1
|
Moomivand S, Nikbakht M, Majd A, Bikhof Torbati M, Mousavi SA. Synergistic Enhancement of Apo2L/TRAIL and DR4-Induced Apoptosis by Arsenic Trioxide in Triple-Negative Breast Cancer Cells: A Comparison to Conventional Chemotherapy. Cell Biochem Biophys 2025:10.1007/s12013-025-01764-9. [PMID: 40293700 DOI: 10.1007/s12013-025-01764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype lacking hormonal and HER2 receptors, making it highly resistant to treatment. Apo2L/TRAIL, a tumor necrosis factor-related ligand, induces apoptosis in cancer cells via the death receptor DR4. However, TNBC often develops resistance to TRAIL-mediated apoptosis, limiting its therapeutic potential. This study investigates whether arsenic trioxide (ATO) can overcome TRAIL resistance by modulating the Apo2L/TRAIL pathway and enhancing the effects of carboplatin (CP) and cyclophosphamide (CY). TNBC cell lines BT-20 and MDA-MB-231 were treated with ATO, CP, CY, and their combinations. Cell viability was measured using the MTT assay, while real-time PCR and Western blot analysis assessed Apo2L/TRAIL and DR4 expression. Statistical analysis was performed using ANOVA with Dunnett's post hoc test. ATO induced dose-dependent cytotoxicity in TNBC cells, which was significantly enhanced in combination treatments. The highest reductions in cell viability were observed with 3 µM ATO plus 5000 µM CP or 500 µM CY (p < 0.0001). ATO markedly upregulated Apo2L/TRAIL and DR4 at both mRNA and protein levels, with the most pronounced effects seen in ATO-CY combinations. These findings indicate that ATO sensitizes TNBC cells to TRAIL-mediated apoptosis by upregulating DR4 and Apo2L/TRAIL, while also exhibiting strong synergistic cytotoxicity with CP and CY. This highlights ATO's potential as an adjuvant therapy to improve TNBC treatment efficacy and overcome chemoresistance, warranting further clinical exploration.
Collapse
Affiliation(s)
- Soraya Moomivand
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohsen Nikbakht
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran.
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran.
| | - Ahmad Majd
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Bikhof Torbati
- Department of Biology, Yadegar-e-Imam Khomeini (RAH) Shahre rey branch, Islamic Azad University, Tehran, Iran
| | - Seyed Asadoullah Mousavi
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Qiao X, Guo S, Meng Z, Gan H, Wu Z, Sun Y, Liu S, Dou G, Gu R. Advances in the study of death receptor 5. Front Pharmacol 2025; 16:1549808. [PMID: 40144653 PMCID: PMC11936945 DOI: 10.3389/fphar.2025.1549808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
DR5, a receptor with the highest affinity for TRAIL under physiological conditions, selectively induces apoptosis in specific target cells such as tumor and aberrant immune cells, while minimally affecting normal cells. The TRAIL-DR5 signaling pathway is a crucial regulatory mechanism when the body responds to various exogenous interference factors, including viruses, chemicals, and radiation. This pathway plays a vital role in maintaining physiological homeostasis and in the pathological development of various diseases. Different modulations of DR5, such as upregulation, activation, and antagonism, hold significant potential for therapeutic applications in tumors, cardiovascular diseases, autoimmune diseases, viral infections, and radiation injuries. This article provides an overview of the current research progress on DR5, including the status and prospects of its clinical applications.
Collapse
Affiliation(s)
- Xuan Qiao
- Graduate Collaborative Training Base of Academy of Military Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuang Guo
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhiyun Meng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Gan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhuona Wu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yunbo Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Guifang Dou
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Ruolan Gu
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
3
|
Pan H, Jing C. Exploring druggable targets and inflammation-mediated pathways in cancer: a Mendelian randomization analysis integrating transcriptomic and proteomic data. Inflamm Res 2025; 74:46. [PMID: 40038097 DOI: 10.1007/s00011-025-02011-w] [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: 11/15/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Cancer remains a predominant global health challenge, necessitating the ongoing exploration of novel biomarkers and therapeutic targets to improve diagnosis and treatment. METHODS By integrating expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL) data with genome-wide association studies (GWAS) data, we performed Mendelian randomization (MR) analysis to identify potential druggable targets at the gene expression and protein levels for multiple cancers. We conducted mediation analysis to explore whether inflammatory factors mediate the pathways linking identified druggable targets to cancer. Phenome-wide MR analysis, drug prediction, and molecular docking were employed to evaluate the medicinal potential. RESULTS We finally identified five druggable targets: CDKN1A, FES, and PDIA3 were associated with breast cancer, whereas TP53 and VAMP8 were associated with prostate cancer. Mediation analysis identified six inflammatory proteins as potential mediators in the causal pathways from these druggable targets to cancer: caspase 8, interleukin-1-alpha, C-X-C motif chemokine 1, C-C motif chemokine 23, TNF-related apoptosis-inducing ligand, and interleukin-6. Subsequent analyses further provided evidence supporting the pharmaceutical potential of these five targets. CONCLUSIONS Our study identified five druggable targets causally associated with breast and prostate cancers, with six inflammatory proteins acting as potential mediators, providing novel insights into the treatment of these cancers.
Collapse
Affiliation(s)
- Hao Pan
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, No.324 Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, People's Republic of China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, No.324 Jingwu Road, Huaiyin District, Jinan, 250021, Shandong, People's Republic of China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Mengyuan H, Aixue L, Yongwei G, Qingqing C, Huanhuan C, Xiaoyan L, Jiyong L. Biomimetic nanocarriers in cancer therapy: based on intercellular and cell-tumor microenvironment communication. J Nanobiotechnology 2024; 22:604. [PMID: 39370518 PMCID: PMC11456251 DOI: 10.1186/s12951-024-02835-4] [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: 07/04/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Inspired by the concept of "natural camouflage," biomimetic drug delivery systems have emerged to address the limitations of traditional synthetic nanocarriers, such as poor targeting, susceptibility to identification and clearance, inadequate biocompatibility, low permeability, and systemic toxicity. Biomimetic nanocarriers retain the proteins, nucleic acids, and other components of the parent cells. They not only facilitate drug delivery but also serve as communication media to inhibit tumor cells. This paper delves into the communication mechanisms between various cell-derived biomimetic nanocarriers, tumor cells, and the tumor microenvironment, as well as their applications in drug delivery. In addition, the additional communication capabilities conferred on the modified biomimetic nanocarriers, such as targeting and environmental responsiveness, are outlined. Finally, we propose future development directions for biomimetic nanocarriers, hoping to inspire researchers in their design efforts and ultimately achieve clinical translation.
Collapse
Affiliation(s)
- He Mengyuan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Li Aixue
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Gu Yongwei
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Chai Qingqing
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Cai Huanhuan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China
| | - Liu Xiaoyan
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| | - Liu Jiyong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Kim SL, Shin M, Jin BC, Seo S, Ha GW, Kim SW. Acquired Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Resistance of Human Colorectal Cancer Cells Is Linked to Histone Acetylation and Is Synergistically Ameliorated by Combination with HDAC Inhibitors. Dig Dis Sci 2024; 69:3305-3317. [PMID: 39090444 DOI: 10.1007/s10620-024-08569-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive target for the treatment of various malignancies; however, its therapeutic potential is limited because of the frequent occurrence of tumor cell resistance. In this study, we determined whether TRAIL resistance acquired by repeated administration could be overcome by HDAC inhibition in human colorectal cancer cells. METHODS TRAIL-resistant HCT116 human colorectal cancer cells (HCT116-TR) were generated by repeated treatment with 10 and 25 ng/mL TRAIL twice weekly for 28 days. RESULTS The resulting TRAIL-resistant cells were noncross-resistant to other chemotherapeutic agents. The levels of histone acetylation-related proteins, such as ac-histone H4 and HDAC1, were altered in HCT116-TR cells compared with the parental HCT116 cell line. The combined treatment with TRAIL and HDAC inhibitors significantly increased apoptosis in HCT116-TR cells and indicated a synergistic effect. The mechanism by which HDAC inhibition sensitizes HCT116-TR cells to TRAIL is dependent on the intrinsic pathway. In addition, we found that HDAC inhibition enhanced the sensitivity of cells to TRAIL through mitogen-activated protein kinases/CCAAT/enhancer-binding protein homologs of protein-dependent upregulation of death receptor 5. CONCLUSION These results suggest that histone acetylation is responsible for acquired TRAIL resistance after repeated exposure and acquired resistance to TRAIL may be overcome by combination therapies with HDAC inhibitors.
Collapse
Affiliation(s)
- Se Lim Kim
- Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, 20, Geonji-Ro, Deokjin-Gu, Jeonju, Jeonbuk, 54907, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - MinWoo Shin
- Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, 20, Geonji-Ro, Deokjin-Gu, Jeonju, Jeonbuk, 54907, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Byung Chul Jin
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - SeungYoung Seo
- Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, 20, Geonji-Ro, Deokjin-Gu, Jeonju, Jeonbuk, 54907, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Gi Won Ha
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
- Department of Surgery, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, 20, Geonji-Ro, Deokjin-Gu, Jeonju, Jeonbuk, 54907, Republic of Korea
| | - Sang Wook Kim
- Department of Internal Medicine, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, 20, Geonji-Ro, Deokjin-Gu, Jeonju, Jeonbuk, 54907, Republic of Korea.
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea.
| |
Collapse
|
6
|
Grisendi G, Dall'Ora M, Casari G, Spattini G, Farshchian M, Melandri A, Masciale V, Lepore F, Banchelli F, Costantini RC, D'Esposito A, Chiavelli C, Spano C, Spallanzani A, Petrachi T, Veronesi E, Ferracin M, Roncarati R, Vinet J, Magistri P, Catellani B, Candini O, Marra C, Eccher A, Bonetti LR, Horwtiz EM, Di Benedetto F, Dominici M. Combining gemcitabine and MSC delivering soluble TRAIL to target pancreatic adenocarcinoma and its stroma. Cell Rep Med 2024; 5:101685. [PMID: 39168103 PMCID: PMC11384958 DOI: 10.1016/j.xcrm.2024.101685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) still has a poor response to therapies, partly due to their cancer-associated fibroblasts (CAFs). Here, we investigate the synergistic impact of a combinatory approach between a known chemotherapy agent, such as gemcitabine (GEM), and gene-modified human mesenchymal stromal/stem cells (MSCs) secreting the pro-apoptotic soluble (s)TRAIL (sTRAIL MSCs) on both PDAC cells and CAFs. The combo significantly impacts on PDAC survival in 2D and 3D models. In orthotopic xenograft models, GEM and sTRAIL MSCs induce tumor architecture shredding with a reduction of CK7- and CK8/18-positive cancer cells and the abrogation of spleen metastases. A cytotoxic effect on primary human CAFs is also observed along with an alteration of their transcriptome and a reduction of the related desmoplasia. Collectively, we demonstrate a promising therapeutic profile of combining GEM and sTRAIL MSCs to target both tumoral and stromal compartments in PDAC.
