1
|
Zhang S, Li M, Zeng J, Zhou S, Yue F, Chen Z, Ma L, Wang Y, Wang F, Luo J. Somatostatin receptor-targeted polymeric nanoplatform for efficient CRISPR/Cas9 gene editing to enhance synergistic hepatocellular carcinoma therapy. J Nanobiotechnology 2025; 23:127. [PMID: 39979929 DOI: 10.1186/s12951-025-03214-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION The CRISPR/Cas9 system-based gene therapy can fundamentally address the issues of cancer occurrence, development, progression, and metastasis. However, the lack of targeting and effectiveness hinders gene therapy from entering clinical application. Herein, a somatostatin receptor-targeted polymeric nanoplatform is developed for the delivery of a PD-L1-targeted CRISPR/Cas9 system and synergistic treatment of hepatocellular carcinoma. This nanoplatform can effectively incorporate the CRISPR/Cas9 system and the chemotherapeutic drug paclitaxel to simultaneously address the biological safety and packaging capacity issues of viral vectors. After the octreotide-modified polymer (LNA-PEG-OCT) guided the nanoparticle into hepatoma carcinoma cells, the nanoparticle protected the CRISPR/Cas9 ribonucleoprotein complex (RNP) and achieved lysosomal escape. Then, the RNP reached the target gene (PD-L1) under the guidance of the single guide RNA (sgRNA) in the RNP. The PD-L1 gene editing efficiency reached up to 55.8% for HepG2 cells in vitro and 46.0% for tumor tissues in vivo, leading to effective suppression of PD-L1 protein expression. Substantial inhibition of hepatocellular carcinoma cell proliferation and further 79.45% growth repression against subcutaneous xenograft tumors were achieved. Overall, this somatostatin receptor-targeted polymeric nanoplatform system not only provides a promising nanocarrier for CRISPR/Cas9 system delivery, but also expands the potential of combining gene editing and chemotherapy.
Collapse
Affiliation(s)
- Suqin Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Meng Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Jingyi Zeng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Songli Zhou
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Feifan Yue
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Zhaoyi Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Yang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Fei Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Jingwen Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Province Key Laboratory of industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
2
|
Hu Q, Chen S, Deng R, Deng H, Peng M, Wang X, Deng S, Wang J, Xu B, Xu Y, Zhu H, Zheng J, Xia M, Zuo C. Exosomal PDL1 Suppresses the Anticancer Activity of CD8 + T Cells in Hepatocellular Carcinoma. Anal Cell Pathol (Amst) 2024; 2024:1608582. [PMID: 39421264 PMCID: PMC11483647 DOI: 10.1155/2024/1608582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/23/2024] [Accepted: 08/17/2024] [Indexed: 10/19/2024] Open
Abstract
Tumor microenvironment (TME) is essential for the development and progression of hepatocellular carcinoma (HCC). Exosomes participate in constructing TME by passing biological information, but the regulatory effect of PDL1 in exosomes on anticancer activity of CD8+ T cells in HCC still needs to be further explored. In this study, high level of PDL1 was found in plasma exosomes of HCC patients, which turned out to be significantly associated with the increased number of tumor nodules, the upregulated level of serum AFP, the raised tendency of TNM stage, and the poor prognosis of HCC. The expression of CD8 may be inhibited in HCC that is characterized with high level of PDL1, and the protein level of exosomal PDL1 was determined by intracellular PDL1 abundance. High level of exosomal PDL1 inhibited the proliferation and activation of CD8+ T cells, but exhibited limited effect on the proliferation of hepatic cancer cells. Moreover, the growth of tumors formed by hepatic cancer cells Hepa1-6 in C57L mice was significantly promoted by the exosomal PDL1, which might be caused by the inhibitory effect of exosomal PDL1 on CD8+ T cells. Thus, exosomal PDL1 promotes the development and progression of HCC through inhibiting the anticancer activity of CD8+ T cells. This study provides sights for understanding the oncogenic role of PDL1 and a reasonable explanation for the low efficacy of anti-PD1/PDL1 immunotherapies in HCC.
