1
|
Gao Y, Yu B, Li L, Zhang J, Zhao T, Feng X, Hirayama R, Di C, Zhang Y, Ye Y, Li Y, Li Q, Jin X. mtDNA/RNA boosts radiation-induced abscopal effect via M1 macrophage polarization-promoted IFN-β-dependent inflammatory response. Int Immunopharmacol 2025; 155:114673. [PMID: 40245773 DOI: 10.1016/j.intimp.2025.114673] [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: 01/20/2025] [Revised: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
The radiation-induced abscopal effect (RIAE) can suppress distal metastatic lesions and elicit a systemic anti-tumor response; however, the underlying mechanisms remain to be fully elucidated. Current research has shown that autophagy promotes the production of IFN-β by regulating mitochondrial DNA (mtDNA), thereby contributing to the modulation of RIAE. Nevertheless, the downstream pathways through which IFN-β influences RIAE require further investigation. In this study, we observed accumulation of an abundance of mtDNA in the cytosol of mammary tumor cells following RT, along with the presence of mitochondrial RNA (mtRNA). These molecules activated the cGAS-STING and RIG-I-MAVS signaling pathways, respectively, thereby synergistically promoting the production of IFN-β and secretion into the extracellular matrix. Subsequently, IFN-β facilitated the polarization of macrophages in distant non-irradiated tumor microenvironment towards the M1 phenotype through activating STAT1. Furthermore, our findings indicate that high linear energy transfer (LET) carbon ions are significantly more effective in inducing the production of IFN-β and promoting macrophage polarization compared to low-LET X-rays. Thus, our findings provide insights into the intricate mechanisms by which mtDNA/RNA and IFN-β mediate RIAE, suggesting that IFN-β could be a promising target for provoking RT immunogenicity in patients with breast cancer and high-LET radiation might effectively elicit RIAE.
Collapse
Affiliation(s)
- Yuting Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; School of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China
| | - Boyi Yu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linjing Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahao Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ting Zhao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Xianglong Feng
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ryoichi Hirayama
- National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanshan Zhang
- Gansu Wuwei Tumor Hospital, Wuwei 733000, Gansu Province, China
| | - Yancheng Ye
- Gansu Wuwei Tumor Hospital, Wuwei 733000, Gansu Province, China
| | - Yuan Li
- School of Life Sciences, Northwest Normal University, Gansu Province, Lanzhou 730070, China.
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Zhao H, Gao H, Kang J, Chen J, Ouyang H, Chen P, Althaf HS, Alrubie TM, Narendra M, Wang K. Enhancement of radiation effects by Plantago major in MCF-7 human breast cancer: an animal study. Discov Oncol 2025; 16:764. [PMID: 40366497 DOI: 10.1007/s12672-025-02436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
PURPOSE We aimed to evaluate the potential effects of Plantago major (PM) to enhance the radiosensitivity of human breast adenocarcinoma cell line (MCF-7) in an animal model. MATERIALS AND METHODS Seventy-two female Balb/c mice were divided into nine groups (8 mice per group) as follows: MCF-7 breast cancer control group, MCF-7+low dose of PM (1000 mg/kg), MCF-7+high dose of PM (1500 mg/kg), MCF-7+3 Gy superficial X-ray, MCF-7+5 Gy superficial X-ray, MCF-7+1000&1500 mg/kg of PM with 3 and 5 Gy irradiations. For each treatment group, micronuclei in 500 binucleate MCF-7 cells from a minimum of three experiments were counted. The alkaline comet assay was used to calculate % DNA in the tail and % of apoptotic comets. The tumor size in the treated groups (3 mice per group) was assessed at 4- and 8-weeks post-treatment. RESULTS The number of total micronuclei and binucleated micronuclei in the PM and/or irradiation treated groups was significantly higher than in the control group (P < 0.05). Irradiation+PM resulted in a significant increase treatment effect compared to the irradiation-only groups (P < 0.01). In addition, the higher PM concentration had a significantly higher number of micronucleated binucleate cells when combined with 5 Gy irradiation (P = 0.022). Irradiation alone or in combination with PM resulted in significant increases in percentages of DNA in tail and apoptotic comet values compared to the PM-only treatment groups (P < 0.02). Combining 5 Gy of irradiation with 1000 mg/kg of PM led to a 26% reduction in tumor size (0.28±0.04 vs. 0.38 ± 0.03) after 4 weeks, and combining 5 Gy of irradiation with 1500 mg/kg of PM resulted in a 40% decrease in tumor volume after 4 weeks (0.21±0.04 vs. 0.35±0.03). CONCLUSION PM extract at both doses demonstrated an antitumor effect on induced MCF-7 breast cancer tumors, with this effect being enhanced when combined with irradiation.
Collapse
Affiliation(s)
- Hongliang Zhao
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Hongxiang Gao
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Jing Kang
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Jiexin Chen
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Huiling Ouyang
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Peiyao Chen
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China
| | - Hussain Shaik Althaf
- Department of Zoology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Turki Mayudh Alrubie
- Department of Zoology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Maddu Narendra
- Department of Biochemistry, Sri Krishnadevaraya University, Anantapur, Andhra Pradesh, India
| | - Kai Wang
- Department of Hematology & Oncology, Honghui Hospital, Xi An Jiao Tong University, Xian, 710054, People's Republic of China.
| |
Collapse
|
3
|
Sang R, Nixdorf S, Hung T, Power C, Deng F, Bui TA, Engel A, Goldys EM, Deng W. Unlocking the in vivo therapeutic potential of radiation-activated photodynamic therapy for locally advanced rectal cancer with lymph node involvement. EBioMedicine 2025; 116:105724. [PMID: 40359628 DOI: 10.1016/j.ebiom.2025.105724] [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: 10/31/2024] [Revised: 03/14/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Rectal cancer is a leading cause of cancer-related mortality worldwide. The recurrence of locally advanced rectal cancer (LARC), particularly in cases involving lymph node-positive tumours, remains a critical challenge in rectal cancer management. In this study, a therapeutic strategy, radiation-activated photodynamic therapy (RA-PDT), for the treatment of LARC with lymph node-positive tumours was developed and evaluated. METHODS RA-PDT was achieved by using a lipid-polymer hybrid nanoplatform loaded with verteporfin (VP) and functionalised with folic acid (FA) as a targeting molecule. Upon receiving a single 4 Gy fraction of radiation, VP was effectively activated, generating sufficient reactive oxygen species (ROS) to induce cancer cell death-however surrounding tissue was less affected and was spared. The efficacy of this strategy was assessed through in vitro cytotoxicity studies in HCT116 cells, as well as in orthotopic and subcutaneous mouse models. In vivo lymph node tumour progression was also evaluated. FINDINGS RA-PDT effectively generated ROS following 4 Gy irradiation and exhibited significant cytotoxicity in HCT116 cells. In vivo, this strategy largely inhibited primary tumour growth in both orthotopic and subcutaneous mouse models while also suppressing lymph node tumour progression. Surrounding tissues were minimally affected, highlighting the precision and safety of this approach. INTERPRETATION RA-PDT demonstrates potential as a safe therapeutic strategy for LARC, paving the way for its clinical translation. FUNDING This study was supported by the Australian National Health and Medical Research Council (GNT1181889), fellowship award (2019/CDF1013) from Cancer Institute NSW, Australia, the Australian Research Council Centre of Excellence for Nanoscale Biophotonics (CE140100003), UNSW SHARP funding, project grant from National Foundation for Medical Research and Innovation, Australia, International Research Training Program Scholarship (IRTP) from Australian Government, PhD Research Scholar Award from Sydney Vital Translational Cancer Research, and Translational Cancer Research Network PhD Scholarship Top-up award.
Collapse
Affiliation(s)
- Rui Sang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia; School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Sheri Nixdorf
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Tzongtyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fei Deng
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thuy Anh Bui
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Alexander Engel
- Sydney Medical School, University of Sydney, Sydney, NSW, 2050, Australia; Department of Colorectal Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
| | - Ewa M Goldys
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, University of New South Wales, Sydney, NSW, 2052, Australia.
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| |
Collapse
|
4
|
Xu S, Lin X, Jia F. NAT10-mediated N4-acetylcytosine modification promotes the progression of retinoblastoma by improving the HK1 mRNA stability to enhance glycolysis. Clinics (Sao Paulo) 2025; 80:100678. [PMID: 40344913 DOI: 10.1016/j.clinsp.2025.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 03/05/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVE Retinoblastoma (RB) is a type of intraocular tumor in childhood with a high lethality rate. N4-acetylcytosine (ac4C) modification is known to regulate multiple cancers, which is mediated by the only known ac4C writer N-Acetyltransferase 10 (NAT10). In this study, the authors aimed to reveal the mechanism of RB progression regulated by ac4C modification. METHOD Phenotypically, dot blot assay and quantitative real-time PCR were used to detect the ac4C levels and NAT10 expression in clinical samples and RB cell lines. Then, NAT10 was knocked down to assess its effect on glycolysis. Mechanically, RNA immunoprecipitation assay, immunofluorescence assay, and dual luciferase report were used to explore the mRNA modified by NAT10-mediated ac4C modification. Mice xenograft model was used to determine the effect of NAT10 on tumor growth in vivo. RESULTS The present results demonstrated that the levels of ac4C and NAT10 were increased in cancer tissues and RB cell lines. Furthermore, NAT10 knockdown inhibited the glycolysis in RB cell lines. Moreover, the authors revealed that NAT10 knockdown decreased the ac4C modification and mRNA stability of HK1, while the inhibition of glycolysis by NAT10 knockdown was reversed by HK1 overexpression. Finally, NAT10 knockdown relieved the growth of tumors in mice models. CONCLUSION The authors illustrated that NAT10 plays an important role in the progression of RB by regulating the ac4C modification on HK1 mRNA and affects its stability, which may provide a novel theoretical basis for the treatment of RB.
Collapse
Affiliation(s)
- Shan Xu
- Department of Ophthalmology, Yantai Yuhuangding Hospital, Yantai, Shandong, China.
| | - Xuming Lin
- Department of Ophthalmology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Fengling Jia
- Department of Ophthalmology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
5
|
Kovács SA, Kovács T, Lánczky A, Paál Á, Hegedűs ZI, Sayour NV, Szabó L, Kovács A, Bianchini G, Ferdinandy P, Ocana A, Varga ZV, Fekete JT, Győrffy B. Unlocking the power of immune checkpoint inhibitors: Targeting YAP1 reduces anti-PD1 resistance in skin cutaneous melanoma. Br J Pharmacol 2025. [PMID: 40324810 DOI: 10.1111/bph.70052] [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: 07/02/2024] [Revised: 02/02/2025] [Accepted: 03/15/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND PURPOSE Immune checkpoint inhibitors, such as anti-PD1, revolutionized melanoma treatment. However, resistance and low response rates remain problems. Our goal was to pinpoint actionable biomarkers of resistance to anti-PD1 treatment and verify therapeutic effectiveness in vivo. EXPERIMENTAL APPROACH Using receiver operating characteristic (ROC) and survival analysis in a database of 1434 samples, we identified the strongest resistance-associated genes. Inhibitors were evaluated in C57BL/6J mice using wild-type B16-F10, and BRAF, -PTEN, -CDKN2A-mutant YUMM1.7 melanoma cell lines. We investigated the synergistic impact of anti-PD1 therapy and yes-associated protein 1 (YAP1) inhibition by non-photoactivated Verteporfin. Tumour volume was determined at fixed cutoff points, normalized to body weights. KEY RESULTS In the anti-PD1-treated melanoma cohort, YAP1 was the strongest druggable candidate overexpressed in non-responder patients (ROC AUC = 0.699, FC = 1.8, P=1.1E-8). The baseline YAP1 expression correlated with worse progression-free survival (HR = 2.51, P=1.2E-6, FDR = 1%), and overall survival (HR = 2.15, P = 1.2E-5, FDR = 1%). In YUMM1.7, combination of Verteporfin plus anti-PD1 reduced tumour size more than anti-PD1 monotherapy (P=0.008), or control (P=0.021). There was no difference between the cohorts in B16-F10 inoculated mice. We found increased expression of YAP1 in YUMM1.7 mice compared to B16-F10. The combination therapy induced a more-immune-inflamed phenotype characterized by increased expression of T cell and M1 macrophage markers. CONCLUSIONS AND IMPLICATIONS Verteporfin with anti-PD1 exhibited antitumor potential by promoting a pro-inflammatory tumour microenvironment in melanoma. We believe that YAP1 acts as a master regulator of anti-PD1 resistance.
Collapse
Affiliation(s)
- Szonja Anna Kovács
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Oncology Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | | | - Ágnes Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsombor I Hegedűs
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Lilla Szabó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Andrea Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Alberto Ocana
- START Madrid-Fundación Jiménez Díaz (FJD) Early Phase Program, Fundación Jiménez Díaz Hospital, Madrid, Spain
- Experimental Therapeutics in Cancer Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - János Tibor Fekete
- Department of Bioinformatics, Semmelweis University, Budapest, Hungary
- Oncology Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Balázs Győrffy
- Department of Biophysics, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
6
|
Mustafa A, ArumughamIndiran M, Perumal E, Ponnala A, Rasheed DA, Ramalingam K, Shanmugham R, Karobari MI. Chemopreventive effects of chitosan nanogel with thiocolchicoside and lauric acid in chemically induced oral carcinogenesis, in a rodent model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04185-w. [PMID: 40314765 DOI: 10.1007/s00210-025-04185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025]
Abstract
There is always a quest for newer, more effective chemopreventive agents to prevent and manage oral cancer. A novel nanogel prepared using thiocolchicoside, lauric acid, and chitosan showed promising anticancer activity in KB-1 cell lines. The current manuscript aims to investigate the chemopreventive activity of chitosan nanogel with thiocolchicoside and lauric acid in chemically induced oral carcinogenesis using Wistar rats. Forty-six male Wistar rats were divided into three different groups: group I (control group), group II (cancer induction group), and group III (cancer induction with a chemopreventive agent). Male Wistar rats were given 20 μl/ml 4NQO solutions daily in their drinking water. One group received a daily oral application of CTL nanogel and carcinogen in drinking water. After the 23-week carcinogen treatment, the rats were euthanized; the tongues of the rats were dissected and histopathologically examined. Additionally, RT-PCR was employed to assess the gene expression of various signaling molecules involved in cancer progression like Erk1/2, β-catenin, Ki-67, Cyclin D1, TNF-α, NFκB, COX-2, and RAC1. Wistar rats developed white lesions and growth in the tongue in the cancer induction group. At the same time, the incidence and size of tumors were significantly less in the CTL nanogel-treated group. There was a significant increase in p53, Caspase-3, and Bax expression levels, while Bcl-2 showed a decreased expression in the CTL nanogel-treated group. There was also a significant decrease in the expression of EGFR and VEGFR signaling molecules in the CTL nanogel-treated group (p < 0.05 level). CTL nanogel shows potent chemopreventive efficiency in reducing the occurrence and severity of 4NQO-induced oral cancer in Wistar rats, marking it a promising candidate for further investigation in cancer prevention strategies.