Collapse
Affiliation(s)
- Giulia Grisendi
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy.
| | | | - Giulia Casari
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, Ancona
| | | | - Moein Farshchian
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Aurora Melandri
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Valentina Masciale
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Fabio Lepore
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Federico Banchelli
- Center of Statistic, Department of Medical and Surgical Sciences, UNIMORE, Modena, Italy
| | | | - Angela D'Esposito
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Chiara Chiavelli
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy
| | - Carlotta Spano
- Department of Biomedical, Metabolic, and Neural Sciences, UNIMORE, Modena, Italy
| | | | | | | | - Manuela Ferracin
- Department of Medical and Surgical Sciences, University of Bologna, Bologna; IRCCS AOU di Bologna, Policlinico S. Orsola-Malpighi, Bologna
| | | | | | - Paolo Magistri
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | - Barbara Catellani
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | | | - Caterina Marra
- Division of Plastic Surgery, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Edwin M Horwtiz
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Fabrizio Di Benedetto
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, UNIMORE, Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia (UNIMORE), Modena, Italy; Division of Oncology, University-Hospital of Modena, Modena, Italy; Division of Medical Oncology, Residency School of Medical Oncology, Program in Cellular Therapy and Immuno-oncology, Laboratory of Cellular Therapy, University Hospital of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
7
|
Masum AA, Aoki S, Rahman MM, Hisamatsu Y. Chemical synthetic approaches to mimic the TRAIL: promising cancer therapeutics. RSC Med Chem 2024; 15:d4md00183d. [PMID: 39246747 PMCID: PMC11376135 DOI: 10.1039/d4md00183d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Apoptosis is programmed cell death that eliminates undesired cells to maintain homeostasis in metazoan. Aberration of this process may lead to cancer genesis. The tumor necrosis factor related apoptosis inducing ligand (TRAIL) induces apoptosis in cancer cells after ligation with death receptors (DR4/DR5) while sparing most normal cells. Therefore, strategies to induce apoptosis in cancer cells by mimicking the TRAIL emerge as a promising therapeutic tool. Hence, approaches are taken to develop TRAIL/DR-based cancer therapeutics. The recombinant soluble TRAIL (rhTRAIL) and death receptor agonistic antibodies were produced and tested pre-clinically and clinically. Pre-clinical and clinical trial data demonstrate that these therapeutics are safe and relatively well tolerated. But some of these therapeutics failed to exert adequate efficacy in clinical settings. Besides these biotechnologically derived therapeutics, a few chemically synthesized therapeutics are reported. Some of these therapeutics exert considerable efficacy in vitro and in vivo. In this review, we will discuss chemically synthesized TRAIL/DR-based therapeutics, their chemical and biological behaviour, design concepts and strategies that may contribute to further improvement of TRAIL/DR-based therapeutics.
Collapse
Affiliation(s)
- Abdullah-Al Masum
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Science and Technology, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
| | - Md Mahbubur Rahman
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Yosuke Hisamatsu
- Graduate School of Pharmaceutical Sciences, Nagoya City University Mizuho-Ku Nagoya 467-8603 Japan
| |
Collapse
|
8
|
Lee HY, Hsu MJ, Chang HH, Chang WC, Huang WC, Cho EC. Enhancing anti-cancer capacity: Novel class I/II HDAC inhibitors modulate EMT, cell cycle, and apoptosis pathways. Bioorg Med Chem 2024; 109:117792. [PMID: 38897139 DOI: 10.1016/j.bmc.2024.117792] [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: 03/12/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Cancer has been a leading cause of death over the last few decades in western countries as well as in Taiwan. However, traditional therapies are limited by the adverse effects of chemotherapy and radiotherapy, and tumor recurrence may occur. Therefore, it is critical to develop novel therapeutic drugs. In the field of HDAC inhibitor development, apart from the hydroxamic acid moiety, 2-aminobenzamide also functions as a zinc-binding domain, which is shown in well-known HDAC inhibitors such as Entinostat and Chidamide. With recent successful experiences in synthesizing 1-(phenylsulfonyl)indole-based compounds, in this study, we further combined two features of the above chemical compounds and generated indolyl benzamides. Compounds were screened in different cancer cell lines, and enzyme activity was examined to demonstrate their potential for anti-HDAC activity. Various biological functional assays evidenced that two of these compounds could suppress cancer growth and migration capacity, through regulating epithelial-mesenchymal transition (EMT), cell cycle, and apoptosis mechanisms. Data from 3D cancer cells and the in vivo zebrafish model suggested the potential of these compounds in cancer therapy in the future.
Collapse
Affiliation(s)
- Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| | - Min-Jung Hsu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Hao-Hsien Chang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Wei-Chiao Chang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Wan-Chen Huang
- Single-Molecule Biology Core Lab, Institute of Cellular and Organismic Biology (ICOB), Academia Sinica, Taipei, Taiwan.
| | - Er-Chieh Cho
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Cancer Center, Wan Fang Hospital, Taipei Medical University, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
9
|
Yin Y, Shi X, Cai X, Liu F, Ni W, Li B, Wan X, Ren M. Isolation Techniques, Structural Characteristics, and Pharmacological Effects of Phellinus Polysaccharides: A Review. Molecules 2024; 29:3047. [PMID: 38998999 PMCID: PMC11243265 DOI: 10.3390/molecules29133047] [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: 05/14/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Phellinus is a precious perennial medicinal fungus. Its polysaccharides are important bioactive components, and their chemical composition is complex. The polysaccharides are mainly extracted from the fruiting body and mycelium. The yield of the polysaccharides is dependent on the extraction method. They have many pharmacological activities, such as antitumor, immunomodulatory, antioxidant, hypoglycemic, anti-inflammatory, etc. They are also reported to show minor toxic and side effects. Many studies have reported the anticancer activity of Phellinus polysaccharides. This review paper provides a comprehensive examination of the current methodologies for the extraction and purification of Phellinus polysaccharides. Additionally, it delves into the structural characteristics, pharmacological activities, and mechanisms of action of these polysaccharides. The primary aim of this review is to offer a valuable resource for researchers, facilitating further studies on Phellinus polysaccharides and their potential applications.
Collapse
Affiliation(s)
- Yiming Yin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
- College of Pharmacy, Shandong University, Jinan 250100, China
| | - Xiaolin Shi
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
| | - Xiaoqing Cai
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
| | - Fangrui Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
| | - Wenting Ni
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
| | - Baohong Li
- Innovative Institute of Chinse Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China;
| | - Xinhuan Wan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Y.); (X.S.); (X.C.); (F.L.); (W.N.)
| | - Meng Ren
- College of Physical Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
10
|
Bravo Perina L, Faria Gomes IN, Alcantara Pelloso AR, Silva VAO, Rebolho Batista Arantes LM, Eliseo Melendez M. Combined effect of the pro-apoptotic rhTRAIL protein and HSV-1 virus in head and neck cancer cell lines. Sci Rep 2023; 13:18023. [PMID: 37865660 PMCID: PMC10590400 DOI: 10.1038/s41598-023-44888-9] [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: 02/03/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
Knowledge on the molecular and clinical characteristics of head and neck squamous cell carcinoma (HNSCC) is vast. However, an effective therapy that increases the life expectancy of these patients, with a 5-year overall survival of 50%, is still unknown. Here we evaluated the combined effect of the pro-apoptotic protein rhTRAIL with the replication-competent wild-type HSV-1 virus in head and neck cancer cell lines. We observed a difference in the modulation profile of proteins related to apoptotic pathways in the studied cell lines. The HCB289 exhibited caspase-9 activation in the presence of the HSV-1 virus, while the UD-SCC-2 exhibited caspase-8 activation in the presence of rhTRAIL. Both cell lines exhibited PARP activation by combining rhTRAIL and HSV-1 virus treatment. Flow cytometry analysis exhibited greater induction of late apoptosis for the HCB289 and UD-SCC-2 after the combination treatment of the HSV-1 and rhTRAIL. However, the UD-SCC-2 also presented induction of late apoptosis by the presence of rhTRAIL in monotherapy. These data suggest an enhancement of the effect of the combination treatment of the rhTRAIL and the HSV-1 on reducing viability and induction of cell death.
Collapse
Affiliation(s)
- Lucas Bravo Perina
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, 14784-400, Brazil
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, RJ, 20230-240, Brazil
| | | | - Ana Rúbia Alcantara Pelloso
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, 14784-400, Brazil
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, RJ, 20230-240, Brazil
| | - Viviane Aline Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, 14784-400, Brazil
- Department of Pathology and Legal Medicine, Medical School of the Federal University of Bahia, Salvador, BA, 40026-010, Brazil
- Laboratory of Pathology and Molecular Biology, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, 40296-710, Brazil
| | | | - Matias Eliseo Melendez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, 14784-400, Brazil.
- Molecular Carcinogenesis Program, National Cancer Institute (INCA), Rio de Janeiro, RJ, 20230-240, Brazil.
| |
Collapse
|
11
|
Cheng Q, Kang Y, Yao B, Dong J, Zhu Y, He Y, Ji X. Genetically Engineered-Cell-Membrane Nanovesicles for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302131. [PMID: 37409429 PMCID: PMC10502869 DOI: 10.1002/advs.202302131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
The advent of immunotherapy has marked a new era in cancer treatment, offering significant clinical benefits. Cell membrane as drug delivery materials has played a crucial role in enhancing cancer therapy because of their inherent biocompatibility and negligible immunogenicity. Different cell membranes are prepared into cell membrane nanovesicles (CMNs), but CMNs have limitations such as inefficient targeting ability, low efficacy, and unpredictable side effects. Genetic engineering has deepened the critical role of CMNs in cancer immunotherapy, enabling genetically engineered-CMN (GCMN)-based therapeutics. To date, CMNs that are surface modified by various functional proteins have been developed through genetic engineering. Herein, a brief overview of surface engineering strategies for CMNs and the features of various membrane sources is discussed, followed by a description of GCMN preparation methods. The application of GCMNs in cancer immunotherapy directed at different immune targets is addressed as are the challenges and prospects of GCMNs in clinical translation.
