1
|
Jang JH, Kim H, Jun H, Park CY, Kim JY, Yeo M, Kim H, Shin Y, Kang S, Kim E, Lee TJ. Targeting RBM39 with Tasisulam enhances TRAIL-induced apoptosis through DR5 upregulation and Bcl-2 downregulation in renal cell carcinoma. Biochem Pharmacol 2025; 236:116877. [PMID: 40112928 DOI: 10.1016/j.bcp.2025.116877] [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: 01/06/2025] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in tumor cells but faces limitations due to resistance mechanisms involving anti-apoptotic regulators such as Bcl-2. This study investigates Tasisulam, a molecular glue degrader, that degrades RNA-binding motif protein 39 (RBM39), as a sensitizer for TRAIL-mediated apoptosis in renal cell carcinoma (RCC). Tasisulam enhances TRAIL-induced apoptosis by activating both extrinsic and intrinsic apoptotic pathways, achieved through upregulation of death receptor 5 (DR5) and downregulation of B-cell lymphoma 2 (Bcl-2). Importantly, Tasisulam selectively sensitizes RCC cells to TRAIL-induced apoptosis without affecting normal cells.RBM39 knockdown mimicked the effects of Tasisulam by upregulating DR5, downregulating Bcl-2, and enhancing TRAIL-induced apoptosis, suggesting RBM39 as a critical regulator of these pathways. To address TRAIL instability in vivo, AaLS/TRAIL nanoparticles were employed in combination with Tasisulam in a Caki-1 xenograft model. This combination significantly reduced tumor volume and weight compared to single treatments, without observed toxicity. These findings demonstrate that Tasisulam sensitizes RCC cells to TRAIL-induced apoptosis through RBM39-dependent DR5 upregulation and Bcl-2 downregulation. This combination strategy holds significant promise as a potential solution to overcoming TRAIL resistance and advancing more effective treatment outcomes for RCC.
Collapse
Affiliation(s)
- Ji Hoon Jang
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Haein Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heejin Jun
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Cho-Young Park
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Joo-Young Kim
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Mirae Yeo
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hunmin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yerim Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eunhee Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-Gu, Daegu 42415, Republic of Korea.
| |
Collapse
|
2
|
Oh YT, Chen Z, Wang D, Ramalingam SS, Sun SY. Induction of IL6/STAT3-dependent TRAIL expression that contributes to the therapeutic efficacy of osimertinib in EGFR mutant NSCLC cells. Oncogene 2025:10.1038/s41388-025-03381-5. [PMID: 40247082 DOI: 10.1038/s41388-025-03381-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/19/2025]
Abstract
The third-generation mutation-selective EGFR tyrosine kinase inhibitor (EGFR-TKI) osimertinib (or AZD9291) effectively induces apoptosis in EGFR mutant (EGFRm) non-small cell lung cancer (NCSLC) cells. However, the underlying mechanisms have not been fully elucidated. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or TNFSF10) is known as a death ligand that initiates apoptosis via binding to its cell surface death receptors such as DR5. In this study, we found that osimertinib and other EGFR-TKIs increased the expression of TRAIL primarily in EGFRm NSCLC cell lines. This effect was accompanied with increased IL6 expression and STAT3 activation. Inhibition of STAT3 with either protein degradation or gene knockout abrogated the ability of osimertinib or recombinant human IL6 to elevate TRAIL levels. Moreover, osimertinib increased STAT3-dependent transcription of TRAIL via two STAT3 novel binding sites present in the TRAIL 5'flanking region. Hence, osimertinib induces IL6/STAT3-mediated TRAIL expression in EGFRm NSCLC cells. While osimertinib lost the ability to induce TRAIL expression in osimertinib-resistant EGFRm NSCLC, knockdown or knockout of TRAIL in sensitive EGFRm NSCLC cells rendered them less sensitive to osimertinib both in vitro and in vivo. Thus, TRAIL elevation contributes to the induction of apoptosis by osimertinib in EGFRm NSCLC cells. Furthermore, osimertinib increased membrane-bound TRAIL and DR5 membrane clustering and DR5 knockdown significantly compromised the cell-killing effect of osimertinib, together suggesting a DR5-dependent effect. Collectively, this study has revealed a previously undiscovered connection between TRAIL induction and osimertinib-induced apoptosis in EGFRm NSCLC cells, increasing our understanding of mechanisms accounting for apoptosis induced by osimertinib.
Collapse
Affiliation(s)
- You-Take Oh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Zhen Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Dongsheng Wang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
3
|
Kim Y, Kim J, Eom S, Jun H, Lee HB, Jeong D, Kang S. Protein Nanoparticles Simultaneously Displaying TRAIL and EGFR-Binding Ligands Effectively Induce Apoptotic Cancer Cell Death and Overcome EGFR-TKI Resistance in Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40237189 DOI: 10.1021/acsami.5c04021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Lung cancer remains one of the most lethal cancers globally, with nonsmall cell lung cancer (NSCLC) representing the predominant subtype. Despite significant advancements in targeted therapies, overcoming therapeutic resistance in NSCLC remains a significant challenge, particularly in cases resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs). Here, we developed target-specific, apoptosis-inducing protein nanoparticles using Aquifex aeolicus lumazine synthase (AaLS), which were engineered to simultaneously display multiple TRAIL molecules and EGFR-binding ligands, including EGFR affibody (Afb) or anti-EGFR nanobodies (7D12, 9G8, and EgB4). These nanoparticles utilize the EGFR-binding ligand to enhance selective targeting of EGFR-overexpressing lung adenocarcinoma (PC9, HCC827, A549) and squamous cell carcinoma (H226) cells, regardless of mutations within the intracellular kinase domain of EGFR, which are primarily driven by tyrosine kinase inhibitors commonly used as first-line treatments in lung cancer therapy. The codisplayed EGFR-binding ligands enhance the attachment of TRAIL-displaying protein nanoparticles to cancer cells by stabilizing interactions with EGFR, promoting cell surface clustering of TRAIL molecules and improving TRAIL engagement with death receptors (DRs). This sustained interaction significantly amplifies TRAIL-mediated apoptotic cancer cell death signaling, effectively overcoming both TRAIL and EGFR-TKI resistance in NSCLC cells. Our findings suggest that dual ligand-displaying protein nanoparticles targeting DRs and EGFR represent a promising therapeutic strategy to potentiate TRAIL efficacy and circumvent EGFR-TKI resistance in NSCLC.
Collapse
Affiliation(s)
- Yunjung Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jiwoo Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Soomin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heejin Jun
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hyun Bin Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Diane Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
4
|
Attia A, Habel A, Xu W, Stayoussef M, Mezlini A, Larbi A, Yaacoubi-Loueslati B. Serum Protein Profiling as theranostic biomarkers for Left- and Right-Sided Colon Cancer using Luminex ® technology. Cancer Biomark 2025; 42:18758592251329321. [PMID: 40232184 DOI: 10.1177/18758592251329321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
BackgroundGiven the differences between malignancies arising from different segments of the colon, specific theranostic biomarkers can be linked to either Right-sided (RCC) or Left-sided colon cancer (LCC).ObjectiveAnalysis of 65 serum proteins to identify panels of theranostic biomarkers for LCC and RCC.MethodsSerum levels of 65 immunomodulators were measured in CC, LCC, and RCC patients, as well as healthy controls with the ProcartaPlex Human Immune Monitoring 65-Plex Panel.ResultsIL-27 may be used for early detection in LCC. CD-30 was up-regulated in metastatic CC, BLC was up-regulated in metastatic LCC and CD-40L was down-regulated in metastatic RCC. MDC and MMP-1 were positively associated, while IL-9 and VEGF-A were negatively associated with lymph nodes invasion in CC. Up-regulation of IL-12p70 and MMP-1 in LCC with lymph nodes invasion contrasted with down-regulation of IL-9 and MIP-1beta. IL-23, I-TAC, and SDF-1α were negatively associated with resistant CC to Folfox chemotherapy, and I-TAC was down-regulated in resistant LCC. IL-2 and FGF-2 were down-regulated, while APRIL was up-regulated in resistant RCC.ConclusionsOur study revealed significant differences in serum protein levels between LCC and RCC emphasizing the importance to explore novel theranostic biomarkers for CC, associated with resistance or sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Amani Attia
- Department of Biology, Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Azza Habel
- Department of Biology, Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Weili Xu
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Mouna Stayoussef
- Department of Biology, Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Amel Mezlini
- Medical Oncology Department, Salah Azaiez Oncology Institute, Tunis, Tunisia
| | - Anis Larbi
- Beckman Coulter Life Sciences, Ville pinte, France
| | - Besma Yaacoubi-Loueslati
- Department of Biology, Faculty of Sciences of Tunis (FST), Laboratory of Mycology, Pathologies and Biomarkers (LR16ES05), University of Tunis El Manar (UTM), Tunis, Tunisia
| |
Collapse
|
5
|
Li M, Sun T, Chang M, Liu T, Feng L, Zhang D, Wu Y, Ma Y, Ma H, Meng G, Ji C, Ye J. Impact of single nucleotide polymorphisms of immunomodulatory factors on treatment response and prognosis in acute myeloid leukemia. Front Immunol 2025; 16:1571332. [PMID: 40230861 PMCID: PMC11994645 DOI: 10.3389/fimmu.2025.1571332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background Acute myeloid leukemia (AML) is a hematologic malignancy characterized by poor overall survival (OS). The impaired function, altered phenotype, and abnormal distribution of T cells create an immunosuppressive microenvironment in AML, affecting the efficacy of chemotherapy. Studies have shown that differentiated monocyte-like AML cells can express various immunomodulatory factors, resulting in T cell phenotypic changes and the development of an immunosuppressive AML microenvironment. Methods Seven single nucleotide polymorphisms (SNPs) of four immunomodulatory factors-HMOX1, TXNIP, TNSF10/TRAIL, and TNFAIP2-were selected and analyzed in 255 non-M3 AML patients and 316 healthy controls. SNP genotyping was conducted using the MassARRAY platform. Furthermore, we analyzed the relationship between AML susceptibility, bone marrow (BM) blast percentage, clinical characteristics, treatment response, and prognosis with the selected SNPs. Results The study indicated that HMOX1 rs2071746 and TNFAIP2 rs1132339 are associated with BM blasts at the diagnosis of AML patients. TXNIP rs7211 is associated with sensitivity to cytarabine- and anthracycline-induced chemotherapy in AML, while TXNIP rs9245 is associated with AML relapse. Moreover, TRAIL/TNFSF10 rs12488654 is associated with the overall survival of AML patients, and the AA genotype of TRAIL/TNFSF10 rs12488654 may be an independent favorable factor for AML prognosis. Conclusions Our results on the association between AML and SNPs in HMOX1, TXNIP, TNSF10/TRAIL, and TNFAIP2 genes provide an important reference for predicting the treatment response and prognosis of AML patients.
Collapse
MESH Headings
- Humans
- Polymorphism, Single Nucleotide
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/diagnosis
- Middle Aged
- Male
- Female
- Prognosis
- Adult
- Aged
- Young Adult
- Carrier Proteins/genetics
- Treatment Outcome
- Adolescent
- Genetic Predisposition to Disease
- Genotype
Collapse
Affiliation(s)
- Mingying Li
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tao Sun
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mengyuan Chang
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tingting Liu
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Feng
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Di Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuyan Wu
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuechan Ma
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huixian Ma
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guangqiang Meng
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jingjing Ye
- Department of Hematology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
6
|
Lee CE, Noh KM, Kim S, Hong J, Kim K. Recent Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Engineering Strategies for Precise Strike Therapy against Tumor. Biomater Res 2025; 29:0170. [PMID: 40110051 PMCID: PMC11922527 DOI: 10.34133/bmr.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025] Open
Abstract
Effective drug delivery relies on the selection of suitable carriers, which is crucial for protein-based therapeutics such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). One of the key advantages of TRAIL is its ability to selectively induce apoptosis in cancer cells excluding healthy tissues by binding to death receptors DR4 and DR5, which are highly expressed in various cancer cells. Despite this promise, the clinical application of TRAIL has been limited by its short half-life, limited stability, and inefficient delivery to tumor sites. To overcome currently available clinical and engineering approaches, a series of sophisticated strategies is required: (a) the design of biomaterial-mediated carriers for enhanced targeting efficacy, particularly via optimizing selected materials, composition, formulation, and surface modulation. Moreover, (b) development of genetically modified cellular products for augmented TRAIL secretion toward tumor microenvironments and (c) cell surface engineering techniques for TRAIL immobilization onto infusible cell populations are also discussed in the present review. Among these approaches, living cell-based carriers offer the distinct advantage of systemically administered TRAIL-functionalized cells capturing circulating tumor cells in the bloodstream, thereby preventing secondary tumor formation. This review provides insight into the development of novel TRAIL delivery platforms, discusses considerations for clinical translation, and suggests future directions and complementary strategies to advance the field of TRAIL-based cancer therapeutics.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyung Mu Noh
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Jiyeon Hong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
7
|
Haghir-Sharif-Zamini Y, Khosravi A, Hassan M, Zarrabi A, Vosough M. c-FLIP/Ku70 complex; A potential molecular target for apoptosis induction in hepatocellular carcinoma. Arch Biochem Biophys 2025; 765:110306. [PMID: 39818348 DOI: 10.1016/j.abb.2025.110306] [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: 11/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide and the most common form of liver cancer. Despite global efforts toward early diagnosis and effective treatments, HCC is often diagnosed at advanced stages, where conventional therapies frequently lead to resistance and/or high recurrence rates. Therefore, novel biomarkers and promising medications are urgently required. Epi-drugs, or epigenetic-based medicines, have recently emerged as a promising therapeutic modality. Since the epigenome of the cancer cells is always dysregulated and this is followed by apoptosis-resistance, reprogramming the epigenome of cancer cells by epi-drugs (such as HDAC inhibitors (HDACis), and DNMT inhibitors (DNMTis)) could be an alternative approach to use in concert with established treatment protocols. C-FLIP, an anti-apoptotic protein, and Ku70, a member of the DNA repair system, bind together and make a cytoplasmic complex in certain cancers and induce resistance to apoptosis. Many epi-drugs, such as HDACis, can dissociate this complex through Ku70 acetylation and activate cellular apoptosis. The novel compounds for dissociating this complex could provide an innovative insight into molecular targeted HCC treatments. In this review, we address the innovative therapeutic potential of targeting c-FLIP/Ku70 complex by epi-drugs, particularly HDACis, to overcome apoptosis resistance of HCC cells. This review will cover the mechanisms by which the c-FLIP/Ku70 complex facilitates cancer cell survival, the impact of epigenetic alterations on the complex dissociation, and highlight HDACis potential in combination therapies, biomarker developments and mechanistic overviews. This review highlights c-FLIP ubiquitination and Ku70 acetylation levels as diagnostic and prognostic tools in HCC management.