Collapse
Affiliation(s)
- Ameena Mustafa
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Oral Pathology and Microbiology, Azeezia College of Dental Sciences & Research, Kollam, Kerala, India
| | - Meignana ArumughamIndiran
- Department of Public Health Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Elumalai Perumal
- Department of Biochemistry, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Anandakumar Ponnala
- Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Dinsha Abdul Rasheed
- Department of Pediatric Dentistry, City University College of Ajman, Ajman, United Arab Emirates
| | - Karthikeyan Ramalingam
- Department of Oral Pathology and Microbiology, Malla Reddy Institute of Dental Sciences, Malla Reddy Vishwa Vidyapeeth, Suraram, Hyderabad, Telangana, India
| | - Rajeshkumar Shanmugham
- Department of Biochemistry, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Mohmed Isaqali Karobari
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
- Department of Restorative Dentistry & Endodontics, Faculty of Dentistry, University of Puthisastra, Phnom Penh, 12211, Cambodia.
| |
Collapse
|
7
|
Ngô TM, Vágner A, Nagy G, Ország G, Nagy T, Szoboszlai Z, Csikos C, Váradi B, Trencsényi G, Tircsó G, Garai I. HER2 expression in different cell lines at different inoculation sites assessed by [ 52Mn]Mn-DOTAGA(anhydride)-trastuzumab. Pathol Oncol Res 2025; 31:1611999. [PMID: 40365451 PMCID: PMC12069034 DOI: 10.3389/pore.2025.1611999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025]
Abstract
Purpose Positron emission tomography (PET) hybrid imaging targeting HER2 requires antibodies labelled with longer half-life isotopes. With a suitable radiation profile, 52Mn coupled with DOTAGA as a bifunctional chelator is a potential candidate. In this study, we investigated the tumor HER2 specificity and the temporal biodistribution of the [52Mn]Mn-DOTAGA(anhydride)-trastuzumab in preclinical models. Methods PET/MRI and PET/CT were performed on SCID mice bearing orthotopic and ectopic HER2-positive and ectopic HER2-negative tumors at 4, 24, 48, 72, and 120 h post-injection with [52Mn]Mn-DOTAGA(anhydride)-trastuzumab. Melanoma xenografts were included for comparison of specificity. Results In vivo biodistribution demonstrated strong contrast in HER2-positive tumors, particularly in orthotopic tumors, where uptake was significantly higher than in the blood pool and other organs from 24 h onwards and consistently higher than in ectopic HER2-positive tumors at all time points. Significantly higher tumor-to-blood and tumor-to-muscle ratios were observed in HER2-positive ectopic tumors compared to HER2-negative tumors but only at 4 and 24 h; the differences were likely due to non-specific binding of the tracer. The ratios for orthotopic HER2-positive tumors were significantly higher than those for ectopic HER2-negative tumors and melanoma at all time points. However, the differences between HER2-positive and HER2-negative tumors decreased at later time points. Conclusion These results suggest that [52Mn]Mn-DOTAGA(anhydride)-trastuzumab demonstrates efficient tumor-to-background contrast, emphasize the higher tumor uptake observed in orthotopic tumors, and highlight the influence of tumor environment characteristics on uptake.
Collapse
Affiliation(s)
- Toàn Minh Ngô
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | | | | | | | - Tamás Nagy
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| | | | - Csaba Csikos
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Balázs Váradi
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - György Trencsényi
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| | - Gyula Tircsó
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Ildikó Garai
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Nuclear Medicine, Medical Imaging Clinic, Clinical Centre, University of Debrecen, Debrecen, Hungary
- Scanomed Ltd., Debrecen, Hungary
| |
Collapse
|
8
|
Smith KA, Batatinha H, Niemiro GM, Baker FL, Zúñiga TM, Diak D, Mylabathula PL, Kistner TM, Davini D, Hoffman E, Colombo JN, Seckeler M, Bond RA, Katsanis E, Simpson RJ. Exercise-induced β 2-adrenergic receptor activation enhances effector lymphocyte mobilization in humans and suppresses lymphoma growth in mice through NK-cells. Brain Behav Immun 2025:S0889-1591(25)00176-X. [PMID: 40311885 DOI: 10.1016/j.bbi.2025.04.040] [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: 02/05/2025] [Revised: 04/06/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025] Open
Abstract
Signaling through the β2-adrenergic receptor (β2-AR) mobilizes immune cells during exercise and is implicated in tumor lymphocyte infiltration. We investigated mechanisms governing immune cell mobilization in humans and the role of adrenergic signaling in anti-cancer responses to a murine lymphoma. Human studies included double-blind, placebo-controlled, crossover trials with beta blocker drugs and a phosphodiesterase inhibitor during steady-state and graded exercise. β1 + β2-AR blockade reduced lymphocyte and NK-cell mobilization during steady-state exercise, while β1-AR blockade enhanced the mobilization of NK-cells. Combining a β1-AR antagonist with a phosphodiesterase-4 (PDE4) inhibitor during graded exercise further increased mobilization of CD8 + T-cells, γδ T-cells, and monocytes. Isoproterenol infusion also elevated lymphocyte and NK-cell levels similarly to exercise at 70 % VO2max. Single cell RNA sequencing revealed complex signaling downstream of cAMP that relate to lymphocyte activation and effector function. In murine models of voluntary wheel running, β2-AR signaling and NK-cells were critical for exercise-induced protection against B-cell lymphoma, as β2-AR blockade or NK-cell depletion abrogated these effects. These findings highlight the pivotal role of β2-AR signaling in mobilizing cytotoxic immune cells and protecting against tumor progression through exercise, suggesting potential therapeutic strategies combining exercise with adrenergic modulation to enhance immune responses.
Collapse
Affiliation(s)
- Kyle A Smith
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA
| | - Helena Batatinha
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA
| | - Grace M Niemiro
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA; Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Forrest L Baker
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA; Department of Pediatrics, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Tiffany M Zúñiga
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA
| | - Douglass Diak
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA
| | | | - Timothy M Kistner
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Dan Davini
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Emely Hoffman
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Jamie N Colombo
- Department of Pediatrics (Cardiology), University of Arizona, Tucson, AZ, USA
| | - Michael Seckeler
- Department of Pediatrics (Cardiology), University of Arizona, Tucson, AZ, USA
| | - Richard A Bond
- College of Pharmacy, Science, and Engineering Research Center, University of Houston, Houston, TX, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA; Department of Medicine, University of Arizona, Tucson, AZ, USA; Department of Pathology, University of Arizona, Tucson, AZ, USA
| | - Richard J Simpson
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, USA; Department of Pediatrics, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
9
|
Wang Y, Tan Y, Zhang T, Wang Z, Gong J, Du Z, Mei Y, Ma J. TRUB1 is a novel biomarker for promoting malignancy in colorectal cancer via NFκB signaling. Gastroenterol Rep (Oxf) 2025; 13:goaf027. [PMID: 40260225 PMCID: PMC12011359 DOI: 10.1093/gastro/goaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/16/2024] [Accepted: 11/27/2024] [Indexed: 04/23/2025] Open
Abstract
Background Colorectal cancer (CRC) is one of the most aggressive malignancies of the digestive tract, characterized by aberrant post-transcriptional RNA modifications, including pseudouridine (Ψ). TruB pseudouridine synthase family member 1 (TRUB1) is a key pseudouridine synthase but its role in CRC progression remains unclear. Methods Public databases and CRC cell lines were analysed to assess TRUB1 expression in CRC. Receiver-operating characteristic (ROC) curve analysis and survival analysis were performed to evaluate the diagnostic and prognostic significance of TRUB1. The impact of TRUB1 on tumor proliferation and Ψ modification was examined in TRUB1-knock-down HCT116 cell lines. Mechanistically, RNA sequencing of control and TRUB1-knock-down HCT116 cells was conducted to identify potential pathways, which were validated by using real-time polymerase chain reaction (PCR), Western blot, and immunofluorescence assays. Results TRUB1 was significantly upregulated in CRC tumor tissues and cell lines. ROC analysis showed that TRUB1 had strong diagnostic potential and its overexpression was associated with poorer overall survival in CRC patients. In TRUB1-knock-down HCT116 cells, apoptosis increased and tumor growth slowed in nude mice, with a corresponding increase in apoptosis-related proteins and decreased Ψ modification. Mechanistically, RNA sequencing indicated that tumor necrosis factor α signaling via the nuclear factor kappa B (NFκB) pathway was activated in TRUB1-knock-down HCT116 cells. Further analysis identified Baculoviral inhibitor of apoptosis proteins repeat-containing 3 (BIRC3) as a potential downstream target gene that was regulated by TRUB1 in the NFκB pathway. Conclusions TRUB1 serves as a potential biomarker for CRC diagnosis and prognosis, and it can inhibit apoptosis in CRC cells via BIRC3-mediated NFκB signaling.
Collapse
Affiliation(s)
- Yingzhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yonghuang Tan
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Tianhao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Zhaoliang Wang
- School of Modern Information Industry, Guangzhou College of Commerce, Guangzhou, Guangdong, P. R. China
| | - Jingru Gong
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Zhenshuang Du
- Department of Gastrointestinal Surgery, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, P. R. China
| | - Yong Mei
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Jinping Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
10
|
Dalal N, Dhandapani H, Ingle A, Sharma D, Tayalia P. Functionalized Poly(ethylene Glycol) Diacrylate Scaffolds for In Situ Immunomodulation of Dendritic Cells Targeting Melanoma Tumor. ACS Biomater Sci Eng 2025; 11:2396-2407. [PMID: 40048381 DOI: 10.1021/acsbiomaterials.4c02036] [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: 04/15/2025]
Abstract
Various immunotherapeutic strategies are being developed to fight cancer, which is one of the leading causes of mortality. Dendritic cells (DCs), being professional antigen-presenting cells, after efficient manipulation with tumor-associated antigens, can lead to effective T-cell recruitment and activation at the tumor site, resulting in cytotoxic T-cell-mediated cancer cell killing. To circumvent the inefficiencies of ex vivo DC modification and patient infusion, an alternative strategy involving in situ DC activation has been explored here. Here, the vaccine components are tumor lysates, as antigens, and polyinosinic:polycytidylic acid (poly(I:C)), a toll-like receptor-3 (TLR3) agonist, as an adjuvant. Our in vitro studies demonstrate that complexing poly(I:C) with a carrier molecule, chitosan, enhances its stability and accessibility to TLR3 in the DC endosomal membrane. Material-based localized delivery of immunomodulatory factors is known to improve their stability and reduce their off-target side effects. Here, PEGDA-PLL-based macroporous scaffolds allow easy recruitment of host cells, thereby enabling effective interaction between the vaccine components loaded on them and the infiltrating immune cells. The vaccine components present in the scaffold facilitate efficient DC activation and migration, leading to subsequent T-cell activation and antitumor response, as shown by our in vivo studies.
Collapse
Affiliation(s)
- Neha Dalal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Hemavathi Dhandapani
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Arvind Ingle
- Tata Memorial Centre Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Deepak Sharma
- Radiation Biology and Health Science Division, Bhabha Atomic Research Center, Mumbai 400085, India
| | - Prakriti Tayalia
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
11
|
Park JH, Lee YK, Lee H, Choi DH, Rhee KJ, Kim HS, Seo JB. Sonoporation with Echogenic Liposomes: The Evaluation of Glioblastoma Applicability Using In Vivo Xenograft Models. Pharmaceutics 2025; 17:509. [PMID: 40284504 PMCID: PMC12030003 DOI: 10.3390/pharmaceutics17040509] [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: 02/25/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Objective: In previous studies, echogenic liposomes with liquid and gas cores were analyzed as alternative carriers of drug molecules and cavitation nuclei for sonoporation. The possibility of small interfering RNA (si-RNA) encapsulation has also been presented. In this study, the usability of echogenic liposomes as drug carriers and cavitation seeds was evaluated using an in vivo model. Methods: A doxorubicin-loaded echogenic liposome was synthesized as a drug carrier. The size distribution and the number of formed echogenic liposomes were measured. Five comparative in vivo experiments were conducted with and without doxorubicin-loaded echogenic liposomes, and the results were statically analyzed. Results: Sonoporation with doxorubicin-loaded echogenic liposomes at 3.05 W/cm2 of ISPTA ultrasound sonication and 0.98 MHz results in an average tumor volume growth of less than 25% of that following the simple administration of doxorubicin. Considering the p-value between the two groups is approximately 0.03, doxorubicin-loaded echogenic liposomes were effectively applicable as cavitation nuclei for sonoporation. Conclusions: Although further studies are needed to clarify the responses to incident ultrasound fields, the proposed echogenic liposome appears to be a promising alternative cavitation nuclei/carrier for sonoporation.
Collapse
Affiliation(s)
- Ju-Hyun Park
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Yoo-Kyung Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Dong-Hyun Choi
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, Yonsei University, Wonju 26493, Republic of Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Jong-Bum Seo
- Department of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| |
Collapse
|
12
|
Ulus G, Özbek EN, Yılmaz H, Keselik E, Sarıcaoğlu M, Akyol Bahçeci S, İşel E, Debeleç Bütüner B, Yetik Anacak G, Koparal AT. Borax pentahydrate as a promising boron-based angiogenesis inhibitor. J Trace Elem Med Biol 2025; 89:127640. [PMID: 40203787 DOI: 10.1016/j.jtemb.2025.127640] [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: 01/27/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Boron, a trace element, is involved in various physiological and metabolic processes, and recent studies suggest that boron compounds may have potential in cancer prevention and treatment. In this study, the antiangiogenic effects of a boron compound, borax pentahydrate (BPH), were investigated. Angiogenesis is a tightly regulated biological process responsible for the formation of new blood vessels from existing vasculatures. This process plays a critical role in cancer progression, making it an important target for cancer therapy. Pancreatic and kidney cancers are difficult to treat because they are aggressive and resistant to chemotherapy. METHODS The antiproliferative activity was evaluated using the MTT assay, while antiangiogenic effects were tested through in vitro tube formation assays and in ovo chick chorioallantoic membrane (CAM) assay. The effect of BPH on VEGF levels was determined using Western blot analysis in HUVEC, ACHN, PANC-1 cells. The effect of BPH on tumor angiogenesis was investigated with an in vivo Ehrlich ascites carcinoma model (EAC). RESULTS BPH exhibited potent antiproliferative and antiangiogenic activities, inhibiting the proliferation of ACHN, PANC-1, and HUVECs, disrupting endothelial tube formation, and inhibiting vascular formation on the CAM surface in a dose-dependent manner. VEGF levels were significantly decreased in ACHN, PANC-1 and HUVECs. There was also a decrease in VEGF and TGF-β1 levels in BPH-treated tumor groups. In addition, BPH caused a decrease in tumor size. CONCLUSION These findings suggest that BPH may be a new antiangiogenic and antitumoral agent. BPH may contribute to drug development studies targeting angiogenesis-related diseases as a promising new therapeutic agent.
Collapse
Affiliation(s)
- G Ulus
- Republic of Türkiye, Ministry of Education, Şerife Bacı Vocational and Technical High School, Izmir 35090, Turkiye.
| | - E N Özbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - H Yılmaz
- Republic of Türkiye, Ministry of Education, Mimar Sinan Vocational and Technical High School, Izmir 35090, Turkiye
| | - E Keselik
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - M Sarıcaoğlu
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - S Akyol Bahçeci
- Department of Histology and Embryology, Faculty of Medicine, Katip Çelebi University, Izmir 35100, Turkiye
| | - E İşel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - B Debeleç Bütüner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir 35100, Turkiye
| | - G Yetik Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkiye
| | - A T Koparal
- Yunus Emre Vocational School of Health Services, Anadolu University, Eskisehir 26470, Turkiye
| |
Collapse
|
13
|
Wang Q, Suo Y, Tian X. 5-Aminolaevulinic Acid-Mediated Photodynamic Therapy Combined with Tirapazamine Enhances Efficacy in Ovarian Cancer. Biomedicines 2025; 13:724. [PMID: 40149700 PMCID: PMC11939993 DOI: 10.3390/biomedicines13030724] [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: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Ovarian cancer is a common gynaecological malignancy. Photodynamic therapy (PDT) mediated by 5-aminolaevulinic acid (5-ALA-PDT) is widely used in clinical practice. However, hypoxia may impact the efficacy of this treatment. In the present study, we combined the bioreductively active drug tirapazamine (TPZ) with PDT to explore its potential in enhancing ovarian cancer cell death. Methods: A cell counting kit-8 assay was used to determine cytotoxicity under different intervention conditions. The distribution of protoporphyrin IX, a metabolite of 5-ALA, was observed using in vivo fluorescence imaging. The effect of the combined treatment was assessed by measuring changes in tumour size following the corresponding interventions and by haematoxylin and eosin staining of tumour tissues. Immunohistochemical staining was used to detect the expression levels of relevant proteins. Results: TPZ exhibited no cytotoxicity under normoxic conditions but was activated under hypoxic conditions, inducing cytotoxic effects that were enhanced when combined with PDT. Over time, protoporphyrin IX achieved systemic distribution, and high drug concentrations were maintained within the tumour. The combination therapy suppressed tumour growth, and pathological staining showed that necrotic tumour areas were significantly enlarged after treatment. The enhanced therapeutic effect may be attributable to the inhibition of the hypoxia-inducible factor-1α/vascular endothelial growth factor axis and PI3K/Akt/mTOR pathway. Conclusions: 5-ALA-PDT combined with TPZ can overcome both the hypoxic state of ovarian cancer tissues and the increased hypoxia induced by PDT, thereby inhibiting tumour growth.