Collapse
Affiliation(s)
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jinrui Dong
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yalan Zhu
- Jinhua Municipal Central HospitalJinhua321000China
| | - Yiling He
- Jinhua Municipal Central HospitalJinhua321000China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
12
|
Targeting TRAIL Death Receptors in Triple-Negative Breast Cancers: Challenges and Strategies for Cancer Therapy. Cells 2022; 11:cells11233717. [PMID: 36496977 PMCID: PMC9739296 DOI: 10.3390/cells11233717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily member TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis in cancer cells via death receptor (DR) activation with little toxicity to normal cells or tissues. The selectivity for activating apoptosis in cancer cells confers an ideal therapeutic characteristic to TRAIL, which has led to the development and clinical testing of many DR agonists. However, TRAIL/DR targeting therapies have been widely ineffective in clinical trials of various malignancies for reasons that remain poorly understood. Triple negative breast cancer (TNBC) has the worst prognosis among breast cancers. Targeting the TRAIL DR pathway has shown notable efficacy in a subset of TNBC in preclinical models but again has not shown appreciable activity in clinical trials. In this review, we will discuss the signaling components and mechanisms governing TRAIL pathway activation and clinical trial findings discussed with a focus on TNBC. Challenges and potential solutions for using DR agonists in the clinic are also discussed, including consideration of the pharmacokinetic and pharmacodynamic properties of DR agonists, patient selection by predictive biomarkers, and potential combination therapies. Moreover, recent findings on the impact of TRAIL treatment on the immune response, as well as novel strategies to address those challenges, are discussed.
Collapse
|
13
|
Alizadeh Zeinabad H, Szegezdi E. TRAIL in the Treatment of Cancer: From Soluble Cytokine to Nanosystems. Cancers (Basel) 2022; 14:5125. [PMID: 36291908 PMCID: PMC9600485 DOI: 10.3390/cancers14205125] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/23/2022] Open
Abstract
The death ligand tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF cytokine superfamily, has long been recognized for its potential as a cancer therapeutic due to its low toxicity against normal cells. However, its translation into a therapeutic molecule has not been successful to date, due to its short in vivo half-life associated with insufficient tumor accumulation and resistance of tumor cells to TRAIL-induced killing. Nanotechnology has the capacity to offer solutions to these limitations. This review provides a perspective and a critical assessment of the most promising approaches to realize TRAIL's potential as an anticancer therapeutic, including the development of fusion constructs, encapsulation, nanoparticle functionalization and tumor-targeting, and discusses the current challenges and future perspectives.
Collapse
Affiliation(s)
- Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Biomedical Sciences Building, School of Biological and Chemical Sciences, University of Galway, H91 W2TY Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, Biomedical Sciences Building, School of Biological and Chemical Sciences, University of Galway, H91 W2TY Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
14
|
Qin R, You FM, Zhao Q, Xie X, Peng C, Zhan G, Han B. Naturally derived indole alkaloids targeting regulated cell death (RCD) for cancer therapy: from molecular mechanisms to potential therapeutic targets. J Hematol Oncol 2022; 15:133. [PMID: 36104717 PMCID: PMC9471064 DOI: 10.1186/s13045-022-01350-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/03/2022] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) is a critical and active process that is controlled by specific signal transduction pathways and can be regulated by genetic signals or drug interventions. Meanwhile, RCD is closely related to the occurrence and therapy of multiple human cancers. Generally, RCD subroutines are the key signals of tumorigenesis, which are contributed to our better understanding of cancer pathogenesis and therapeutics. Indole alkaloids derived from natural sources are well defined for their outstanding biological and pharmacological properties, like vincristine, vinblastine, staurosporine, indirubin, and 3,3′-diindolylmethane, which are currently used in the clinic or under clinical assessment. Moreover, such compounds play a significant role in discovering novel anticancer agents. Thus, here we systemically summarized recent advances in indole alkaloids as anticancer agents by targeting different RCD subroutines, including the classical apoptosis and autophagic cell death signaling pathways as well as the crucial signaling pathways of other RCD subroutines, such as ferroptosis, mitotic catastrophe, necroptosis, and anoikis, in cancer. Moreover, we further discussed the cross talk between different RCD subroutines mediated by indole alkaloids and the combined strategies of multiple agents (e.g., 3,10-dibromofascaplysin combined with olaparib) to exhibit therapeutic potential against various cancers by regulating RCD subroutines. In short, the information provided in this review on the regulation of cell death by indole alkaloids against different targets is expected to be beneficial for the design of novel molecules with greater targeting and biological properties, thereby facilitating the development of new strategies for cancer therapy.
Collapse
|
15
|
Qian L, Vallega KA, Yao W, Wang D, Zhai Y, He X, Sun SY. Therapeutic potential of the novel Bcl-2/Bcl-X L dual inhibitor, APG1252, alone or in combination against non-small cell lung cancer. Mol Carcinog 2022; 61:1031-1042. [PMID: 36066010 DOI: 10.1002/mc.23458] [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: 07/04/2022] [Revised: 08/08/2022] [Accepted: 08/21/2022] [Indexed: 11/08/2022]
Abstract
Targeting the induction of apoptosis is a promising cancer therapeutic strategy with some clinical success. This study focused on evaluating the therapeutic efficacy of the novel Bcl-2/Bcl-XL dual inhibitor, APG1252-M1 (also named APG-1244; an in vivo active metabolite of APG1252 or pelcitoclax), as a single agent or in combination, against non-small cell lung cancer (NSCLC) cells. APG1252-M1 effectively decreased the survival of some NSCLC cell lines expressing low levels of Mcl-1 and induced apoptosis. Overexpression of ectopic Mcl-1 in the sensitive cells substantially compromised APG1252-M1's cell-killing effects, whereas inhibition of Mcl-1 greatly sensitized insensitive cell lines to APG1252-M1, indicating the critical role of Mcl-1 levels in impacting cell response to APG1252-M1. Moreover, APG1252-M1, when combined with the third generation epidermal growth factor receptor (EGFR) inhibitor, osimertinib, synergistically decreased the survival of EGFR-mutant NSCLC cell lines including those resistant to osimertinib with enhanced induction of apoptosis and abrogated emergence of acquired resistance to osimertinib. Importantly, the combination was effective in inhibiting the growth of osimertinib-resistant tumors in vivo. Collectively, these results demonstrate the efficacy of APG1252 alone or in combination against human NSCLC cells.
Collapse
Affiliation(s)
- Luxi Qian
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China.,Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Karin A Vallega
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Weilong Yao
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA.,Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Dongsheng Wang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co., Ltd, Suzhou, Jiangsu, P. R. China
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, P. R. China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Sevastre AS, Manea EV, Popescu OS, Tache DE, Danoiu S, Sfredel V, Tataranu LG, Dricu A. Intracellular Pathways and Mechanisms of Colored Secondary Metabolites in Cancer Therapy. Int J Mol Sci 2022; 23:ijms23179943. [PMID: 36077338 PMCID: PMC9456420 DOI: 10.3390/ijms23179943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022] Open
Abstract
Despite the great advancements made in cancer treatment, there are still many unsatisfied aspects, such as the wide palette of side effects and the drug resistance. There is an obvious increasing scientific attention towards nature and what it can offer the human race. Natural products can be used to treat many diseases, of which some plant products are currently used to treat cancer. Plants produce secondary metabolites for their signaling mechanisms and natural defense. A variety of plant-derived products have shown promising anticancer properties in vitro and in vivo. Rather than recreating the natural production environment, ongoing studies are currently setting various strategies to significantly manipulate the quantity of anticancer molecules in plants. This review focuses on the recently studied secondary metabolite agents that have shown promising anticancer activity, outlining their potential mechanisms of action and pathways.
Collapse
Affiliation(s)
- Ani-Simona Sevastre
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Elena Victoria Manea
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Oana Stefana Popescu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Daniela Elise Tache
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Suzana Danoiu
- Department of Pathophysiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Veronica Sfredel
- Department of Physiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Clinical Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania
- Correspondence: ; Tel.: +40-21-334-30-25
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 200349 Craiova, Romania
| |
Collapse
|
17
|
Yao W, Bai L, Wang S, Zhai Y, Sun SY. Mcl-1 levels critically impact the sensitivities of human colorectal cancer cells to APG-1252-M1, a novel Bcl-2/Bcl-X L dual inhibitor that induces Bax-dependent apoptosis. Neoplasia 2022; 29:100798. [PMID: 35462114 PMCID: PMC9046866 DOI: 10.1016/j.neo.2022.100798] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/08/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022]
Abstract
New treatment options, such as targeted therapies, are urgently needed for the treatment of colorectal cancer (CRC), the third leading cause of cancer-related deaths worldwide. The current study focuses on demonstrating the therapeutic efficacies of APG-1252-M1 (an active form of the prodrug, APG-1252 or pelcitoclax), a highly potent Bcl-2/Bcl-XL dual inhibitor in clinical trials, against CRC and understanding the underlying mechanisms. APG-1252-M1 effectively decreased the survival of CRC cell lines, particularly those expressing relatively low levels of Mcl-1, with the induction of apoptosis. High levels of Mcl-1 were significantly correlated with decreased sensitivity of CRC cell lines to APG-1252-M1. When combined with an Mcl-1 inhibitor, APG-1252-M1 synergistically decreased the survival and induced apoptosis of APG-1252-M1-insensitive cell lines with high levels of Mcl-1. This combination further decreased the survival and enhanced apoptosis even in sensitive cell lines with relatively low levels of Mcl-1, whereas enforced expression of ectopic Mcl-1 in these cells abrogated APG-1252-M1's effects on decreasing cell survival and inducing apoptosis, which could be reversed by Mcl-1 inhibition. APG-1252-M1 rapidly induced cytochrome C and Smac release from mitochondria with caspase-3 and PARP cleavage. Deficiency of Bax in CRC cells abolished APG-1252-M1's ability to induce apoptosis, indicating that APG-1252-M1 induces Bax-dependent apoptosis. The current study thus demonstrates the potential of APG-1252-M1 as a monotherapy in the treatment of CRC, particularly those with low Mcl-1 expression, or in combination with an Mcl-1 inhibitor, warranting further evaluation in vivo and in the clinic.
