res 2022; 12:4977-4987. [PMID: 36504891 PMCID: PMC9729896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/25/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer (PC) is the fourth-most-deadly cancer in the United States with a 5-year survival rate of only 8%. The majority of patients with locally advanced pancreatic cancer undergo chemotherapy and/or radiation therapy (RT). However, current treatments are inadequate and novel strategies are desperately required. 3-Bromopyruvate (3-BP) is a promising anticancer drug against pancreatic cancer. It exerts potent anticancer effects by inhibiting hexokinase II enzyme (HK2) of the glycolytic pathway in cancer cells while not affecting the normal cells. 3-BP killed 95% of Panc-2 cells at 15 μM concentration and severely inhibited ATP production by disrupting the interaction between HK2 and mitochondrial Voltage Dependent Anion Channel-1 (VDAC1) protein. Electron microscopy data revealed that 3-BP severely damaged mitochondrial membrane in cancer cells. We further examined therapeutic effect of 3-BP in syngeneic mouse pancreatic cancer model by treating animals with 10, 15 and 20 mg/kg dose. 3-BP at 15 & 20 mg/kg dose level significantly reduced tumor growth by approximately 75-80% in C57BL/6 female mice. Immunohistochemistry data showed complete inhibition of hexokinase II (HK2) and TGFβ, in animals treated with 3-BP drug. We also observed enhanced expression of active caspase-3 in tumor tissues exhibited apoptotic death. Flow Cytometry analysis showed significant inhibition in MDSC (CD11b) population in treated tumor which may have allowed infiltration of CD8+ T cells and inhibited tumor growth. Notably, metabolomic data also revealed severe inhibition in glycolysis, NADP, ATP and lactic acid production in cancer cells treated with 40 μM 3-BP. Importantly, we also observed inhibition in lactic acid production responsible for tumor aggression. These results provide new evidence that 3-BP severely inhibit glucose metabolism in cancer cells by blocking hexokinase II, and disrupting mitochondria by suppressing BCL2L1 in pancreatic cancer.
Collapse
|
10
|
Kang Y, Yang X, Feng Z, Lv R, Zhu J. Tumor-associated macrophages improve hypoxia-induced endoplasmic reticulum stress response in colorectal cancer cells by regulating TGF-β1/SOX4. Cell Signal 2022; 99:110430. [PMID: 35933032 DOI: 10.1016/j.cellsig.2022.110430] [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] [Received: 09/02/2021] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a common feature of solid tumors that can induce endoplasmic reticulum stress (ERS). This study aimed to explore the mechanism behind tumor-associated macrophages (TAMs) improving the ERS response of colorectal cancer (CRC) under hypoxic conditions. Herein, it was demonstrated that TAMs reduce ERS by secreting TGF-β1 and activating SOX4/TMEM2 signaling in CRC cells. The expression levels of TGF-β1, SOX4, and TMEM2 in 20 pairs of tumor tissues and para-carcinoma tissues were assessed. A co-culture system of CRC cells with THP-1-derived macrophages under hypoxic conditions in vitro was investigated to determine the protective effect of TAMs on CRC cells. Moreover, to further verify the underlying mechanism, TGF-β1 and SOX4 were knocked down in the TAMs and CRC cells, respectively. The results exposed that TGF-β1, SOX4, and TMEM2 were abundantly expressed in tumor tissues. Moreover, the co-culture system revealed that macrophages stimulate TGF-β1 secretion under hypoxia, which depresses the CRC cells' ERS, further promoting cell proliferation and inhibiting apoptosis. Furthermore, increased TGF-β1 levels promoted the expression of SOX4 and TMEM2 in CRC cells. Conversely, the knockdown of SOX4 attenuated the protective effect of TAMs on TGF-β1-stimulated CRC cells. In conclusion, these results suggest that the elevated ERS induced by hypoxia in CRC cells could be relieved by TAMs via the secretion of TGF-β1. Finally, TGF-β1 suppresses undue ERS response in CRC cells by activating the SOX4-TMEM2 axis.