Collapse
Affiliation(s)
- Yasamin Haghir-Sharif-Zamini
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, 34959, Turkiye
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
8
|
Collins VG, Hutton D, Hossain-Ibrahim K, Joseph J, Banerjee S. The abscopal effects of sonodynamic therapy in cancer. Br J Cancer 2025; 132:409-420. [PMID: 39537767 PMCID: PMC11876350 DOI: 10.1038/s41416-024-02898-y] [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: 07/21/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The abscopal effect is a phenomenon wherein localised therapy on the primary tumour leads to regression of distal metastatic growths. Interestingly, various pre-clinical studies utilising sonodynamic therapy (SDT) have reported significant abscopal effects, however, the mechanism remains largely enigmatic. SDT is an emerging non-invasive cancer treatment that uses focussed ultrasound (FUS) and a sonosensitiser to induce tumour cell death. To expand our understanding of abscopal effects of SDT, we have summarised the preclinical studies that have found SDT-induced abscopal responses across various cancer models, using diverse combination strategies with nanomaterials, microbubbles, chemotherapy, and immune checkpoint inhibitors. Additionally, we shed light on the molecular and immunological mechanisms underpinning SDT-induced primary and metastatic tumour cell death, as well as the role and efficacy of different sonosensitisers. Notably, the observed abscopal effects underscore the need for continued investigation into the SDT-induced 'vaccine-effect' as a potential strategy for enhancing systemic anti-tumour immunity and combating metastatic disease. The results of the first SDT human clinical trials are much awaited and are hoped to enable the further evaluation of the safety and efficacy of SDT, paving the way for future studies specifically designed to explore the potential of translating SDT-induced abscopal effects into clinical reality.
Collapse
Affiliation(s)
- Victoria G Collins
- Department of Neurosurgery, Ninewells Hospital, Dundee, UK
- Department of Neurosurgery, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Dana Hutton
- The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | | | - James Joseph
- Department of Biomedical Engineering, School of Science and Engineering, University of Dundee, Dundee, UK.
| | - Sourav Banerjee
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
9
|
Zappa F, Muniozguren NL, Conrad JE, Acosta-Alvear D. The integrated stress response engages a cell-autonomous, ligand-independent, DR5-driven apoptosis switch. Cell Death Dis 2025; 16:101. [PMID: 39955274 PMCID: PMC11830069 DOI: 10.1038/s41419-025-07403-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
The integrated stress response (ISR) is a fundamental signaling network that leverages the cell's biosynthetic capacity against different stresses to restore homeostasis. However, when homeostasis is unattainable, the ISR switches to drive cell death and eliminate irreparably damaged cells. Previous work has shown that persistent activity of the ISR kinase PERK during unyielding endoplasmic reticulum (ER) stress induces apoptosis downstream of death receptor 5 (DR5) [1]. ER stress provides activating signals that engage the ectodomain (ED) of DR5 to drive its unconventional activation in the Golgi apparatus [1, 2]. Here, using chemical genetics to uncouple stress sensing from ISR activation, we found that DR5 signaling from the Golgi apparatus is integral to the ISR and not specific to ER stress. Furthermore, we show that DR5 activation can be driven solely by increased expression and does not require its ED. These findings indicate that a general ISR kill switch eliminates irreversibly injured cells.
Collapse
Affiliation(s)
- Francesca Zappa
- Department of Cellular, Molecular, and Developmental Biology, University of California, Santa Barbara, USA
- Altos Labs Bay Area Institute of Science, Altos Labs, Inc., Redwood City, USA
| | - Nerea L Muniozguren
- Department of Cellular, Molecular, and Developmental Biology, University of California, Santa Barbara, USA
| | - Julia E Conrad
- Altos Labs Bay Area Institute of Science, Altos Labs, Inc., Redwood City, USA
| | - Diego Acosta-Alvear
- Department of Cellular, Molecular, and Developmental Biology, University of California, Santa Barbara, USA.
- Altos Labs Bay Area Institute of Science, Altos Labs, Inc., Redwood City, USA.
| |
Collapse
|
10
|
Carlson S, Lin TL, Larson SM. Allogeneic chimeric antigen receptors (CARs) as an "off-the-shelf" therapy in multiple myeloma. Immunotherapy 2025; 17:211-222. [PMID: 39961649 PMCID: PMC11951714 DOI: 10.1080/1750743x.2025.2461987] [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: 10/01/2024] [Accepted: 01/30/2025] [Indexed: 03/28/2025] Open
Abstract
The success of autologous chimeric antigen receptor (CAR)-T cells has changed the treatment landscape in relapsed and refractory multiple myeloma (MM) resulting in potential movement of CAR-T cells to the frontline treatment setting. However, one of the greatest weaknesses of this therapy is its autologous nature, which makes it time-consuming, labor intensive, and dependent on the patient's T cell fitness. The development of allogeneic CARs is critical to overcome these challenges and provide patients with an off-the-shelf alternative that is readily available. This review will investigate the current landscape and future perspectives of allogeneic CAR research in MM, exploring both pre-clinical research and active clinical trials. More specifically, it will focus on the advantages and disadvantages of various CAR cellular candidates including CAR-T, CAR-NK, and CAR-iNKT cells, among other more novel candidates.
Collapse
Affiliation(s)
- Sophie Carlson
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Tasha L. Lin
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Sarah M. Larson
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
11
|
Uffenorde J, Hariri M, Papalanis E, Staffas A, Berg J, Stenerlöw B, Berglund H, Malmberg C, Spiegelberg D. Enhancing glioblastoma therapy: unveiling synergistic anticancer effects of Onalespib - radiotherapy combination therapy. Front Oncol 2025; 15:1451156. [PMID: 39949745 PMCID: PMC11821960 DOI: 10.3389/fonc.2025.1451156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 01/06/2025] [Indexed: 02/16/2025] Open
Abstract
Background Glioblastoma (GBM) is the deadliest form of brain cancer, impacting both adults and children, marked by exceptionally high morbidity and mortality rates, even with current standard treatments such as surgery, radiation therapy, and chemotherapy. Therefore, there is a pressing need for new therapeutic strategies to improve survival and reduce treatment side effects. In this study, we investigated the effect of HSP90 inhibition in combination with radiotherapy in established and patient-derived glioblastoma cell lines. Methods Potential radiosensitizing effects of the HSP90 inhibitor Onalespib were studied in XTT and clonogenic survival assays as well as in tumor-mimicking multicellular spheroid models. Further, migration capacity and effects on protein expression were studied after exposure to Onalespib and radiation using Proximity Extension Assay analysis. Results HSP90 inhibition with Onalespib synergistically enhanced the radiosensitivity of glioblastoma cells grown in 2D and 3D models, resulting in increased cell death, reduced migration capacity and activation of the apoptotic signaling pathway. The proteomic analysis of glioblastoma cells treated with Onalespib, radiation, and their combination revealed significant alterations in protein expression profiles, involved in growth signaling, immune modulation pathways and angiogenesis. Moreover, the combination treatment indicated potential for enhancing cell cycle arrest and apoptosis, suggesting promising anti-tumor effects. Conclusion These findings demonstrate that HSP90 inhibition may be a promising strategy to enhance the efficacy of radiotherapy in the treatment of GBM, potentially leading to improved outcomes for patients battling this challenging disease.
Collapse
Affiliation(s)
- Julia Uffenorde
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Mehran Hariri
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Eleftherios Papalanis
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Annika Staffas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Josefine Berg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hanna Berglund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Diana Spiegelberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
12
|
Radhi JH, El-Hagrasy AMA, Almosawi SH, Alhashel A, Butler AE. The Role of Osteoprotegerin in Breast Cancer: Genetic Variations, Tumorigenic Pathways, and Therapeutic Potential. Cancers (Basel) 2025; 17:337. [PMID: 39941709 PMCID: PMC11815763 DOI: 10.3390/cancers17030337] [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: 12/16/2024] [Revised: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
INTRODUCTION Osteoprotegerin (OPG), encoded by the TNFRSF11B gene, is linked to the development of breast cancer via several pathways, including interactions with the receptor activator of nuclear factor-κB (RANK) ligands, apoptosis-inducing proteins like TRAIL, and genetic variations such as single nucleotide polymorphisms (SNPs), directly altering gene expression. This review aims to investigate the role of OPG expression in breast cancer. METHODS A comprehensive literature search was conducted using PubMed Medline, Google Scholar, and ScienceDirect. Only full-text English publications from inception to September 2024 were included. RESULTS Studies have demonstrated that certain SNPs in the OPG gene, specifically rs3102735 and rs2073618, are linked to a higher risk of breast cancer development. Additionally, OPG's function as a TRAIL decoy receptor may inhibit the death of cancer cells. Furthermore, OPG in the serum and its interactions with BRCA mutations are being investigated for their potential influence on breast cancer progression. Studies have found that OPG promotes tumorigenesis by enhancing cell proliferation, angiogenesis, and aneuploidy in normal mammary epithelial cells. Moreover, OPG mediates the tumor-promoting effects of interleukin-1 beta and may serve as a biomarker for breast cancer risk, particularly in BRCA1 mutation carriers, through its role in dysregulated RANK signaling. Lastly, the use of recombinant OPG in mouse models has been found to exert anti-tumor effects. CONCLUSIONS In this review, the role of OPG in breast cancer is examined. OPG has a multifaceted role in breast cancer tumorigenesis and exerts its effects through genetic variations (SNPs), interactions with TNF-related apoptosis-inducing ligand (TRAIL), and the modulation of the pro-tumorigenic microenvironment effects of angiogenesis, cell survival, and metastasis. Additionally, OPG's dual role as a tumor suppressor and promoter serves as a possible therapeutic target to enhance apoptosis, limit bone metastasis, and modulate the tumor microenvironment. Whilst much is now known, further studies are necessary to fully delineate the role of OPG.
Collapse
Affiliation(s)
- Janan Husain Radhi
- School of Medicine, Royal College of Surgeons in Ireland—Medical University of Bahrain (RCSI Bahrain), Building No. 2441, Road 2835, Busaiteen P.O. Box 15503, Bahrain; (J.H.R.); (A.M.A.E.-H.); (S.H.A.)
| | - Ahmed Mohsen Abbas El-Hagrasy
- School of Medicine, Royal College of Surgeons in Ireland—Medical University of Bahrain (RCSI Bahrain), Building No. 2441, Road 2835, Busaiteen P.O. Box 15503, Bahrain; (J.H.R.); (A.M.A.E.-H.); (S.H.A.)
| | - Sayed Husain Almosawi
- School of Medicine, Royal College of Surgeons in Ireland—Medical University of Bahrain (RCSI Bahrain), Building No. 2441, Road 2835, Busaiteen P.O. Box 15503, Bahrain; (J.H.R.); (A.M.A.E.-H.); (S.H.A.)
| | - Abdullatif Alhashel
- School of Medicine, Royal College of Surgeons in Ireland—Medical University of Bahrain (RCSI Bahrain), Building No. 2441, Road 2835, Busaiteen P.O. Box 15503, Bahrain; (J.H.R.); (A.M.A.E.-H.); (S.H.A.)