Collapse
Affiliation(s)
- Qian Wang
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| | - Yuping Suo
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
- Department of Gynaecology and Obstetrics, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Xiaojuan Tian
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| |
Collapse
|
14
|
Gari MK, Lee HJ, Inman DR, Burkel BM, Highland MA, Kwon GS, Gupta N, Ponik SM. Inhibiting fibronectin assembly in the breast tumor microenvironment increases cell death and improves response to doxorubicin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637963. [PMID: 40161788 PMCID: PMC11952368 DOI: 10.1101/2025.02.12.637963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Purpose Effective therapies for solid tumors, including breast cancers, are hindered by several roadblocks that can be largely attributed to the fibrotic extracellular matrix (ECM). Fibronectin (FN) is a highly upregulated ECM component in the fibrotic tumor stroma and is associated with poor patient prognosis. This study aimed to investigate the therapeutic potential of an anti-fibrotic peptide that specifically targets FN and blocks the fibrillar assembly of FN. Methods To target FN, we used PEGylated Functional Upstream Domain (PEG-FUD), which binds to the 70 kDa N-terminal region of FN with high affinity, localizes to mammary tumors, and potently inhibits FN assembly in vitro and in vivo. Here, we used the 4T1 tumor model to investigate the efficacy and mechanisms of PEG-FUD to inhibit tumor growth. Results Our data demonstrates that PEG-FUD monotherapy reduces tumor growth without systemic toxicity. Analysis of the tumor microenvironment revealed that PEG-FUD effectively inhibited FN matrix assembly within tumors and reduced adhesion-mediated signaling through α5 integrin and FAK leading to enhanced tumor cell death. Notably, signaling through FAK has been associated with resistance mechanisms to doxorubicin (DOX). Therefore, we tested the combination of PEG-FUD and Dox, which significantly reduced tumor growth by 60% compared to vehicle control and 30% compared to Dox monotherapy. Conclusions Our findings demonstrate that PEG-FUD significantly modifies the peritumoral ECM of breast cancer, leading to increased tumor cell death, and potentiates the efficacy of conventional breast cancer therapy.
Collapse
Affiliation(s)
- Metti K. Gari
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Hye Jin Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - David R. Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian M. Burkel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Margaret A. Highland
- Wisconsin Veterinary Diagnostic Laboratory, University of Wisconsin - Madison, WI, USA
| | - Glen S. Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, WI, USA
| | - Nikesh Gupta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin - Madison, WI, USA
| | - Suzanne M. Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
15
|
Pougoue Ketchemen J, Njotu FN, Babeker H, Monzer A, Nwangele E, Tikum AF, Henning N, Hassani N, Frye S, Perron R, Byrne C, Didychuk C, Qi Q, Bannister L, Doroudi A, Fonge H. Complete Remissions of HER2-Positive Trastuzumab-Resistant Xenografts Using a Potent [225Ac]Ac-Labeled Anti-HER2 Antibody-Drug Radioconjugate. Clin Cancer Res 2025; 31:685-696. [PMID: 39670857 DOI: 10.1158/1078-0432.ccr-24-1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/09/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
PURPOSE There is overwhelming interest to use actinium-225 ([225Ac]Ac) to develop targeted α therapies. Antibody-drug conjugates (ADC) are highly cytotoxic. Combining [225Ac]Ac with an ADC to develop an antibody-drug radioconjugate [225Ac]Ac-macropa-trastuzumab(T)-PEG6-emtansine (DM1), is expected to be more effective than its ADC (T-PEG6-DM1) against breast cancer. EXPERIMENTAL DESIGN [89Zr]Zr-p-isothiocyanatobenzyl desferrioxamine (DFO)-T-PEG6-DM1 (imaging) and [225Ac]Ac-macropa-T-PEG6-DM1 (radiotherapy) were developed. Biodistribution and safety evaluations of [225Ac]Ac-macropa-T-PEG6-DM1 were carried out in non-tumor-bearing BALB/c mice. MicroPET imaging and biodistribution were done using [89Zr]Zr-DFO-T-PEG6-DM1, and radiotherapy using [225Ac]Ac-macropa-T-PEG6-DM1 was carried out in athymic BALB/c nude mice bearing trastuzumab-resistant HCC1954 and trastuzumab-DM1 (T-DM1)/trastuzumab-resistant JIMT-1 tumor-bearing mice. RESULTS After 7 days of incubation at 37°C, [225Ac]Ac-macropa-T-PEG6-DM1 was stable in both human serum (89.2% ± 0.9%) and PBS (82.8% ± 0.4%). T-PEG6-DM1 (8 mg/kg) and [225Ac]Ac-macropa-T-PEG6-DM1 (3 × 18 kBq) administered separately in non-tumor-bearing mice 10 days apart were well tolerated biochemically and hematologically. Imaging and biodistribution showed high tumor uptake of [89Zr]Zr-DFO-T-PEG6-DM1 in tumor-bearing mice at 120 hours after injection: 38.1% ± 2.8% IA/g (HCC1954) and 14.6% ± 1% IA/g (JIMT-1). In HCC1954 tumor-bearing mice, all treatment groups had complete remission (8/8), indicative of the responsiveness of the xenograft to T-DM1-based treatments, whereas for JIMT-1 xenografts (having 1/8 complete remission) at 23 days after treatment, tumor volumes were 332.1 ± 77.5 vs. 244.6 ± 63 vs. 417.9 ± 62.1 vs. 102.4 ± 18.5 for the saline (negative control), T-DM1 (positive control), T-PEG6-DM1, and [225Ac]Ac-macropa-T-PEG6-DM1, respectively. CONCLUSIONS [225Ac]Ac-macropa-T-PEG6-DM1 is more potent than ADC against trastuzumab-resistant breast cancer and necessitates clinical translation.
Collapse
Affiliation(s)
- Jessica Pougoue Ketchemen
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
| | - Fabrice Ngoh Njotu
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Hanan Babeker
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Alissar Monzer
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Emmanuel Nwangele
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Anjong Florence Tikum
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Nikita Henning
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Nava Hassani
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Sarah Frye
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Randy Perron
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Chris Byrne
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Candice Didychuk
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Qi Qi
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Laura Bannister
- Canadian Nuclear Laboratories, Radiobiology and Health Branch, Chalk River, Canada
| | - Alireza Doroudi
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, Canada
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Department of Medical Imaging, Royal University Hospital Saskatoon, Saskatoon, Canada
| |
Collapse
|
16
|
Liu Y, Liu R, Dong J, Xia X, Yang H, Wei S, Fan L, Fang M, Zou Y, Zheng M, Leong KW, Shi B. Targeted protein degradation via cellular trafficking of nanoparticles. NATURE NANOTECHNOLOGY 2025; 20:296-302. [PMID: 39468359 DOI: 10.1038/s41565-024-01801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/04/2024] [Indexed: 10/30/2024]
Abstract
Strategies that selectively bind proteins of interest and target them to the intracellular protein recycling machinery for targeted protein degradation have recently emerged as powerful tools for undruggable targets in biomedical research and the pharmaceutical industry. However, targeting any new protein of interest with current degradation tools requires a laborious case-by-case design for different diseases and cell types, especially for extracellular targets. Here we observe that nanoparticles can mediate specific receptor-independent internalization of a bound protein and further develop a general strategy for degradation of extracellular proteins of interest by making full use of clinically approved components. This extremely flexible strategy aids in targeted protein degradation tool development and provides knowledge for targeted drug therapies and nanomedicine design.
Collapse
Affiliation(s)
- Yang Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
- Huaihe Hospital of Henan University, Henan University, Kaifeng, China
| | - Runhan Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Jiawei Dong
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Xue Xia
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Haoying Yang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Sijun Wei
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Linlin Fan
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Mengke Fang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
| | - Yan Zou
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China.
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences, Henan University, Kaifeng, China.
- Huaihe Hospital of Henan University, Henan University, Kaifeng, China.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China.
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
17
|
Wei H, Zhao D, Zhi Y, Wu Q, Ma J, Xu J, Liu T, Zhang J, Wang P, Hu Y, He X, Guo F, Jiang M, Zhang D, Nie W, Yang R, Zhao T, Dong Z, Liu K. RTN4IP1 Contributes to ESCC via Regulation of Amino Acid Transporters. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406220. [PMID: 39757767 PMCID: PMC11848606 DOI: 10.1002/advs.202406220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/19/2024] [Indexed: 01/07/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for about 90% of esophageal cancer cases. The lack of effective therapeutic targets makes it difficult to improve the overall survival of patients with ESCC. Reticulon 4 Interacting Protein 1 (RTN4IP1) is a novel mitochondrial oxidoreductase. Here, a notable upregulation of RTN4IP1 is demonstrated, which is associated with poor survival in patients with ESCC. RTN4IP1 depletion impairs cell proliferation and induces apoptosis of ESCC cells. Furthermore, c-Myc regulates RTN4IP1 expression via iron regulatory protein 2 (IRP2) at the post-transcriptional level. Mechanistically, RTN4IP1 mRNA harbors functional iron-responsive elements (IREs) in the 3' UTR, which can be targeted by IRP2, resulting in increased mRNA stability. Finally, RTN4IP1 depletion abrogates amino acid uptake and induces amino acid starvation via downregulation of the amino acid transporters SLC1A5, SLC3A2, and SLC7A5, indicating a possible pathway through which RTN4IP1 contributes to ESCC carcinogenesis and progression. In vivo studies using cell-derived xenograft and patient-derived xenograft mouse models as well as a 4-nitroquinoline 1-oxide-induced ESCC model in esophageal-specific Rtn4ip1 knockout mice demonstrate the essential role of RTN4IP1 in ESCC development. Thus, RTN4IP1 emerges as a key cancer-promoting protein in ESCC, suggesting therapeutic RTN4IP1 suppression as a promising strategy for ESCC treatment.
Collapse
Affiliation(s)
- Huifang Wei
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Dengyun Zhao
- Department of PathophysiologySchool of Basic Medical Sciences, Zhengzhou UniversityChina‐US (Henan) Hormel Cancer InstituteChest Hospital of Zhengzhou UniversityZhengzhou450000China
| | - Yafei Zhi
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Qiong Wu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Jing Ma
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhou450000China
| | - Jialuo Xu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhou450000China
| | - Tingting Liu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Jing Zhang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Penglei Wang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Yamei Hu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Xinyu He
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Fangqin Guo
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou University, China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Ming Jiang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Dandan Zhang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Wenna Nie
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Ran Yang
- China‐US (Henan) Hormel Cancer InstituteZhengzhou450000China
| | - Tongjin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
- State Key Laboratory of Genetic EngineeringShanghai Key Laboratory of Metabolic Remodeling and HealthInstitute of Metabolism and Integrative BiologyZhongshan HospitalShanghai Qi Zhi InstituteFudan UniversityShanghai200438China
| | - Zigang Dong
- Department of PathophysiologySchool of Basic Medical SciencesThe Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, State Key Laboratory of EsophagealCancer Prevention and TreatmentProvincial Cooperative Innovation Center for Cancer ChemopreventionChina‐US (Henan) Hormel Cancer Institute, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
| | - Kangdong Liu
- Department of PathophysiologySchool of Basic Medical SciencesThe Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, State Key Laboratory of EsophagealCancer Prevention and TreatmentProvincial Cooperative Innovation Center for Cancer ChemopreventionChina‐US (Henan) Hormel Cancer Institute, Tianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhou450000China
| |
Collapse
|
18
|
Zhang G, Wu J, Ji M, Liu X, Shi M. SLC25A1 promotes lymph node metastasis of esophageal squamous cell carcinoma by regulating lipid metabolism. Int J Oncol 2025; 66:15. [PMID: 39821659 PMCID: PMC11753767 DOI: 10.3892/ijo.2025.5721] [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: 08/07/2024] [Accepted: 12/06/2024] [Indexed: 01/19/2025] Open
Abstract
Solute carrier family 25 member 1 (SLC25A1) affects lipid metabolism and energy regulation in multiple types of tumor cell, affecting their proliferation and survival. To the best of our knowledge, however, the impact of SLC25A1 on the proliferation and survival of esophageal squamous cell carcinoma (ESCC) cells has yet to be explored. Here, SLC25A1 expression was detected in ESCC tissues and cell lines. SLC25A1 was silenced or blocked by lentivirus transfection or 2‑[(4‑chloro‑3‑nitrophenyl)sulfonylamino]benzoic acid in ESCC cells. To evaluate the impact of SLC25A1 on in vivo and in vitro proliferation, invasion and migration of ESCC cells, Cell Counting‑Kit, wound healing, colony formation, Transwell, EdU, flow cytometry, tumor xenograft in nude mice, lipid metabolism and energy metabolism detection assays were performed. Reverse transcription‑quantitative PCR and western blot analysis were performed to determine expression of downstream molecules and pathway proteins following the silencing and blockade of SLC25A1. SLC25A1 was significantly overexpressed in ESCC tissue and cell lines. The targeted silencing of SLC25A1 or inhibition of its protein led to a significant decrease in proliferative, invasive and migratory capabilities of ESCC cells, accompanied by increased apoptosis. Additionally, silencing of the SLC25A1 gene significantly inhibited xenograft tumor growth in vivo. The present results indicate that knockdown or blockade of SLC25A1 can significantly impede the malignant biological behavior of ESCC.