Collapse
Affiliation(s)
- Weilong Yao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China; Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road, C3088, Atlanta, GA 30322, USA
| | - Longchuan Bai
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yifan Zhai
- Ascentage Pharma (Suzhou) Co., Ltd, Suzhou, Jiangsu, PR China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road, C3088, Atlanta, GA 30322, USA.
| |
Collapse
|
18
|
Li Z, Han Z, Stenzel MH, Chapman R. A High Throughput Approach for Designing Polymers That Mimic the TRAIL Protein. NANO LETTERS 2022; 22:2660-2666. [PMID: 35312327 DOI: 10.1021/acs.nanolett.1c04469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We have leveraged a high throughput approach to design a fully synthetic polymer mimic of the chemotherapeutic protein "TRAIL". Our design enables the synthesis of libraries of star-shaped polymers presenting exactly one receptor binding peptide at the end of each arm with no purification steps. Clear structure-activity relationships in screening for receptor binding and the apoptotic activity on colon cancer lines (COLO205) led us to identify trivalent structures, ∼1.5 nm in hydrodynamic radius as the best mimics. These showed IC50 values ∼2 μM and resulted in the elevated levels of caspase-8 expected from this mechanism of cell death. Our results demonstrate the potential for HTP screening methods to be used in the design of polymers that can mimic a whole range of complex therapeutic proteins.
Collapse
Affiliation(s)
- Zihao Li
- Centre for Advanced Macromolecular Design, School of Chemistry, Univeristy of New South Wales Sydney, Kensington, New South Wales 2052, Australia
| | - Zifei Han
- Centre for Advanced Macromolecular Design, School of Chemistry, Univeristy of New South Wales Sydney, Kensington, New South Wales 2052, Australia
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, Univeristy of New South Wales Sydney, Kensington, New South Wales 2052, Australia
| | - Robert Chapman
- Centre for Advanced Macromolecular Design, School of Chemistry, Univeristy of New South Wales Sydney, Kensington, New South Wales 2052, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia
| |
Collapse
|
19
|
The tricks for fighting against cancer using CAR NK cells: A review. Mol Cell Probes 2022; 63:101817. [DOI: 10.1016/j.mcp.2022.101817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 01/07/2023]
|
20
|
Xu W, Fang F, Wang Y, Qin L, Han Y, Huang Y, Li B, Liu Y, Wang Z. Co-overexpression of TRAIL and Smac sensitizes MDA-MB-231 cells to radiation through apoptosis depending on mitochondrial pathway. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:37-48. [PMID: 35006369 DOI: 10.1007/s00411-021-00961-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Pro-apoptosis in cancer cells has been proposed as a beneficial therapeutic strategy for potentiating the anticancer effects of radiotherapy. TNF-related apoptosis inducing ligand (TRAIL) and Second mitochondria derived activator of caspase (Smac) can induce cell apoptosis. Herein, we designed a conditionally replicating adenoviral co-overexpression vector of TRAIL and Smac regulated by the Egr1 promoter, in which hTERT, E1A-E1B and E1B55K genes were inserted to achieve enhanced tumor targeting characteristics. After breast cancer MDA-MB-231 cells were infected and irradiated, cellular proliferation and colony formation were measured, apoptotic rate was detected by FCM after AnnexinV-FITC/PI staining. To explore the molecular mechanisms of apoptosis, mRNA and protein levels of TRAIL, Smac, Cytochrome c (Cyt c), death receptor 5 (DR5), caspase-8, -9 and -3 were measured by quantitative real-time PCR, ELISA and Western blot, and caspase-3 activity was detected using caspase-3 activity kits. The results showed that TRAIL and/or Smac overexpression enhanced proliferation inhibition and radio-sensitivity through apoptosis. In addition, the combination of IR and overexpression of TRAIL and/or Smac can activate more apoptosis in tumor cells, and the transcriptional levels and protein expressions of Cyt c, DR5, caspase-8, -9 and -3 had similar regularity with apoptotic changes, indicating the molecular mechanisms of TRAIL and Smac involves the mitochondrial pathway. Our findings may have implications for novel radiotherapy plans for breast tumor treatment.
Collapse
Affiliation(s)
- Weiqiang Xu
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Fang Fang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yuexuan Wang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Lijing Qin
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yu Han
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yuwei Huang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Bin Li
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China
| | - Yang Liu
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China.
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
21
|
Golinelli G, Grisendi G, Dall'Ora M, Casari G, Spano C, Talami R, Banchelli F, Prapa M, Chiavelli C, Rossignoli F, Candini O, D'Amico R, Nasi M, Cossarizza A, Casarini L, Dominici M. Anti-GD2 CAR MSCs against metastatic Ewing's sarcoma. Transl Oncol 2022; 15:101240. [PMID: 34649148 PMCID: PMC8517927 DOI: 10.1016/j.tranon.2021.101240] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Ewing's sarcoma (ES) is an aggressive cancer affecting children and young adults. We pre-clinically demonstrated that mesenchymal stromal/stem cells (MSCs) can deliver tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) against primary ES after local injection. However, ES is often metastatic calling for approaches able to support MSC targeting to the ES multiple remote sites. Considering that the disialoganglioside GD2 is expressed by ES and to optimise MSC tumour affinity, bi-functional (BF) MSCs expressing both TRAIL and a truncated anti-GD2 chimeric antigen receptor (GD2 tCAR) were generated and challenged against ES. METHODS The anti-GD2 BF MSCs delivering a soluble variant of TRAIL (sTRAIL) were tested in several in vitro ES models. Tumour targeting and killing by BF MSCs was further investigated by a novel immunodeficient ES metastatic model characterized by different metastatic sites, including lungs, liver and bone, mimicking the deadly clinical scenario. FINDINGS In vitro data revealed both tumour affinity and killing of BF MSCs. In vivo, GD2 tCAR molecule ameliorated the tumour targeting and persistence of BF MSCs counteracting ES in lungs but not in liver. INTERPRETATION We here generated data on the potential effects of BF MSCs within a complex ES metastatic in vivo model, exploring also the biodistribution of MSCs. Our BF MSC-based strategy promises to pave the way for potential improvements in the therapeutic delivery of TRAIL for the treatment of metastatic ES and other deadly GD2-positive malignancies.
Collapse
Affiliation(s)
- Giulia Golinelli
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy.
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; Rigenerand Srl, Medolla, Modena, Italy
| | | | - Giulia Casari
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | | | - Rebecca Talami
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Medical Statistic, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Malvina Prapa
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Chiavelli
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Filippo Rossignoli
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, United States of America; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Roberto D'Amico
- Center of Medical Statistic, Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy; National Institute for Cardiovascular Research - INRC, Bologna, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy; Rigenerand Srl, Medolla, Modena, Italy.
| |
Collapse
|
22
|
Wang BT, Kothambawala T, Wang L, Matthew TJ, Calhoun SE, Saini AK, Kotturi MF, Hernandez G, Humke EW, Peterson MS, Sinclair AM, Keyt BA. Multimeric Anti-DR5 IgM Agonist Antibody IGM-8444 Is a Potent Inducer of Cancer Cell Apoptosis and Synergizes with Chemotherapy and BCL-2 Inhibitor ABT-199. Mol Cancer Ther 2021; 20:2483-2494. [PMID: 34711645 PMCID: PMC9398157 DOI: 10.1158/1535-7163.mct-20-1132] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 07/07/2021] [Accepted: 09/15/2021] [Indexed: 01/07/2023]
Abstract
Death receptor 5 (DR5) is an attractive target for cancer therapy due to its broad upregulated expression in multiple cancers and ability to directly induce apoptosis. Though anti-DR5 IgG antibodies have been evaluated in clinical trials, limited efficacy has been attributed to insufficient receptor crosslinking. IGM-8444 is an engineered, multivalent agonistic IgM antibody with 10 binding sites to DR5 that induces cancer cell apoptosis through efficient DR5 multimerization. IGM-8444 bound to DR5 with high avidity and was substantially more potent than an IgG with the same binding domains. IGM-8444 induced cytotoxicity in a broad panel of solid and hematologic cancer cell lines but did not kill primary human hepatocytes in vitro, a potential toxicity of DR5 agonists. In multiple xenograft tumor models, IGM-8444 monotherapy inhibited tumor growth, with strong and sustained tumor regression observed in a gastric PDX model. When combined with chemotherapy or the BCL-2 inhibitor ABT-199, IGM-8444 exhibited synergistic in vitro tumor cytotoxicity and enhanced in vivo efficacy, without augmenting in vitro hepatotoxicity. These results support the clinical development of IGM-8444 in solid and hematologic malignancies as a monotherapy and in combination with chemotherapy or BCL-2 inhibition.
Collapse
Affiliation(s)
| | | | - Ling Wang
- IGM Biosciences Inc., Mountain View, California
| | | | | | | | | | | | | | | | | | - Bruce A Keyt
- IGM Biosciences Inc., Mountain View, California.
| |
Collapse
|
23
|
Kaempferol sensitizes tumor necrosis factor-related apoptosis-inducing ligand-resistance chronic myelogenous leukemia cells to apoptosis. Mol Biol Rep 2021; 49:19-29. [PMID: 34820749 DOI: 10.1007/s11033-021-06778-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The tumor necrosis factor (TNF)-related apoptosis-inducing ligand, TRAIL, an apoptosis-inducing cytokine, has attracted much attention in the treatment of cancer for its selective toxicity to malignant rather than normal cells. However, the apoptosis-inducing ability of TRAIL is weaker than expected primarily due to cancer cell resistance. As one of the dietary flavonoids, kaempferol, has been shown to be antiproliferative and might have a protective effect against TRAIL resistance, particularly for hematologic malignancies. METHODS AND RESULTS Here, we studied the potential of kaempferol to enhance the TRAIL-induced cytotoxicity and apoptosis in human chronic myelogenous leukemia (CML) cell line K-562, as well as the expression of specific genes with impact on TRAIL signal regulation. Analysis of flowcytometry data showed that treatment with kaempferol did enhance sensitivity of CML cells to pro-apoptotic effects of anti-TRAIL antibody. Although the gene expression levels were heterogeneous, cFLIP, cIAP1 and cIAP2 expression were generally downregulated where co-treatment of kaempferol and TRAIL was employed and these effects appeared to be dose-dependent. We further demonstrated that the expression of death receptors 4 and 5 tended to increase subsequent to the combination treatment. CONCLUSIONS Consequently, it is reasonable to conclude that sensitization of chronic leukemia cells to TRAIL by kaempferol in vitro should be considered as a way of focusing clinical attention on leukemia therapy.