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland—Medical University of Bahrain (RCSI Bahrain), Building No. 2441, Road 2835, Busaiteen P.O. Box 15503, Bahrain
| |
Collapse
|
13
|
Louie AD, Huntington KE, Lee Y, Mompoint J, Wu LJ, Lee S, Miner TJ, El-Deiry WS. TRAIL receptor agonist TLY012 in combination with PD-1 inhibition promotes tumor regression in an immune-competent mouse model of pancreatic ductal adenocarcinoma. Am J Cancer Res 2025; 15:286-298. [PMID: 39949937 PMCID: PMC11815385 DOI: 10.62347/roat5658] [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: 08/29/2024] [Accepted: 01/12/2025] [Indexed: 02/16/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an immunosuppressed, apoptosis-resistant phenotype. TLY012 is pegylated recombinant Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL), an orphan drug for chronic pancreatitis and systemic sclerosis. Innate immune TRAIL signaling suppresses cancer. We hypothesized that the combination of immune checkpoint-blocking anti-PD-1 antibody and TLY012 would have synergistic anti-tumor efficacy in immune-competent PDAC-bearing mice. PDAC tumor-bearing C57Bl/6 mice treated with 10 mg/kg anti-mouse PD-1 antibody twice weekly and 10 mg/kg TLY012 three times weekly had reduced tumor growth and tumor volume at 70 days compared to either drug alone (all P < 0.005). B-cell activating factor (BAFF), which promotes PDAC tumors, decreased to 44% of control mice with dual treatment at 7 days and remained decreased at 3 months. Long-term dual treatment showed the highest plasma levels of proinflammatory cytokines interferon-gamma (average 5.6 times control level, P=0.046), CCL5 (average 14.1 times control level, P=0.048), and interleukin-3 (IL-3, average 71.1 times control level, P=0.0053). Flow cytometry showed trends toward decreased circulating regulatory T cells, increased NK cells, and a higher proportion of CD8+ T cells within tumors in the dual treatment group. In summary, the combination of anti-PD-1 and TLY012 prevented the growth of PDAC in an immunocompetent mouse model while increasing tumor-infiltrating CD8+ T cells, decreasing circulating T-regulatory cells and altering plasma cytokine expression of CCL5, interferon-gamma, and IL-3 to promote proinflammatory, antitumor effects. Combining TLY012 and anti-mouse PD-1 modifies immune cell and cytokine levels to induce a more proinflammatory immune environment that contributes to decreased PDAC tumor growth.
Collapse
Affiliation(s)
- Anna D Louie
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
- Department of Surgery, Warren Alpert Medical School of Brown University and Lifespan Health SystemProvidence, RI 02912, USA
- Legorreta Cancer Center, Brown UniversityProvidence, RI 02912, USA
- Joint Program in Cancer Biology, Brown University and Lifespan Cancer InstituteProvidence, RI 02912, USA
| | - Kelsey E Huntington
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
- Legorreta Cancer Center, Brown UniversityProvidence, RI 02912, USA
- Joint Program in Cancer Biology, Brown University and Lifespan Cancer InstituteProvidence, RI 02912, USA
| | - Young Lee
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
| | - Jared Mompoint
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
| | - Laura Jinxuan Wu
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
| | - Seulki Lee
- D&D Pharmatech4th Floor, I&C Building 24, Pangyo-ro 255 beon-gil, Bundang-gu Seongnam-si, Gyeonggi-do 13486, South Korea
| | - Thomas J Miner
- Department of Surgery, Warren Alpert Medical School of Brown University and Lifespan Health SystemProvidence, RI 02912, USA
- Legorreta Cancer Center, Brown UniversityProvidence, RI 02912, USA
- Joint Program in Cancer Biology, Brown University and Lifespan Cancer InstituteProvidence, RI 02912, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Translational Cancer Therapeutics, Warren Alpert Medical School of Brown UniversityProvidence, RI 02912, USA
- Legorreta Cancer Center, Brown UniversityProvidence, RI 02912, USA
- Joint Program in Cancer Biology, Brown University and Lifespan Cancer InstituteProvidence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Brown UniversityProvidence, RI 02912, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan and Brown UniversityProvidence, RI 02912, USA
| |
Collapse
|
14
|
Lu X, Zhan X, Xia G, Wang F, Lv M, Liu R, Liu Y, Zi C, Li G, Wang R, Li J, Yuan F, Jia D. Improving Targeted Delivery and Antitumor Efficacy of TRAIL through Fusion with a B7H3-Antagonistic Affibody. Mol Pharm 2025; 22:284-294. [PMID: 39620978 DOI: 10.1021/acs.molpharmaceut.4c00891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an attractive candidate for anticancer therapeutics due to its efficient pro-apoptotic activity against tumor cells and its well-tolerated safety profile. However, the in vivo antitumor efficacy of TRAIL is often limited by its poor tumor targeting capacity. Nowadays, the B7 homologue 3 (B7-H3) immune checkpoint has emerged as a promising target for tumor immunotherapy and drug delivery. Here, we report the achievement of tumor-targeted delivery of TRAIL by genetically fusing it with a B7H3-antagonistic affibody. The affibody-TRAIL fusion protein, named ACT, was easily expressed in Escherichia coli with a high yield and could form the active trimeric state. In vitro ACT showed significantly increased cellular binding to multiple B7H3-positive tumor cells and improved cytotoxicity by 2-3 times compared to the parent TRAIL. In vivo ACT demonstrated a 2.4-fold higher tumor uptake than TRAIL in mice bearing B7H3-positive A431 tumor grafts. More importantly, ACT exhibited significantly improved antitumor efficacy against tumors in vivo. In addition, ACT treatment did not cause body weight loss or histopathological changes in the major organs of mice, indicating its good safety profile. Overall, our findings demonstrate that targeting B7H3 to enhance TRAIL delivery is a viable approach to improve its therapeutic efficacy, and ACT may be a potential agent for targeted therapy of B7H3-positive tumors.
Collapse
Affiliation(s)
- Xiaomeng Lu
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Xinyu Zhan
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Guozi Xia
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Feifei Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Mingjia Lv
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Renwei Liu
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Yuxue Liu
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Chen Zi
- Department of Clinical Laboratory, Linyi People's Hospital, Linyi 276034, China
| | - Guangyong Li
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Rui Wang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Jun Li
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Dianlong Jia
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
15
|
König C, Ivanisenko NV, Ivanisenko VA, Kulms D, Lavrik IN. Pharmacological targeting of caspase-8/c-FLIP L heterodimer enhances complex II assembly and elimination of pancreatic cancer cells. Commun Biol 2025; 8:4. [PMID: 39753884 PMCID: PMC11698904 DOI: 10.1038/s42003-024-07409-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/17/2024] [Indexed: 01/06/2025] Open
Abstract
Extrinsic apoptotic network is driven by Death Ligand (DL)-mediated activation of procaspase-8. Recently, we have developed the first-in class small molecule, FLIPinB, which specifically targets the key regulator of extrinsic apoptosis, the protein c-FLIPL, in the caspase-8/c-FLIPL heterodimer. We have shown that FLIPinB enhances DL-induced caspase-8 activity and apoptosis. However, the effects of FLIPinB action in combination with other cell death inducers have only just begun to be elucidated. Here, we show that FLIPinB enhances the cell death in pancreatic cancer cells induced by combinatorial treatment with DL, gemcitabine and Mcl-1 inhibitor S63845. Further, we found that these effects are mediated via an increase in the complex II assembly. Collectively, our study shows that targeting the caspase-8/c-FLIPL heterodimer in combination with the other drugs in pancreatic cancer cells is a promising direction that may provide a basis for further therapeutic strategies.
Collapse
Affiliation(s)
- Corinna König
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University (OvGU), Magdeburg, Magdeburg, Germany
| | - Nikita V Ivanisenko
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University (OvGU), Magdeburg, Magdeburg, Germany
| | - Vladimir A Ivanisenko
- Institute of Cytology and Genetics, Novosibirsk, Russia
- State Novosibirsk University, Novosibirsk, Russia
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
- National Center for Tumor Diseases, TU-Dresden, Dresden, Germany
| | - Inna N Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University (OvGU), Magdeburg, Magdeburg, Germany.
| |
Collapse
|
16
|
Nguyen TTK, Woo SM, Seo SU, Banstola A, Kim H, Duwa R, Vu ATT, Hong IS, Kwon TK, Yook S. Enhanced anticancer efficacy of TRAIL-conjugated and odanacatib-loaded PLGA nanoparticles in TRAIL resistant cancer. Biomaterials 2025; 312:122733. [PMID: 39106819 DOI: 10.1016/j.biomaterials.2024.122733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Seon Min Woo
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea
| | - Seung Un Seo
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea
| | - Asmita Banstola
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Haesoo Kim
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of medicine, Stanford University, Stanford, CA, 94305, USA
| | - An Thi Thanh Vu
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
17
|
El-Zehery IM, El-Mesery M, El-Sherbiny M, El Gayar AM, Eisa NH. Carbenoxolone upregulates TRAIL\TRAILR2 expression and enhances the anti-neoplastic effect of doxorubicin in experimentally induced hepatocellular carcinoma in rats. Biochem Biophys Res Commun 2024; 741:150876. [PMID: 39579528 DOI: 10.1016/j.bbrc.2024.150876] [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: 07/26/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/25/2024]
Abstract
AIMS This study investigates the in vivo anticancer activity of carbenoxolone (CBX) and its role in fighting hepatocellular carcinoma (HCC) progression and alleviating resistance against doxorubicin (DOX). Moreover, the molecular mechanism of action of CBX is explored. METHODS HCC was induced in Sprague Dawley rats via biweekly administration of thioacetamide (TAA) (200 mg/kg) intraperitoneally (i.p.) for 16 weeks after administering a single dose of diethylnitrosamine (DEN) (200 mg/kg, i.p.). A prophylactic model was established by treating rats with i.p. CBX (20 mg/kg/day) for 4 weeks starting on week 13 post-TAA injection. A therapeutic model was established by treating rats with CBX, DOX, or their combination for 7 weeks following 16 weeks of TAA administration. Serum Alpha-fetoprotein (AFP) and biochemical markers of hepatic functions were assessed. Histopathological examinations of hepatic tissues were performed. Immunohistochemical and qRT-PCR analyses were applied to assess the differential expressions of TRAIL/TRAILR2, Bcl-2, TGF-β1, and caspases 3, 8, and 9. RESULTS CBX markedly improved hepatic functions, reduced serum AFP levels, and alleviated TAA-induced hepatic histopathological alterations. CBX triggered apoptosis as evident by upregulating apoptotic markers: TRAIL/TRAILR2, caspases 3, 8, and 9, and downregulating the antiapoptotic protein Bcl-2. CBX downregulated TGF-β1. Interestingly, CBX/DOX combination mitigated hepatic damage and induced apoptosis in a way that surpassed DOX-only treatment. CONCLUSION The current study proposes that CBX is a promising anti-tumor compound, which can work effectively under prophylactic and therapeutic modes. Interestingly, CBX enhanced the anti-tumor effect of DOX. CBX exerted these effects via, in part, stimulating TRAIL-induced apoptosis along with attenuating fibrosis.
Collapse
MESH Headings
- Animals
- Doxorubicin
- Rats, Sprague-Dawley
- Male
- Carbenoxolone/pharmacology
- Rats
- Up-Regulation/drug effects
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/genetics
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Liver Neoplasms, Experimental/drug therapy
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/chemically induced
- Apoptosis/drug effects
- Antibiotics, Antineoplastic
- Drug Synergism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/chemically induced
- Liver Neoplasms/genetics
- Antineoplastic Agents/pharmacology
- TNF-Related Apoptosis-Inducing Ligand
Collapse
Affiliation(s)
- Iman M El-Zehery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Division of Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Amal M El Gayar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
18
|
Guo R, Wei Y, Du Y, Liu L, Zhang H, Ren R, Sun R, Zhang T, Xiong X, Zhao L, Wang H, Guo X, Zhu X. EX527, a sirtuins 1 inhibitor, sensitizes T-cell leukemia to death receptor-mediated apoptosis by downregulating cellular FLICE inhibitory protein. Cancer Biol Ther 2024; 25:2402588. [PMID: 39286953 PMCID: PMC11409494 DOI: 10.1080/15384047.2024.2402588] [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: 11/02/2023] [Revised: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Death receptor-mediated extrinsic apoptosis system had been developed as a promising therapeutic strategy in clinical oncology, such as TRAIL therapy. However, multiple studies have demonstrated that TRAIL resistance is the biggest problem for disappointing clinical trials despite preclinical success. Targeting cellular FLICE inhibitory protein (cFLIP) is one strategy of combinatorial therapies to overcome resistance to DR-mediated apoptosis due to its negative regulator of extrinsic apoptosis. E × 527 (Selisistat) is a specific inhibitor of SIRT1 activity with safe and well tolerance in clinical trials. Here, we show that E × 527 could strengthen significantly activation of rhFasL-mediated apoptotic signaling pathway and increased apoptotic rate of T leukemia cells with high expression of cFLIP. Mechanically, Inhibition of SIRT1 by E × 527 increased polyubiquitination level of cFLIP via increasing acetylation of Ku70, which could promote proteosomal degradation of cFLIP protein. It implied that combinatorial therapies of E × 527 plus TRAIL may have a potential as a novel clinical application for TRAIL-resistant hematologic malignancies.