Collapse
Affiliation(s)
- Guoquan Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Jingru Wu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Minghao Ji
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiangyan Liu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Mo Shi
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
19
|
Claridge SE, Nath S, Baum A, Farias R, Cavallo J, Rizvi NM, De Boni L, Park E, Granados GL, Hauesgen M, Fernandez‐Rodriguez R, Kozan EN, Kanshin E, Huynh KQ, Chen P, Wu K, Ueberheide B, Mosquera JM, Hirsch FR, DeVita RJ, Elemento O, Pauli C, Pan Z, Hopkins BD. Functional genomics pipeline identifies CRL4 inhibition for the treatment of ovarian cancer. Clin Transl Med 2025; 15:e70078. [PMID: 39856363 PMCID: PMC11761363 DOI: 10.1002/ctm2.70078] [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: 01/30/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The goal of precision oncology is to find effective therapeutics for every patient. Through the inclusion of emerging therapeutics in a high-throughput drug screening platform, our functional genomics pipeline inverts the common paradigm to identify patient populations that are likely to benefit from novel therapeutic strategies. APPROACH Utilizing drug screening data across a panel of 46 cancer cell lines from 11 tumor lineages, we identified an ovarian cancer-specific sensitivity to the first-in-class CRL4 inhibitors KH-4-43 and 33-11. CRL4 (i.e., Cullin-4 RING E3 ubiquitin ligase) is known to be dysregulated in a variety of cancer contexts, making it an attractive therapeutic target. Unlike proteasome inhibitors that are associated with broad toxicity, CRL4 inhibition offers the potential for tumor-specific effects. RESULTS We observed that CRL4 inhibition negatively regulates core gene signatures that are upregulated in ovarian tumors and significantly slowed tumor growth as compared to the standard of care, cisplatin, in OVCAR8 xenografts. Building on this, we performed combination drug screening in conjunction with proteomic and transcriptomic profiling to identify ways to improve the antitumor effects of CRL4 inhibition in ovarian cancer models. CRL4 inhibition consistently resulted in activation of the mitogen-activated protein kinase (MAPK) signaling cascade at both the transcriptomic and protein levels, suggesting that survival signaling is induced in response to CRL4 inhibition. These observations were concordant with the results of the combination drug screens in seven ovarian cancer cell lines that showed CRL4 inhibition cooperates with MEK inhibition. Preclinical studies in OVCAR8 and A2780 xenografts confirmed the therapeutic potential of the combination of KH-4-43 and trametinib, which extended overall survival and slowed tumor progression relative to either single agent or the standard of care. CONCLUSIONS Together, these data demonstrate the prospective utility of functional modeling pipelines for therapeutic development and underscore the clinical potential of CRL4 inhibition in the ovarian cancer context. HIGHLIGHTS A precision medicine pipeline identifies ovarian cancer sensitivity to CRL4 inhibitors. CRL4 inhibition induces activation of MAPK signalling as identified by RNA sequencing, proteomics, and phosphoproteomics. Inhibitor combinations that target both CRL4 and this CRL4 inhibitor-induced survival signalling enhance ovarian cancer sensitivity to treatment.
Collapse
Affiliation(s)
- Sally E. Claridge
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNew YorkUSA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Shalini Nath
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNew YorkUSA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Anneliese Baum
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Richard Farias
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Julie‐Ann Cavallo
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Nile M. Rizvi
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Lamberto De Boni
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNew YorkUSA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Eric Park
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Genesis Lara Granados
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Matthew Hauesgen
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ruben Fernandez‐Rodriguez
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Eda Nur Kozan
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNew YorkUSA
| | - Evgeny Kanshin
- Department of Biochemistry and Molecular PharmacologyNew York University School of MedicineNew YorkNew YorkUSA
- Proteomics LaboratoryNew York University School of MedicineNew YorkNew YorkUSA
| | - Khoi Q. Huynh
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Drug Discovery Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Peng‐Jen Chen
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Drug Discovery Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Kenneth Wu
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Beatrix Ueberheide
- Department of Biochemistry and Molecular PharmacologyNew York University School of MedicineNew YorkNew YorkUSA
- Proteomics LaboratoryNew York University School of MedicineNew YorkNew YorkUSA
- Department of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Juan Miguel Mosquera
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNew YorkUSA
| | - Fred R. Hirsch
- Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Medicine, Hematology, and Medical OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of Pathology, Molecular and Cell‐Based MedicineIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Robert J. DeVita
- Proteomics LaboratoryNew York University School of MedicineNew YorkNew YorkUSA
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
- Institute for Computational Biomedicine, Weill Cornell MedicineNew YorkNew YorkUSA
- Clinical and Translational Science Center, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Chantal Pauli
- Department of Pathology and Molecular PathologyUniversity Hospital ZurichZurichSwitzerland
| | - Zhen‐Qiang Pan
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Benjamin D. Hopkins
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNew YorkUSA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York Presbyterian HospitalNew YorkNew YorkUSA
| |
Collapse
|
20
|
Song M, Yuan J, Zhang G, Sun M, Zhang Y, Su X, Lv R, Zhao Y, Shi Y, Zhao L. Mitochondrial transfer of drug-loaded artificial mitochondria for enhanced anti-Glioma therapy through synergistic apoptosis/ferroptosis/immunogenic cell death. Acta Biomater 2025; 193:514-530. [PMID: 39674237 DOI: 10.1016/j.actbio.2024.12.027] [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: 09/18/2024] [Revised: 11/20/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Mitochondrial targeting in gliomas represents a novel therapeutic strategy with significant potential to enhance drug sensitivity by effectively killing glioma cells at the mitochondrial level. In this study, we developed artificial mitochondria derived from mitochondrial membrane-based nanovesicles, enabling precise mitochondrial targeting of doxorubicin (Dox) to selectively eradicate cancer cells by amplifying multiple cell death pathways. It was found that Dox-encapsulating mitochondria-based nanovesicles (DOX-MitoNVs) exhibited an extraordinary ability to penetrate the blood-brain barrier (BBB), specifically targeting gliomas. By targeting mitochondria instead of locating at the nucleus, DOX-MitoNVs not only amplified Dox mediated apoptosis effects through the overloading of intracellular Ca2+ but also intensified ferroptosis by generating reactive oxygen species (ROS). Furthermore, DOX-MitoNVs demonstrated a significant ability to modulate the tumor immune microenvironment, thereby inducing pronounced immunogenic cell death (ICD) effects. In summary, it presents a novel therapeutic strategy utilizing DOX-MitoNVs for precise mitochondrial targeting in gliomas, enhancing drug sensitivity, inducing multiple cell death pathways, and modulating the tumor immune microenvironment to promote immunogenic cell death. STATEMENT OF SIGNIFICANCE: Mitochondrial targeting in gliomas is a promising therapeutic strategy that enhances drug sensitivity by exploiting glioma cells' mitochondrial vulnerabilities. We engineered mitochondrial membrane-based nanovesicles as artificial mitochondria for precise mitochondrial targeting of Dox. This approach facilitates selective cancer cell eradication and amplifies multiple cell death pathways alongside immunogenic chemotherapy. Notably, DOX-MitoNVs effectively cross the BBB and specifically target gliomas. By focusing on mitochondria, Dox induces apoptosis and intensifies ferroptosis through ROS generation. Additionally, DOX-MitoNVs can transform the tumor immune microenvironment, promoting ICD. Overall, DOX-MitoNVs offer a promising platform for enhanced glioma therapy.
Collapse
Affiliation(s)
- Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Jiayu Yuan
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ge Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Mengdi Sun
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yifei Zhang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Xiangchen Su
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Ruizhen Lv
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yuting Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China; Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China; Key Laboratory of Neurodegenerative Diseases of Liaoning Province, Jinzhou Medical University, Jinzhou, China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
21
|
Matsubara J, Li YF, Koul S, Mukohyama J, Salazar LEV, Isobe T, Qian D, Clarke MF, Sahoo D, Altman RB, Dalerba P. The E2F4 transcriptional repressor is a key mechanistic regulator of colon cancer resistance to irinotecan (CPT-11). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.633435. [PMID: 39896677 PMCID: PMC11785039 DOI: 10.1101/2025.01.22.633435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background Colorectal carcinomas (CRCs) are seldom eradicated by cytotoxic chemotherapy. Cancer cells with stem-like functional properties, often referred to as "cancer stem cells" (CSCs), display preferential resistance to several anti-tumor agents used in cancer chemotherapy, but the molecular mechanisms underpinning their selective survival remain only partially understood. Methods In this study, we used Transcription Factor Target Genes (TFTG) enrichment analysis to identify transcriptional regulators (activators or repressors) that undergo preferential activation by chemotherapy in CRC cells with a "bottom-of-the-crypt" phenotype (EPCAM+/CD44+/CD166+; CSC-enriched) as compared to CRC cells with a "top-of-the-crypt" phenotype (EPCAM+/CD44neg/CD166neg; CSC-depleted). The two cell populations were purified in parallel by fluorescence-activated cell sorting (FACS) from a patient-derived xenograft (PDX) line representative of a moderately differentiated human CRC, following in vivo chemotherapy with irinotecan (CPT-11). The transcriptional regulators identified as differentially activated were tested for differential expression in normal vs. cancer tissues, and in cell populations enriched in stem/progenitor cell-types as compared to differentiated lineages (goblet cells, enterocytes) in the mouse colon epithelium. Finally, the top candidate was tested for mechanistic contribution to drug-resistance by selective down-regulation using short-hairpin RNAs (shRNAs). Results Our analysis identified E2F4 and TFDP1, two core components of the DREAM transcriptional repression complex, as transcriptional modulators preferentially activated by irinotecan in EPCAM+/CD44+/CD166+ as compared to EPCAM+/CD44neg/CD166neg cancer cells. The expression levels of both genes (E2F4, TFDP1) were found up-regulated in CRCs as compared to human normal colon tissues, and in a sub-population of mouse colon epithelial cells enriched in stem/progenitor elements (Epcam+/Cd44+/Cd66alow/Kitneg) as compared to other sub-populations enriched in either goblet cells (Epcam+/Cd44+/Cd66alow/Kit+) or enterocytes (Epcam+/Cd44neg/Cd66ahigh). Most importantly, E2F4 down-regulation using shRNAs dramatically enhanced the sensitivity of human CRCs to in vivo treatment with irinotecan, across three independent PDX models. Conclusions Our data identified E2F4 and the DREAM repressor complex as critical regulators of human CRC resistance to irinotecan, and as candidate targets for the development of chemo-sensitizing agents.
Collapse
Affiliation(s)
- Junichi Matsubara
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA (USA)
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto (Japan)
| | - Yong Fuga Li
- Department of Genetics, Stanford University, Stanford, CA (USA)
- Department of Bioengineering, Stanford University, Stanford, CA (USA)
- Illumina Inc., San Diego, CA (USA)
| | - Sanjay Koul
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health (HMH), Nutley, NJ (USA)
- Department of Biological Sciences and Geology, Queensborough Community College (QCC), The City University of New York (CUNY), Bayside, NY (USA)
- Department of Pathology and Cell Biology, Columbia University, New York, NY (USA)
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY (USA)
- Columbia Stem Cell Initiative (CSCI), Columbia University, New York, NY (USA)
| | - Junko Mukohyama
- Department of Pathology and Cell Biology, Columbia University, New York, NY (USA)
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY (USA)
- Columbia Stem Cell Initiative (CSCI), Columbia University, New York, NY (USA)
- Department of Surgery, Institute of Medical Science, University of Tokyo, Tokyo (Japan)
| | - Luis E. Valencia Salazar
- Department of Pathology and Cell Biology, Columbia University, New York, NY (USA)
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY (USA)
- Columbia Stem Cell Initiative (CSCI), Columbia University, New York, NY (USA)
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY (USA)
| | - Taichi Isobe
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA (USA)
- Department of Comprehensive Oncology, Graduate School of Medicine, Kyushu University, Fukuoka (Japan)
| | - Dalong Qian
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA (USA)
| | - Michael F. Clarke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA (USA)
| | - Debashis Sahoo
- Department of Computer Science and Engineering, University of California San Diego (UCSD), San Diego, CA (USA)
- Department of Pediatrics, University of California San Diego (UCSD), San Diego, CA (USA); Department of Medicine (Division of Digestive and Liver Diseases), Columbia University, New York, NY (USA)
| | - Russ B. Altman
- Department of Genetics, Stanford University, Stanford, CA (USA)
- Department of Bioengineering, Stanford University, Stanford, CA (USA)
| | - Piero Dalerba
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health (HMH), Nutley, NJ (USA)
- Department of Pathology and Cell Biology, Columbia University, New York, NY (USA)
- Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY (USA)
- Columbia Stem Cell Initiative (CSCI), Columbia University, New York, NY (USA)
- Digestive and Liver Disease Research Center (DLDRC), Columbia University, New York, NY (USA)
- Department of Medical Sciences, Hackensack Meridian School of Medicine (HMSOM), Nutley, NJ (USA)
- Lombardi Comprehensive Cancer Center (LCCC), Georgetown University, Washington, DC (USA)
| |
Collapse
|
22
|
Wang X, Li S, Li Z, Lin Z, Wang Z. SRT3025-loaded cell membrane hybrid liposomes (3025@ML) enhanced anti-tumor activity of Oxaliplatin via inhibiting pyruvate kinase M2 and fatty acid synthase. Lipids Health Dis 2025; 24:14. [PMID: 39825408 PMCID: PMC11740399 DOI: 10.1186/s12944-025-02431-x] [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: 07/05/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Bladder cancer is one of the most common malignancies of the urinary system. Despite significant advances in diagnosis and treatment, the compromised therapeutic effect of chemotherapeutic agents, such as Oxaliplatin (OXA), remains a major clinical challenge. Thus, a combination therapy is required to enhance the OXA's therapeutic effectiveness and improve patient outcomes. METHODS The thin film hydration method was used to prepare the liposomes. Drug encapsulation efficiency and loading capacity were determined to investigate the advantages of the SRT3025-loaded cell membrane hybrid liposomes (3025@ML). Bladder cancer cell lines T24 and 5637 were cultured in McCoy's 5 A and RPMI 1640 medium, respectively. The Cell Counting Kit-8 assay was used to determine the cell viability by treating cells with a medium containing either the vehicle solution (control), the cell membrane hybrid liposomes (ML), 3025@ML, or compound 3 K. The antiproliferative activities were investigated after treating cells with OXA + 3025@ML and compound 3 K + OXA. Cell death and apoptosis were quantified by trypan blue and Annexin V-APC/PI apoptosis assay after treating cells with control, OXA, OXA + 3025@ML, and 3025@ML. Western blot analysis was performed after treating cells with 3025@ML, OXA, 3 K, 3025@ML + OXA, and 3 K + OXA to determine the protein levels of pyruvate kinase M2 (PKM2) and fatty acid synthase (FASN), etc. RESULTS: The present study demonstrated that 3025@ML enhances the chemotherapeutic effect of OXA. 3025@ML + OXA treated T24 and 5637 cells showed that combination therapy significantly reduced cell viability and increased cell death rate. Flow cytometry analysis showed that the combination of 3025@ML and OXA significantly increased the percentage of apoptotic cells in T24 cells. 3025@ML and compound 3 K reduced the levels of FASN in T24 and 5637 cells and increased the anti-tumor activity of OXA. Mechanistic studies showed that 3025@ML inhibited the PI3K/AKT/mTOR signaling pathway and reduced the expression of key metabolic regulators PKM2 and FASN. Furthermore, this study demonstrated that targeting lipid metabolism and inhibiting FASN can effectively overcome the compromised therapeutic effect of OXA. CONCLUSION The study demonstrated that 3025@ML significantly enhances the anti-tumor activity of OXA. This novel drug delivery system inhibits key metabolic pathways, which increase DNA damage and tumor cell apoptosis. The results indicate that 3025@ML is a promising therapeutic strategy for overcoming OXA's compromised therapeutic effect and potentially improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, 518052, China.
| | - Shulin Li
- Department of Urology, Southern University of Science and Technology Hospital, Shenzhen, 518052, China
| | - Zichen Li
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Zhuona Lin
- Medical School of Basic Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
23
|
Pragyandipta P, Naik E, Reddy PK, Nayek A, Kantevari S, Naik PK. In silico inspired design of urea noscapine congeners as anticancer agents: Chemical synthesis and experimental evaluation using breast cancer cells and a xenograft mouse model. Eur J Med Chem 2025; 282:117091. [PMID: 39602993 DOI: 10.1016/j.ejmech.2024.117091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
A series of semisynthetic noscapine-urea congeners (7a-7h) as potential tubulin-binding agents are being developed by integrating a urea pharmacophore at the C-9 position of the noscapine scaffold. Their binding affinity to tubulin was predicted through molecular docking, molecular dynamics (MD) simulations, and the MM-PBSA approach. These molecules were subsequently chemically synthesized and assessed using breast cancer cell lines (MCF-7 and MDA-MB-231) and normal human embryonic kidney cells (HEK). Both the docking score and the predicted binding free energy (ΔGbind,pred) revealed that urea congeners had a stronger affinity towards tubulin than noscapine and effectively inhibited the proliferation of all cancer cell types without affecting normal healthy cells. The results indicated that compound 7g exhibited the most promise and was chosen for further studies. Moreover, MDA-MB-231 cells treated with 7g at its IC50 concentration showed morphological changes such as membrane blebbing, fragmented nuclei, and the presence of apoptotic bodies. Apoptosis induction was further confirmed by flow cytometry. Moreover, the tubulin binding assay revealed a greater binding affinity with an equilibrium dissociation constant (KD) of 42 ± 2.4 μM for compound 7g. The number of MCF-7 cells engrafted as breast tumors in nude mice was found to be reduced significantly without any adverse effects. Noscapine is already in clinical trials, but the urea noscapine congener offers an advantage because of its increased potency without impacting the nontoxic profile of noscapine.