Collapse
|
24
|
Crucial Role of Reactive Oxygen Species (ROS) for the Proapoptotic Effects of Indirubin Derivatives in Cutaneous SCC Cells. Antioxidants (Basel) 2021; 10:antiox10101514. [PMID: 34679649 PMCID: PMC8532942 DOI: 10.3390/antiox10101514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Efficient drugs are needed for countering the worldwide high incidence of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis. Indirubin derivatives represent promising candidates, but their effects in cSCC cells have not been reported before. Here, we investigated the efficacy of three indirubin derivatives (DKP-071, -073 and -184) in four cSCC cell lines. High efficacy was seen in SCL-I, SCL-II, SCC-12 and SCC-13, resulting in up to 80% loss of cell proliferation, 60% loss of cell viability and 30% induced apoptosis (10 µM). Apoptosis was further enhanced in combinations with TNF-related apoptosis-inducing ligand (TRAIL). Induction of reactive oxygen species (ROS) appeared as critical for these effects. Thus, antioxidative pretreatment completely abolished apoptosis as well as restored cell proliferation and viability. Concerning the pathways, complete activation of caspases cascades (caspases-3, -4, -6, -7, -8 and -9), loss of mitochondrial membrane potential, activation of proapoptotic PKCδ (protein kinase C delta), inhibition of STAT3 (signal transducer and activator of transcription 3), downregulation of antiapoptotic XIAP (X-linked inhibitor of apoptosis protein) and survivin as well as upregulation of the proapoptotic Bcl-2 protein Puma and the cell cycle inhibitor p21 were obtained. Importantly, all activation steps were prevented by antioxidants, thus proving ROS as a master regulator of indirubins' antitumor effects. ROS induction presently develops as an important issue in anticancer therapy.
Collapse
|
25
|
Sano E, Kazaana A, Tadakuma H, Takei T, Yoshimura S, Hanashima Y, Ozawa Y, Yoshino A, Suzuki Y, Ueda T. Interleukin-6 sensitizes TNF-α and TRAIL/Apo2L dependent cell death through upregulation of death receptors in human cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119037. [PMID: 33839168 DOI: 10.1016/j.bbamcr.2021.119037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Interleukin-6 (IL-6) enhanced TNF-α and TRAIL/Apo2L induced cell death in various human cancer cells derived from malignant glioma, melanoma, breast cancer and leukemia, although the effect was not detected with IL-6 alone. The effects of IL-6 using SKBR3 cells were associated with the generation of apoptotic cells as analyzed by fluorescence microscopy and flow cytometry. IL-6 activated p53 and upregulated TRAIL death receptors (DR-4 and DR-5) and stimulated the TNF-α and TRAIL dependent extrinsic apoptotic pathway without activation of the p53 mediated intrinsic apoptotic pathway. TNF-α and TRAIL induced cleavage of caspase-8 and caspase-3 was more enhanced by IL-6, although these caspases were not cleaved by IL-6 alone. The dead cell generation elicited by the combination with IL-6 was blocked by anti-human TRAIL R2/TNFRSF10B Fc chimera antibody which can neutralize the DR-5 mediated death signal. These findings indicate that IL-6 could contribute to the enhancement of TNF-α or TRAIL induced apoptosis through p53 dependent upregulation of DR-4 and DR-5. The data suggest that a favorable therapeutic interaction could occur between TNF-α or TRAIL and IL-6, and provide an experimental basis for rational clinical treatments in various cancers.
Collapse
Affiliation(s)
- Emiko Sano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan.
| | - Akira Kazaana
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Hisashi Tadakuma
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Toshiaki Takei
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Sodai Yoshimura
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yuya Hanashima
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoshinari Ozawa
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Atsuo Yoshino
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan; Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
26
|
High ROS Production by Celecoxib and Enhanced Sensitivity for Death Ligand-Induced Apoptosis in Cutaneous SCC Cell Lines. Int J Mol Sci 2021; 22:ijms22073622. [PMID: 33807213 PMCID: PMC8036359 DOI: 10.3390/ijms22073622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Incidence of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis has increased worldwide, and non-steroidal anti-inflammatory drugs as celecoxib are considered for treatment. We show here strong anti-proliferative effects of celecoxib in four cSCC cell lines, while apoptosis and cell viability largely remained unaffected. Impeded apoptosis was overcome in combinations with agonistic CD95 antibody or TNF-related apoptosis-inducing ligand (TRAIL), resulting in up to 60% apoptosis and almost complete loss of cell viability. Proapoptotic caspase cascades were activated, and apoptosis was suppressed by caspase inhibition. TRAIL receptor (DR5) and proapoptotic Bcl-2 proteins (Puma and Bad) were upregulated, while anti-apoptotic factors (survivin, XIAP, cFLIP, Mcl-1, and Bcl-w) were downregulated. Strongly elevated levels of reactive oxygen species (ROS) turned out as particularly characteristic for celecoxib, appearing already after 2 h. ROS production alone was not sufficient for apoptosis induction but may play a critical role in sensitizing cancer cells for apoptosis and therapy. Thus, the full therapeutic potential of celecoxib may be better used in combinations with death ligands. Furthermore, the immune response against cSCC/AK may be improved by celecoxib, and combinations with checkpoint inhibitors, recently approved for the treatment of cSCC, may be considered.
Collapse
|
27
|
Oh YT, Sun SY. Regulation of Cancer Metastasis by TRAIL/Death Receptor Signaling. Biomolecules 2021; 11:499. [PMID: 33810241 PMCID: PMC8065657 DOI: 10.3390/biom11040499] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/17/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Death ligands such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL; TNFSF10) and their corresponding death receptors (e.g., DR5) not only initiate apoptosis through activation of the extrinsic apoptotic pathway but also exert non-apoptotic biological functions such as regulation of inflammation and cancer metastasis. The involvement of the TRAIL/death receptor signaling pathway in the regulation of cancer invasion and metastasis is complex as both positive and negative roles have been reported. The underlying molecular mechanisms are even more complicated. This review will focus on discussing current knowledge in our understanding of the involvement of TRAIL/death receptor-mediated signaling in the regulation of cancer cell invasion and metastasis.
Collapse
Affiliation(s)
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA;
| |
Collapse
|
28
|
Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol 2021; 17:581-596. [PMID: 33401962 DOI: 10.2217/fon-2020-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, can induce apoptosis in cancer cells, sparing normal cells when bound to its associated death receptors (DR4/DR5). This unique mechanism makes TRAIL a potential anticancer therapeutic agent. However, clinical trials of recombinant TRAIL protein and TRAIL receptor agonist monoclonal antibodies have shown disappointing results due to its short half-life, poor pharmacokinetics and the resistance of the cancer cells. This review summarizes TRAIL-induced apoptotic and survival pathways as well as mechanisms leading to apoptotic resistance. Recent development of methods to overcome cancer cell resistance to TRAIL-induced apoptosis, such as protein modification, combination therapy and TRAIL-based gene therapy, appear promising. We also discuss the challenges and opportunities in the development of TRAIL-based therapies for the treatment of human cancers.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, 221005, India
| | - Gopeshwar Narayan
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
29
|
Tao Z, Liu Y, Yang H, Feng Y, Li H, Shi Q, Li S, Cheng J, Lu X. Customizing a Tridomain TRAIL Variant to Achieve Active Tumor Homing and Endogenous Albumin-Controlled Release of the Molecular Machine In Vivo. Biomacromolecules 2020; 21:4017-4029. [PMID: 32804484 DOI: 10.1021/acs.biomac.0c00785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive antitumor drug candidate for precision cancer therapy due to its superior selective cytotoxicity in a variety of tumor cells. However, the clinical application of TRAIL in cancer therapy has been limited by its poor tumor-homing capacities and short half-life. Herein, we designed a tridomain TRAIL variant, Z-ABD-TRAIL, by sequentially fusing the platelet-derived growth factor receptor beta (PDGFRβ)-specific affibody ZPDGFRβ and an albumin-binding domain (ABD) to the N-terminus of TRAIL. The fusion protein Z-ABD-TRAIL was produced as a soluble protein with high yield in Escherichia coli (E. coli). The ZPDGFRβ domain provided Z-ABD-TRAIL with PDGFRβ-binding properties and thus promoted its tumor homing via the engagement of PDGFRβ-expressing pericytes on tumor microvessels. ABD-mediated binding of Z-ABD-TRAIL to albumin in the blood endowed TRAIL with long-lasting (>72 h for Z-ABD-TRAIL vs <0.5 h for TRAIL) abilities to kill tumor cells. Although the in vitro cytotoxicity of Z-ABD-TRAIL in tumor cells was similar to that of the parent TRAIL, the in vivo tumor uptake, apoptosis-inducing ability, and antitumor effect of Z-ABD-TRAIL were much greater than those of TRAIL, indicating that ZPDGFRβ-mediated tumor homing and ABD-introduced albumin binding significantly improved the pharmacodynamics of TRAIL. In addition, repeated injection of high-dose Z-ABD-TRAIL showed no obvious acute toxicity in mice. These results demonstrate that the newly designed tridomain Z-ABD-TRAIL is a promising agent for precision cancer therapy.
Collapse
Affiliation(s)
- Ze Tao
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuehua Liu
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanru Feng
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Heng Li
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuxiao Shi
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengfu Li
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.,Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaofeng Lu
- Key Lab of Transplant Engineering and Immunology, MOH, Regenerative Medical Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.,Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
30
|
Muthu M, Chun S, Gopal J, Park GS, Nile A, Shin J, Shin J, Kim TH, Oh JW. The MUDENG Augmentation: A Genesis in Anti-Cancer Therapy? Int J Mol Sci 2020; 21:E5583. [PMID: 32759789 PMCID: PMC7432215 DOI: 10.3390/ijms21155583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/18/2022] Open
Abstract
Despite multitudes of reports on cancer remedies available, we are far from being able to declare that we have arrived at that defining anti-cancer therapy. In recent decades, researchers have been looking into the possibility of enhancing cell death-related signaling pathways in cancer cells using pro-apoptotic proteins. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and Mu-2/AP1M2 domain containing, death-inducing (MUDENG, MuD) have been established for their ability to bring about cell death specifically in cancer cells. Targeted cell death is a very attractive term when it comes to cancer, since most therapies also affect normal cells. In this direction TRAIL has made noteworthy progress. This review briefly sums up what has been done using TRAIL in cancer therapeutics. The importance of MuD and what has been achieved thus far through MuD and the need to widen and concentrate on applicational aspects of MuD has been highlighted. This has been suggested as the future perspective of MuD towards prospective progress in cancer research.