Collapse
Affiliation(s)
- Rongqi Guo
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Yihui Wei
- Henan Red Cross Blood Center, Xinxiang, China
| | - Yating Du
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Luyue Liu
- Departments of Laboratory Medicine, Zhoukou Central Hospital, Zhoukou, China
| | - Haoqi Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Ruiying Ren
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Ruili Sun
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Tingting Zhang
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, China
| | - Lijun Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Hongfei Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| | - Xiaofang Guo
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
- Department of Microbiology, School of Basic Medical Sciences, Xinxiang
Medical University, Xinxiang, China
| | - Xiaofei Zhu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Tumor Microenvironment and Immunotherapy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
19
|
Kumar M, Kumar A, Srivastav A, Ghosh A, Kumar D. Genomic and molecular landscape of gallbladder cancer elucidating pathogenic mechanisms novel therapeutic targets and clinical implications. Mutat Res 2024; 830:111896. [PMID: 39754821 DOI: 10.1016/j.mrfmmm.2024.111896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
Gallbladder cancer (GBC) is an aggressive malignancy with a poor prognosis, often diagnosed at advanced stages due to subtle early symptoms. Recent studies have provided a comprehensive view of GBC's genetic and mutational landscape, uncovering crucial pathways involved in its pathogenesis. Environmental exposures, particularly to heavy metals, have been linked to elevated GBC risk. Established signaling pathways, including hormonal, apoptotic, metabolic, inflammatory, and DNA damage repair pathways, are integral to GBC progression, and evidence points to the involvement of specific germline and somatic mutations in its development. Key mutations in genes such as KRAS, TP53, IDH1/2, ERBB, PIK3CA, MET, MYC, BRAF, MGMT, CDKN2A and p16 have been identified as contributors to tumorigenesis, with additional alterations including chromosomal aberrations and epigenetic modifications. These molecular insights reveal several potential therapeutic targets that could address the limited treatment options for GBC. Promising therapeutic avenues under investigation include immune checkpoint inhibitors, tyrosine kinase inhibitors, tumor necrosis factor-related apoptosis-inducing ligands (TRAIL), and phytochemicals. Numerous clinical trials are assessing the efficacy of these targeted therapies. This review provides a detailed examination of GBC's genetic and mutational underpinnings, highlighting critical pathways and emerging therapeutic strategies. We discuss the implications of germline and somatic mutations for early detection and individualized treatment, aiming to bridge current knowledge gaps. By advancing our understanding of GBC's molecular profile, we hope to enhance diagnostic accuracy and improve treatment outcomes, ultimately paving the way for precision medicine approaches in managing GBC.
Collapse
Affiliation(s)
- Manishankar Kumar
- School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand 248007, India
| | - Arun Kumar
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Abhinav Srivastav
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Ashok Ghosh
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Dhruv Kumar
- School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand 248007, India.
| |
Collapse
|
20
|
Zhang Y, Kang T, Wang Y, Song C, Li H, Mi H, Li Y, Dong M, Ma X, Zhu H, Cheng L, Zhang P, Chen Z, Zhou L, Wu Q, Mao F, Wang B, Zhang S, Shu K, Wan F, Zhou W, Rich JN, Shen J, Xiao Q, Yu X. A low level of tumor necrosis factor α in tumor microenvironment maintains the self-renewal of glioma stem cells by Vasorin-mediated glycolysis. Neuro Oncol 2024; 26:2256-2271. [PMID: 39093693 PMCID: PMC11630517 DOI: 10.1093/neuonc/noae147] [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: 10/10/2023] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Self-renewal of glioma stem cells (GSCs) is responsible for glioblastoma (GBM) therapy resistance and recurrence. Tumor necrosis factor α (TNFα) and TNF signaling pathway display an antitumor activity in preclinical models and in tumor patients. However, TNFα exhibits no significance for glioma clinical prognosis based on the Glioma Genome Atlas database. This study aimed to explore whether TNFα of tumor microenvironment maintains self-renewal of GSCs and promotes worse prognosis in glioma patients. METHODS Spatial transcriptomics, immunoblotting, sphere formation assay, extreme limiting dilution, and gene expression analysis were used to determine the role of TNFα on GSC's self-renewal. Mass spectrometry, RNA-sequencing detection, bioinformatic analyses, qRT-RNA, immunofluorescence, immunohistochemistry, single-cell RNA sequencing, in vitro and in vivo models were used to uncover the mechanism of TNFα-induced GSC self-renewal. RESULTS A low level of TNFα displays a promoting effect on GSC self-renewal and worse glioma prognosis. Mechanistically, Vasorin (VASN) mediated TNFα-induced self-renewal by potentiating glycolysis. Lactate produced by glycolysis inhibits the TNFα secretion of tumor-associated macrophages (TAMs) and maintains TNFα at a low level. CONCLUSIONS TNFα-induced GSC self-renewal mediated by VASN provides a possible explanation for the failures of endogenous TNFα effect on GBM. A combination of targeting VASN and TNFα antitumor effect may be an effective approach for treating GBM.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianxu Kang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxi Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Song
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Li
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailong Mi
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yachao Li
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minhai Dong
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Ma
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lidong Cheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Po Zhang
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zhiye Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Zhou
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiulian Wu
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Feng Mao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wan
- Department of Neurosurgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenchao Zhou
- Division of Life Sciences and Medicine, Intelligent Pathology Institute, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jianying Shen
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qungen Xiao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjiang Yu
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, China
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discov 2024; 23:939-961. [PMID: 39448880 DOI: 10.1038/s41573-024-01053-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/26/2024]
Abstract
The first anti-tumour necrosis factor (TNF) monoclonal antibody, infliximab (Remicade), celebrated its 25th anniversary of FDA approval in 2023. Inhibitors of TNF have since proved clinically efficacious at reducing inflammation associated with several autoimmune diseases, including rheumatoid arthritis, psoriasis and Crohn's disease. The success of TNF inhibitors raised unrealistic expectations for targeting other members of the TNF superfamily (TNFSF) of ligands and their receptors, with difficulties in part related to their more limited, variable expression and potential redundancy. However, there has been a resurgence of interest and investment, with many of these cytokines or their cognate receptors now under clinical investigation as targets for modulation of autoimmune and inflammatory diseases, as well as cancer. This Review assesses TNFSF-targeted biologics currently in clinical development for immune system-related diseases, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | | | - Carl F Ware
- Laboratory of Molecular Immunology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
22
|
Ye M, Liu T, Miao L, Ji H, Xu Z, Wang H, Zhang J, Zhu X. Cisplatin-encapsulated TRAIL-engineered exosomes from human chorion-derived MSCs for targeted cervical cancer therapy. Stem Cell Res Ther 2024; 15:396. [PMID: 39497209 PMCID: PMC11536590 DOI: 10.1186/s13287-024-04006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Cisplatin (DDP) is an efficacious and widely applied chemotherapeutic drug for cervical cancer patients who are diagnosed as metastatic and inoperable, or desiring fertility preservation. Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) selectively triggers cancer cells apoptosis by binding to cognate death receptors (DR4 and DR5). Mesenchymal stem cells-derived exosomes (MSCs-Exo) have been regarded as ideal drug carriers on account of their nanoscale, low toxicity, low immunogenicity, high stability, biodegradability, and abundant sources. METHODS Human chorion-derived mesenchymal stem cells (hCD-MSCs) were isolated by adherent culture method. TRAIL-engineered hCD-MSCs (hCD-MSCsTRAIL) were constructed by lentivirus transfection, and its secreted Exo (hCD-MSCs-ExoTRAIL) were acquired by differential centrifugation and confirmed to overexpress TRAIL by western blotting. Next, nanoscale drug delivery systems (DDP & hCD-MSCs-ExoTRAIL) were fabricated by loading DDP into hCD-MSCs-ExoTRAIL via electroporation. The CCK-8 assay and flow cytometry were conducted to explore the proliferation and apoptosis of cervical cancer cells (SiHa and HeLa), respectively. Cervical cancer-bearing nude mice were constructed to examine the antitumor activity and biosafety of DDP & hCD-MSCs-ExoTRAIL in vivo. RESULTS Compared with hCD-MSCs-Exo, hCD-MSCs-ExoTRAIL weakened proliferation and enhanced apoptosis of cervical cancer cells. DDP & hCD-MSCs-ExoTRAIL were proved to retard cervical cancer cell proliferation and propel cell apoptosis more effectively than DDP or hCD-MSCs-ExoTRAIL alone in vitro. In cervical cancer-bearing mice, DDP & hCD-MSCs-ExoTRAIL evidently hampered tumor growth, and its role in inducing apoptosis was mechanistically associated with JNK/p-c-Jun activation and survivin suppression. Moreover, DDP & hCD-MSCs-ExoTRAIL showed favorable biosafety in vivo. CONCLUSIONS DDP & hCD-MSCs-ExoTRAIL nanoparticles exhibited great promise for cervical cancer treatment as an Exo-based chemo-gene combinational therapy in clinical practice.
Collapse
Affiliation(s)
- Miaomiao Ye
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Tingxian Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Liqing Miao
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Zhihui Xu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Huihui Wang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China
| | - Jian'an Zhang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, No. 109 Xueyuan Xi Road, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
23
|
Steinmetz AR, Pierce M, Martini A, Tholomier C, Manyam G, Chen Y, Sood A, Duplisea JJ, Johnson BA, Czerniak BA, Lee BH, Jagannath C, Yla-Herttuala S, Parker NR, McConkey DJ, Dinney CP, Mokkapati S. Single-cell RNA sequencing analysis identifies acute changes in the tumor microenvironment induced by interferon α gene therapy in a murine bladder cancer model. Front Immunol 2024; 15:1387229. [PMID: 39559365 PMCID: PMC11570268 DOI: 10.3389/fimmu.2024.1387229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Nadofaragene firadenovec (Ad-IFNα/Syn3) is now approved for BCG-unresponsive bladder cancer (BLCA). IFNα is a pleiotropic cytokine that causes direct tumor cell killing via TRAIL-mediated apoptosis, angiogenesis inhibition, and activation of the innate and adaptive immune system. We established an immunocompetent murine BLCA model to study the effects of murine adenoviral IFNα (muAd-Ifnα) gene therapy on cancer cells and the tumor microenvironment using a novel murine equivalent of Nadofaragene firadenovec (muAd-Ifnα). Methods Tumors were induced by instilling MB49 cells into the bladders of mice; luciferase imaging confirmed tumor development. Mice were treated with adenovirus control (Ad-Ctrl; empty vector), or muAd-Ifnα (3x1011 VP/mL), and survival analysis was performed. For single-cell sequencing (scRNAseq) analysis (72h), bladders were harvested and treated with collagenase/hyaluronidase and TrypLE for cell dissociation. Single cells were suspended in PBS/1% FBS buffer; viability was assessed with Vicell cell counter. scRNAseq analysis was performed using 10X genomics 3' sequencing. Raw RNAseq data were pre-processed using Cell Ranger single-cell software. Seurat (R package) was used to normalize and cluster the scRNA data. Pooled differential gene expression analysis in specific cell clusters was performed with DESeq2. Results We identified 16 cell clusters based on marker expression which were grouped into epithelial (tumor), uroplakin-enriched, endothelial, T-cells, neutrophils, and macrophage clusters. Top differentially expressed genes between muAd-Ifnα and Ad-Ctrl were identified. Within the specific cell clusters, IPA analysis revealed significant differences between muAd-Ifnα and control. IFNα signaling and hypercytokinemia/chemokinemia were upregulated in all clusters. Cell death pathways were upregulated in tumor and endothelial clusters. T-cells demonstrated upregulation of the immunogenic cell death signaling pathway and a decrease in the Th2 pathway genes. Macrophages showed upregulation of PD1/PD-L1 pathways along with downregulation of macrophage activation pathways (alternate and classical). Multiplex immunofluorescence confirmed increased infiltration with macrophages in muAd-Ifnα treated tumors compared to controls. PD1/PD-L1 expression was reduced at 72h. Discussion This single-cell analysis builds upon our understanding of the impact of Ad-IFNα on tumor cells and other compartments of the microenvironment. These data will help identify mechanisms to improve patient selection and therapeutic efficacy of Nadofaragene firadenovec.
Collapse
Affiliation(s)
- Alexis R. Steinmetz
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Morgan Pierce
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alberto Martini
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Come Tholomier
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ganiraju Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yan Chen
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Akshay Sood
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jonathan J. Duplisea
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Burles A. Johnson
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Bogdan A. Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Byron H. Lee
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Nigel R. Parker
- A.I. Virtanen Institute for Molecular Sciences, Kuopio, Finland
| | - David J. McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Colin P. Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sharada Mokkapati
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
24
|
Zhou X, Wang Y, Dou Z, Delfanti G, Tsahouridis O, Pellegry CM, Zingarelli M, Atassi G, Woodcock MG, Casorati G, Dellabona P, Kim WY, Guo L, Savoldo B, Tsagaratou A, Milner JJ, Metelitsa LS, Dotti G. CAR-redirected natural killer T cells demonstrate superior antitumor activity to CAR-T cells through multimodal CD1d-dependent mechanisms. NATURE CANCER 2024; 5:1607-1621. [PMID: 39354225 PMCID: PMC12002392 DOI: 10.1038/s43018-024-00830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/28/2024] [Indexed: 10/03/2024]
Abstract
Human natural killer T (NKT) cells have been proposed as a promising cell platform for chimeric antigen receptor (CAR) therapy in solid tumors. Here we generated murine CAR-NKT cells and compared them with CAR-T cells in immune-competent mice. Both CAR-NKT cells and CAR-T cells showed similar antitumor effects in vitro, but CAR-NKT cells showed superior antitumor activity in vivo via CD1d-dependent immune responses in the tumor microenvironment. Specifically, we show that CAR-NKT cells eliminate CD1d-expressing M2-like macrophages. In addition, CAR-NKT cells promote epitope spreading and activation of endogenous T cell responses against tumor-associated neoantigens. Finally, we observed that CAR-NKT cells can co-express PD1 and TIM3 and show an exhaustion phenotype in a model of high tumor burden. PD1 blockade as well as vaccination augmented the antitumor activity of CAR-NKT cells. In summary, our results demonstrate the multimodal function of CAR-NKT cells in solid tumors, further supporting the rationale for developing CAR-NKT therapies in the clinic.