Collapse
Affiliation(s)
- Pratyush Pragyandipta
- Centre of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Sambalpur, 768019, Odisha, India
| | - Eeshara Naik
- Centre of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Sambalpur, 768019, Odisha, India
| | - Praveen Kumar Reddy
- Fluoro-Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
| | - Arnab Nayek
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Srinivas Kantevari
- Fluoro-Agrochemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
| | - Pradeep K Naik
- Centre of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Sambalpur, 768019, Odisha, India.
| |
Collapse
|
24
|
Khairani AF, Harmonia S, Chou Y, Alfarafisa NM, Ramadhanti J. Optimizing Xenograft Models for Breast Cancer: A Comparative Analysis of Cell-Derived and Patient-Derived Implantation Techniques in Pre-Clinical Research. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:1-10. [PMID: 39811602 PMCID: PMC11727321 DOI: 10.2147/bctt.s490532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Purpose The high mortality rate of breast cancer motivates researchers to search for effective treatments. Due to their ability to simulate human conditions, xenograft models such as CDX (Cell line-Derived Xenografts) and PDX (Patient-Derived Xenografts) have gained popularity in pre-clinical research. The choice of xenograft technique is influenced by the type of tumor employed, particularly in more aggressive tumor models like TNBC with metastases. Subcutaneous or orthotopic implantation may influence tumor engraftment rates and the applicability of the models for drug testing. To optimize xenograft models and support the development of breast cancer drugs, selecting a suitable transplantation technique is essential to attaining the best results. Methods This scoping review used PRISMA-Scr methodology to summarize findings from eleven articles published between 2012 and 2024 on pre-clinical trials related to xenograft models for breast cancer considering PDX began traction after 2010. Using specific criteria, the review included studies from electronic platforms. The inclusion criteria ensured relevant English sources were available in full text, while the exclusion criteria eliminated certain types of articles and inadequately comprehensive studies. Results Subcutaneous and orthotopic implantation are critical methods for xenograft models in cancer research. Subcutaneous implantation is less invasive and more manageable but does not fully mimic the tumor's natural environment. Orthotopic implantation accurately mimic the migration, invasion, and molecular characteristics of the original tumor, although the procedure is more complex and requires specialized techniques. The specific research objectives determine their choice, the need for accurate tumor replication, and the testing convenience. Conclusion Orthotopic implantation is the preferable method for developing PDX and CDX models of breast cancer because it closely mimics the tumor microenvironment and metastatic behavior, yielding clinically relevant results for drug testing. Subcutaneous implantation may result in higher engraftment rates, but it cannot accurately represent the complexity of tumors.
Collapse
Affiliation(s)
- Astrid Feinisa Khairani
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Shella Harmonia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Julia Ramadhanti
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
25
|
Ermakova NN, Skurikhin EG, Zharkikh IL, Zhukova MA, Pan VY, Pan ES, Minakova MY, Morozov SG, Kubatiev AA, Dygai AM. A Model of Combination Therapy of Lewis Lung Carcinoma using Resection and Cytostatics. Bull Exp Biol Med 2025; 178:393-398. [PMID: 39951229 DOI: 10.1007/s10517-025-06343-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Indexed: 02/28/2025]
Abstract
We propose a model of combination treatment of Lewis lung carcinoma (LLC) in C57BL/6 mice that includes tumor resection and chemotherapy. A single injection of 5×106 LLC cells into the right lateral subcostal region caused the growth of the primary tumor and its metastasis to the lung. For reducing metastasis and mortality after resection, the primary tumor should be removed with subcutaneous fat on day 8 after inoculation. Antitumor and antimetastatic effects and reduced mortality were achieved by intraperitoneal injection of carboplatin (63.3 mg/kg) and paclitaxel (13.3 mg/kg); chemotherapy was administered twice. The combination of the two approaches increased the survival: the antitumor and antimetastatic effects were observed in 60% of mice with LLC relative to resection alone or treatment with cytostatics alone. However, chemoresistance was formed in 40% of mice, the values of the tumor growth inhibition index and the metastasis inhibition index decreased. This was accompanied by the appearance of cancer stem cells in the circulation with the potential for the formation of spheroids in vitro. The presented model (combination of tumor resection and chemotherapy) can be used in the development of new approaches to improve treatment efficiency in patients with lung cancer.
Collapse
Affiliation(s)
- N N Ermakova
- Institute of General Pathology and Pathophysiology, Moscow, Russia.
| | - E G Skurikhin
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - I L Zharkikh
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - M A Zhukova
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - V Yu Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - E S Pan
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - M Yu Minakova
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - S G Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - A A Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - A M Dygai
- Institute of General Pathology and Pathophysiology, Moscow, Russia
- Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
26
|
Monzavi SM, Muhammadnejad S, Mansouri V, Ashraf H, Ahmadbeigi N. Unwanted disorders and xenogeneic graft-versus-host disease in experimental immunodeficient mice: How to evaluate and how to report. Animal Model Exp Med 2025; 8:20-29. [PMID: 39601130 PMCID: PMC11798742 DOI: 10.1002/ame2.12509] [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: 11/11/2023] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Human-derived tumor models are essential for preclinical development of new anticancer drug entities. Generating animal models bearing tumors of human origin, such as patient-derived or cell line-derived xenograft tumors, is dependent on immunodeficient strains. Tumor-bearing immunodeficient mice are susceptible to developing unwanted disorders primarily irrelevant to the tumor nature; and if get involved with such disorders, reliability of the study results will be undermined, inevitably confounding the research in general. Therefore, a rigorous health surveillance and clinical monitoring system, along with the establishment of a strictly controlled barrier facility to maintain a pathogen-free state, are mandatory. Even if all pathogen control and biosafety measures are followed, there are various noninfectious disorders capable of causing tissue and multiorgan damage in immunodeficient animals. Therefore, the researchers should be aware of sentinel signs to carefully monitor and impartially report them. This review discusses clinical signs of common unwanted disorders in experimental immunodeficient mice, and how to examine and report them.
Collapse
Affiliation(s)
- Seyed Mostafa Monzavi
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
- INSERM U981Institut Gustave RoussyVillejuifFrance
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Hami Ashraf
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD)Shahid Beheshti University of Medical SciencesTehranIran
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
27
|
Hsu CY, Lin J, Wei MF, Chen LH, Liang HKT, Lin FH. Local delivery of carboplatin-loaded hydrogel and calcium carbonate enables two-stage drug release for limited-dose radiation to eliminate mouse malignant glioma. Biomaterials 2025; 312:122746. [PMID: 39106816 DOI: 10.1016/j.biomaterials.2024.122746] [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: 09/13/2022] [Revised: 11/07/2023] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Postoperative radiotherapy remains the gold standard for malignant glioma treatment. Clinical limitations, including tumor growth between surgery and radiotherapy and the emergence of radioresistance, reduce treatment effectiveness and result in local disease progression. This study aimed to develop a local drug delivery system to inhibit tumor growth before radiotherapy and enhance the subsequent anticancer effects of limited-dose radiotherapy. We developed a compound of carboplatin-loaded hydrogel (CPH) incorporated with carboplatin-loaded calcium carbonate (CPCC) to enable two-stage (peritumoral and intracellular) release of carboplatin to initially inhibit tumor growth and to synergize with limited-dose radiation (10 Gy in a single fraction) to eliminate malignant glioma (ALTS1C1 cells) in a C57BL/6 mouse subcutaneous tumor model. The doses of carboplatin in CPH and CPCC treatments were 150 μL (carboplatin concentration of 5 mg/mL) and 15 mg (carboplatin concentration of 4.1 μg/mg), respectively. Mice receiving the combination of CPH-CPCC treatment and limited-dose radiation exhibited significantly reduced tumor growth volume compared to those receiving double-dose radiation alone. Furthermore, combining CPH-CPCC treatment with limited-dose radiation resulted in significantly longer progression-free survival than combining CPH treatment with limited-dose radiation. Local CPH-CPCC delivery synergized effectively with limited-dose radiation to eliminate mouse glioma, offering a promising solution for overcoming clinical limitations.
Collapse
Affiliation(s)
- Cheng-Yi Hsu
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan.
| | - Jason Lin
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan.
| | - Ming-Feng Wei
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, No. 7, Chung Shan South Rd., Zhongzheng Dist., Taipei 10002, Taiwan.
| | - Liang-Hsin Chen
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Division of Proton Therapy, Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei 10672, Taiwan.
| | - Hsiang-Kuang Tony Liang
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, No. 7, Chung Shan South Rd., Zhongzheng Dist., Taipei 10002, Taiwan; Division of Proton Therapy, Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, No.57, Ln. 155, Sec. 3, Keelung Rd., Da'an Dist., Taipei 10672, Taiwan.
| | - Feng-Huei Lin
- Department of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, Taipei 10617, Taiwan; Institute of Biomedical Engineering and Nano-medicine, National Health Research Institutes, No. 35, Keyan Road, Zhunan Town, Miaoli County 35053, Miaoli County, Taiwan.
| |
Collapse
|
28
|
Isaguliants M, Zhitkevich A, Petkov S, Gorodnicheva T, Mezale D, Fridrihsone I, Kuzmenko Y, Kostyushev D, Kostyusheva A, Gordeychuk I, Bayurova E. Enzymatic activity of HIV-1 protease defines migration of tumor cells in vitro and enhances their metastatic activity in vivo. Biochimie 2025; 228:32-43. [PMID: 39128490 DOI: 10.1016/j.biochi.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/09/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Overexpression of aspartic proteases, as cathepsin D, is an independent marker of poor prognosis in breast cancer, correlated with the incidence of clinical metastasis. We aimed to find if HIV-1 aspartic protease (PR) can play a similar role. Murine adenocarcinoma 4T1luc2 cells were transduced with lentivirus encoding inactivated drug-resistant PR, generating subclones PR20.1 and PR20.2. Subclones were assessed for production of reactive oxygen species (ROS), expression of epithelial-mesenchymal transition (EMT) factors, and in vitro migratory activity in the presence or absence of antioxidant N-acetyl cysteine and protease inhibitors. Tumorigenic activity was evaluated by implanting cells into BALB/c mice and following tumor growth by calipering and bioluminescence imaging in vivo, and metastases, by organ imaging ex vivo. Both subclones expressed PR mRNA, and PR20.2, also the protein detected by Western blotting. PR did not induce production of ROS, and had no direct effect on cell migration rate, however, treatment with inhibitors of drug-resistant PR suppressed the migratory activity of both subclones. Furthermore, expression of N-cadherin and Vimentin in PR20.2 cells and their migration were enhanced by antioxidant treatment. Sensitivity of in vitro migration to protease inhibitors and to antioxidant, known to restore PR activity, related the effects to the enzymatic activity of PR. In vivo, PR20.2 cells demonstrated higher tumorigenic and metastatic activity than PR20.1 or parental cells. Thus, HIV-1 protease expressed in breast cancer cells determines their migration in vitro and metastatic activity in vivo. This effect may aggravate clinical course of cancers in people living with HIV-1.
Collapse
Affiliation(s)
- M Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - A Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - S Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - T Gorodnicheva
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| | - D Mezale
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - I Fridrihsone
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Y Kuzmenko
- Engelhardt Institute of Molecular Biology, Academy of Sciences of the Russian Federation, 119991, Moscow, Russia.
| | - D Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - A Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - I Gordeychuk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - E Bayurova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| |
Collapse
|
29
|
Lomeli N, Pearre DC, Lepe J, Argueta DA, Arellano MA, Ricks-Oddie JL, Gupta K, Bota DA. N-acetylcysteine prevents cisplatin-induced cognitive impairments in an ovarian cancer rat model. Cancer Lett 2024; 611:217405. [PMID: 39706252 DOI: 10.1016/j.canlet.2024.217405] [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: 09/05/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Cancer-related cognitive impairment (CRCI) is prevalent among cancer patients. A critical disparity in the CRCI field is that most pre-clinical studies have been conducted on young cancer-free male rodents, although CRCI predominantly affects breast cancer and ovarian cancer women survivors. Since oxidative stress is widely implicated in the development of CRCI, we developed an ovarian cancer xenograft rat model of CRCI in Cr:NIH-RNU female rats to examine whether administration of the antioxidant N-acetylcysteine (NAC) prevents cisplatin-induced CRCI without altering its anti-cancer efficacy. In vitro, delayed treatment with NAC (10 h) following cisplatin treatment in the human ovarian cancer cell line SKOV3.ip1 did not decrease cisplatin's anti-cancer efficacy while mitigating hippocampal dendritic branching damage and neuronal apoptosis. Rats received subcutaneous and intraperitoneal implantation of SKOV3.ip1 cells. Rats received one cisplatin (5 mg/kg) injection every two weeks for a total of four cycles, with or without NAC (250 mg/kg/day), given for five consecutive days during cisplatin treatment. NAC was administered 10 h after cisplatin, based on our in vitro data. Cognitive testing was performed six to seven weeks after treatment cessation. In vivo, cognitive impairments were observed in tumor-bearing rats in the vehicle and cisplatin-treatment groups, while delayed NAC prevented cognitive impairments. Delayed NAC administration did not affect cisplatin-induced tumor volume reduction. Our study supports using NAC to mitigate cisplatin-induced CRCI through the novel development of an ovarian cancer rodent model. This study highlights the importance of developing clinically relevant tumor-bearing models to elucidate the underlying mechanisms associated with CRCI, which will aid in identifying potential therapeutic agents for preventing CRCI.
Collapse
Affiliation(s)
- Naomi Lomeli
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Diana C Pearre
- Gynecologic Oncology, Providence Cancer Institute, Burbank, CA, USA
| | - Javier Lepe
- Department of Pathology, University of California Irvine, Irvine, CA, USA
| | - Donovan A Argueta
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA, USA
| | - Mya A Arellano
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA, USA
| | - Joni L Ricks-Oddie
- Center for Statistical Consulting, Department of Statistics, University of California Irvine, Irvine, CA, USA; Biostatistics, Epidemiology and Research Design Unit, Institute for Clinical and Translational Sciences, University of California Irvine, Irvine, CA, USA
| | - Kalpna Gupta
- Department of Medicine, Division of Hematology/Oncology, University of California Irvine, Irvine, CA, USA
| | - Daniela A Bota
- Department of Neurology, University of California Irvine, Irvine, CA, USA; Department of Pathology, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
30
|
Yuan J, Wang J, Song M, Zhao Y, Shi Y, Zhao L. Brain-targeting biomimetic disguised manganese dioxide nanoparticles via hybridization of tumor cell membrane and bacteria vesicles for synergistic chemotherapy/chemodynamic therapy of glioma. J Colloid Interface Sci 2024; 676:378-395. [PMID: 39032420 DOI: 10.1016/j.jcis.2024.07.121] [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: 03/28/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Glioma is a prevalent brain malignancy associated with poor prognosis. Although chemotherapy serves as the primary treatment for brain tumors, its effectiveness is hindered by the limited ability of drugs to traverse the blood-brain barrier (BBB) and the development of drug resistance linked to tumor hypoxia. Herein, we report the creation of hybrid camouflaged multifunctional nanovesicles comprising membranes of tumor C6 cells (mT) and bacterial outer membrane vesicles (OMVs) and co-loaded with manganese dioxide nanoparticles (MnO2 NPs) and doxorubicin (DOX) to synergistically enhance the chemotherapy/chemodynamic therapy (CDT) of glioma. Owing to OMV-mediated BBB penetration and mT-inherited tumor-homing properties, MnO2-DOX@mT/OMVs can penetrate the BBB and enhance the tumor cell-specific uptake of DOX via "proton sponge effect"-mediated lysosomal escape. This enhances the apoptotic effect induced by DOX and minimizing DOX-associated cardiotoxicity by facilitating the accumulation of DOX at the tumor site. Furthermore, the MnO2 NPs in MnO2-DOX@mT/OMVs can generate potent CDT by accelerating the Fenton-like reaction with DOX-generated H2O2 and achieving glutathione (GSH)-depletion-induced glutathione peroxidase 4 (GPX4) inactivation. These results showed that MnO2-DOX@mT/OMVs, designed for brain tumor targeting, significantly inhibited tumor growth and exhibited favorable biological safety. This innovative approach offers the augmentation of anticancer treatment efficacy via a potential combination of chemotherapy and CDT.