Collapse
Affiliation(s)
- Yuqing Kang
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai, China
| | - Xuezhong Yang
- Department of Anesthesiology, Zhoupu Hospital of Pudong New Area, Shanghai, China
| | - Zhaoming Feng
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai, China
| | - Ranxu Lv
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai, China
| | - Junfeng Zhu
- Department of Anesthesiology, Jinshan Branch Hospital of Shanghai Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
11
|
Yu B, Zhu Z, Hu T, Lu J, Shen B, Wu T, Guo K, Chaudhary SK, Feng H, Zhao W, Wu D. Construction of a circular RNA-based competing endogenous RNA network to screen biomarkers related to intervertebral disc degeneration. BMC Musculoskelet Disord 2022; 23:675. [PMID: 35840955 PMCID: PMC9284696 DOI: 10.1186/s12891-022-05579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Intervertebral disc degeneration (IDD) is a leading cause of disability with limited treatment strategies. A better understanding of the mechanism of IDD might enable less invasive and more targeted treatments. This study aimed to identify the circular RNA (circRNA)–microRNA (miRNA)–messenger RNA (mRNA) competing endogenous RNA (ceRNA) regulatory mechanisms in IDD. Methods The GSE67567 microarray dataset was downloaded from the Gene Expression Omnibus database. After data preprocessing, differentially expressed circRNAs, miRNAs and mRNAs between IDD and controls were identified. A ceRNA network was constructed on the basis of the interaction between circRNAs and miRNAs, and miRNAs and mRNAs. Pathway enrichment analysis was performed on the mRNAs in the ceRNA network. Then, with ‘intervertebral disc degeneration’ as keywords, IDD-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were searched for in the Comparative Toxicogenomics Database. Results A total of 105 differentially expressed circRNAs, 84 miRNAs and 967 mRNAs were identified. After analysis, 86 circRNA–miRNA, and 126 miRNA–mRNA regulatory relationship pairs were obtained to construct a ceRNA network. The mRNAs were enriched in six KEGG signalling pathways, and four were associated with IDD: the hsa04350: TGF-beta signalling pathway, hsa04068: FoxO signalling pathway, hsa05142: Chagas disease (American trypanosomiasis) and hsa04380: Osteoclast differentiation. An IDD-related ceRNA network was constructed involving four circRNAs, three miRNAs and 11 mRNAs. Auxiliary validation showed that the expression levels of miR-185-5p, miR-486-5p, ACVR1B, FOXO1, SMAD2 and TGFB1 were consistent in different databases. Conclusions Our study identified some circRNA–miRNA–mRNA interaction axes potentially associated with the progression of IDD, viz.: circRNA_100086–miR-509-3p–MAPK1, circRNA_000200–miR-185-5p–TGFB1, circRNA_104308–miR-185-5p–TGFB1, circRNA_400090–miR-486-5p–FOXO1 and circRNA_400090–miR-486-5p–SMAD2. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05579-0. 1. An IDD-related ceRNA network involving four circRNAs, three miRNAs and 11 mRNAs was constructed. 2. The expression levels of miR-185-5p, miR-486-5p, FOXO1, SMAD2 and TGFB1 were consistent in different databases. 3. Our study identified IDD-related circRNA–miRNA–mRNA interaction axes, including circRNA_100086–miR-509-3p–MAPK1.
Collapse
Affiliation(s)
- Bin Yu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Ziqi Zhu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Tao Hu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Jiawei Lu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Beiduo Shen
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Tongde Wu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Kai Guo
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Surendra Kumar Chaudhary
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China
| | - Hang Feng
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450003, Henan, China
| | - Weidong Zhao
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China.
| | - Desheng Wu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Road, Shanghai, 200092, China.
| |
Collapse
|
12
|
Chen J, Ding Q, Jiao X, Wang B, Sun Z, Zhang Y, Zhao J. Dexmedetomidine attenuates hippocampal neuroinflammation in postoperative neurocognitive disorders by inhibiting microRNA-329-3p and activating the CREB1/IL1RA axis. Psychopharmacology (Berl) 2022; 239:2171-2186. [PMID: 35412062 DOI: 10.1007/s00213-022-06091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/14/2022] [Indexed: 11/24/2022]
Abstract
RATIONALE Due to its anti-inflammatory effect, dexmedetomidine (DEX) can confer neuroprotection in postoperative neurocognitive disorders (NCD). Here, the mechanism responsible for this effect of DEX is rarely ascertained. OBJECTIVES Our research was implemented to figure out mechanism governing the protection of DEX against hippocampal neuroinflammation in postoperative NCD. METHODS Exploratory laparotomy was applied for generating a postoperative NCD mouse model before bilateral hippocampal injection with microRNA (miR)-329-3p-agomir and intraperitoneal injection with DEX. Cognitive function of mice was evaluated by water maze test and fear conditioning test. Immunofluorescence was performed to assess microglial activation in hippocampus. After cell transfection and DEX treatment, mouse microglial cells (BV-2) were stimulated by lipopolysaccharide (LPS). IL-1β, IL-6, and TNF-α levels and the number of phagocytes were assessed by ELISA and flow cytometry. Dual-luciferase reporter assay was adopted to assess the relationship between miR-329-3p and CREB1. RESULTS miR-329-3p expression was reduced in the postoperative NCD mice after DEX treatment. DEX treatment or miR-329-3p downregulation caused attenuated cognitive dysfunction and microglia activation as well as reduced IL-1β, IL-6, and TNF-α levels in the hippocampus of the postoperative NCD mice. Mechanistically, miR-329-3p inversely targeted CREB1 that activated IL1RA in LPS-induced BV-2 cells. DEX treatment, miR-329-3p inhibition, or CREB1 or IL1RA upregulation curtailed the release of proinflammatory proteins and the number of phagocytes in LPS-induced BV-2 cells. CONCLUSIONS Collectively, our data provided the novel insight of the neuroprotective mechanism of DEX in postoperative NCD pertaining to the miR-329-3p/CREB1/IL1RA axis.