Collapse
Affiliation(s)
- Manikandan Muthu
- Department of Environmental Health Sciences, Konkuk University, Seoul 143-701, Korea; (M.M.); (S.C.); (J.G.)
| | - Sechul Chun
- Department of Environmental Health Sciences, Konkuk University, Seoul 143-701, Korea; (M.M.); (S.C.); (J.G.)
| | - Judy Gopal
- Department of Environmental Health Sciences, Konkuk University, Seoul 143-701, Korea; (M.M.); (S.C.); (J.G.)
| | - Gyun-Seok Park
- Department of Bioresources and Food Science, Konkuk University, Seoul 143-701, Korea; (G.-S.P.); (A.N.)
| | - Arti Nile
- Department of Bioresources and Food Science, Konkuk University, Seoul 143-701, Korea; (G.-S.P.); (A.N.)
| | - Jisoo Shin
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (J.S.); (J.S.)
| | - Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (J.S.); (J.S.)
| | - Tae-Hyoung Kim
- Department of Biochemistry, Chosun University School of Medicine, 309 Pilmoondaero, Dong-gu, Gwangju 501-759, Korea;
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea; (J.S.); (J.S.)
| |
Collapse
|
31
|
Elmallah MIY, Cogo S, Constantinescu AA, Elifio-Esposito S, Abdelfattah MS, Micheau O. Marine Actinomycetes-Derived Secondary Metabolites Overcome TRAIL-Resistance via the Intrinsic Pathway through Downregulation of Survivin and XIAP. Cells 2020; 9:cells9081760. [PMID: 32708048 PMCID: PMC7464567 DOI: 10.3390/cells9081760] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 01/03/2023] Open
Abstract
Resistance of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis represents the major hurdle to the clinical use of TRAIL or its derivatives. The discovery and development of lead compounds able to sensitize tumor cells to TRAIL-induced cell death is thus likely to overcome this limitation. We recently reported that marine actinomycetes’ crude extracts could restore TRAIL sensitivity of the MDA-MB-231 resistant triple negative breast cancer cell line. We demonstrate in this study, that purified secondary metabolites originating from distinct marine actinomycetes (sharkquinone (1), resistomycin (2), undecylprodigiosin (3), butylcyclopentylprodigiosin (4), elloxizanone A (5) and B (6), carboxyexfoliazone (7), and exfoliazone (8)), alone, and in a concentration-dependent manner, induce killing in both MDA-MB-231 and HCT116 cell lines. Combined with TRAIL, these compounds displayed additive to synergistic apoptotic activity in the Jurkat, HCT116 and MDA-MB-231 cell lines. Mechanistically, these secondary metabolites induced and enhanced procaspase-10, -8, -9 and -3 activation leading to an increase in PARP and lamin A/C cleavage. Apoptosis induced by these compounds was blocked by the pan-caspase inhibitor QvD, but not by a deficiency in caspase-8, FADD or TRAIL agonist receptors. Activation of the intrinsic pathway, on the other hand, is likely to explain both their ability to trigger cell death and to restore sensitivity to TRAIL, as it was evidenced that these compounds could induce the downregulation of XIAP and survivin. Our data further highlight that compounds derived from marine sources may lead to novel anti-cancer drug discovery.
Collapse
Affiliation(s)
- Mohammed I. Y. Elmallah
- LNC, INSERM, UMR1231, F-21079 Dijon, France; (S.C.); (A.A.C.)
- UFR Science de Santé, Université de Bourgogne Franche-Comté, F-21079 Dijon, France
- Chemistry Department, Faculty of Science, Helwan University, 11795 Ain Helwan, Cairo 11795, Egypt;
- Correspondence: (M.I.Y.E.); (O.M.)
| | - Sheron Cogo
- LNC, INSERM, UMR1231, F-21079 Dijon, France; (S.C.); (A.A.C.)
- UFR Science de Santé, Université de Bourgogne Franche-Comté, F-21079 Dijon, France
- Graduate Programme in Health Sciences, Pontifícia Universidade Catolica do Parana, Curitiba 80215–901, Parana, Brazil;
| | - Andrei A. Constantinescu
- LNC, INSERM, UMR1231, F-21079 Dijon, France; (S.C.); (A.A.C.)
- UFR Science de Santé, Université de Bourgogne Franche-Comté, F-21079 Dijon, France
| | - Selene Elifio-Esposito
- Graduate Programme in Health Sciences, Pontifícia Universidade Catolica do Parana, Curitiba 80215–901, Parana, Brazil;
| | - Mohammed S. Abdelfattah
- Chemistry Department, Faculty of Science, Helwan University, 11795 Ain Helwan, Cairo 11795, Egypt;
- Marine Natural Products Unit (MNPRU), Faculty of Science, Helwan University, 11795 Ain Helwan, Cairo 11795, Egypt
| | - Olivier Micheau
- LNC, INSERM, UMR1231, F-21079 Dijon, France; (S.C.); (A.A.C.)
- UFR Science de Santé, Université de Bourgogne Franche-Comté, F-21079 Dijon, France
- Correspondence: (M.I.Y.E.); (O.M.)
| |
Collapse
|
32
|
Liang R, Yao Y, Wang G, Yue E, Yang G, Qi X, Wang Y, Zhao L, Zheng T, Zhang Y, Wenge Wang E. Repositioning Quinacrine Toward Treatment of Ovarian Cancer by Rational Combination With TRAIL. Front Oncol 2020; 10:1118. [PMID: 32766144 PMCID: PMC7379129 DOI: 10.3389/fonc.2020.01118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/04/2020] [Indexed: 11/21/2022] Open
Abstract
Quinacrine has been identified as a potent DR5-inducing agent that sensitizes cancer cells to TRAIL-induced apoptosis. In the current study, we found that quinacrine increased DR5 mRNA levels significantly in ovarian cancer cell lines regardless of p53 status. Further study showed the half-life of DR5 in quinacrine-treated cells was significantly prolonged, indicating that DR5 protein degradation was inhibited by quinacrine. We tested if the combination of TRAIL and quinacrine could be effective in ovarian cancer treatment in vitro and in ovarian cancer xenograft mouse models. We found that quinacrine enhanced TRAIL sensitivity or reversed TRAIL resistance in all the ovarian cancer cell lines tested. Mice treated with quinacrine and TRAIL remained disease-free for up to 20 weeks, however, mice treated with TRAIL or quinacrine alone and in control group died within ~8 weeks after treatment. Intraperitoneal delivery of quinacrine and TRAIL is rational and practical with extraordinary synergistic anti-cancer effects in preclinical models of ovarian cancer. Clinical investigation of combining quinacrine with TRAIL for ovarian cancer treatment is warranted.
Collapse
Affiliation(s)
- Rui Liang
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States.,Department of Pharmacy, Suzhou Vocational Health College, Suzhou, China
| | - Yuanfei Yao
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States.,Cancer Hospital, Harbin Medical University, Harbin, China
| | - Guangyu Wang
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States.,Cancer Hospital, Harbin Medical University, Harbin, China
| | - Er Yue
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Guangchao Yang
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Xiuying Qi
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Yang Wang
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Ling Zhao
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| | - Tongsen Zheng
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States.,Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yanqiao Zhang
- Cancer Hospital, Harbin Medical University, Harbin, China
| | - Edward Wenge Wang
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA, United States
| |
Collapse
|
33
|
Han Y, Zou R, Xiang L, Chen C, Cai C. Engineering a label- and enzyme-free detection of HIV-DNA on a cyclic DNA self-assembling strategy using G-triplexes as the signal reporter. Microchem J 2020. [DOI: 10.1016/j.microc.2020.104656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
34
|
Genetically Modified DR5-Specific TRAIL Variant DR5-B Revealed Dual Antitumor and Protumoral Effect in Colon Cancer Xenografts and an Improved Pharmacokinetic Profile. Transl Oncol 2020; 13:100762. [PMID: 32224450 PMCID: PMC7110358 DOI: 10.1016/j.tranon.2020.100762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 11/25/2022] Open
Abstract
Despite the weak clinical efficacy of TRAIL death receptor agonists, a search is under way for new agents that more efficiently activate apoptotic signaling. We previously created a TRAIL DR5-selective variant DR5-B without affinity for the DR4, DcR1, DcR2, and OPG receptors and increased proapoptotic activity in tumor cells. Here we showed that DR5-B significantly inhibited tumor growth in HCT116 and Caco-2 but not in HT-29 xenografts. The antitumor activity of DR5-B was 2.5 times higher in HCT116 xenografts compared to TRAIL. DR5-B at a dose of 2 or 10 mg/kg/d for 10 days inhibited tumor growth in HCT116 xenografts by 26% or 50% respectively, and increased animal survival. Unexpectedly, DR5-B at a higher dose (25 mg/kg/d) inhibited tumor growth only during the first 8 days of drug exposure, while at the end of the monitoring, no effect or even slight stimulation of tumor growth was observed. The pharmacokinetic parameters of DR5-B were comparable to those of TRAIL, except that the half-life was 3.5 times higher. Thus, enhancing TRAIL selectivity to DR5 may increase both antitumor and proliferative activities depending on the concentration and administration regimens.
Collapse
|
35
|
Yoshimura S, Sano E, Hanashima Y, Yamamuro S, Sumi K, Ueda T, Nakayama T, Hara H, Yoshino A, Katayama Y. IFN‑β sensitizes TRAIL‑induced apoptosis by upregulation of death receptor 5 in malignant glioma cells. Oncol Rep 2019; 42:2635-2643. [PMID: 31638255 PMCID: PMC6859459 DOI: 10.3892/or.2019.7383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor necrosis factor‑related apoptosis‑inducing ligand (TRAIL), a member of the tumor necrosis factor (TNF) family, induces apoptosis in cancer cells by binding to its receptors, death receptor 4 (DR4) and DR5, without affecting normal cells, and is therefore considered to be a promising antitumor agent for use in cancer treatment. However, several studies have indicated that most glioma cell lines display resistance to TRAIL‑induced apoptosis. To overcome such resistance and to improve the efficacy of TRAIL‑based therapies, identification of ideal agents for combinational treatment is important for achieving rational clinical treatment in glioblastoma patients. The main aim of this study was to investigate whether interferon‑β (IFN‑β) (with its pleiotropic antitumor activities) could sensitize malignant glioma cells to TRAIL‑induced apoptosis using glioma cell lines. TRAIL exhibited a dose‑dependent antitumor effect in all of the 7 types of malignant glioma cell lines, although the intensity of the effect varied among the cell lines. In addition, combined treatment with TRAIL (low clinical dose: 1 ng/ml) and IFN‑β (clinically relevant concentration: 10 IU/ml) in A‑172, AM‑38, T98G, U‑138MG and U‑251MG demonstrated a more marked antitumor effect than TRAIL alone. Furthermore, the antitumor effect of the combined treatment with TRAIL and IFN‑β may be enhanced via an extrinsic apoptotic system, and upregulation of DR5 was revealed to play an important role in this process in U‑138MG cells. These findings provide an experimental basis to suggest that combined treatment with TRAIL and IFN‑β may offer a new therapeutic strategy for malignant gliomas.