Collapse
Affiliation(s)
- Jiayu Yuan
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Jingchen Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Mingzhu Song
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Yuting Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China.
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou 121000, PR China; Collaborative Innovation Center for Age-related Disease, Jinzhou Medical University, Jinzhou 121001, Liaoning, China.
| |
Collapse
|
31
|
Kannan R, Koh AJ, Kent RN, Bhutada K, Wasi F, Wagner L, Kozloff K, Baker BM, Roca H, McCauley LK. CCL2/CCR2 Signalling in Mesenchymal Stem/Progenitor Cell Recruitment and Fracture Healing in Mice. J Cell Mol Med 2024; 28:e70300. [PMID: 39721002 DOI: 10.1111/jcmm.70300] [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: 08/27/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024] Open
Abstract
Macrophage efferocytosis (clearance of apoptotic cells) is crucial for tissue homeostasis and wound repair, where macrophages secrete factors that promote resolution of inflammation and regenerative signalling. This study examined the role of efferocytic macrophage-associated CCL2 secretion, its influence on mesenchymal stem/progenitor cell (MSPC) chemotaxis, and in vivo cell recruitment using Ccr2-/- (KO) mice with disrupted CCL2 receptor signalling in two regenerative models: ossicle implants and ulnar stress fractures. Single cell RNA sequencing and PCR validation indicated that efferocytosis of various apoptotic cells at bone injury sites (osteoblasts, pre-osteoblasts, MSPC) upregulated CCL2. CCL2 gradients enhanced MSPC migration through type I collagen matrices. In vivo, MSPC (LepR+) infiltration was significantly reduced while macrophage (F4/80+) infiltration increased in KO ossicle implants versus WT. In ulnar stress fractures, micro-CT revealed increased mineralized callus incidence in CCR2 KO male mice 5 days post injury (dpi) versus WT. By 7-dpi callus fractional bone volume, trabecular thickness, and bone mineral density were increased versus WT. Immunohistochemistry of mice 5-dpi confirmed an increase in callus area (including soft tissue); however, the percent of osteoprogenitors (%Osx+) within the callus was not different. These findings suggest that CCL2 differentially impacts MSPC recruitment depending on bone wound healing model.
Collapse
Affiliation(s)
- Rahasudha Kannan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy J Koh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Kaira Bhutada
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Fatima Wasi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Leon Wagner
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Kenneth Kozloff
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Hernan Roca
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Mizutani T, Boretto M, Lim S, Drost J, González DM, Oka R, Geurts MH, Begthel H, Korving J, van Es JH, van Boxtel R, Clevers H. Recapitulating the adenoma-carcinoma sequence by selection of four spontaneous oncogenic mutations in mismatch-repair-deficient human colon organoids. NATURE CANCER 2024; 5:1852-1867. [PMID: 39487295 PMCID: PMC11663794 DOI: 10.1038/s43018-024-00841-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/23/2024] [Indexed: 11/04/2024]
Abstract
Carcinogenesis results from the sequential acquisition of oncogenic mutations that convert normal cells into invasive, metastasizing cancer cells. Colorectal cancer exemplifies this process through its well-described adenoma-carcinoma sequence, modeled previously using clustered regularly interspaced short palindromic repeats (CRISPR) to induce four consecutive mutations in wild-type human gut organoids. Here, we demonstrate that long-term culture of mismatch-repair-deficient organoids allows the selection of spontaneous oncogenic mutations through the sequential withdrawal of Wnt agonists, epidermal growth factor (EGF) agonists and the bone morphogenetic protein (BMP) antagonist Noggin, while TP53 mutations were selected through the addition of Nutlin-3. Thus, organoids sequentially acquired mutations in AXIN1 and AXIN2 (Wnt pathway), TP53, ACVR2A and BMPR2 (BMP pathway) and NRAS (EGF pathway), gaining complete independence from stem cell niche factors. Quadruple-pathway (Wnt, EGF receptor, p53 and BMP) mutant organoids formed solid tumors upon xenotransplantation. This demonstrates that carcinogenesis can be recapitulated in a DNA repair-mutant background through in vitro selection that targets four consecutive cancer pathways.
Collapse
Affiliation(s)
- Tomohiro Mizutani
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Matteo Boretto
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Sangho Lim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jarno Drost
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Diego Montiel González
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rurika Oka
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Maarten H Geurts
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Ruben van Boxtel
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland.
| |
Collapse
|
33
|
Song JH, Kim SJ, Kwon S, Jeon SY, Park SE, Choi SJ, Oh SY, Jeon HB, Chang JW. Nervonic acid improves fat transplantation by promoting adipogenesis and angiogenesis. Int J Mol Med 2024; 54:108. [PMID: 39364738 PMCID: PMC11517738 DOI: 10.3892/ijmm.2024.5432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 10/05/2024] Open
Abstract
Adipose tissue engraftment has become a promising strategy in the field of regenerative surgery; however, there are notable challenges associated with it, such as resorption of 50‑90% of the transplanted fat or cyst formation due to fat necrosis after fat transplantation. Therefore, identifying novel materials or methods to improve the engraftment efficiency is crucial. The present study investigated the effects of nervonic acid (NA), a monounsaturated very long‑chain fatty acid, on adipogenesis and fat transplantation, as well as its underlying mechanisms. To assess this, NA was used to treat cells during adipogenesis in vitro, and the expression levels of markers, including PPARγ and CEBPα, and signaling molecules were detected through reverse transcription‑quantitative PCR and western blotting. In addition, NA was mixed with fat grafts in in vivo fat transplantation, followed by analysis through Oil Red O staining, hematoxylin & eosin staining and immunohistochemistry. It was demonstrated that NA treatment accelerated adipogenesis through activation of the Akt/mTOR pathway and inhibition of Wnt signaling. NA treatment enriched the expression of Akt/mTOR signaling‑related genes, and increased the expression of genes involved in angiogenesis and fat differentiation in human mesenchymal stem cells (MSCs). Additionally, NA effectively improved the outcome of adipose tissue engraftment in mice. Treatment of grafts with NA at transplantation reduced the resorption of transplanted fat and increased the proportion of perilipin‑1+ adipocytes with a lower portion of vacuoles in mice. Moreover, the NA‑treated group exhibited a reduced pro‑inflammatory response and had more CD31+ vessel structures, which were relatively evenly distributed among viable adipocytes, facilitating successful engraftment. In conclusion, the present study demonstrated that NA may not only stimulate adipogenesis by regulating signaling pathways in human MSCs, but could improve the outcome of fat transplantation by reducing inflammation and stimulating angiogenesis. It was thus hypothesized that NA could serve as an adjuvant strategy to enhance fat engraftment in regenerative surgery.
Collapse
Affiliation(s)
- Jae Hoon Song
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soojin Kwon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Su Yeon Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Republic of Korea
| |
Collapse
|
34
|
Zhu X, Potterfield R, Gruber KA, Zhang E, Newton SD, Norgard MA, Levasseur PR, Bai P, Chen X, Gu Q, Grossberg AJ, Marks DL. Melanocortin-4 receptor antagonist TCMCB07 alleviates chemotherapy-induced anorexia and weight loss in rats. J Clin Invest 2024; 135:e181305. [PMID: 39509261 DOI: 10.1172/jci181305] [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: 03/21/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
Cancer patients undergoing chemotherapy often experience anorexia and weight loss that substantially deteriorates overall health, reduces treatment tolerance and quality of life, and worsens oncologic outcomes. There are currently few effective therapeutic options to mitigate these side effects. The central melanocortin system, which plays a pivotal role in regulating appetite and energy homeostasis, presents a logical target for treating anorexia and weight loss. In this preclinical study, we evaluated the efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor, in mitigating anorexia and weight loss in several rat models of chemotherapy: cisplatin, 5-fluorouracil, cyclophosphamide, vincristine, doxorubicin, and a combination of irinotecan and 5-fluorouracil. Our results indicate that peripheral administration of TCMCB07 improved appetite, stabilized body weight, preserved fat and heart mass, and slightly protected lean mass after multiple cycles of chemotherapy. Furthermore, combining TCMCB07 with a growth differentiation factor 15 antibody enhanced treatment effectiveness. Similar effects from TCMCB07 treatment were observed in a rat tumor model following combination chemotherapy. No notable adverse effects nor increased chemotherapy-related toxicities were observed with TCMCB07 treatment. These findings suggest that peripheral administration of TCMCB07 holds promise as a therapeutic approach for alleviating chemotherapy-induced anorexia and weight loss, potentially benefiting numerous patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute and
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | | | - Kenneth A Gruber
- Endevica Bio, Northbrook, Illinois, USA
- Department of Medical Pharmacology and Physiology and the Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | | | | | | | - Peter R Levasseur
- Papé Family Pediatric Research Institute and
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| | - Peng Bai
- In Vivo Pharmacology Unit, WuXi App Tec, Nantong, Jiangsu, China
| | - Xu Chen
- In Vivo Pharmacology Unit, WuXi App Tec, Shanghai, China
| | - Qingyang Gu
- In Vivo Pharmacology Unit, WuXi App Tec, Shanghai, China
| | - Aaron J Grossberg
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
- Department of Radiation Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | | |
Collapse
|
35
|
Chen L, Xing J, Lv J, Si S, Wang H, Yu W. Corynoxine suppresses lung adenocarcinoma proliferation and metastasis via inhibiting PI3K/AKT pathway and suppressing Cyclooxygenase-2 expression. Hereditas 2024; 161:41. [PMID: 39511658 PMCID: PMC11542349 DOI: 10.1186/s41065-024-00343-x] [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: 06/06/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is the most common lung cancer subtype, and the prognosis of affected patients is generally poor. The traditional Chinese medicine Uncaria rhychophaylla has been reported to exhibit anti-lung cancer properties. Accordingly, the main bioactive ingredient in Uncaria rhychophaylla, Corynoxine, may hold great value as a treatment for lung cancer. METHODS The impact of Corynoxine on the viability of LUAD cells was assessed using the Cell Counting Kit-8 (CCK-8) assay. Apoptosis in A549 cells was evaluated via flow cytometry. Migration and invasion capabilities were determined through wound healing and Transwell assays, respectively. The key pathways targeted by Corynoxine in LUAD were identified using a network pharmacology approach. Additionally, Western immunoblotting, quantitative real-time PCR (qRT-PCR), and ELISA assays were conducted to validate the underlying mechanisms. The in vivo anti-tumor efficacy of Corynoxine was assessed in xenograft nude mice. RESULTS In this study, Corynoxine treatment was found to markedly suppress in vitro LUAD cell proliferative, migratory, and invasive activity. It additionally downregulated Vimentin and promoted E-cadherin upregulation consistent with the disruption of epithelial-mesenchymal transition (EMT) induction while also accelerating apoptotic death. Furthermore, network pharmacology analysis revealed that the PI3K/AKT pathway is a potential target of Corynoxine in LUAD. In vitro assays demonstrated that treatment with Corynoxine resulted in the suppression of PI3K/AKT signaling and a consequent drop in cyclooxygenase-2 (COX-2) expression. These findings were further confirmed in vivo in mice harboring A549 tumor xenografts in which Corynoxine was able to interfere with the PI3K/AKT/COX-2 signaling axis. CONCLUSION This study elucidated the potential effects of Corynoxine in suppressing proliferation and metastasis in LUAD, along with investigating the underlying mechanisms. These data highlight the promise of Corynoxine as a novel therapeutic tool for the treatment of individuals diagnosed with LUAD.
Collapse
Affiliation(s)
- Liping Chen
- Department of Central Laboratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Jing Xing
- Department of Central Laboratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiapei Lv
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Sainv Si
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Huaying Wang
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wanjun Yu
- Department of Respiratory, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
36
|
Parpex G, Chassaing B, Bourdon M, Santulli P, Doridot L, Thomas M, Batteux F, Chouzenoux S, Chapron C, Nicco C, Marcellin L. Western diet promotes endometriotic lesion growth in mice and induces depletion of Akkermansia muciniphila in intestinal microbiota. BMC Med 2024; 22:513. [PMID: 39501247 PMCID: PMC11539706 DOI: 10.1186/s12916-024-03738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Endometriosis, affecting 10% of women in their reproductive years, remains poorly understood. Both individual and environmental unexplained factors are implicated in this heterogenous condition. This study aims to examine the influence of a Western diet on endometriosis lesion development in mice and to uncover the mechanisms involved. METHODS Mice were fed either a control diet or a Western diet (high in fatty acids and low in fiber) for 4 weeks. Endometriosis was then surgically induced, and lesion development was monitored by ultrasound. After 7 weeks, the mice were sacrificed for analysis of lesion characteristics through RT-qPCR, immunohistochemistry, and flow cytometry. Additionally, the intestinal microbiota was assessed using 16S rRNA gene sequencing. RESULTS Mice on the Western diet developed lesions that were significantly twice as large compared to those on the control diet. These lesions exhibited greater fibrosis and proliferation, alongside enhanced macrophage activity and leptin pathway expression. Changes in the intestinal microbiota were significantly noted after endometriosis induction, regardless of diet. Notably, mice on the Western diet with the most substantial lesions showed a loss of Akkermansia Muciniphila in their intestinal microbiota. CONCLUSIONS A Western diet significantly exacerbates lesion size in a mouse model of endometriosis, accompanied by metabolic and immune alterations. The onset of endometriosis also leads to substantial shifts in intestinal microbiota, suggesting a potential link between diet, intestinal health, and endometriosis development.
Collapse
Affiliation(s)
- Guillaume Parpex
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France.
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France.
| | - Benoît Chassaing
- Institut Pasteur, Université Paris Cité, Microbiome-Host Interaction Group, INSERM U1306, Paris, France
| | - Mathilde Bourdon
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Pietro Santulli
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Ludivine Doridot
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Marine Thomas
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Frédéric Batteux
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | | | - Charles Chapron
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Carole Nicco
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Louis Marcellin
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| |
Collapse
|
37
|
Fu JF, Hsu CL, Hsu PC. The antitumor activity of osimertinib plus palbociclib in non-small cell lung cancer patient-derived xenograft (PDX)/2D/3D culture models harboring EGFR amplification and CDKN2A/2B homozygous deletions. Neoplasia 2024; 57:101039. [PMID: 39146623 PMCID: PMC11375314 DOI: 10.1016/j.neo.2024.101039] [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: 07/03/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Non-small cell lung cancer (NSCLC) patients without targetable driver mutation have limited treatment options. In this study, we aimed to explore a new therapeutic strategy by using established nine patient-derived xenograft (PDX) and two-dimensional (2D) /3D culture models with specific genetic alternations. The gene mutations and copy number aberrations were detected by next-generation sequencing and confirmed using polymerase chain reaction (PCR) followed by DNA sequencing, and genomic DNA quantitative PCR. Protein expression was evaluated by immunohistochemistry. Drug sensitivities of PDX/2D/3D models were evaluated by in vivo and in vitro antitumor assays. RNA interference was performed to silence gene expression. Our study found that 44.4 % (4/9) of cases had CDKN2A homozygous deletion (homdel), while 33.3 % (3/9) had CDKN2B homdel. Additionally, 22.2 % (2/9) had amplification (amp) in wildtype CDK4, 44.4 % (4/9) in CDK6, and 44.4 % (4/9) in EGFR. Among the cases, 77.8 % (7/9) lacked CDKN2A, and 33.3 % (3/9) had high CDK4, CDK6, and EGFR had high protein expression. Moreover, 33.3 % (3/9) had KRAS mutations, and 66.7 % (6/9) had TP53 mutations. Antitumor activity of osimertinib plus palbociclib was assessed in four PDX/2D/3D models, two of which had simultaneous EGFR amp and CDKN2A/2B homdel. The data showed that NSCLC with EGFR amp and CDKN2A/2B homdel were sensitive to combined drugs. Additional oncogenic KRAS mutation reduced the drug's antitumor effect. EGFR amp is responsible for osimertinib sensitivity. Osimertinib plus palbociclib effectively treat NSCLC with wildtype EGFR and CDK6 amp and CDKN2A/2B homdel in the absence of oncogenic KRAS mutation.