Collapse
Affiliation(s)
- Jinquan Chen
- Anesthesia Operation Center, The First Peoples Hospital of Xianyang, Xianyang, 712000, People's Republic of China
| | - Qian Ding
- Anesthesia Operation Center, Xi´an International Medical Center Hospital, No. 777, Xitai Road, 710100, Xi´an, People's Republic of China
| | - Xiangxue Jiao
- Anesthesia Operation Center, The First Peoples Hospital of Xianyang, Xianyang, 712000, People's Republic of China
| | - Binrong Wang
- Anesthesia Operation Center, Xi´an International Medical Center Hospital, No. 777, Xitai Road, 710100, Xi´an, People's Republic of China
| | - Zhenzhong Sun
- Department of Anesthesiology, Guangdong Armed Police Corps Hospital, Guangzhou, 510507, People's Republic of China
| | - Yutao Zhang
- Anesthesia Operation Center, Xi´an International Medical Center Hospital, No. 777, Xitai Road, 710100, Xi´an, People's Republic of China
| | - Juan Zhao
- Anesthesia Operation Center, Xi´an International Medical Center Hospital, No. 777, Xitai Road, 710100, Xi´an, People's Republic of China.
| |
Collapse
|
13
|
Tewari D, Priya A, Bishayee A, Bishayee A. Targeting transforming growth factor-β signalling for cancer prevention and intervention: Recent advances in developing small molecules of natural origin. Clin Transl Med 2022; 12:e795. [PMID: 35384373 PMCID: PMC8982327 DOI: 10.1002/ctm2.795] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cancer is the world's second leading cause of death, but a significant advancement in cancer treatment has been achieved within the last few decades. However, major adverse effects and drug resistance associated with standard chemotherapy have led towards targeted treatment options. OBJECTIVES Transforming growth factor-β (TGF-β) signaling plays a key role in cell proliferation, differentiation, morphogenesis, regeneration, and tissue homeostasis. The prime objective of this review is to decipher the role of TGF-β in oncogenesis and to evaluate the potential of various natural and synthetic agents to target this dysregulated pathway to confer cancer preventive and anticancer therapeutic effects. METHODS Various authentic and scholarly databases were explored to search and obtain primary literature for this study. The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria was followed for the review. RESULTS Here we provide a comprehensive and critical review of recent advances on our understanding of the effect of various bioactive natural molecules on the TGF-β signaling pathway to evaluate their full potential for cancer prevention and therapy. CONCLUSION Based on emerging evidence as presented in this work, TGF-β-targeting bioactive compounds from natural sources can serve as potential therapeutic agents for prevention and treatment of various human malignancies.