Collapse
Affiliation(s)
- Sodai Yoshimura
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Emiko Sano
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Yuya Hanashima
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shun Yamamuro
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Koichiro Sumi
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Tomohiro Nakayama
- Division of Companion Diagnostics, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroyuki Hara
- Division of Functional Morphology, Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Atsuo Yoshino
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoichi Katayama
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
- Center for Brain and Health Science, Aomori University, Aomori 030-8505, Japan
| |
Collapse
|
36
|
Moyer TJ, Chen F, Toft DJ, Ruff Y, Cryns VL, Stupp SI. Self-assembled peptide nanostructures targeting death receptor 5 and encapsulating paclitaxel as a multifunctional cancer therapy. ACS Biomater Sci Eng 2019; 5:6046-6053. [PMID: 33304996 PMCID: PMC7725269 DOI: 10.1021/acsbiomaterials.9b01259] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of tumor-targeted nanoscale carriers for the delivery of cancer therapeutics offers the ability to increase efficacy while limiting off-target toxicity. In this work we focused on targeting death receptor 5 (DR5), which is highly expressed by cancer cells, and upon binding, triggers programmed cell death. Hence, a nanostructure targeting DR5 would act as a dual targeting and therapeutic agent. We report here on a peptide amphiphile (PA) containing a dimeric, cyclic peptide that self-assembles into cylindrical supramolecular nanofibers and targets DR5. Coassembly of the DR5-targeting PA and a pegylated PA creates a supramolecular nanoscale construct with enhanced binding affinity to DR5 relative to a monomeric targeting PA, and was found to be cytotoxic in vitro. When combined with the chemotherapy paclitaxel, DR5-targeting carriers showed potent antitumor activity in vivo, demonstrating the multifunctional capabilities of peptide-based supramolecular nanostructures.
Collapse
Affiliation(s)
- Tyson J Moyer
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611
- Department of Materials Science, Northwestern University, Evanston, Illinois 60208
| | - Feng Chen
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Daniel J Toft
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yves Ruff
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208
| | - Vincent L Cryns
- Department of Medicine, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | - Samuel I Stupp
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611
- Department of Materials Science, Northwestern University, Evanston, Illinois 60208
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
37
|
Ghaemdoust F, Keshavarz-Fathi M, Rezaei N. Natural killer cells and cancer therapy, what we know and where we are going. Immunotherapy 2019; 11:1231-1251. [DOI: 10.2217/imt-2019-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells are among the significant components of innate immune system and they have come to the first line of defense against tumor cells developing inside the body. CD56lo/CD16+ NK cells are highly cytotoxic and CD56hi NK cells can produce cytokines and perform a regulatory function. Specific features of NK cells have made them a unique choice for cancer immunotherapy. Simple interventions like cytokine-injection to boost the internal NK cells were the first trials to target these cells. Nowadays, many other types of intervention are under investigation, such as adoptive NK cell immunotherapy. In this paper, we will discuss the biology and function of NK cells in cancer immunosurveillance and therapeutic approaches against cancer via using NK cells.
Collapse
Affiliation(s)
- Faezeh Ghaemdoust
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| |
Collapse
|
38
|
Xu Y, Liu D, Hu J, Ding P, Chen M. Hyaluronic acid-coated pH sensitive poly (β-amino ester) nanoparticles for co-delivery of embelin and TRAIL plasmid for triple negative breast cancer treatment. Int J Pharm 2019; 573:118637. [PMID: 31550511 DOI: 10.1016/j.ijpharm.2019.118637] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 02/04/2023]
Abstract
Triple negative breast cancer (TNBC) still lacks an effective targeted treatment. In this study, hyaluronic acid (HA)-mediated tumor targeting and pH-sensitive amphiphilic polymeric nanoparticles were designed and prepared to co-deliver the anticancer drug embelin (EMB) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plasmid (pTRAIL) (EMB/TRAIL-HA/PBAE-PEI) for synergistic anti-breast cancer efficacy. These pH-sensitive amphiphilic polymeric nanoparticles were formed using the amphiphilic polymers polyethyleneimine (PEI)-poly[(1,6-hexanediol)-diacrylate-β-5-hydroxyamylamine] (PBAE), which was synthesized via Michael addition polymerization. Taking advantage of the specific binding between HA and CD44, which is highly expressed in MDA-MB-231 TNBC cells, the HA-coated nanoparticles increased drug uptake in MDA-MB-231 TNBC cells compared with MCF-7 non-TNBC cells with lower CD44 expression. Moreover, EMB/TRAIL-HA/PBAE-PEI exhibited enhanced cytotoxic and pro-apoptotic effects against MDA-MB-231 cells compared with free EMB and EMB- or pTRAIL-loaded nanoparticles via activation of caspase 3/7, an increase in reactive oxygen species levels, and inhibition of the expressions of apoptosis-related proteins. These results demonstrated that EMB/TRAIL-HA/PBAE-PEI exerted enhanced cytotoxic and pro-apoptotic effects against MDA-MB-231 cells and showed great potential for TNBC treatment.
Collapse
Affiliation(s)
- Yingqi Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Dingxin Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Jie Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau
| | - Peirong Ding
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
39
|
Kocik J, Machula M, Wisniewska A, Surmiak E, Holak TA, Skalniak L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers (Basel) 2019; 11:E1014. [PMID: 31331108 PMCID: PMC6678622 DOI: 10.3390/cancers11071014] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023] Open
Abstract
The protein p53, known as the "Guardian of the Genome", plays an important role in maintaining DNA integrity, providing protection against cancer-promoting mutations. Dysfunction of p53 is observed in almost every cancer, with 50% of cases bearing loss-of-function mutations/deletions in the TP53 gene. In the remaining 50% of cases the overexpression of HDM2 (mouse double minute 2, human homolog) protein, which is a natural inhibitor of p53, is the most common way of keeping p53 inactive. Disruption of HDM2-p53 interaction with the use of HDM2 antagonists leads to the release of p53 and expression of its target genes, engaged in the induction of cell cycle arrest, DNA repair, senescence, and apoptosis. The induction of apoptosis, however, is restricted to only a handful of p53wt cells, and, generally, cancer cells treated with HDM2 antagonists are not efficiently eliminated. For this reason, HDM2 antagonists were tested in combinations with multiple other therapeutics in a search for synergy that would enhance the cancer eradication. This manuscript aims at reviewing the recent progress in developing strategies of combined cancer treatment with the use of HDM2 antagonists.
Collapse
Affiliation(s)
- Justyna Kocik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Monika Machula
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Aneta Wisniewska
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewa Surmiak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Tad A Holak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Lukasz Skalniak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
40
|
de Looff M, de Jong S, Kruyt FAE. Multiple Interactions Between Cancer Cells and the Tumor Microenvironment Modulate TRAIL Signaling: Implications for TRAIL Receptor Targeted Therapy. Front Immunol 2019; 10:1530. [PMID: 31333662 PMCID: PMC6617985 DOI: 10.3389/fimmu.2019.01530] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/19/2019] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) signaling is far more complex than initially anticipated and can lead to either anti- or protumorigenic effects, hampering the successful clinical use of therapeutic TRAIL receptor agonists. Cell autonomous resistance mechanisms have been identified in addition to paracrine factors that can modulate apoptosis sensitivity. The tumor microenvironment (TME), consisting of cellular and non-cellular components, is a source for multiple signals that are able to modulate TRAIL signaling in tumor and stromal cells. Particularly immune effector cells, also part of the TME, employ the TRAIL/TRAIL-R system whereby cell surface expressed TRAIL can activate apoptosis via TRAIL receptors on tumor cells, which is part of tumor immune surveillance. In this review we aim to dissect the impact of the TME on signaling induced by endogenous and exogenous/therapeutic TRAIL, thereby distinguishing different components of the TME such as immune effector cells, neutrophils, macrophages, and non-hematopoietic stromal cells. In addition, also non-cellular biochemical and biophysical properties of the TME are considered including mechanical stress, acidity, hypoxia, and glucose deprivation. Available literature thus far indicates that tumor-TME interactions are complex and often bidirectional leading to tumor-enhancing or tumor-reducing effects in a tumor model- and tumor type-dependent fashion. Multiple signals originating from different components of the TME simultaneously affect TRAIL receptor signaling. We conclude that in order to unleash the full clinical potential of TRAIL receptor agonists it will be necessary to increase our understanding of the contribution of different TME components on outcome of therapeutic TRAIL receptor activation in order to identify the most critical mechanism responsible for resistance, allowing the design of effective combination treatments.
Collapse
Affiliation(s)
- Margot de Looff
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
41
|
Su XY, Zhao JQ, Li N, Kumar M, yang AMO. Chemoprotective Effects of Resveratrol Against Diethylnitrosamine Induced Hepatocellular Carcinoma in Wistar Rats. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.549.559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
42
|
Wang JR, Shen GN, Luo YH, Piao XJ, Zhang Y, Wang H, Li JQ, Xu WT, Zhang Y, Wang SN, Zhang T, Xue H, Cao LK, Jin CH. 2-(4-methoxyphenylthio)-5,8-dimethoxy-1,4-naphthoquinone induces apoptosis via ROS-mediated MAPK and STAT3 signaling pathway in human gastric cancer cells. J Chemother 2019; 31:214-226. [PMID: 31074342 DOI: 10.1080/1120009x.2019.1610832] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The 1,4-naphthoquinones and their derivatives have garnered great interest due to their antitumor pharmacological properties in various cancers; however, their clinical application is limited by side effects. In this study, to reduce side effects and improve therapeutic efficacy, a novel 1,4-naphthoquinone derivative-2-(4-methoxyphenylthio)-5,8-dimethoxy-1,4-naphthoquinone (MPTDMNQ) was synthesized. We investigated the effects and underlying mechanisms of MPTDMNQ on cell viability, apoptosis, and reactive oxygen species (ROS) generation in human gastric cancer cells. Our results showed that MPTDMNQ decreased cell viability in nine human gastric cancer cell lines. MPTDMNQ significantly induced apoptosis accompanied by the accumulation of ROS in GC cells. However, pre-treatment with the ROS scavenger N-acetyl-L-cysteine (NAC) attenuated the MPTDMNQ-induced apoptosis. Moreover, MPTDMNQ decreased the phosphorylation levels of extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription 3 (STAT3); and increased the phosphorylation levels of c-Jun N-terminal kinase (JNK) and p38 kinase. However, phosphorylation was inhibited by NAC and a mitogen-activated protein kinase (MAPK) inhibitor. These findings showed that MPTDMNQ induced AGS cell apoptosis via ROS-mediated MAPK and STAT3 signaling pathways. Thus, MPTDMNQ may be a promising candidate for treating gastric cancer.