Collapse
Affiliation(s)
- Qi Hu
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Shuai Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Rilin Deng
- Hunan Normal University School of Medicine, Changsha 410013, Hunan, China
| | - Hongyu Deng
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Joint Research Center of Liver Cancer, Laboratory of Digestive Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Provincial, Central South University, Changsha 410013, Hunan, China
| | - Mingjing Peng
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Joint Research Center of Liver Cancer, Laboratory of Digestive Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Provincial, Central South University, Changsha 410013, Hunan, China
| | - Xiaohong Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The Second Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, Hainan, China
| | - Shun Deng
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Joint Research Center of Liver Cancer, Laboratory of Digestive Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Provincial, Central South University, Changsha 410013, Hunan, China
| | - Jinfeng Wang
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Joint Research Center of Liver Cancer, Laboratory of Digestive Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Provincial, Central South University, Changsha 410013, Hunan, China
| | - Biaoming Xu
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yan Xu
- Hunan Normal University School of Medicine, Changsha 410013, Hunan, China
| | - Haizhen Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Pathogen Biology, School of Basic Medicine and Life Science, Department of Clinical Laboratory of the Second Affiliated Hospital, The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The Second Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, Hainan, China
| | - Jinhai Zheng
- School of Biomedical Sciences, Hunan University, Changsha 410082, Hunan, China
| | - Man Xia
- Department of Gynecological Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School Medicine, Central South University, Changsha 410013, Hunan, China
| | - Chaohui Zuo
- Graduate Collaborative Training Base of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Joint Research Center of Liver Cancer, Laboratory of Digestive Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Clinical Research Center for Tumor of Pancreaticobiliary Duodenal Junction in Hunan Provincial, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
3
|
Cordani M, Strippoli R, Trionfetti F, Barzegar Behrooz A, Rumio C, Velasco G, Ghavami S, Marcucci F. Immune checkpoints between epithelial-mesenchymal transition and autophagy: A conflicting triangle. Cancer Lett 2024; 585:216661. [PMID: 38309613 DOI: 10.1016/j.canlet.2024.216661] [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: 11/21/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 02/05/2024]
Abstract
Inhibitory immune checkpoint (ICP) molecules are pivotal in inhibiting innate and acquired antitumor immune responses, a mechanism frequently exploited by cancer cells to evade host immunity. These evasion strategies contribute to the complexity of cancer progression and therapeutic resistance. For this reason, ICP molecules have become targets for antitumor drugs, particularly monoclonal antibodies, collectively referred to as immune checkpoint inhibitors (ICI), that counteract such cancer-associated immune suppression and restore antitumor immune responses. Over the last decade, however, it has become clear that tumor cell-associated ICPs can also induce tumor cell-intrinsic effects, in particular epithelial-mesenchymal transition (EMT) and macroautophagy (hereafter autophagy). Both of these processes have profound implications for cancer metastasis and drug responsiveness. This article reviews the positive or negative cross-talk that tumor cell-associated ICPs undergo with autophagy and EMT. We discuss that tumor cell-associated ICPs are upregulated in response to the same stimuli that induce EMT. Moreover, ICPs themselves, when overexpressed, become an EMT-inducing stimulus. As regards the cross-talk with autophagy, ICPs have been shown to either stimulate or inhibit autophagy, while autophagy itself can either up- or downregulate the expression of ICPs. This dynamic equilibrium also extends to the autophagy-apoptosis axis, further emphasizing the complexities of cellular responses. Eventually, we delve into the intricate balance between autophagy and apoptosis, elucidating its role in the broader interplay of cellular dynamics influenced by ICPs. In the final part of this article, we speculate about the driving forces underlying the contradictory outcomes of the reciprocal, inhibitory, or stimulatory effects between ICPs, EMT, and autophagy. A conclusive identification of these driving forces may allow to achieve improved antitumor effects when using combinations of ICIs and compounds acting on EMT and/or autophagy. Prospectively, this may translate into increased and/or broadened therapeutic efficacy compared to what is currently achieved with ICI-based clinical protocols.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy.