Collapse
Affiliation(s)
- Devesh Tewari
- Department of PharmacognosySchool of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
| | - Anu Priya
- Department of PharmacologySchool of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
| | | | - Anupam Bishayee
- College of Osteopathic MedicineLake Erie College of Osteopathic MedicineBradentonFloridaUSA
| |
Collapse
|
14
|
Tschernia NP, Gulley JL. Tumor in the Crossfire: Inhibiting TGF-β to Enhance Cancer Immunotherapy. BioDrugs 2022; 36:153-180. [PMID: 35353346 PMCID: PMC8986721 DOI: 10.1007/s40259-022-00521-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 02/04/2023]
Abstract
Cancer immunotherapy using monoclonal antibodies targeting immune checkpoints has undoubtedly revolutionized the cancer treatment landscape in the last decade. Immune checkpoint inhibitors can elicit long-lasting, previously unheard-of responses in a number of tumor entities. Yet, even in such tumors as metastatic melanoma and non-small cell-lung cancer, in which immune checkpoint inhibition has become the first-line treatment of choice, only a minority of patients will benefit considerably from these treatments. This has been attributed to a number of factors, including an immune-suppressive tumor microenvironment (TME). Using different modalities to break these barriers is of utmost importance to expand the population of patients that benefit from immune checkpoint inhibition. The multifunctional cytokine transforming growth factor-β (TGF-β) has long been recognized as an immune-suppressive factor in the TME. A considerable number of drugs have been developed to target TGF-β, yet most of these have since been discontinued. The combination of anti-TGF-β agents with immune checkpoint inhibitors now has the potential to revive this target as a viable immunomodulatory therapeutic approach. Currently, a limited number of small molecular inhibitor and monoclonal antibody candidates that target TGF-β are in clinical development in combination with the following immune checkpoint inhibitors: SRK 181, an antibody inhibiting the activation of latent TGF-β1; NIS 793, a monoclonal antibody targeting TGF-β; and SHR 1701, a fusion protein consisting of an anti-PD-L1 monoclonal antibody fused with the extracellular domain of human TGF-β receptor II. Several small molecular inhibitors are also in development and are briefly reviewed: LY364947, a pyrazole-based small molecular inhibitor of the serine-threonine kinase activity of TGFβRI; SB-431542, an inhibitor targeting several TGF-β superfamily Type I activin receptor-like kinases as well as TGF-β1-induced nuclear Smad3 localization; and galunisertib, an oral small molecular inhibitor of the TGFβRI kinase. One of the most advanced agents in this area is bintrafusp alfa, a bifunctional fusion protein composed of the extracellular domain of TGF-β receptor II fused to a human IgG1 mAb blocking PD-L1. Bintrafusp alfa is currently in advanced clinical development and as an agent in this space with the most clinical experience, is a focused highlight of this review.
Collapse
Affiliation(s)
- Nicholas P Tschernia
- Genitourinary Malignancies Branch, Medical Oncology Service, National Cancer Institute, Building 10, Room 13N240, Bethesda, MD, 20892, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Medical Oncology Service, National Cancer Institute, Building 10, Room 13N240, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022; 24:456-472. [DOI: 10.1016/j.jcyt.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
|
16
|
Understanding and improving cellular immunotherapies against cancer: From cell-manufacturing to tumor-immune models. Adv Drug Deliv Rev 2021; 179:114003. [PMID: 34653533 DOI: 10.1016/j.addr.2021.114003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment (TME) is shaped by dynamic metabolic and immune interactions between precancerous and cancerous tumor cells and stromal cells like epithelial cells, fibroblasts, endothelial cells, and hematopoietically-derived immune cells. The metabolic states of the TME, including the hypoxic and acidic niches, influence the immunosuppressive phenotypes of the stromal and immune cells, which confers resistance to both host-mediated tumor killing and therapeutics. Numerous in vitro TME platforms for studying immunotherapies, including cell therapies, are being developed. However, we do not yet understand which immune and stromal components are most critical and how much model complexity is needed to answer specific questions. In addition, scalable sourcing and quality-control of appropriate TME cells for reproducibly manufacturing these platforms remain challenging. In this regard, lessons from the manufacturing of immunomodulatory cell therapies could provide helpful guidance. Although immune cell therapies have shown unprecedented results in hematological cancers and hold promise in solid tumors, their manufacture poses significant scale, cost, and quality control challenges. This review first provides an overview of the in vivo TME, discussing the most influential cell populations in the tumor-immune landscape. Next, we summarize current approaches for cell therapies against cancers and the relevant manufacturing platforms. We then evaluate current immune-tumor models of the TME and immunotherapies, highlighting the complexity, architecture, function, and cell sources. Finally, we present the technical and fundamental knowledge gaps in both cell manufacturing systems and immune-TME models that must be addressed to elucidate the interactions between endogenous tumor immunity and exogenous engineered immunity.
Collapse
|
17
|
Principe DR, Timbers KE, Atia LG, Koch RM, Rana A. TGFβ Signaling in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2021; 13:5086. [PMID: 34680235 PMCID: PMC8533869 DOI: 10.3390/cancers13205086] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with poor clinical outcomes, largely attributed to incomplete responses to standard therapeutic approaches. Recently, selective inhibitors of the Transforming Growth Factor β (TGFβ) signaling pathway have shown early promise in the treatment of PDAC, particularly as a means of augmenting responses to chemo- and immunotherapies. However, TGFβ is a potent and pleiotropic cytokine with several seemingly paradoxical roles within the pancreatic tumor microenvironment (TME). Although TGFβ signaling can have potent tumor-suppressive effects in epithelial cells, TGFβ signaling also accelerates pancreatic tumorigenesis by enhancing epithelial-to-mesenchymal transition (EMT), fibrosis, and the evasion of the cytotoxic immune surveillance program. Here, we discuss the known roles of TGFβ signaling in pancreatic carcinogenesis, the biologic consequences of the genetic inactivation of select components of the TGFβ pathway, as well as past and present attempts to advance TGFβ inhibitors in the treatment of PDAC patients.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.E.T.); (L.G.A.); (R.M.K.)