Collapse
Affiliation(s)
- Jen-Fen Fu
- Department of Medical Research, Chang Gung Memorial Hospital, Guishan, Taoyuan, Taiwan.
| | - Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Guishan, Taoyuan, Taiwan.
| | - Ping-Chih Hsu
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Chang Gung University, Guishan, Taoyuan, Taiwan.
| |
Collapse
|
38
|
Simanullang RH, Siahaan JM, Situmorang PC. Histological Alterations of Cervical Cancer Following Zanthoxylum acanthopodium DC Therapy in Relation to E7, pRb, EGFR and p16 Expression. Pak J Biol Sci 2024; 27:602-612. [PMID: 39731430 DOI: 10.3923/pjbs.2024.602.612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
<b>Background and Objective:</b> Cervical cancer is the second most common cancer in Indonesia, where traditional herbal treatments like <i>Zanthoxylum acanthopodium</i> (andaliman) are culturally used. Investigating protein biomarkers such as E7, pRb, EGFR and p16 can help assess the efficacy of these treatments. <b>Materials and Methods:</b> There were 5 groups in this study: 2 control groups (C- and C+) and 3 treatment groups (each receiving one of three doses). Oral administration of andaliman was performed for 30 days in cancer model rats, after which the cervix was dissected, cervical tissue was taken and immunohistochemistry repair was performed. Statistical analysis was performed using the Kruskal-Wallis test with a p<0.05. <b>Results:</b> As <i>Zanthoxylum acanthopodium</i> DC dose rose, cervical tissue E7, EGFR and p16 expression decreased. However, greater doses of this plant increased cervical tissue pRb protein. Cervical cancer histology exhibited increased nuclear size, irregular cellular structure, atypical cell shape, higher nuclear-cytoplasmic ratio and various nuclear shape variants. This herb induced tissue to show well-organized non-hyperchromatic cells that resembled normal clusters. <b>Conclusion:</b> <i>Zanthoxylum acanthopodium</i> DC improved cervical tissue and balanced cervical cancer biomarker proteins such E7, EGFR, pRB and p16.
Collapse
|
39
|
Fang X, Yu WY, Zhu CM, Zhao N, Zhao W, Xie TT, Wei LJ, Sun XR, Xie J, Zhao Y. Chromosome instability functions as a potential therapeutic reference by enhancing chemosensitivity to BCL-XL inhibitors in colorectal carcinoma. Acta Pharmacol Sin 2024; 45:2420-2431. [PMID: 39187678 PMCID: PMC11489767 DOI: 10.1038/s41401-024-01372-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Chromosome instability (CIN) and subsequent aneuploidy are prevalent in various human malignancies, influencing tumor progression such as metastases and relapses. Extensive studies demonstrate the development of chemoresistance in high-CIN tumors, which poses significant therapeutic challenges. Given the association of CIN with poorer prognosis and suppressed immune microenvironment observed in colorectal carcinoma (CRC), here we aimed to discover chemotherapeutic drugs exhibiting increased inhibition against high-CIN CRC cells. By using machine learning methods, we screened out two BCL-XL inhibitors Navitoclax and WEHI-539 as CIN-sensitive reagents in CRC. Subsequent analyses using a CIN-aneuploidy cell model confirmed the vulnerability of high-CIN CRC cells to these drugs. We further revealed the critical role of BCL-XL in the viability of high-CIN CRC cells. In addition, to ease the evaluation of CIN levels in clinic, we developed a three-gene signature as a CIN surrogate to predict prognosis, chemotherapeutic and immune responses in CRC samples. Our results demonstrate the potential value of CIN as a therapeutic target in CRC treatment and the importance of BCL-XL in regulating survival of high-CIN CRC cells, therefore representing a valuable attempt to translate a common trait of heterogeneous tumor cells into an effective therapeutic target.
Collapse
Affiliation(s)
- Xiao Fang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Clinical Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Wen-Ying Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Chun-Miao Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Nan Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Wei Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Ting-Ting Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Li-Jie Wei
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Xi-Ran Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Juan Xie
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China
| | - Ya Zhao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, 225001, China.
| |
Collapse
|
40
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024; 25:5080-5112. [PMID: 39415049 PMCID: PMC11549407 DOI: 10.1038/s44319-024-00288-2] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
41
|
Sadasivam P, Hartimath SV, Khanapur S, Ramasamy B, Cheng P, Feng CZ, Green D, Goggi JL, Robins EG, Yan R. Novel [ 18F]FPG-interleukin-2 conjugate for monitoring immune checkpoint therapy with positron emission tomography. Biomed Pharmacother 2024; 180:117617. [PMID: 39471651 DOI: 10.1016/j.biopha.2024.117617] [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: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024] Open
Abstract
18F-interleukin-2 based PET imaging of activated T cells serves as a potential tool for non-invasive response prediction, treatment evaluation, and patient stratification in cancer immune checkpoint therapy. Herein, we report the radiolabelling of interleukin-2 (IL-2) with a novel arginine selective bioconjugation reagent, 4-[18F]fluorophenylglyoxal ([18F]FPG). Good non-decay corrected bioconjugation efficiencies of 29 ± 4 % (n = 5) were obtained for the [18F]FPG-IL-2. [18F]FPG-IL-2 uptake by the phytohemagglutinin-activated Jurkat cells (50.5 ± 1.2 %, n = 3) was significantly higher compared to the non-activated Jurkat cells (12.9 ± 1.1 %, n = 3). The [18F]FPG-IL-2 uptake was blocked by the pre-treatment of activated Jurkat cells with excess native IL-2 (22.3 ± 2.2 %, n = 3). Dynamic PET imaging and ex vivo biodistribution study of [18F]FPG-IL-2 in healthy and CT26 tumour bearing mice demonstrated hepatobiliary and renal clearance with minimal uptake in other organs and CT26 tumours. [18F]FPG-IL-2 PET imaging was applied to non-invasively monitor immune checkpoint therapy in CT26 tumour bearing mice, treated with IgG (control), ⍺PD-1 (monotherapy), and ⍺PD-1+⍺CTLA-4 (combination therapy). Significant uptake was observed in the spleens and tumours of the mice in the combination therapy group, which was associated with increased cytotoxic CD8+ T-cell infiltration and reduced tumour volumes. [18F]FPG-IL-2 based PET imaging has the potential to monitor immune checkpoint therapy.
Collapse
Affiliation(s)
- Pragalath Sadasivam
- School of Biomedical Engineering and Imaging Sciences, Department of Imaging Chemistry and Biology, King's College London, UK; Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore; Clinical Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Minerva Imaging ApS, Lyshøjvej 21, Ølstykke 3650, Denmark
| | - Siddesh V Hartimath
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Shivashankar Khanapur
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Boominathan Ramasamy
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Peter Cheng
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - Chin Zan Feng
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore
| | - David Green
- Clinical Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Julian L Goggi
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore; Minerva Imaging ApS, Lyshøjvej 21, Ølstykke 3650, Denmark
| | - Edward G Robins
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology, and Research (A⁎ STAR), 11 Biopolis Way, #01-02 Helios, Singapore 138667, Singapore; Clinical Imaging Research Centre, 14 Medical Drive, #B01-01 Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Molecular Imaging and Therapy Research Unit, South Australian Health, and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace & George Street, Adelaide, SA 5000, Australia
| | - Ran Yan
- School of Biomedical Engineering and Imaging Sciences, Department of Imaging Chemistry and Biology, King's College London, UK.
| |
Collapse
|
42
|
Liu Y, Zhang D, Kong M, Wang Y, Mei H, Shan C, Meng J, Zou Y, Wang J. Synaptic vesicle protein 2-targeted doxorubicin-loaded liposome for effective neuroblastoma therapy. Biomed Pharmacother 2024; 180:117548. [PMID: 39413621 DOI: 10.1016/j.biopha.2024.117548] [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: 07/08/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
Neuroblastoma, a pediatric cancer originating from neural crest tissues of the sympathetic nervous system, poses significant treatment challenges due to its molecular diversity and restricted druggable targets. While chemotherapy is a common treatment, its drawbacks, including poor targeting of cancer cells and nonspecific cytotoxicity, highlight the urgent need for innovative and effective therapeutic strategies. Herein, we developed a novel drug by coupling the receptor binding domain of botulinum neurotoxin type A (Hc) fused with monomeric streptavidin (mSA) to biotin coated doxorubicin (Dox)-loaded liposome, via interaction between mSA and biotin. The resultant Hc-coated liposome (Hc-Lipo@Dox) actively targeted the recycling synaptic vesicle 2 protein (SV2) abundantly expressed on the surface of neuroblastoma cells. Our results revealed that Hc-Lipo@Dox more effectively entered the neuroblastoma SH-SY5Y cells, inducing apoptosis compared to non-targeted liposome and free Dox. Moreover, Hc-Lipo@Dox rapidly enriched Dox in the subcutaneously implanted neuroblastoma tumor in nude mice, resulting potent anti-neuroblastoma effect compared to non-targeted liposomes or free Dox. Importantly, Hc-Lipo@Dox significantly improved the survival rate of treated mice, while also exhibiting a favorable safety profile with no discernible impact on mobility or observable side effects. These findings highlight the potential of SV2-targeted Dox liposome as a promising and well-tolerated chemotherapy approach for neuroblastoma treatment. Moreover, the technology established here has broader applications for various cancer therapies by substituting the Hc moiety with other tumor-specific targeting moieties.
Collapse
Affiliation(s)
- Yang Liu
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Dongya Zhang
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Miaomiao Kong
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Yibin Wang
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Huiyuan Mei
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Chunxu Shan
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| | - Jianghui Meng
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| | - Yan Zou
- School of Life Sciences, Henan University, Kaifeng 475001, China.
| | - Jiafu Wang
- School of Biotechnology, Dublin City University, Collins Avenue, Dublin, Ireland.
| |
Collapse
|
43
|
Dellalibera-Joviliano R, Garcia ME, Marins M, Fachin ALÚ, Couto LB, Mesquita E, Komoto TT, Silva G, Neto WC, Orlando L, Durand M, França SC, Bestetti RB. Interleukin-12 treatment reduces tumor growth and modulates the expression of CASKA and MIR-203 in athymic mice bearing tumors induced by the HGC-27 gastric cancer cell line. Pathol Res Pract 2024; 263:155625. [PMID: 39393266 DOI: 10.1016/j.prp.2024.155625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
Gastric cancer (GC) is one of the most common malignant tumors in the digestive system and due to its poor prognosis, there is an increase in the demand for more effective anticancer therapies. Interleukins are potential anticancer agents which can modulate expression of cancer related genes and have therapeutic effects. Interleukin 12 (IL-12) exhibits potent anti-tumor, anti-angiogenic and anti-metastatic activities and represents the ideal candidate for tumor immunotherapy, due to its ability to activate both innate and adaptive immunities. The aim of this study was to evaluate the effect of IL-12 administration on GC tumor growth induced in the cancer xenograft nude mouse model. Tumor development was analyzed weekly and after 8 weeks, the animals were sacrificed for cytokine analysis (IL-4, TNF-alfa, IL-2, INF-gamma, IL-12, IL-10, TGF-beta) by ELISA. The tumor cells in the implanted areas of the animals that developed solid growth of the tumor (anatomopathological analysis was performed). We have also evaluated CASK and miR203 expression, two related cell invasion factors, in the induced tumors after administration of 6 n/kg IL-12. The development of tumor masses was observed in all groups of animals inoculated with HGC-27 neoplastic cells. In animals treated with 6 n/kg IL-12, there was no tumor development confirmed by anatomopathological analysis. Changes in the levels of pro and anti-inflammatory cytokines were also observed. Our results indicated that miR203 expression was elevated while CASK was downregulated. These results suggest that IL-12 treatment repress the tumor growth by induction of miR203 expression which in turn repress CASK expression.
Collapse
Affiliation(s)
| | - Marcelo E Garcia
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil.
| | - Mozart Marins
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil; Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Ana L Úcia Fachin
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil; Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Lucélio B Couto
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Edgar Mesquita
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil; Syrian Lebanese Hospital, São Paulo, Brazil
| | - Tatiana T Komoto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Gabriel Silva
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto-USP, Ribeirão Preto, SP, Brazil
| | - Walter Campos Neto
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Leonardo Orlando
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Marina Durand
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Suzelei C França
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| | - Reinaldo B Bestetti
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP 2201, Brazil
| |
Collapse
|
44
|
Islam TMT, Mahat NC, Shaker IA, Rahman SA, Kabir MH, Shohel MA, Kamruzzaman M, Tang AK. Investigation of the Relationship Between Brown HT Dye Exposure and Mammary Tumor Development in Female Rats: An Assessment of the Potential Risk of Breast Cancer. Cureus 2024; 16:e73351. [PMID: 39659309 PMCID: PMC11631162 DOI: 10.7759/cureus.73351] [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] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Azo dyes featuring one (monoazo) or several intramolecular NQN bonds are utilized in the food, pharmaceutical, and textile industries. The food azo dye chocolate brown HT (E155) adversely affects hepatic and renal function upon prolonged consumption. This study aimed to assess the carcinogenic potential of E155 in the development of mammary tumors and breast cancer. METHODS A total of 20 female Long-Evans rats (eight to nine weeks old) were randomly assigned to five groups, each consisting of four rats. The control (female control) group received a regular diet, whereas the positive control (female positive control) group received 7,12-dimethylbenz(a)anthracene. The remaining three groups received 200, 400, or 600 mg/kg body weight (BW)/day E155 for 40 weeks. Tumor development, BW, and biochemical, hematological, and histological data were monitored. RESULTS BW decreased significantly with increasing dosages in the female moderate dose (FMD) group. Blood counts indicated potential microcytic anemia and inflammation in the treatment groups, especially in the female high-dose (FHD) group. E155 dose-dependently impaired renal function and increased blood creatinine and uric acid levels. Elevated serum glutamic pyruvic transaminase (SGPT) and serum glutamic-oxaloacetic transaminase levels indicate abnormal liver function. FHD animals had more tumors and larger sizes. Higher alpha-fetoprotein (AFP) and cancer antigen levels were detected even at low doses. Histopathological analysis revealed that E155 causes mammary gland fibroadenomas, ductal carcinoma in situ, and hyperplasia. It also causes circular layer granulomas, fibrosis, and crypt abscesses in the intestines of FMD and FHD. CONCLUSION The current study suggests that prolonged exposure to E155 may result in a higher incidence of mammary tumors, indicating an elevated risk for the onset of breast cancer.