| |
Collapse
|
4
|
Chen Z, Yue Z, Yang K, Shen C, Cheng Z, Zhou X, Li S. Four Ounces Can Move a Thousand Pounds: The Enormous Value of Nanomaterials in Tumor Immunotherapy. Adv Healthc Mater 2023; 12:e2300882. [PMID: 37539730 DOI: 10.1002/adhm.202300882] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 07/17/2023] [Indexed: 08/05/2023]
Abstract
The application of nanomaterials in healthcare has emerged as a promising strategy due to their unique structural diversity, surface properties, and compositional diversity. In particular, nanomaterials have found a significant role in improving drug delivery and inhibiting the growth and metastasis of tumor cells. Moreover, recent studies have highlighted their potential in modulating the tumor microenvironment (TME) and enhancing the activity of immune cells to improve tumor therapy efficacy. Various types of nanomaterials are currently utilized as drug carriers, immunosuppressants, immune activators, immunoassay reagents, and more for tumor immunotherapy. Necessarily, nanomaterials used for tumor immunotherapy can be grouped into two categories: organic and inorganic nanomaterials. Though both have shown the ability to achieve the purpose of tumor immunotherapy, their composition and structural properties result in differences in their mechanisms and modes of action. Organic nanomaterials can be further divided into organic polymers, cell membranes, nanoemulsion-modified, and hydrogel forms. At the same time, inorganic nanomaterials can be broadly classified as nonmetallic and metallic nanomaterials. The current work aims to explore the mechanisms of action of these different types of nanomaterials and their prospects for promoting tumor immunotherapy.
Collapse
Affiliation(s)
- Ziyin Chen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Ziqi Yue
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Kaiqi Yang
- Clinical Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Congrong Shen
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Zhe Cheng
- Department of Forensic Medicine, Harbin Medical University, 150001, Harbin, P. R. China
| | - Xiaofeng Zhou
- Department of Urology, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 110042, Shenyang, P. R. China
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, 110042, China
| |
Collapse
|
5
|
Xu W, Chen Y, Zhang Z, Jiang Y, Wang Z. Exosomal PIK3CB promotes PD-L1 expression and malignant transformation in esophageal squamous cell carcinoma. Med Oncol 2023; 40:221. [PMID: 37402056 DOI: 10.1007/s12032-023-02093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC), which accounts for 90% of esophageal carcinomas, seriously endangers human health. Worse still, the 5-year overall survival of ESCC is approximately 20%. Elucidation of the potential mechanism and exploration of promising drugs for ESCC are urgently needed. In this study, a high level of exosomal PIK3CB protein was found in the plasma of ESCC patients, which might indicate a poor prognosis. Moreover, a significant Pearson's correlation was observed at the protein level between exosomal PIK3CB and exosomal PD-L1. Further study revealed that cancer cell-intrinsic and exosome-derived PIK3CB promoted the transcriptional activity of the PD-L1 promoter in ESCC cells. Moreover, treatment with exosomes with lower levels of exosomal PIK3CB decreased the protein level of the mesenchymal marker β-catenin while increasing that of the epithelial marker claudin-1, indicating the potential regulation of epithelial-mesenchymal transition. Consequently, the migratory ability and cancer stemness of ESCC cells and the growth of tumors formed by ESCC cells were decreased with the downregulation of exosomal PIK3CB. Therefore, exosomal PIK3CB plays an oncogenic role by promoting PD-L1 expression and malignant transformation in ESCC. This study may provide new insight into the inherent biological aggressiveness and the poor response to currently available therapies of ESCC. Exosomal PIK3CB may be a promising target for the diagnosis and therapy of ESCC in the future.
Collapse
Affiliation(s)
- Wei Xu
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing, China
| | - Yang Chen
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing, China
| | - Zhi Zhang
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing, China
| | - Yuequan Jiang
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing, China.
| | - Zhiqiang Wang
- Department of Thoracic Surgery, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|