| | - Kaytlin E. Timbers
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.E.T.); (L.G.A.); (R.M.K.)
| | - Luke G. Atia
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.E.T.); (L.G.A.); (R.M.K.)
| | - Regina M. Koch
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (K.E.T.); (L.G.A.); (R.M.K.)
| | - Ajay Rana
- Jesse Brown Veterans Affairs Hospital, Chicago, IL 60612, USA
| |
Collapse
|
18
|
Faruk EM, Sabry D, Morsi AA, El-Din YA, Taha NM, Medhat E. The anti-tumour effect of induced pluripotent stem cells against submandibular gland carcinoma in rats is achieved via modulation of the apoptotic response and the expression of Sirt-1, TGF-β, and MALAT-1 in cancer cells. Mol Cell Biochem 2021; 477:53-65. [PMID: 34533647 DOI: 10.1007/s11010-021-04255-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/31/2021] [Indexed: 11/09/2022]
Abstract
The era of induced pluripotent stem cells (iPSCs) was used as novel biotechnology to replace embryonic stem cells bypassing the ethical concerns and problems of stem cell transplant rejection. The anti-tumour potential of iPSCs against many tumours including salivary cancer was proven in previous studies. The current study aimed to investigate the contribution of the Bax, Sirt-1, TGF-β, and MALAT genes and/or their protein expression to the pathogenesis of submandibular carcinogenesis before and after iPSCs treatment. Thirty Wistar albino rats were equally assigned into three groups: group I (control), group II (Squamous cell carcinoma (SCC)): submandibular glands were injected SCC cells, and group III (SCC/iPSCs): SCC rats were treated by 5 × 106 iPSCs. Submandibular gland sections were subjected to histological and immunohistochemical analyses to detect mucopolysaccharides, Bax, and TGF-β expression as well as PCR quantification for TGF-β, Sirt-1, and lncRNA MALAT-1 gene expressions. Western blotting was also used to detect Sirt-1 and TGF-β protein expressions. SCC group revealed infiltration by sheets of malignant squamous cells with or without keratin pearls and inflammatory cells, in addition to upregulation of TGF-β, Sirt-1, MALAT-1, and Bax, whereas SCC/iPSCs group showed an improved submandibular histoarchitecture with the maintenance of the secretory function. Bax and TGF-β immunoexpression were significantly reduced. The upregulated TGF-β, Sirt-1, and MALAT-1 genes were significantly decreased. iPSCs protected against the experimentally induced submandibular gland carcinoma that might be achieved via their regenerative potential and their regulatory modulation of Sirt-1, TGF-β, and MALAT-1 gene/protein expressions and of the apoptotic response in cancer cells.
Collapse
Affiliation(s)
- Eman Mohamed Faruk
- Department of Histology and Cytology, Faculty of Medicine, Benha University, Benha, Egypt.,Department of Anatomy, College of Medicine, Umm Al-Qura University, Mecca, Kingdom of Saudi Arabia
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt.,Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Badr University in Cairo, Badr City, 11829, Egypt
| | - Ahmed A Morsi
- Department of Histology and Cell Biology, Faculty of Medicine, Fayoum University, Fayoum, Fayoum Governorate, Egypt.
| | - Yasmine Alaa El-Din
- Department of Oral Pathology, Faculty of Dentistry, October 6 University, Giza, Egypt
| | - Neama M Taha
- Department of Physiology, Umm Al-Qura University, Mecca, Kingdom of Saudi Arabia
| | - Engy Medhat
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Shi X, Luo J, Weigel KJ, Hall SC, Du D, Wu F, Rudolph MC, Zhou H, Young CD, Wang XJ. Cancer-Associated Fibroblasts Facilitate Squamous Cell Carcinoma Lung Metastasis in Mice by Providing TGFβ-Mediated Cancer Stem Cell Niche. Front Cell Dev Biol 2021; 9:668164. [PMID: 34527666 PMCID: PMC8435687 DOI: 10.3389/fcell.2021.668164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/21/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) have been shown to enhance squamous cell carcinoma (SCC) growth, but it is unclear whether they promote SCC lung metastasis. We generated CAFs from K15.KrasG12D.Smad4-/- mouse SCCs. RNA expression analyses demonstrated that CAFs had enriched transforming growth factor-beta (TGFβ) signaling compared to normal tissue-associated fibroblasts (NAFs), therefore we assessed how TGFβ-enriched CAFs impact SCC metastasis. We co-injected SCC cells with CAFs to the skin, tail vein, or the lung to mimic sequential steps of lung metastasis. CAFs increased SCC volume only in lung co-transplantations, characterized with increased proliferation and angiogenesis and decreased apoptosis compared to NAF co-transplanted SCCs. These CAF effects were attenuated by a clinically relevant TGFβ receptor inhibitor, suggesting that CAFs facilitated TGFβ-dependent SCC cell seeding and survival in the lung. CAFs also increased tumor volume when co-transplanted to the lung with limiting numbers of SCC cancer stem cells (CSCs). In vitro, CSC sphere formation and invasion were increased either with co-cultured CAFs or with CAF conditioned media (which contains the highest TGFβ1 concentration) and these CAF effects were blocked by TGFβ inhibition. Further, TGFβ activation was higher in primary human oral SCCs with lung metastasis than SCCs without lung metastasis. Similarly, TGFβ activation was detected in the lungs of mice with micrometastasis. Our data suggest that TGFβ-enriched CAFs play a causal role in CSC seeding and expansion in the lung during SCC metastasis, providing a prognostic marker and therapeutic target for SCC lung metastasis.