Collapse
Affiliation(s)
- Xin Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Ying Wang
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zhangqi Dou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Gloria Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ourania Tsahouridis
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | - Manuela Zingarelli
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Gatphan Atassi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Division of Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Division of Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Linjie Guo
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Ageliki Tsagaratou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - J Justin Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid S Metelitsa
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
Chen X, Wu J, Zhou B, Zhu M, Zhang J, Zhou N, Zhu YZ, Zhang X, Duan X, Men K. Bacterial Lysate-Based Bifunctional mRNA Nanoformulation for Efficient Colon Cancer Immunogene Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56580-56598. [PMID: 39397736 DOI: 10.1021/acsami.4c07684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
mRNA-based nonviral gene therapy has played an important role in cancer therapy, however, the limited delivery efficiency and therapeutic capacity still require further exploration and enhancement. Immunogene therapy provides a strategy for cancer treatment. Bacteria are tiny single-celled living organisms, many of which can be found in and on the human body and are beneficial to humans. Lactobacillus reuteri is a bacterial member of the gut flora, and recent research has shown that it can reduce intestinal inflammation by stimulating an immunomodulatory response. L. reuteri lysate represents an ideal resource for constructing advanced mRNA delivery systems with immune stimulation potential. Here, we prepared a bifunctional mRNA delivery system DMP-Lac (DOTAP-mPEG-PCL-L. reuteri lysate), which successfully codelivered L. reuteri lysate and IL-23A mRNA, exhibited a high mRNA delivery efficiency of 75.56% ± 0.85%, and strongly promoted the maturation and activation of the immune system in vivo. Both the CT26 abdominal metastasis model and the lung metastasis model also exhibited a good therapeutic effect, and the tumor inhibition rate of DMP-Lac/IL-23A group reached 97.92%. Protein chip technology verified that DMP acted as an immune adjuvant, demonstrating that the L. reuteri lysate could regulate the related immune cells, while IL-23 mRNA caused changes in downstream factors, thus producing the corresponding tumor treatment effect. The DMP-Lac/IL-23A complex exhibited strong anticancer immunotherapeutic effects. Our results demonstrated that this bifunctional mRNA formulation served as a tumor-specific nanomedicine, providing an advanced strategy for colon cancer immunogene therapy.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jieping Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Na Zhou
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Yi Zhun Zhu
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xin Zhang
- State Key Laboratory for Quality Research of Chinese Medicines and School of Pharmacy, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
26
|
Kurmangaliyeva S, Baktikulova K, Tkachenko V, Seitkhanova B, Shapambayev N, Rakhimzhanova F, Almagambetova A, Kurmangaliyev K. An Overview of Hexavalent Chromium-Induced Necroptosis, Pyroptosis, and Ferroptosis. Biol Trace Elem Res 2024:10.1007/s12011-024-04376-1. [PMID: 39287767 DOI: 10.1007/s12011-024-04376-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Heavy metals are common environmental industrial pollutants. Due to anthropogenic activity, chromium, especially its hexavalent form [Cr(VI)], is a widespread environmental contaminant that poses a threat to human health. In this review paper, we summarize the currently reported molecular mechanisms involved in chromium toxicity with a focus on the induction of pro-inflammatory non-apoptotic cell death pathways such as necroptosis, pyroptosis, and ferroptosis. The review highlights the ability of chromium to induce necroptosis, pyroptosis, and ferroptosis revealing the signaling pathways involved. Cr(VI) can induce RIPK1/RIPK3-dependent necroptosis both in vitro and in vivo. Chromium toxicity is associated with pyroptotic NLRP3 inflammasome/caspase-1/gasdermin D-dependent secretion of IL-1β and IL-18. Furthermore, this review emphasizes the role of redox imbalance and intracellular iron accumulation in Cr(VI)-induced ferroptosis. Of note, the crosstalk between the investigated lethal subroutines in chromium-induced toxicity is primarily mediated by reactive oxygen species (ROS), which are suggested to act as a rheostat determining the cell death pathway in cells exposed to chromium. The current study provides novel insights into the pro-inflammatory effects of chromium, since necroptosis, pyroptosis, and ferroptosis affect inflammation owing to their immunogenic properties linked primarily with damage-associated molecular patterns. Inhibition of these non-apoptotic lethal subroutines can be considered a therapeutic strategy to reduce the toxicity of heavy metals, including chromium.
Collapse
Affiliation(s)
- Saulesh Kurmangaliyeva
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kristina Baktikulova
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan.
| | - Viktoriya Tkachenko
- State Institution "Republican Scientific and Practical Centre of Sports, " 8 Narochanskaya St, Minsk, Republic of Belarus
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Al-Farabi Sq, Shymkent, Republic of Kazakhstan
| | - Nasriddin Shapambayev
- Department of General Practitioner - 1, Khoja Akhmet Yasawi International Kazakh-Turkish University, 7/7 Baitursynov St, Shymkent, Republic of Kazakhstan
| | - Farida Rakhimzhanova
- Department of Microbiology, NCJSC "Semey Medical University, " 103 Abay St, Semey, Republic of Kazakhstan
| | - Altyn Almagambetova
- Department of Phthisiology and Dermatovenerology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| | - Kairat Kurmangaliyev
- Department of Microbiology, Virology and Immunology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev St, Aktobe, Republic of Kazakhstan
| |
Collapse
|
27
|
Gamie Z, Krippner-Heidenreich A, Gerrand C, Rankin KS. Targeting Death Receptor 5 (DR5) for the imaging and treatment of primary bone and soft tissue tumors: an update of the literature. Front Mol Biosci 2024; 11:1384795. [PMID: 39286782 PMCID: PMC11402684 DOI: 10.3389/fmolb.2024.1384795] [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: 02/10/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Background Death Receptor 5 (DR5) is expressed on the surface of primary bone and soft tissue sarcoma cells, and its activation induces cell death primarily through apoptosis. The combination of DR5 agonists and commonly used chemotherapeutic agents, such as doxorubicin, can promote cell death. Currently, clinical trials are investigating the effectiveness of DR5 activation using new biological agents, such as bi-specific or tetravalent antibodies, in improving the survival of patients with relapsed or refractory cancers. Furthermore, investigations continue into the use of novel combination therapies to enhance DR5 response, for example, with inhibitor of apoptosis protein (IAP) antagonist agents [such as the second mitochondria-derived activator of caspase (SMAC) mimetics] and with immune checkpoint inhibitor anti-programmed death-ligand 1 (anti-PD-L1) or anti-programmed cell death-1 (anti-PD-1) antibodies. Other therapies include nanoparticle-mediated delivery of TRAIL plasmid DNA or TRAIL mRNA and stem cells as a vehicle for the targeted delivery of anti-cancer agents, such as TRAIL, to the tumor. Methods Scoping review of the literature from November 2017 to March 2024, utilizing PubMed and Google Scholar. Results New agents under investigation include nanoTRAIL, anti-Kv10.1, multimeric IgM, and humanized tetravalent antibodies. Developments have been made to test novel agents, and imaging has been used to detect DR5 in preclinical models and patients. The models include 3D spheroids, genetically modified mouse models, a novel jaw osteosarcoma model, and patient-derived xenograft (PDX) animal models. There are currently two ongoing clinical trials focusing on the activation of DR5, namely, IGM-8444 and INBRX-109, which have progressed to phase 2. Further modifications of TRAIL delivery with fusion to single-chain variable fragments (scFv-TRAIL), directed against tumor-associated antigens (TAAs), and in the use of stem cells focus on targeted TRAIL delivery to cancer cells using bi-functional strategies. Conclusion In vitro, in vivo, and clinical trials, as well as advances in imaging and theranostics, indicate that targeting DR5 remains a valid strategy in the treatment of some relapsed and refractory cancers.
Collapse
Affiliation(s)
- Zakareya Gamie
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Craig Gerrand
- Department of Orthopaedic Oncology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, United Kingdom
| | - Kenneth Samora Rankin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
28
|
Seyrek K, Ivanisenko NV, König C, Lavrik IN. Modulation of extrinsic apoptotic pathway by intracellular glycosylation. Trends Cell Biol 2024; 34:728-741. [PMID: 38336591 DOI: 10.1016/j.tcb.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
The importance of post-translational modifications (PTMs), particularly O-GlcNAcylation, of cytoplasmic proteins in apoptosis has been neglected for quite a while. Modification of cytoplasmic proteins by a single N-acetylglucosamine sugar is a dynamic and reversible PTM exhibiting properties more like phosphorylation than classical O- and N-linked glycosylation. Due to the sparse information existing, we have only limited understanding of how GlcNAcylation affects cell death. Deciphering the role of GlcNAcylation in cell fate may provide further understanding of cell fate decisions. This review focus on the modulation of extrinsic apoptotic pathway via GlcNAcylation carried out by O-GlcNAc transferase (OGT) or by other bacterial effector proteins.
Collapse
Affiliation(s)
- Kamil Seyrek
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Nikita V Ivanisenko
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Corinna König
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - Inna N Lavrik
- Translational Inflammation Research, Medical Faculty, Center of Dynamic Systems (CDS), Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany.
| |
Collapse
|
29
|
Ulucan O. Expanding Beyond Genetic Subtypes in B-Cell Acute Lymphoblastic Leukemia: A Pathway-Based Stratification of Patients for Precision Oncology. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:470-477. [PMID: 39158364 DOI: 10.1089/omi.2024.0145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Precision oncology promises individually tailored drugs and clinical care for patients with cancer: That is, "the right drug, for the right patient, at the right dose, and at the right time." Although stratification of the risk for treatment resistance and toxicity is key to precision oncology, there are multiple ways in which such stratification can be achieved, for example, genetic, functional pathway based, among others. Moving toward precision oncology is sorely needed in the case of acute lymphoblastic leukemia (ALL) wherein adult patients display survival rates ranging from 30% to 70%. The present study reports on the pathway activity signature of adult B-ALL, with an eye to precision oncology. Transcriptome profiles from three different expression datasets, comprising 346 patients who were adolescents or adults with B-ALL, were harnessed to determine the activity of signaling pathways commonly disrupted in B-ALL. Pathway activity analyses revealed that Ph-like ALL closely resembles Ph-positive ALL. Although this was the case at the average pathway activity level, the pathway activity patterns in B-ALL differ from genetic subtypes. Importantly, clustering analysis revealed that five distinct clusters exist in B-ALL patients based on pathway activity, with each cluster displaying a unique pattern of pathway activation. Identifying pathway-based subtypes thus appears to be crucial, considering the inherent heterogeneity among patients with the same genetic subtype. In conclusion, a pathway-based stratification of the B-ALL could potentially allow for simultaneously targeting highly active pathways within each ALL subtype, and thus might open up new avenues of innovation for personalized/precision medicine in this cancer that continues to have poor prognosis in adult patients compared with the children.
Collapse
Affiliation(s)
- Ozlem Ulucan
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Bilgi University, Istanbul, Turkiye
| |
Collapse
|
30
|
Zhang X, Taylor H, Valdivia A, Dasari R, Buckley A, Bonacquisti E, Nguyen J, Kanchi K, Corcoran DL, Herring LE, Steindler DA, Baldwin A, Hingtgen S, Satterlee AB. Auto-loaded TRAIL-exosomes derived from induced neural stem cells for brain cancer therapy. J Control Release 2024; 372:433-445. [PMID: 38908756 PMCID: PMC11283351 DOI: 10.1016/j.jconrel.2024.06.048] [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: 02/01/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Transdifferentiation (TD), a somatic cell reprogramming process that eliminates pluripotent intermediates, creates cells that are ideal for personalized anti-cancer therapy. Here, we provide the first evidence that extracellular vesicles (EVs) from TD-derived induced neural stem cells (Exo-iNSCs) are an efficacious treatment strategy for brain cancer. We found that genetically engineered iNSCs generated EVs loaded with the tumoricidal gene product TRAIL at nearly twice the rate of their parental fibroblasts, and TRAIL produced by iNSCs was naturally loaded into the lumen of EVs and arrayed across their outer membrane (Exo-iNSC-TRAIL). Uptake studies in ex vivo organotypic brain slice cultures showed that Exo-iNSC-TRAIL selectively accumulates within tumor foci, and co-culture assays demonstrated that Exo-iNSC-TRAIL killed metastatic and primary brain cancer cells more effectively than free TRAIL. In an orthotopic mouse model of brain cancer, Exo-iNSC-TRAIL reduced breast-to-brain tumor xenografts by approximately 3000-fold compared to treatment with free TRAIL, with all Exo-iNSC-TRAIL treated animals surviving through 90 days post-treatment. In additional in vivo testing against aggressive U87 and invasive GBM8 glioblastoma tumors, Exo-iNSC-TRAIL also induced a statistically significant increase in survival. These studies establish a novel, easily generated, stable, tumor-targeted EV to efficaciously treat multiple forms of brain cancer.