Collapse
Affiliation(s)
- Jia-Ru Wang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Gui-Nan Shen
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Ying-Hua Luo
- b College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Xian-Ji Piao
- c Department of Gynaecology and Obstetrics , The Fifth Affiliated Hospital of Harbin Medical University , Daqing , China
| | - Yi Zhang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Hao Wang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Jin-Qian Li
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Wan-Ting Xu
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Yu Zhang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Shi-Nong Wang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Tong Zhang
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Hui Xue
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Long-Kui Cao
- d College of Food Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| | - Cheng-Hao Jin
- a Department of Biochemistry and Molecular Biology , College of Life Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China.,d College of Food Science & Technology, Heilongjiang Bayi Agricultural University , Daqing , China
| |
Collapse
|
43
|
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can initiate the apoptosis pathway by binding to its associated death receptors DR4 and DR5. The activation of the TRAIL pathway in inducing tumor-selective apoptosis leads to the development of TRAIL-based cancer therapies, which include recombinant forms of TRAIL, TRAIL receptor agonists, and other therapeutic agents. Importantly, TRAIL, DR4, and DR5 can all be induced by synthetic and natural agents that activate the TRAIL apoptosis pathway in cancer cells. Thus, understanding the regulation of the TRAIL apoptosis pathway can aid in the development of TRAIL-based therapies for the treatment of human cancer.
Collapse
|
44
|
Han Y, Zhang F, Gong H, Cai C. Multifunctional G-quadruplex-based fluorescence probe coupled with DNA-templated AgNCs for simultaneous detection of multiple DNAs and MicroRNAs. Anal Chim Acta 2019; 1053:105-113. [DOI: 10.1016/j.aca.2018.11.062] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
|
45
|
Poondla N, Chandrasekaran AP, Heese K, Kim KS, Ramakrishna S. CRISPR-mediated upregulation of DR5 and downregulation of cFLIP synergistically sensitize HeLa cells to TRAIL-mediated apoptosis. Biochem Biophys Res Commun 2019; 512:60-65. [PMID: 30862357 DOI: 10.1016/j.bbrc.2019.03.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/03/2019] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has received attention as an anticancer therapy because it mediates apoptosis of several cancer cell types but not normal human cell types. In this study, we implemented genome editing techniques to upregulate DR5 and downregulate cFLIP in HeLa cells to stimulate TRAIL-induced apoptosis. We designed and validated sgRNAs to enrich the endogenous level of DR5 by dead Cas9 (dCas9). Similarly, we designed two sgRNAs to disrupt the cFLIP gene by CRISPR/Cas9. We analyzed the effect of TRAIL on tumor cells by co-transfecting HeLa cells with the best combinations of sgRNAs regulating DR5 and cFLIP genes. TRAIL-induced apoptosis in HeLa cells was evaluated by the γH2AX foci formation assay to check for double-strand break and propidium iodide and Annexin V staining to quantify apoptotic cells. Viable cells were identified by CCK-8 assay, and cleaved-PARP level was evaluated by Western blot. This is the first study to demonstrate that genome editing techniques can be used as an effective combinatorial treatment strategy to induce apoptosis of cancer cells. In particular, enhancement of DR5 expression and inhibition of cFLIP expression by genome editing had a synergistic effect of inhibiting proliferation and inducing apoptosis in TRAIL-resistant HeLa cells. These results suggest that combinatorial treatment strategies mediated by the CRISPR/Cas9 system may be effective for design of other human TRAIL-resistant cell types.
Collapse
Affiliation(s)
- Naresh Poondla
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | | | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| |
Collapse
|
46
|
Dubuisson A, Favreau C, Fourmaux E, Lareure S, Rodrigues-Saraiva R, Pellat-Deceunynck C, El Alaoui S, Micheau O. Generation and characterization of novel anti-DR4 and anti-DR5 antibodies developed by genetic immunization. Cell Death Dis 2019; 10:101. [PMID: 30718507 PMCID: PMC6362131 DOI: 10.1038/s41419-019-1343-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022]
Abstract
Development of therapeutic antibodies in oncology has attracted much interest in the past decades. More than 30 of them have been approved and are being used to treat patients suffering from cancer. Despite encouraging results, and albeit most clinical trials aiming at evaluating monoclonal antibodies directed against TRAIL agonist receptors have been discontinued, DR4 or DR5 remain interesting targets, since these receptors are overexpressed by tumour cells and are able to trigger their death. In an effort to develop novel and specific anti-DR4 and anti-DR5 antibodies with improved properties, we used genetic immunization to express native proteins in vivo. Injection of DR4 and DR5 cDNA into the tail veins of mice elicited significant humoral anti-DR4 and anti-DR5 responses and fusions of the corresponding spleens resulted in numerous hybridomas secreting antibodies that could specifically recognize DR4 or DR5 in their native forms. All antibodies bound specifically to their targets with a very high affinity, from picomolar to nanomolar range. Among the 21 anti-DR4 and anti-DR5 monoclonal antibodies that we have produced and purified, two displayed proapoptotic properties alone, five induced apoptosis after cross-linking, four were found to potentiate TRAIL-induced apoptosis and three displayed antiapoptotic potential. The most potent anti-DR4 antibody, C#16, was assessed in vivo and was found, alone, to inhibit tumour growth in animal models. This is the first demonstration that DNA-based immunization method can be used to generate novel monoclonal antibodies targeting receptors of the TNF superfamily that may constitute new therapeutic agents.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Female
- HEK293 Cells
- Humans
- Immunization
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists
- Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Agathe Dubuisson
- Université Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079, Dijon, France
- Research Department, CovalAb, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France
- INSERM, UMR1231, « Equipe labellisée Ligue contre le Cancer » and Laboratoire d'Excellence LipSTIC, F-21079, Dijon, France
| | - Cécile Favreau
- Université Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079, Dijon, France
- INSERM, UMR1231, « Equipe labellisée Ligue contre le Cancer » and Laboratoire d'Excellence LipSTIC, F-21079, Dijon, France
| | - Eric Fourmaux
- Research Department, CovalAb, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France
| | - Sabrina Lareure
- Research Department, CovalAb, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France
| | - Rafael Rodrigues-Saraiva
- Université Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079, Dijon, France
- INSERM, UMR1231, « Equipe labellisée Ligue contre le Cancer » and Laboratoire d'Excellence LipSTIC, F-21079, Dijon, France
| | | | - Said El Alaoui
- Research Department, CovalAb, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France
| | - Olivier Micheau
- Université Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079, Dijon, France.
- Research Department, CovalAb, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, « Equipe labellisée Ligue contre le Cancer » and Laboratoire d'Excellence LipSTIC, F-21079, Dijon, France.
| |
Collapse
|
47
|
Kamalabadi-Farahani M, Vasei M, Ahmadbeigi N, Ebrahimi-Barough S, Soleimani M, Roozafzoon R. Anti-tumour effects of TRAIL-expressing human placental derived mesenchymal stem cells with curcumin-loaded chitosan nanoparticles in a mice model of triple negative breast cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1011-S1021. [DOI: 10.1080/21691401.2018.1527345] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Mohammad Kamalabadi-Farahani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Vasei
- Department of Pathology, Molecular and Cell Biology Laboratory, Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Naser Ahmadbeigi
- Cell Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Tarbiat Modares University, Tehran, Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
48
|
Kamalabadi-Farahani M, Vasei M, Ahmadbeigi N, Ebrahimi-Barough S, Soleimani M, Roozafzoon R. Anti-tumour effects of TRAIL-expressing human placental derived mesenchymal stem cells with curcumin-loaded chitosan nanoparticles in a mice model of triple negative breast cancer. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2018. [DOI: https://doi.org/10.1080/21691401.2018.1527345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Mohammad Kamalabadi-Farahani
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Vasei
- Department of Pathology, Molecular and Cell Biology Laboratory, Shariati Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Naser Ahmadbeigi
- Cell Based Therapies Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Tarbiat Modares University, Tehran, Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
49
|
Brin E, Wu K, Dagostino E, Meng-Chiang Kuo M, He Y, Shia WJ, Chen LC, Stempniak M, Hickey R, Almassy R, Showalter R, Thomson J. TRAIL stabilization and cancer cell sensitization to its pro-apoptotic activity achieved through genetic fusion with arginine deiminase. Oncotarget 2018; 9:36914-36928. [PMID: 30651925 PMCID: PMC6319333 DOI: 10.18632/oncotarget.26398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) binds to death receptors and induces apoptosis in various cancer cell lines while sparing normal cells. Recombinant TRAIL has shown good safety and efficacy profiles in preclinical cancer models. However, clinical success has been limited due to poor PK and development of resistance to death receptor-induced apoptosis. We have addressed these issues by creating a fusion protein of TRAIL and arginine deiminase (ADI). The fusion protein benefits from structural and functional synergies between its two components and has an extended half-life in vivo. ADI downregulates survivin, upregulates DR5 receptor and sensitizes cancer cells to TRAIL induced apoptosis. ADI-TRAIL fusion protein was efficacious in a number of cell lines and synergized with some standard of care drugs. In an HCT116 xenograft model ADI-TRAIL localized to the tumor and induced dose-dependent tumor regression, the fusion protein was superior to rhTRAIL administered at the same molar amounts.
Collapse
Affiliation(s)
- Elena Brin
- Polaris Pharmaceuticals, San Diego, CA, USA
| | | | | | | | - Yudou He
- Polaris Pharmaceuticals, San Diego, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Twomey JD, Zhao L, Luo S, Xu Q, Zhang B. Tubulin couples death receptor 5 to regulate apoptosis. Oncotarget 2018; 9:36804-36815. [PMID: 30613368 PMCID: PMC6298406 DOI: 10.18632/oncotarget.26407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 11/16/2018] [Indexed: 12/30/2022] Open
Abstract
Activation of death receptor 5 (DR5) to induce apoptosis in cancer cells is an attractive strategy for cancer therapy. However, many tumor cell lines and primary tumors are resistant to DR5 targeted agents including recombinant tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and anti-DR5 agonistic antibodies. Here we identify tubulin proteins - primarily consisting of α and β subunits folded into microtubule polymers - as a crucial modulator of DR5 mediated apoptosis. Using affinity purification coupled with mass spectrometry, we found that DR5 interacts with both α- and β-tubulin proteins in cancer cells. Pharmacological disruption of microtubules increased DR5 protein expression and subsequently sensitized the cells to TRAIL-induced apoptosis. Similar results were observed by selectively silencing tubulin transcript using small RNA interference. We also demonstrate that tubulin/microtubule blockade augments TRAIL induced apoptosis by stabilizing DR5 protein. Together, our results link the tubulin/microtubule network to the stringent regulation of DR5 mediated apoptosis, which could lead to potential therapeutic strategies to enhance cancer therapy efficacy.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Liqun Zhao
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Shen Luo
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Qing Xu
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|