Collapse
Affiliation(s)
- Xueke Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jingjing Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kelsey J. Weigel
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Spencer C. Hall
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Danfeng Du
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Fanglong Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Michael C. Rudolph
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Christian D. Young
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO, United States
| |
Collapse
|
20
|
Kojima K, Ichijo H, Naguro I. Molecular functions of ASK family in diseases caused by stress-induced inflammation and apoptosis. J Biochem 2021; 169:395-407. [PMID: 33377973 DOI: 10.1093/jb/mvaa145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
VCells are constantly exposed to various types of stress, and disruption of the proper response leads to a variety of diseases. Among them, inflammation and apoptosis are important examples of critical responses and should be tightly regulated, as inappropriate control of these responses is detrimental to the organism. In several disease states, these responses are abnormally regulated, with adverse effects. Apoptosis signal-regulating kinase (ASK) family members are stress-responsive kinases that regulate inflammation and apoptosis after a variety of stimuli, such as oxidative stress and endoplasmic reticulum stress. In this review, we summarize recent reports on the ASK family in terms of their involvement in inflammatory diseases, focussing on upstream stimuli that regulate ASK family members.
Collapse
Affiliation(s)
- Kazuki Kojima
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
21
|
Kabel AM, Arab HH, Abd Elmaaboud MA. Attenuation of diethyl nitrosamine-induced hepatocellular carcinoma by taxifolin and/or alogliptin: The interplay between toll-like receptor 4, transforming growth factor beta-1, and apoptosis. Hum Exp Toxicol 2021; 40:1710-1720. [PMID: 33840231 DOI: 10.1177/09603271211008496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver malignancies worldwide. Alogliptin is an anti-diabetic that may have effective anticancer properties against many types of malignancies. Taxifolin is a flavonoid that has potent antioxidant, and anti-inflammatory properties. The objective of this study was to explore the impact of alogliptin and/or taxifolin on diethyl nitrosamine-induced HCC in rats. One hundred male Wistar rats were divided into five equal groups as follows: Control; HCC; HCC + Alogliptin; HCC + Taxifolin; and HCC + Alogliptin + Taxifolin group. The survival rate, liver function tests, tissue antioxidant enzymes, malondialdehyde (MDA), nuclear factor (erythroid derived 2)-like 2 (Nrf2), transforming growth factor beta 1 (TGF-β1), interleukin 1 alpha (IL-1α), and toll-like receptor 4 (TLR4) were measured. Also, hepatic caspase 3, caspase 9, beclin-1, and c-Jun NH2-terminal kinase (JNK) in addition to serum alpha-fetoprotein (AFP) and α-L-Fucosidase (AFU) were assessed. Specimens of the liver were subjected to histopathological examination. Alogliptin and/or taxifolin induced significant improvement of liver function tests with significant increase in the survival rate, tissue antioxidant enzymes, Nrf2, caspase 3, caspase 9, Beclin-1 and JNK activities associated with significant decrease in serum AFP and AFU, tissue MDA, TGF-β1, IL-1α and TLR4 expression compared to HCC group. These results were significant with taxifolin/alogliptin combination when compared to the use of each of these agents alone. In conclusion, taxifolin/alogliptin combination might be used as adjuvant therapy for attenuation of HCC.