Collapse
Affiliation(s)
- Xiaopei Zhang
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah Taylor
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alain Valdivia
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rajaneekar Dasari
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Buckley
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily Bonacquisti
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juliane Nguyen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Krishna Kanchi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David L Corcoran
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dennis A Steindler
- Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Steindler Consulting, Boston, MA, USA
| | - Albert Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Andrew Benson Satterlee
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
31
|
Kelepouras K, Saggau J, Varanda AB, Zrilic M, Kiefer C, Rakhsh-Khorshid H, Lisewski I, Uranga-Murillo I, Arias M, Pardo J, Tonnus W, Linkermann A, Annibaldi A, Walczak H, Liccardi G. The importance of murine phospho-MLKL-S345 in situ detection for necroptosis assessment in vivo. Cell Death Differ 2024; 31:897-909. [PMID: 38783091 PMCID: PMC11239901 DOI: 10.1038/s41418-024-01313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Necroptosis is a caspase-independent modality of cell death implicated in many inflammatory pathologies. The execution of this pathway requires the formation of a cytosolic platform that comprises RIPK1 and RIPK3 which, in turn, mediates the phosphorylation of the pseudokinase MLKL (S345 in mouse). The activation of this executioner is followed by its oligomerisation and accumulation at the plasma-membrane where it leads to cell death via plasma-membrane destabilisation and consequent permeabilisation. While the biochemical and cellular characterisation of these events have been amply investigated, the study of necroptosis involvement in vivo in animal models is currently limited to the use of Mlkl-/- or Ripk3-/- mice. Yet, even in many of the models in which the involvement of necroptosis in disease aetiology has been genetically demonstrated, the fundamental in vivo characterisation regarding the question as to which tissue(s) and specific cell type(s) therein is/are affected by the pathogenic necroptotic death are missing. Here, we describe and validate an immunohistochemistry and immunofluorescence-based method to reliably detect the phosphorylation of mouse MLKL at serine 345 (pMLKL-S345). We first validate the method using tissues derived from mice in which Caspase-8 (Casp8) or FADD are specifically deleted from keratinocytes, or intestinal epithelial cells, respectively. We next demonstrate the presence of necroptotic activation in the lungs of SARS-CoV-infected mice and in the skin and spleen of mice bearing a Sharpin inactivating mutation. Finally, we exclude necroptosis occurrence in the intestines of mice subjected to TNF-induced septic shock. Importantly, by directly comparing the staining of pMLKL-345 with that of cleaved Caspase-3 staining in some of these models, we identify spatio-temporal and functional differences between necroptosis and apoptosis supporting a role of RIPK3 in inflammation independently of MLKL versus the role of RIPK3 in activation of necroptosis.
Collapse
Grants
- Wellcome Trust
- G.L. is funded by the Center for Biochemistry, Univeristy of Cologne - 956400, Köln Fortune, CANcer TARgeting (CANTAR) project NW21-062A, two collaborative research center grants: SFB1399-413326622 Project C06, SFB1530-455784452 Project A03 both funded by the Deutsche Forschungsgemeinschaft (DFG)) and associated to the collaborative SFB1403 also funded by the DFG
- H.W. is funded by the Alexander von Humboldt Foundation, a Wellcome Trust Investigator Award (214342/Z/18/Z), a Medical Research Council Grant (MR/S00811X/1), a Cancer Research UK Programme Grant (A27323) and three collaborative research center grants (SFB1399, Project C06, SFB1530-455784452, Project A03 and SFB1403–414786233) funded by the Deutsche Forschungsgemeinschaft (DFG) and CANcer TARgeting (CANTAR) funded by Netzwerke 2021.
- AA is funded by the Center for Molecular Medine Cologne (CMMC) Junior Research Group program, Deutsche Forschungsgemeinschaft (DFG) (project number AN1717/1-1), the Jürgen Manchot Stiftung foundation, the collaborative research center SFB1530 (Project A5, ID: 455784452)
- JP is funded by FEDER (Fondo Europeo de Desarrollo Regional), Gobierno de Aragón (Group B29_23R), CIBERINFEC (CB21/13/00087), Ministerio de Ciencia, Innovación y Universidades (MCNU)/Agencia Estatal de Investigación (PID2020-113963RBI00)
- MA is funded by a Postdoctoral Juan de la Cierva Contract.
- Work in the Linkermann Lab was funded by the German Research Foundation SFB-TRR205, SFB-TRR 127, SPP2306, and a Heisenberg-Professorship to A.L., project number 324141047, and the international research training group (IRTG) 2251. It was further supported by the BMBF (FERROPath consortium), the TU Dresden / Kings College London transcampus initiative and the DFG-Sachbeihilfe LI 2107/10-1.
Collapse
Affiliation(s)
- Konstantinos Kelepouras
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Julia Saggau
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Ana Beatriz Varanda
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Matea Zrilic
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Christine Kiefer
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Hassan Rakhsh-Khorshid
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Ina Lisewski
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Iratxe Uranga-Murillo
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Maykel Arias
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Julian Pardo
- Department of Microbiology, Radiology, Paediatry and Public Heath, Faculty of Medicine, University of Zaragoza/IIS, Aragon, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alessandro Annibaldi
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Henning Walczak
- Cell Death, Inflammation and Immunity Laboratory, CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Cell Death, Inflammation and Immunity Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, WC1E 6BT, London, UK
| | - Gianmaria Liccardi
- Genome Instability, Inflammation and Cell Death Laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
32
|
Zhang Y, Wang L, Dong C, Zhuang Y, Hao G, Wang F. Licochalcone D exhibits cytotoxicity in breast cancer cells and enhances tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis through upregulation of death receptor 5. J Biochem Mol Toxicol 2024; 38:e23757. [PMID: 38937960 DOI: 10.1002/jbt.23757] [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: 01/28/2024] [Revised: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Anticancer strategies using natural products or derivatives are promising alternatives for cancer treatment. Here, we showed that licochalcone D (LCD), a natural flavonoid extracted from Glycyrrhiza uralensis Fisch, suppressed the growth of breast cancer cells, and was less toxic to MCF-10A normal breast cells. LCD-induced DNA damage, cell cycle arrest, and apoptosis in breast cancer cells. Furthermore, LCD potentiated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cytotoxicity. Mechanistically, LCD was revealed to reduce survival protein expression and to upregulate death receptor 5 (DR5) expressions. Silencing DR5 blocked the ability of LCD to sensitize cells to TRAIL-mediated apoptosis. LCD increased CCAAT/enhancer-binding protein homologous protein (CHOP) expression in breast cancer cells. Knockdown of CHOP attenuated DR5 upregulation and apoptosis triggered by cotreatment with LCD and TRAIL. Furthermore, LCD suppressed the phosphorylation of extracellular signal-regulated kinase and promoted the phosphorylation of c-Jun amino-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Pretreatment with JNK inhibitor SP600125 or p38 MAPK inhibitor SB203580 abolished the upregulation of DR5 and CHOP, and also attenuated LCD plus TRAIL-induced cleavage of poly(ADP-ribose) polymerase. Overall, our results show that LCD exerts cytotoxic effects on breast cancer cells and arguments TRAIL-mediated apoptosis by inhibiting survival protein expression and upregulating DR5 in a JNK/p38 MAPK-CHOP-dependent manner.
Collapse
Affiliation(s)
- Yunyun Zhang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Linlin Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Chuxuan Dong
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Yahui Zhuang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Gangping Hao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Fengze Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
- Center Laboratory, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| |
Collapse
|
33
|
Ren J, Huang J, Yang Z, Sun M, Yang J, Lin C, Jin F, Liu Y, Tang L, Hu J, Wei X, Chen X, Yuan Z, Yang Z, Chen Y, Zhang L. Cytoplasmic TP53INP2 acts as an apoptosis partner in TRAIL treatment: the synergistic effect of TRAIL with venetoclax in TP53INP2-positive acute myeloid leukemia. J Exp Clin Cancer Res 2024; 43:176. [PMID: 38909249 PMCID: PMC11193246 DOI: 10.1186/s13046-024-03100-0] [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: 03/30/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematopoietic malignancy with poor outcomes, especially in older AML patients. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered a promising anticancer drug because it selectively induces the extrinsic apoptosis of tumor cells without affecting normal cells. However, clinical trials have shown that the responses of patients to TRAIL are significantly heterogeneous. It is necessary to explore predictable biomarkers for the preselection of AML patients with better responsiveness to TRAIL. Here, we investigated the critical role of tumor protein p53 inducible nuclear protein 2 (TP53INP2) in the AML cell response to TRAIL treatment. METHODS First, the relationship between TP53INP2 and the sensitivity of AML cells to TRAIL was determined by bioinformatics analysis of Cancer Cell Line Encyclopedia datasets, Cell Counting Kit-8 assays, flow cytometry (FCM) and cell line-derived xenograft (CDX) mouse models. Second, the mechanisms by which TP53INP2 participates in the response to TRAIL were analyzed by Western blot, ubiquitination, coimmunoprecipitation and immunofluorescence assays. Finally, the effect of TRAIL alone or in combination with the BCL-2 inhibitor venetoclax (VEN) on cell survival was explored using colony formation and FCM assays, and the effect on leukemogenesis was further investigated in a patient-derived xenograft (PDX) mouse model. RESULTS AML cells with high TP53INP2 expression were more sensitive to TRAIL in vitro and in vivo. Gain- and loss-of-function studies demonstrated that TP53INP2 significantly enhanced TRAIL-induced apoptosis, especially in AML cells with nucleophosmin 1 (NPM1) mutations. Mechanistically, cytoplasmic TP53INP2 maintained by mutant NPM1 functions as a scaffold bridging the ubiquitin ligase TRAF6 to caspase-8 (CASP 8), thereby promoting the ubiquitination and activation of the CASP 8 pathway. More importantly, simultaneously stimulating extrinsic and intrinsic apoptosis signaling pathways with TRAIL and VEN showed strong synergistic antileukemic activity in AML cells with high levels of TP53INP2. CONCLUSION Our findings revealed that TP53INP2 is a predictor of responsiveness to TRAIL treatment and supported a potentially individualized therapeutic strategy for TP53INP2-positive AML patients.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Animals
- Mice
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Apoptosis/drug effects
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Drug Synergism
- Cell Line, Tumor
- Nucleophosmin
- Xenograft Model Antitumor Assays
- Cytoplasm/metabolism
- Female
- Nuclear Proteins
Collapse
Affiliation(s)
- Jun Ren
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Junpeng Huang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zailin Yang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Minghui Sun
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jing Yang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Can Lin
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Fangfang Jin
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yongcan Liu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Lisha Tang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiayuan Hu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xingyu Wei
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyi Chen
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zihao Yuan
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanmeng Chen
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
34
|
D’Amico M, De Amicis F. Challenges of Regulated Cell Death: Implications for Therapy Resistance in Cancer. Cells 2024; 13:1083. [PMID: 38994937 PMCID: PMC11240625 DOI: 10.3390/cells13131083] [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: 04/22/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Regulated cell death, a regulatory form of cell demise, has been extensively studied in multicellular organisms. It plays a pivotal role in maintaining organismal homeostasis under normal and pathological conditions. Although alterations in various regulated cell death modes are hallmark features of tumorigenesis, they can have divergent effects on cancer cells. Consequently, there is a growing interest in targeting these mechanisms using small-molecule compounds for therapeutic purposes, with substantial progress observed across various human cancers. This review focuses on summarizing key signaling pathways associated with apoptotic and autophagy-dependent cell death. Additionally, it explores crucial pathways related to other regulated cell death modes in the context of cancer. The discussion delves into the current understanding of these processes and their implications in cancer treatment, aiming to illuminate novel strategies to combat therapy resistance and enhance overall cancer therapy.
Collapse
Affiliation(s)
- Maria D’Amico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
35
|
Švajger U, Kamenšek U. Interleukins and interferons in mesenchymal stromal stem cell-based gene therapy of cancer. Cytokine Growth Factor Rev 2024; 77:76-90. [PMID: 38508954 DOI: 10.1016/j.cytogfr.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
The tumor microenvironment is importantly shaped by various cytokines, where interleukins (ILs) and interferons (IFNs) shape the balance of immune activity within tumor niche and associated lymphoid organs. Their importance in activation and tuning of both innate and adaptive immune responses prompted their use in several clinical trials, albeit with limited therapeutic efficacy and risk of toxicity due to systemic administration. Increasing preclinical evidence suggests that local delivery of ILs and IFNs could significantly increase their effectiveness, while simultaneously attenuate the known side effects and issues related to their biological activity. A prominent way to achieve this is to use cell-based delivery vehicles. For this purpose, mesenchymal stromal stem cells (MSCs) are considered an almost ideal candidate. Namely, MSCs can be obtained in large quantities and from obtainable sources (e.g. umbilical cord or adipose tissue), their ex vivo expansion is relatively straightforward compared to other cell types and they possess very low immunogenicity making them suitable for allogeneic use. Importantly, MSCs have shown an intrinsic capacity to respond to tumor-directed chemotaxis. This review provides a focused and detailed discussion on MSC-based gene therapy using ILs and IFNs, engineering techniques and insights on potential future advancements.
Collapse
Affiliation(s)
- Urban Švajger
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Šlajmerjeva Ulica 6, Ljubljana SI-1000, Slovenia; Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, Ljubljana SI-1000, Slovenia.
| | - Urška Kamenšek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, Ljubljana SI-1000, Slovenia; Biotechnical Faculty, University of Ljubljana, Jamnikarjeva Ulica 101, Ljubljana SI-1000, Slovenia
| |
Collapse
|
36
|
Yu Z, Shi J, Fang Y, Zhao Y, Xu A, Li N. Developing innovative strategies of tumor‑infiltrating lymphocyte therapy for tumor treatment. Oncol Rep 2024; 51:85. [PMID: 38666543 PMCID: PMC11082634 DOI: 10.3892/or.2024.8744] [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: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer is the main cause of global mortality, and thus far, effective therapeutic strategies for cancer treatment are in high demand. Adoptive transfer of tumor‑infiltrating lymphocytes (TILs) represents a promising avenue in immunotherapy for the management of malignancies. The clinical safety and efficacy of TIL‑based therapy have been established through numerous rigorous clinical trials. However, the efficacy of TIL infusion in inducing an anti‑tumor response is limited to a subset of clinical patients with cancer. Therefore, there is an urgent need to develop innovative strategies aimed at enhancing the effectiveness of TIL‑based therapy. In the present review, the developmental history of TIL‑based therapy was systematically summarized and analyzed, while also presenting a unique perspective on enhancing the multi‑dimensional anti‑tumor capabilities of TILs. The insight and conclusions presented in this review may contribute to improving the efficacy of TIL‑based therapy and expediting its development.