Collapse
Affiliation(s)
- T M Tawabul Islam
- Department of Food and Nutrition, Faculty of Applied Science, Parul University, Vadodara, IND
| | - Nirmal Chandra Mahat
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, BGD
| | - Ivvala Anand Shaker
- Department of Biochemistry, Swaminarayan Institute of Medical Sciences and Research, Swaminarayan University, Shree Swaminarayan Vishvamangal Gurukul, Kalol, IND
| | - Sheikh Arafat Rahman
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, BGD
| | - Md Humayan Kabir
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, BGD
| | - Mustakin Ahmed Shohel
- Department of Food and Nutrition, Faculty of Applied Science, Parul University, Vadodara, IND
| | - Md Kamruzzaman
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, BGD
| | - Abul Kashem Tang
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, BGD
| |
Collapse
|
45
|
Zhao Y, Gong J, Liu H, Huang H, Tan WS, Cai H. A chemically defined, mechanically tunable, and bioactive hyaluronic acid/alginate double-network hydrogel for liver cancer organoid construction. Int J Biol Macromol 2024; 282:136707. [PMID: 39442832 DOI: 10.1016/j.ijbiomac.2024.136707] [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: 06/04/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer organoids replicate the pathophysiology of primary tumors, making them ideal for drug screening and efficacy evaluation. However, their growth in complex, variable, animal-derived matrices hinders practical application. Here, we designed an easily accessible, chemically defined, biocompatible double-network hydrogel (HADR) using methacrylated hyaluronic acid (HAMA), sodium alginate (SA), methacrylamide dopamine (DMA), and c(RGDFC) for liver cancer organoid culture. By optimizing critical extracellular matrix (ECM) parameters, the HADR hydrogel achieves compatibility with the physiological mechanics of the human liver and fosters the adhesion and proliferation of multiple cell types. In vitro drug efficacy tests showed that HepG2 cell line-derived liver cancer organoids exhibited higher IC50 values than 2D cultures, indicating greater drug resistance. Subcutaneous tumor models in nude mice revealed that HADR hydrogels created a microenvironment for HepG2 cells mirroring the natural tumor ECM, leading to increased tumor volume, denser cell arrangement, and concurrent microvascular development. In vivo drug efficacy evaluations indicated that DOX treatment downregulated Ki-67 and MMP-9 expression, inhibiting HepG2 cell proliferation, invasion, and metastasis. These findings demonstrate the potential of HADR hydrogels for liver cancer organoid culture, offering new strategies for personalized drug screening and efficacy evaluation.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Junjie Gong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hanwen Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Huimin Huang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
46
|
Muñoz-Moreno L, Gómez-Calcerrada MI, Arenas MI, Carmena MJ, Prieto JC, Schally AV, Bajo AM. Antagonist of Growth Hormone-Releasing Hormone Receptor MIA-690 Suppresses the Growth of Androgen-Independent Prostate Cancers. Int J Mol Sci 2024; 25:11200. [PMID: 39456984 PMCID: PMC11508372 DOI: 10.3390/ijms252011200] [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: 08/24/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The development of resistance remains the primary challenge in treating castration-resistant prostate cancer (CRPC). GHRH receptors (GHRH-R), which are coupled to G-proteins (GPCRs), can mediate EGFR transactivation, offering an alternative pathway for tumour survival. This study aimed to evaluate the effects of the GHRH-R antagonist MIA-690, in combination with the EGFR inhibitor Gefitinib, on cell viability, adhesion, gelatinolytic activity, and the cell cycle in advanced prostate cancer PC-3 cells. The findings demonstrate a synergistic effect between MIA-690 and Gefitinib, leading to the inhibition of cell viability, adhesion, and metalloprotease activity. Cell cycle analysis suggests that both compounds induce cell cycle arrest, both individually and in combination. Furthermore, similar effects of the GHRH-R antagonist MIA-690 combined with Gefitinib were observed in PC-3 tumours developed by subcutaneous injection in athymic nude mice 36 days post-inoculation. These results indicate that combined therapy with a GHRH-R antagonist and an EGFR inhibitor exerts a stronger antitumor effect compared to monotherapy by preventing transactivation between EGFR and GHRH-R in CRPC.
Collapse
Affiliation(s)
- Laura Muñoz-Moreno
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. Isabel Gómez-Calcerrada
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. Isabel Arenas
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Biología Celular, Departamento de Biomedicina y Biotecnología, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - M. José Carmena
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - Juan C. Prieto
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| | - Andrew V. Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33125, USA
- Department of Pathology and Medicine, Division of Hematology/Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ana M. Bajo
- Grupo de Investigación Cánceres de Origen Epitelial, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain; (M.I.G.-C.); (M.J.C.); (A.M.B.)
- Unidad de Bioquímica y Biología Molecular, Departamento de Biología de Sistemas, Campus Científico-Tecnológico, Universidad de Alcalá, 28805 Madrid, Spain
| |
Collapse
|
47
|
Sharpe MA, Ijare OB, Raghavan S, Baskin AM, Baskin BN, Baskin DS. Targeting the Leloir Pathway with Galactose-Based Antimetabolites in Glioblastoma. Cancers (Basel) 2024; 16:3510. [PMID: 39456605 PMCID: PMC11506710 DOI: 10.3390/cancers16203510] [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: 08/28/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) uses Glut3 and/or Glut14 and the Leloir pathway to catabolize D-Galactose (Gal). UDP-4-deoxy-4-fluorogalactose (UDP-4DFG) is a potent inhibitor of the two key enzymes, UDP-galactose-4-epimerase (GALE) and UDP-Glucose 6-dehydrogenase (UGDH), involved in Gal metabolism and in glycan synthesis. The Gal antimetabolite 4-deoxy-4-fluorogalactose (4DFG) is a good substrate for Glut3/Glut14 and acts as a potent glioma chemotherapeutic. METHODS Primary GBM cell cultures were used to examine toxicity and alterations in glycan composition via lectin binding in fixed cells and by Western blots. Toxicity/efficacy in vivo data was performed in mouse flank and intracranial models. The effect of 4DFG on D-glucose (Glc) metabolism in GBM cells was assessed by using 13C NMR-based tracer studies. RESULTS 4DFG is moderately potent against GBM cells (IC50: 125-300 µM). GBM glycosylation is disrupted by 4DFG. Survival analysis in an intracranial mouse model showed that treatment with 4DFG (6 × 25 mg/kg of 4DFG, intravenously) improved outcomes by three-fold (p < 0.01). Metabolic flux analysis revealed that both glycolytic and mitochondrial metabolic fluxes of [U-13C]Glc were significantly decreased in the presence of 4DFG in GBM cells. CONCLUSION A functional Gal-scavenging pathway in GBM allows Gal-based antimetabolites to act as chemotherapeutics. 4DFG is metabolized by GBM in vitro and in vivo, is lethal to GBM tumors, and is well tolerated in mice.
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Omkar B. Ijare
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Sudhir Raghavan
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Alexandra M. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - Brianna N. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Houston Methodist Hospital, Houston, TX 77030, USA; (S.R.); (A.M.B.); (B.N.B.); (D.S.B.)
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Houston Methodist Academic Institute, Houston, TX 77030, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
- Texas A & M Medical School, Houston, TX 77030, USA
| |
Collapse
|
48
|
Chen YW, He AC, Huang TY, Lai DH, Wang YP, Liu WW, Kuo WT, Hou HH, Cheng SJ, Lee CY, Chuang WC, Chang CC, Lee BS. Iontophoresis-Enhanced Buccal Delivery of Cisplatin-Encapsulated Chitosan Nanoparticles for Treating Oral Cancer in a Mouse Model. Int J Nanomedicine 2024; 19:10435-10453. [PMID: 39430308 PMCID: PMC11491087 DOI: 10.2147/ijn.s475742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Cisplatin is one of the most effective chemotherapeutic drugs used in oral cancer treatment, but systemic administration has side effects. The purpose of this study was to evaluate the effect of iontophoresis on the enhancement of cisplatin release from cisplatin-encapsulated chitosan nanoparticles. Methods The effect of different mass ratios of chitosan to tripolyphosphate (TPP) (5:1, 10:1, 15:1, 20:1) on the encapsulation efficiency of cisplatin was investigated. Uptake of cisplatin-encapsulated chitosan by cells was observed using a confocal laser scanning microscope. The cell viability at different cisplatin concentrations was examined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Three iontophoresis methods, namely constant-current chronopotentiometry (CCCP), cyclic chronopotentiometry (CCP), and differential pulse voltammetry (DPV), were used to enhance cisplatin release from cisplatin-encapsulated chitosan nanoparticles. In addition, mouse oral squamous cell carcinoma cell lines were implanted into the mouse oral mucosa to induce oral cancer. The effects of enhanced cisplatin release by CCCP, CCP, and DPV on tumor suppression in mice were evaluated. Tumors and lymph nodes were isolated for hematoxylin-eosin staining and immunohistochemistry staining including Ki-67 and pan CK after sacrifice. Inductively coupled plasma mass spectrometry was conducted to quantify the platinum content within the tumors. Results The results showed that nanoparticles with a mass ratio of 15:1 exhibited the highest cisplatin encapsulation efficiency (approximately 15.6%) and longest continued release (up to 35 days) in phosphate buffered saline with a release rate of 100%. Cellular uptake results suggested that chitosan nanoparticles were delivered to the cytoplasm via endocytosis. The results of the MTT assay revealed that the survival rate of cells decreased as the cisplatin concentration increased. The CCP (1 mA, on:off = 1 s: 1 s) and DPV (0-0.06 V) groups were the most effective in inhibiting tumor growth, and both groups exhibited the lowest percentage of Ki-67 positive and pan CK positive. Conclusion This study is the first to investigate and determine the efficacy of DPV in enhancing in vivo drug release from nanoparticles for the treatment of cancer in animals. The results suggest that the CCP and DPV methods have the potential to be combined with surgery for oral cancer treatment.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Ai-Chia He
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Tzu-Yun Huang
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - De-Hao Lai
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Yi-Ping Wang
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
| | - Wei-Wen Liu
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Wei-Ting Kuo
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Hsin-Han Hou
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Shih-Jung Cheng
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| | - Chen-Yi Lee
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
| | - Wei-Chun Chuang
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
| | - Che-Chen Chang
- Department of Chemistry, National Taiwan University, Taipei, 10617, Taiwan
| | - Bor-Shiunn Lee
- Department of Dentistry, National Taiwan University Hospital, Taipei, 100229, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, 100229, Taiwan
| |
Collapse
|
49
|
Puebla-Osorio N, Fowlkes NW, Barsoumian HB, Xega K, Srivastava G, Kettlun-Leyton C, Nizzero S, Voss T, Riad TS, Wong C, Huang A, Hu Y, Mitchell J, Kim M, Rafiq Z, He K, Sezen D, Hsu E, Masrorpour F, Maleki A, Leuschner C, Cortez MA, Oertle P, Loparic M, Plodinec M, Markman JL, Welsh JW. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy. Front Oncol 2024; 14:1407143. [PMID: 39445067 PMCID: PMC11496962 DOI: 10.3389/fonc.2024.1407143] [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: 03/26/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Effective infiltration of chimeric antigen receptor T (CAR-T) cells into solid tumors is critical for achieving a robust antitumor response and improving therapeutic outcomes. While CAR-T cell therapies have succeeded in hematologic malignancies, their efficacy in solid tumors remains limited due to poor tumor penetration and an immunosuppressive tumor microenvironment. This study aimed to evaluate the potential of low-dose radiotherapy (LDRT) administered before T-cell therapy to enhance the antitumor effect by promoting CAR-T cell infiltration. We hypothesized that combining LDRT with T-cell therapy would improve tumor control and survival compared to either treatment alone. Methods We investigated this hypothesis using two NSG mouse models bearing GSU or CAPAN-2 solid tumors. The mice were treated with engineered CAR-T cells targeting guanyl cyclase-C (GCC) or mesothelin as monotherapy or in combination with LDRT. Additionally, we extended this approach to a C57BL/6 mouse model implanted with MC38-gp100+ cells, followed by adoptive transfer of pmel+ T cells before and after LDRT. Tumor growth and survival outcomes were monitored in all models. Furthermore, we employed atomic force microscopy (AFM) in a small cohort to assess the effects of radiotherapy on tumor stiffness and plasticity, exploring the role of tumor nanomechanics as a potential biomarker for treatment efficacy. Results Our results demonstrated enhanced tumor control and prolonged survival in mice treated with LDRT followed by T-cell therapy across all models. The combination of LDRT with CAR-T or pmel+ T-cell therapy led to superior tumor suppression and survival compared to monotherapy, highlighting the synergistic impact of the combined approach. Additionally, AFM analysis revealed significant changes in tumor stiffness and plasticity in response to LDRT, suggesting that the nanomechanical properties of the tumor may be predictive of therapeutic response. Discussion The findings of this study highlight the transformative potential of incorporating LDRT as a precursor to adoptive T-cell therapy in solid tumors. By promoting CAR-T and pmel+ T-cell infiltration into the tumor microenvironment, LDRT enhanced tumor control and improved survival outcomes, offering a promising strategy to overcome the challenges associated with CAR-T therapy in solid tumors. Additionally, the changes in tumor nanomechanics observed through AFM suggest that tumor stiffness and plasticity could be biomarkers for predicting treatment outcomes. These results support further investigation into the clinical application of this combined approach to improve the efficacy of cell-based therapies in patients with solid tumors.
Collapse
Affiliation(s)
- Nahum Puebla-Osorio
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalie Wall Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B. Barsoumian
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kristina Xega
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | | | - Claudia Kettlun-Leyton
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Tiffany Voss
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Thomas S. Riad
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina Wong
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | - Ailing Huang
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yun Hu
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joylise Mitchell
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingee Kim
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Zahid Rafiq
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kewen He
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Duygu Sezen
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ethan Hsu
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fatemeh Masrorpour
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aurian Maleki
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Carola Leuschner
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | - Janet L. Markman
- Takeda Development Centers Americas, Inc, Lexington, MA, United States
| | - James W. Welsh
- Department of Radiation Oncology—Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
50
|
Labra B, Parag-Sharma K, Powers JJ, Srivastava S, Walker JR, Kirkland TA, Brennan CK, Prescher JA, Amelio AL. Optimized in vivo multispectral bioluminescent imaging of tumor biology using engineered BRET reporters. iScience 2024; 27:110655. [PMID: 39252965 PMCID: PMC11381837 DOI: 10.1016/j.isci.2024.110655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/30/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024] Open
Abstract
The ability to visualize and track multiple biological processes in vivo in real time is highly desirable. Bioluminescence imaging (BLI) has emerged as an attractive modality for non-invasive cell tracking, with various luciferase reporters enabling parallel monitoring of several processes. However, simultaneous multiplexed imaging in vivo is challenging due to suboptimal reporter intensities and the need to image one luciferase at a time. We report a multiplexed BLI approach using a single substrate that leverages bioluminescence resonance energy transfer (BRET)-based reporters with distinct spectral profiles for triple-color BLI. These luciferase-fluorophore fusion reporters address light transmission challenges and use optimized coelenterazine substrates. Comparing BRET reporters across two substrate analogs identified a green-yellow-orange combination that allows simultaneous imaging of three distinct cell populations in vitro and in vivo. These tools provide a template for imaging other biological processes in vivo during a single BLI session using a single reporter substrate.
Collapse
Affiliation(s)
- Bryan Labra
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kshitij Parag-Sharma
- Graduate Curriculum in Cell Biology & Physiology, Biological & Biomedical Sciences Program, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John J. Powers
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sonal Srivastava
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | | | - Thomas A. Kirkland
- Promega Biosciences, LLC, San Luis Obispo, CA, USA
- Promega Corporation, Madison, WI, USA
| | | | - Jennifer A. Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Antonio L. Amelio
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Cancer Cell Biology Program, Lineberger Comprehensive Cancer Center, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|