Collapse
Affiliation(s)
- A M Kabel
- Pharmacology Department, 68782Faculty of Medicine, Tanta University, Tanta, Egypt
| | - H H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, 125895Taif University, Taif, Saudi Arabia
| | - M A Abd Elmaaboud
- Pharmacology Department, 68782Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
22
|
Unraveling the Molecular Nexus between GPCRs, ERS, and EMT. Mediators Inflamm 2021; 2021:6655417. [PMID: 33746610 PMCID: PMC7943314 DOI: 10.1155/2021/6655417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent a large family of transmembrane proteins that transduce an external stimulus into a variety of cellular responses. They play a critical role in various pathological conditions in humans, including cancer, by regulating a number of key processes involved in tumor formation and progression. The epithelial-mesenchymal transition (EMT) is a fundamental process in promoting cancer cell invasion and tumor dissemination leading to metastasis, an often intractable state of the disease. Uncontrolled proliferation and persistent metabolism of cancer cells also induce oxidative stress, hypoxia, and depletion of growth factors and nutrients. These disturbances lead to the accumulation of misfolded proteins in the endoplasmic reticulum (ER) and induce a cellular condition called ER stress (ERS) which is counteracted by activation of the unfolded protein response (UPR). Many GPCRs modulate ERS and UPR signaling via ERS sensors, IRE1α, PERK, and ATF6, to support cancer cell survival and inhibit cell death. By regulating downstream signaling pathways such as NF-κB, MAPK/ERK, PI3K/AKT, TGF-β, and Wnt/β-catenin, GPCRs also upregulate mesenchymal transcription factors including Snail, ZEB, and Twist superfamilies which regulate cell polarity, cytoskeleton remodeling, migration, and invasion. Likewise, ERS-induced UPR upregulates gene transcription and expression of proteins related to EMT enhancing tumor aggressiveness. Though GPCRs are attractive therapeutic targets in cancer biology, much less is known about their roles in regulating ERS and EMT. Here, we will discuss the interplay in GPCR-ERS linked to the EMT process of cancer cells, with a particular focus on oncogenes and molecular signaling pathways.
Collapse
|
23
|
de Waal CR, Kleynhans L, Parsons SDC, Goosen WJ, Hausler G, Buss PE, Warren RM, van Helden PD, Landolfi JA, Miller MA, Kerr TJ. Development of a cytokine gene expression assay for the relative quantification of the African elephant (Loxodonta africana) cell-mediated immune responses. Cytokine 2021; 141:155453. [PMID: 33548797 DOI: 10.1016/j.cyto.2021.155453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 11/27/2022]
Abstract
Immunological assays are the basis for many diagnostic tests for infectious diseases in animals and humans. Application in wildlife species, including the African elephant (Loxodonta africana), is limited however due to lack of information on immune responses. Since many immunoassays require both identified biomarkers of immune activation as well as species-specific reagents, it is crucial to have knowledge of basic immunological responses in the species of interest. Cytokine gene expression assays (GEAs) used to measure specific immune responses in wildlife have frequently shown that targeted biomarkers are often species-specific. Therefore, the aim of this study was to identify elephant-specific cytokine biomarkers to detect immune activation and to develop a GEA, using pokeweed mitogen stimulated whole blood from African elephants. This assay will provide the foundation for the development of future cytokine GEAs that can be used to detect antigen specific immune responses and potentially lead to various diagnostic tests for this species.
Collapse
Affiliation(s)
- Candice R de Waal
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sven D C Parsons
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Wynand J Goosen
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Guy Hausler
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Peter E Buss
- South African National Parks, Veterinary Wildlife Services, Kruger National Park, Skukuza, South Africa
| | - Robin M Warren
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Paul D van Helden
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jennifer A Landolfi
- University of Illinois Zoological Pathology Program, 3300 Golf Road, Brookfield, IL, 60153, USA
| | - Michele A Miller
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Tanya J Kerr
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
24
|
Bouhtit F, Najar M, Moussa Agha D, Melki R, Najimi M, Sadki K, Boukhatem N, Bron D, Meuleman N, Hamal A, Lagneaux L, Lewalle P, Merimi M. New Anti-Leukemic Effect of Carvacrol and Thymol Combination through Synergistic Induction of Different Cell Death Pathways. Molecules 2021; 26:molecules26020410. [PMID: 33466806 PMCID: PMC7829697 DOI: 10.3390/molecules26020410] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a cancer of the myeloid lineage of blood cells, and treatment for AML is lengthy and can be very expensive. Medicinal plants and their bioactive molecules are potential candidates for improving human health. In this work, we studied the effect of Ptychotis verticillata (PV) essential oil and its derivatives, carvacrol and thymol, in AML cell lines. We demonstrated that a combination of carvacrol and thymol induced tumor cell death with low toxicity on normal cells. Mechanistically, we highlighted that different molecular pathways, including apoptosis, oxidative, reticular stress, autophagy, and necrosis, are implicated in this potential synergistic effect. Using quantitative RT-PCR, Western blotting, and apoptosis inhibitors, we showed that cell death induced by the carvacrol and thymol combination is caspase-dependent in the HL60 cell line and caspase-independent in the other cell lines tested. Further investigations should focus on improving the manufacturing of these compounds and understanding their anti-tumoral mechanisms of action. These efforts will lead to an increase in the efficiency of the oncotherapy strategy regarding AML.