Collapse
Affiliation(s)
- Zhongjie Yu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Jianhua Shi
- Phase I Clinical Research Center Affiliated, Linyi Tumor Hospital, Linyi, Shandong 276000, P.R. China
| | - Yuan Fang
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Yi Zhao
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Aotian Xu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Ning Li
- Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
37
|
Dhaouadi S, Bouhaouala-Zahar B, Orend G. Tenascin-C targeting strategies in cancer. Matrix Biol 2024; 130:1-19. [PMID: 38642843 DOI: 10.1016/j.matbio.2024.04.002] [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: 12/20/2023] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
Tenascin-C (TNC) is a matricellular and multimodular glycoprotein highly expressed under pathological conditions, especially in cancer and chronic inflammatory diseases. Since a long time TNC is considered as a promising target for diagnostic and therapeutic approaches in anti-cancer treatments and was already extensively targeted in clinical trials on cancer patients. This review provides an overview of the current most advanced strategies used for TNC detection and anti-TNC theranostic approaches including some advanced clinical strategies. We also discuss novel treatment protocols, where targeting immune modulating functions of TNC could be center stage.
Collapse
Affiliation(s)
- Sayda Dhaouadi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis el Manar, Tunis, Tunisia
| | - Gertraud Orend
- INSERM U1109, The Tumor Microenvironment laboratory, Université Strasbourg, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
38
|
Zhang X, Taylor H, Valdivia A, Dasari R, Buckley A, Bonacquisti E, Nguyen J, Kanchi K, Corcoran DL, Herring LE, Steindler DA, Baldwin A, Hingtgen S, Satterlee AB. Auto-loaded TRAIL-exosomes derived from induced neural stem cells for brain cancer therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595724. [PMID: 38854085 PMCID: PMC11160660 DOI: 10.1101/2024.05.24.595724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Transdifferentiation (TD), a somatic cell reprogramming process that eliminates pluripotent intermediates, creates cells that are ideal for personalized anti-cancer therapy. Here, we provide the first evidence that extracellular vesicles (EVs) from TD-derived induced neural stem cells (Exo-iNSCs) are an efficacious treatment strategy for brain cancer. We found that genetically engineered iNSCs generated EVs loaded with the tumoricidal gene product TRAIL at nearly twice the rate as their parental fibroblasts, and the TRAIL produced by iNSCs were naturally loaded into the lumen of EVs and arrayed across their outer membrane (Exo-iNSC-TRAIL). Uptake studies in ex vivo organotypic brain slice cultures showed Exo-iNSC-TRAIL selectively accumulates within tumor foci, and co-culture assays showed that Exo-iNSC-TRAIL killed metastatic and primary brain cancer cells more effectively than free TRAIL. In an orthotopic mouse model of brain cancer, Exo-iNSC-TRAIL reduced breast-to-brain tumor xenografts around 3000-fold greater than treatment with free TRAIL, with all Exo-iNSC-TRAIL treated animals surviving through 90 days post-treatment. In additional in vivo testing against aggressive U87 and invasive GBM8 glioblastoma tumors, Exo-iNSC-TRAIL also induced a statistically significant increase in survival. These studies establish a new easily generated, stable, tumor-targeted EV to efficaciously treat multiple forms of brain cancer.
Collapse
|
39
|
Valdez Capuccino L, Kleitke T, Szokol B, Svajda L, Martin F, Bonechi F, Krekó M, Azami S, Montinaro A, Wang Y, Nikolov V, Kaiser L, Bonasera D, Saggau J, Scholz T, Schmitt A, Beleggia F, Reinhardt HC, George J, Liccardi G, Walczak H, Tóvári J, Brägelmann J, Montero J, Sos ML, Őrfi L, Peltzer N. CDK9 inhibition as an effective therapy for small cell lung cancer. Cell Death Dis 2024; 15:345. [PMID: 38769311 PMCID: PMC11106072 DOI: 10.1038/s41419-024-06724-4] [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: 11/13/2023] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Treatment-naïve small cell lung cancer (SCLC) is typically susceptible to standard-of-care chemotherapy consisting of cisplatin and etoposide recently combined with PD-L1 inhibitors. Yet, in most cases, SCLC patients develop resistance to first-line therapy and alternative therapies are urgently required to overcome this resistance. In this study, we tested the efficacy of dinaciclib, an FDA-orphan drug and inhibitor of the cyclin-dependent kinase (CDK) 9, among other CDKs, in SCLC. Furthermore, we report on a newly developed, highly specific CDK9 inhibitor, VC-1, with tumour-killing activity in SCLC. CDK9 inhibition displayed high killing potential in a panel of mouse and human SCLC cell lines. Mechanistically, CDK9 inhibition led to a reduction in MCL-1 and cFLIP anti-apoptotic proteins and killed cells, almost exclusively, by intrinsic apoptosis. While CDK9 inhibition did not synergise with chemotherapy, it displayed high efficacy in chemotherapy-resistant cells. In vivo, CDK9 inhibition effectively reduced tumour growth and improved survival in both autochthonous and syngeneic SCLC models. Together, this study shows that CDK9 inhibition is a promising therapeutic agent against SCLC and could be applied to chemo-refractory or resistant SCLC.
Collapse
Affiliation(s)
- L Valdez Capuccino
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- CECAD Research Center, University of Cologne, Cologne, Germany
| | - T Kleitke
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- CECAD Research Center, University of Cologne, Cologne, Germany
| | - B Szokol
- Vichem Chemie Research Ltd., Veszprém, Hungary
| | - L Svajda
- Department of Experimental Pharmacology, and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - F Martin
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036, Barcelona, Spain
| | - F Bonechi
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- CECAD Research Center, University of Cologne, Cologne, Germany
| | - M Krekó
- Vichem Chemie Research Ltd., Veszprém, Hungary
- Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, Hungary
| | - S Azami
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- CECAD Research Center, University of Cologne, Cologne, Germany
| | - A Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Y Wang
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- CECAD Research Center, University of Cologne, Cologne, Germany
| | - V Nikolov
- CECAD Research Center, University of Cologne, Cologne, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - L Kaiser
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
| | - D Bonasera
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
- Genome instability, inflammation and cell death laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - J Saggau
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
- Genome instability, inflammation and cell death laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - T Scholz
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
| | - A Schmitt
- University Hospital of Cologne, Medical Faculty, Department I for Internal Medicine, Cologne, Germany
| | - F Beleggia
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University Hospital of Cologne, Medical Faculty, Department I for Internal Medicine, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Mildred Scheel School of Oncology Cologne, Cologne, Germany
| | - H C Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - J George
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine and University Hospital Cologne, University Hospital of Cologne, Cologne, Germany
| | - G Liccardi
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- Genome instability, inflammation and cell death laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - H Walczak
- CECAD Research Center, University of Cologne, Cologne, Germany
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
- Cell death, inflammation and immunity laboratory, Institute of Biochemistry I, Centre for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - J Tóvári
- Department of Experimental Pharmacology, and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - J Brägelmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Mildred Scheel School of Oncology Cologne, Cologne, Germany
| | - J Montero
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08036, Barcelona, Spain
| | - M L Sos
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany
- Division for Translational Oncology, German Cancer Research Center (DKFZ), The German Consortium for Translational Cancer Research (DKTK), München Partner Site, Ludwig-Maximilian University München, Munich, Germany
| | - L Őrfi
- Vichem Chemie Research Ltd., Veszprém, Hungary
- Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, Hungary
| | - N Peltzer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany.
- CECAD Research Center, University of Cologne, Cologne, Germany.
| |
Collapse
|
40
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Extracellular Vesicles in Breast Cancer: From Intercellular Communication to Therapeutic Opportunities. Pharmaceutics 2024; 16:654. [PMID: 38794316 PMCID: PMC11125876 DOI: 10.3390/pharmaceutics16050654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer, a multifaceted and heterogeneous disease, poses significant challenges in terms of understanding its intricate resistance mechanisms and devising effective therapeutic strategies. This review provides a comprehensive overview of the intricate landscape of extracellular vesicles (EVs) in the context of breast cancer, highlighting their diverse subtypes, biogenesis, and roles in intercellular communication within the tumour microenvironment (TME). The discussion spans various aspects, from EVs and stromal cells in breast cancer to their influence on angiogenesis, immune response, and chemoresistance. The impact of EV production in different culture systems, including two dimensional (2D), three dimensional (3D), and organoid models, is explored. Furthermore, this review delves into the therapeutic potential of EVs in breast cancer, presenting emerging strategies such as engineered EVs for gene delivery, nanoplatforms for targeted chemotherapy, and disrupting tumour derived EVs as a treatment approach. Understanding these complex interactions of EV within the breast cancer milieu is crucial for identifying resistance mechanisms and developing new therapeutic targets.
Collapse
Affiliation(s)
- Barathan Muttiah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
41
|
Liu X, Wang LL, Duan CY, Rong YR, Liang YQ, Zhu QX, Hao GP, Wang FZ. Daurisoline inhibits proliferation, induces apoptosis, and enhances TRAIL sensitivity of breast cancer cells by upregulating DR5. Cell Biol Int 2024. [PMID: 38563483 DOI: 10.1002/cbin.12162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
Daurisoline (DS) is an isoquinoline alkaloid that exerts anticancer activities in various cancer cells. However, the underlying mechanisms through which DS affects the survival of breast cancer cells remain poorly understood. Therefore, the present study was undertaken to investigate the potential anticancer effect of DS on breast cancer cells and reveal the mechanism underlying the enhanced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by DS. Cell counting kit-8 (CCK-8) and 5-ethynyl-2-deoxyuridine (EdU) assay were used to evaluate the ability of cell proliferation. Flow cytometry was selected to examine the cell cycle distribution. TUNEL assay was used to detect the cell apoptosis. The protein expression was measured by Western blot analysis. DS was found to reduce the cell viability and suppress the proliferation of MCF-7 and MDA-MB-231 cells by causing G1 phase cell cycle arrest. DS could trigger apoptosis by promoting the cleavage of caspase-8 and PARP. The phosphorylation of ERK, JNK, and p38MAPK was upregulated clearly following DS treatment. Notably, SP600125 (JNK inhibitor) pretreatment significantly abrogated DS-induced PARP cleavage. DS inactivated Akt/mTOR and Wnt/β-catenin signaling pathway and upregulated the expression of ER stress-related proteins. Additionally, DS amplified TRAIL-caused viability reduction and apoptosis in breast cancer cells. Mechanismly, DS upregulated the protein level of DR4 and DR5, and knockdown of DR5 attenuated the cotreatment-induced cleavage of PARP. Inhibition of JNK could block DS-induced upregulation of DR5. This study provides valuable insights into the mechanisms of DS inhibiting cell proliferation, triggering apoptosis, and enhancing TRAIL sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Lin-Lin Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Cun-Yu Duan
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Yan-Ru Rong
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Ya-Qi Liang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Qing-Xiang Zhu
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Gang-Ping Hao
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, People's Republic of China
- Center Laboratory, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| |
Collapse
|
42
|
Dhillon S. Aponermin: First Approval. Drugs 2024; 84:459-466. [PMID: 38441805 DOI: 10.1007/s40265-024-02004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Aponermin () is a recombinant circularly permuted human tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) developed by Beijing Sunbio Biotech (a wholly owned subsidiary of Wuhan Hiteck Biological Pharma CO., LTD) for the treatment of multiple myeloma. Aponermin binds to and activates the death receptors 4 and/or 5 on tumour cells, triggering intracellular caspase reactions and inducing apoptosis, thereby exerting antitumor effects. In November 2023, aponermin in combination with thalidomide and dexamethasone received its first approval in China for the treatment of patients with relapsed or refractory multiple myeloma who have received at least two prior therapies. This article summarizes the milestones in the development of aponermin leading to this first approval for relapsed or refractory multiple myeloma.
Collapse
Affiliation(s)
- Sohita Dhillon
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
43
|
Boccellato C, Rehm M. TRAIL-induced apoptosis and proteasomal activity - Mechanisms, signalling and interplay. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119688. [PMID: 38368955 DOI: 10.1016/j.bbamcr.2024.119688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/20/2024]
Abstract
Programmed cell death, in particular apoptosis, is essential during development and tissue homeostasis, and also is the primary strategy to induce cancer cell death by cytotoxic therapies. Precision therapeutics targeting TRAIL death receptors are being evaluated as novel anti-cancer agents, while in parallel highly specific proteasome inhibitors have gained approval as drugs. TRAIL-dependent signalling and proteasomal control of cellular proteostasis are intricate processes, and their interplay can be exploited to enhance therapeutic killing of cancer cells in combination therapies. This review provides detailed insights into the complex signalling of TRAIL-induced pathways and the activities of the proteasome. It explores their core mechanisms of action, pharmaceutical druggability, and describes how their interplay can be strategically leveraged to enhance cell death responses in cancer cells. Offering this comprehensive and timely overview will allow to navigate the complexity of the processes governing cell death mechanisms in TRAIL- and proteasome inhibitor-based treatment conditions.
Collapse
Affiliation(s)
- Chiara Boccellato
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany.
| | - Markus Rehm
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart 70569, Germany; University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart 70569, Germany.
| |
Collapse
|
44
|
Sittiju P, Wudtiwai B, Chongchai A, Hajitou A, Kongtawelert P, Pothacharoen P, Suwan K. Bacteriophage-based particles carrying the TNF-related apoptosis-inducing ligand (TRAIL) gene for targeted delivery in hepatocellular carcinoma. NANOSCALE 2024; 16:6603-6617. [PMID: 38470366 PMCID: PMC10977282 DOI: 10.1039/d3nr05660k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The TRAIL (Tumour Necrosis Factor-Related Apoptosis-Inducing Ligand) is a promising candidate for cancer treatment due to its unique ability to selectively induce programmed cell death, or apoptosis, in cancer cells while sparing healthy ones. This selectivity arises from the preferential binding of the TRAIL to death receptors on cancer cells, triggering a cascade of events that lead to their demise. However, significant limitations in using the TRAIL for cancer treatment are the administration of the TRAIL protein that can potentially lead to tissue toxicity (off-target) and the short half-life of the TRAIL in the body which may necessitate frequent and sustained administration; these can pose logistical challenges for long-term treatment regimens. We have devised a novel approach for surmounting these limitations by introducing the TRAIL gene directly into cancer cells, enabling them to produce the TRAIL locally and subsequently trigger apoptosis. A novel gene delivery system such as a bacteriophage-based particle TPA (transmorphic phage/AAV) was utilized to address these limitations. TPA is a hybrid M13 filamentous bacteriophage particle encapsulating a therapeutic gene cassette with inverted terminal repeats (ITRs) from adeno-associated viruses (AAVs). The particle also showed a tumour targeting ligand, CDCRGDCFC (RGD4C), on its capsid (RGD4C.TPA) to target the particle to cancer cells. RGD4C selectively binds to αvβ3 and αvβ5 integrins overexpressed on the surface of most of the cancer cells but is barely present on normal cells. Hepatocellular carcinoma (HCC) was chosen as a model because it has one of the lowest survival rates among cancers. We demonstrated that human HCC cell lines (Huh-7 and HepG2) express αvβ5 integrin receptors on their surface. These HCC cells also express death receptors and TRAIL-binding receptors. We showed that the targeted TPA particle carrying the transmembrane TRAIL gene (RGD4C.TPA-tmTRAIL) selectively and efficiently delivered the tmTRAIL gene to HCC cells resulting in the production of tmTRAIL from transduced cells and subsequently induced apoptotic death of HCC cells. This tumour-targeted particle can be an excellent candidate for the targeted gene therapy of HCC.
Collapse
Affiliation(s)
- Pattaralawan Sittiju
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Benjawan Wudtiwai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Aitthiphon Chongchai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Amin Hajitou
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Keittisak Suwan
- Cancer Phage Therapy Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
45
|
Chyuan IT, Liao HJ, Tan TH, Chuang HC, Chu YC, Pan MH, Wu CS, Chu CL, Sheu BC, Hsu PN. Association of TRAIL receptor with phosphatase SHP-1 enables repressing T cell receptor signaling and T cell activation through inactivating Lck. J Biomed Sci 2024; 31:33. [PMID: 38532423 DOI: 10.1186/s12929-024-01023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND T cell receptor (TCR) signaling and T cell activation are tightly regulated by gatekeepers to maintain immune tolerance and avoid autoimmunity. The TRAIL receptor (TRAIL-R) is a TNF-family death receptor that transduces apoptotic signals to induce cell death. Recent studies have indicated that TRAIL-R regulates T cell-mediated immune responses by directly inhibiting T cell activation without inducing apoptosis; however, the distinct signaling pathway that regulates T cell activation remains unclear. In this study, we screened for intracellular TRAIL-R-binding proteins within T cells to explore the novel signaling pathway transduced by TRAIL-R that directly inhibits T cell activation. METHODS Whole-transcriptome RNA sequencing was used to identify gene expression signatures associated with TRAIL-R signaling during T cell activation. High-throughput screening with mass spectrometry was used to identify the novel TRAIL-R binding proteins within T cells. Co-immunoprecipitation, lipid raft isolation, and confocal microscopic analyses were conducted to verify the association between TRAIL-R and the identified binding proteins within T cells. RESULTS TRAIL engagement downregulated gene signatures in TCR signaling pathways and profoundly suppressed phosphorylation of TCR proximal tyrosine kinases without inducing cell death. The tyrosine phosphatase SHP-1 was identified as the major TRAIL-R binding protein within T cells, using high throughput mass spectrometry-based proteomics analysis. Furthermore, Lck was co-immunoprecipitated with the TRAIL-R/SHP-1 complex in the activated T cells. TRAIL engagement profoundly inhibited phosphorylation of Lck (Y394) and suppressed the recruitment of Lck into lipid rafts in the activated T cells, leading to the interruption of proximal TCR signaling and subsequent T cell activation. CONCLUSIONS TRAIL-R associates with phosphatase SHP-1 and transduces a unique and distinct immune gatekeeper signal to repress TCR signaling and T cell activation via inactivating Lck. Thus, our results define TRAIL-R as a new class of immune checkpoint receptors for restraining T cell activation, and TRAIL-R/SHP-1 axis can serve as a potential therapeutic target for immune-mediated diseases.
Collapse
Affiliation(s)
- I-Tsu Chyuan
- School of Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Medical Research, Cathay General Hospital, Taipei, 10630, Taiwan
- Department of Internal Medicine, Cathay General Hospital, Taipei, 10630, Taiwan
| | - Hsiu-Jung Liao
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taipei, 22000, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, 35053, Taiwan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Yu-Chuan Chu
- Department of Medical Research, Cathay General Hospital, Taipei, 10630, Taiwan
| | - Meng-Hsun Pan
- Department of Medical Research, Cathay General Hospital, Taipei, 10630, Taiwan
| | - Chien-Sheng Wu
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taipei, 22000, Taiwan
| | - Ching-Liang Chu
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Bor-Ching Sheu
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, 10002, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Ping-Ning Hsu
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Department of Internal Medicine and Graduate Institute of Immunology, College of Medicine, National Taiwan University, 1 Jen-Ai Rd., Sec. 1, Taipei, 10051, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, 10002, Taiwan.
| |
Collapse
|
46
|
Kobyakova MI, Senotov AS, Krasnov KS, Lomovskaya YV, Odinokova IV, Kolotova AA, Ermakov AM, Zvyagina AI, Fadeeva IS, Fetisova EI, Akatov VS, Fadeev RS. Pro-Inflammatory Activation Suppresses TRAIL-induced Apoptosis of Acute Myeloid Leukemia Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:431-440. [PMID: 38648763 DOI: 10.1134/s0006297924030040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 04/25/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is a promising agent for treatment of AML due to its specific apoptosis-inducing effect on tumor cells but not normal cells. However, emergence of resistance to TRAIL in the AML cells limits its potential as an antileukemic agent. Previously, we revealed increase in the resistance of the human AML THP-1 cells to the TRAIL-induced death during their LPS-dependent proinflammatory activation and in the in vitro model of LPS-independent proinflammatory activation - in a long-term high-density cell culture. In this study, we investigated mechanisms of this phenomenon using Western blot analysis, caspase 3 enzymatic activity analysis, quantitative reverse transcription-PCR, and flow cytometry. The results showed that the increased resistance to the TRAIL-induced cell death of AML THP-1 cells during their pro-inflammatory activation is associated with the decrease in the surface expression of the proapoptotic receptors TRAIL-R1/DR4 and TRAIL-R2/DR5, as well as with the increased content of members of the IAPs family - Livin and cIAP2. The results of this article open up new insights into the role of inflammation in formation of the resistance of AML cells to the action of mediators of antitumor immunity, in particular TRAIL.
Collapse
Affiliation(s)
- Margarita I Kobyakova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
- Institute of Clinical and Experimental Lymphology, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630060, Russia
| | - Anatoly S Senotov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Kirill S Krasnov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Yana V Lomovskaya
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Irina V Odinokova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anastasia A Kolotova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Artem M Ermakov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Alena I Zvyagina
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Irina S Fadeeva
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Elena I Fetisova
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir S Akatov
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
47
|
Garcia-Saez AJ. Dying in self-defense: cell death signaling in animals and plants. Cell Death Differ 2024; 31:263-264. [PMID: 37563260 PMCID: PMC10923845 DOI: 10.1038/s41418-023-01206-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 07/11/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Affiliation(s)
- Ana J Garcia-Saez
- Institute of Genetics, CECAD, University of Cologne, Cologne, Germany.
| |
Collapse
|
48
|
He Z, Li F, Zhang X, Gao D, Zhang Z, Xu R, Cao X, Shan Q, Ren Z, Liu Y, Xu Z. Knockdown of EIF4G1 in NSCLC induces CXCL8 secretion. Front Pharmacol 2024; 15:1346383. [PMID: 38405671 PMCID: PMC10884238 DOI: 10.3389/fphar.2024.1346383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 02/27/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung tumor; however, we lack effective early detection indicators and therapeutic targets. Eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) is vital to initiate protein synthesis, acting as a scaffolding protein for the eukaryotic protein translation initiation factor complex, EIF4F, which regulates protein synthesis together with EIF4A, EIF4E, and other translation initiation factors. However, EIF4G1's function in NSCLC cancer is unclear. Herein, transcriptome sequencing showed that knockdown of EIF4G1 in H1299 NSCLC cells upregulated the expression of various inflammation-related factors. Inflammatory cytokines were also significantly overexpressed in NSCLC tumor tissues, among which CXCL8 (encoding C-X-C motif chemokine ligand 8) showed the most significant changes in both in the transcriptome sequencing data and tumor tissues. We revealed that EIF4G1 regulates the protein level of TNF receptor superfamily member 10a (TNFRSF10A) resulting in activation of the mitogen activated protein kinase (MAPK) and nuclear factor kappa B (NFκB) pathways, which induces CXCL8 secretion, leading to targeted chemotaxis of immune cells. We verified that H1299 cells with EIF4G1 knockdown showed increased chemotaxis compared with the control group and promoted increased chemotaxis of macrophages. These data suggested that EIF4G1 is an important molecule in the inflammatory response of cancer tissues in NSCLC.
Collapse
Affiliation(s)
- Ziyang He
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyi Li
- Shanghai East Hospital, Postgraduate Training Base of Jinzhou Medical University, Shanghai, China
| | - Xinyi Zhang
- Shanghai East Hospital, Postgraduate Training Base of Jinzhou Medical University, Shanghai, China
| | - Dacheng Gao
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiwen Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Xu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xingguo Cao
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiyuan Shan
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Ren
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yali Liu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zengguang Xu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
49
|
Luo Y, Hao H, Wang Z, Ong CY, Dutcher R, Xu Y, Liu J, Pedersen LC, Xu D. Heparan sulfate promotes TRAIL-induced tumor cell apoptosis. eLife 2024; 12:RP90192. [PMID: 38265424 PMCID: PMC10945736 DOI: 10.7554/elife.90192] [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] [Indexed: 01/25/2024] Open
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) is a potent inducer of tumor cell apoptosis through TRAIL receptors. While it has been previously pursued as a potential anti-tumor therapy, the enthusiasm subsided due to unsuccessful clinical trials and the fact that many tumors are resistant to TRAIL. In this report, we identified heparan sulfate (HS) as an important regulator of TRAIL-induced apoptosis. TRAIL binds HS with high affinity (KD = 73 nM) and HS induces TRAIL to form higher-order oligomers. The HS-binding site of TRAIL is located at the N-terminus of soluble TRAIL, which includes three basic residues. Binding to cell surface HS plays an essential role in promoting the apoptotic activity of TRAIL in both breast cancer and myeloma cells, and this promoting effect can be blocked by heparin, which is commonly administered to cancer patients. We also quantified HS content in several lines of myeloma cells and found that the cell line showing the most resistance to TRAIL has the least expression of HS, which suggests that HS expression in tumor cells could play a role in regulating sensitivity towards TRAIL. We also discovered that death receptor 5 (DR5), TRAIL, and HS can form a ternary complex and that cell surface HS plays an active role in promoting TRAIL-induced cellular internalization of DR5. Combined, our study suggests that TRAIL-HS interactions could play multiple roles in regulating the apoptotic potency of TRAIL and might be an important point of consideration when designing future TRAIL-based anti-tumor therapy.
Collapse
Affiliation(s)
- Yin Luo
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New YorkBuffaloUnited States
| | - Huanmeng Hao
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New YorkBuffaloUnited States
| | - Zhangjie Wang
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North CarolinaChapel HillUnited States
| | - Chih Yean Ong
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New YorkBuffaloUnited States
| | - Robert Dutcher
- Macromolecular Structure Group, Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of HealthResearch Triangle ParkUnited States
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North CarolinaChapel HillUnited States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North CarolinaChapel HillUnited States
| | - Lars C Pedersen
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of HealthResearch Triangle ParkUnited States
| | - Ding Xu
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New YorkBuffaloUnited States
| |
Collapse
|
50
|
Zhang Z, King MR. Tuning of TRAIL clustering on the surface of nanoscale liposomes by phase separation. NANOSCALE ADVANCES 2024; 6:402-405. [PMID: 38235079 PMCID: PMC10790813 DOI: 10.1039/d3na00841j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024]
Abstract
Phase-separated liposomes were used to formulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a protein that selectively kills cancer cells while sparing most healthy ones. By controlling the average number of TRAIL molecules per liposome, we demonstrate the ability to tune the formation of TRAIL clusters and their resulting apoptotic activity.
Collapse
Affiliation(s)
- Zhenjiang Zhang
- Department of Biomedical Engineering, Vanderbilt University 2414 Highland Ave Nashville TN 37212 USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University 2414 Highland Ave Nashville TN 37212 USA
| |
Collapse
|