Collapse
Affiliation(s)
- Fatima Bouhtit
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
| | - Mehdi Najar
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), University of Montreal, Montreal, QC H2X 0A9, Canada
| | - Douâa Moussa Agha
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
| | - Rahma Melki
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Khalid Sadki
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Agdal-Rabat 10090, Morocco;
| | - Noureddine Boukhatem
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
| | - Dominique Bron
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
| | - Nathalie Meuleman
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
| | - Abdellah Hamal
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium;
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
| | - Makram Merimi
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.B.); (D.M.A.); (D.B.); (N.M.); (P.L.)
- Genetics and Immune Cell Therapy Unit, LBBES Laboratory, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco; (M.N.); (R.M.); (N.B.); (A.H.)
- Correspondence:
| |
Collapse
|
25
|
Uckun FM, Qazi S, Hwang L, Trieu VN. Recurrent or Refractory High-Grade Gliomas Treated by Convection-Enhanced Delivery of a TGFβ 2-Targeting RNA Therapeutic: A Post-Hoc Analysis with Long-Term Follow-Up. Cancers (Basel) 2019; 11:cancers11121892. [PMID: 31795071 PMCID: PMC6966490 DOI: 10.3390/cancers11121892] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 01/06/2023] Open
Abstract
Background. OT101 is a first-in-class RNA therapeutic designed to abrogate the immunosuppressive actions of transforming growth factor beta 2 (TGFβ2). Here, we report our post-hoc analysis of the single-agent activity of OT101 in adult patients with recurrent and/or refractory (R/R) high-grade gliomas. Methods. In a Phase 2 clinical trial (ClinicalTrials.gov, NCT00431561), OT101 was administered to 89 R/R high-grade glioma (HGG) (anaplastic astrocytoma/AA: 27; glioblastoma multiforme/GBM: 62) patients with an intratumoral catheter using a convection enhanced delivery (CED) system. Seventy-seven patients (efficacy population; GBM: 51; AA: 26) received at least the intended minimum number of four OT101 treatment cycles. Response determinations were based on central review of magnetic resonance imaging (MRI) scans according to the McDonald criteria. Standard statistical methods were applied for the analysis of data. Findings. Nineteen patients had a complete response (CR) or partial response (PR) following a slow but robust size reduction of their target lesions (median time for 90% reduction of the baseline tumor volume = 11.7 months, range: 4.9-57.7 months). The mean log reduction of the tumor volume was 2.2 ± 0.4 (median = 1.4: range: 0.4-4.5) logs. In addition, seven patients had a stable disease (SD) lasting ≥6 months. For the combined group of 26 AA/GBM patients with favorable responses, the median progression-free survival (PFS) of 1109 days and overall survival (OS) of 1280 days were significantly better than the median PFS (p < 0.00001) and OS (p < 0.00001) of the non-responders among the 89 patients or the 77-patient efficacy population. Conclusion. Intratumorally administered OT101 exhibits clinically meaningful single-agent activity and induces durable CR/PR/SD in R/R HGG patients.
Collapse
Affiliation(s)
- Fatih M. Uckun
- Immuno-Oncology Program, Oncotelic Inc., Agoura Hills, CA 91301, USA; (S.Q.); (L.H.); (V.N.T.)
- Department of Immuno-Oncology, Ares Pharmaceuticals, St. Paul, MN 55110, USA
- Correspondence:
| | - Sanjive Qazi
- Immuno-Oncology Program, Oncotelic Inc., Agoura Hills, CA 91301, USA; (S.Q.); (L.H.); (V.N.T.)
- Department of Immuno-Oncology, Ares Pharmaceuticals, St. Paul, MN 55110, USA
- Department of Biology, Bioinformatics Program, Gustavus Adolphus College, St. Peter, MN 56082, USA
| | - Larn Hwang
- Immuno-Oncology Program, Oncotelic Inc., Agoura Hills, CA 91301, USA; (S.Q.); (L.H.); (V.N.T.)
| | - Vuong N. Trieu
- Immuno-Oncology Program, Oncotelic Inc., Agoura Hills, CA 91301, USA; (S.Q.); (L.H.); (V.N.T.)
| |
Collapse
|