1
|
Lee HD, Yuan LY. Nano-revolution in hepatocellular carcinoma: A multidisciplinary odyssey - Are we there yet? World J Hepatol 2024; 16:684-687. [PMID: 38818296 PMCID: PMC11135275 DOI: 10.4254/wjh.v16.i5.684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024] Open
Abstract
In this editorial we comment on the review by Zhou et al reviewing the landscape of nanomedicine in the treatment of hepatocellular carcinoma (HCC). We focus on the immense potential of nanotechnology, particularly ligand-receptor mediated nanotherapy, in revolutionizing the treatment landscape of HCC. Despite advancements in multidisciplinary treatment, HCC remains a significant global health challenge. Ligand-mediated nanotherapy offers the opportunity for precise drug delivery to tumor sites, targeting specific receptors overexpressed in HCC cells, thereby enhancing efficacy and minimizing side effects. Overcoming drug resistance and aggressive tumor biology is facilitated by nanomedicine, bypassing traditional hurdles encountered in chemotherapy. Examples include targeting glypican-3, asialoglycoprotein, transferrin receptor or folic acid receptors, capitalizing on their over-expression in tumor cells. The ability for multi-receptor targeting through dual-ligand nanoparticle modification holds the prospect of further enhancement in specificity and efficacy of directed therapy. However, challenges including immune responses, reproducibility in nanoparticle synthesis, and production scalability remain. Future directions involve refining targeting strategies, improving drug release mechanisms, and streamlining production processes to enable personalized and multifunctional nanotherapies. Overall, the integration of nanotherapy in HCC treatment holds immense promise, but continued partnership and effort are needed in offering hope for more effective, precise, and accessible clinical care in the management of HCC.
Collapse
Affiliation(s)
- Howard D Lee
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, United States..
| | - Li-Yun Yuan
- Department of Medicine, Division of Gastrointestinal and Liver Diseases, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, United States
| |
Collapse
|
2
|
Ren X, Su D, Shi D, Xiang X. The improving strategies and applications of nanotechnology-based drugs in hepatocellular carcinoma treatment. Front Bioeng Biotechnol 2023; 11:1272850. [PMID: 37811369 PMCID: PMC10557528 DOI: 10.3389/fbioe.2023.1272850] [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: 08/04/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of tumor-related death worldwide. Conventional treatments for HCC include drugs, radiation, and surgery. Despite the unremitting efforts of researchers, the curative effect of HCC has been greatly improved, but because HCC is often found in the middle and late stages, the curative effect is still not satisfactory, and the 5-year survival rate is still low. Nanomedicine is a potential subject, which has been applied to the treatment of HCC and has achieved promising results. Here, we summarized the factors affecting the efficacy of drugs in HCC treatment and the strategies for improving the efficacy of nanotechnology-based drugs in HCC, reviewed the recent applications' progress on nanotechnology-based drugs in HCC treatment, and discussed the future perspectives and challenges of nanotechnology-based drugs in HCC treatment.
Collapse
Affiliation(s)
- Xiangyang Ren
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danyang Su
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Doudou Shi
- The Ninth Hospital of Xi’an, Xi’an, Shaanxi, China
| | - Xiaohong Xiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Hu Y, Pan T, Cai X, He QS, Zheng YB, Huang MS, Jiang ZB, Chen JW, Wu C. Addition of transarterial chemoembolization improves outcome of tyrosine kinase and immune checkpoint inhibitors regime in patients with unresectable hepatocellular carcinoma. J Gastrointest Oncol 2023; 14:1837-1848. [PMID: 37720446 PMCID: PMC10502547 DOI: 10.21037/jgo-23-486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/01/2023] [Indexed: 09/19/2023] Open
Abstract
Background Transarterial chemoembolization (TACE) is the standard treatment for hepatocellular carcinoma (HCC); the value of its combination with systemic therapy is worthy of further exploration. This study aimed to investigate the efficacy and safety of TACE combined with tyrosine kinase inhibitor (TKI) and immune checkpoint inhibitor (ICI) in the treatment of unresectable HCC. Methods In this retrospective observational, single-center study, 147 patients with unresectable HCC were divided into a TACE group (n=98) and a non-TACE group (n=49) based on whether TACE was performed during TKI plus ICI therapy. The survival outcomes and adverse events (AEs) of the two groups were compared. Results Data from patients with unresectable HCC who received TKI plus ICI treatment between July 2017 and April 2020 were collected. The median intrahepatic tumor size was 8.7 cm [interquartile range (IQR), 5.9-12.4 cm]. At data cut-off, overall survival (OS) of the TACE group was significantly longer than that of the non-TACE group (19.5 and 10.8 months, respectively, P=0.005). In the high-risk cohort (with main or contralateral portal vein tumor thrombi and/or bile duct invasion and/or a tumor burden >50% of liver), the OS of the TACE group was still longer than that of the non-TACE group (14.9 and 8.7 months, respectively, P=0.031). Major AEs were tolerated in both groups, and there was no significant difference in their incidence (34.7% and 30.6%, respectively, P=0.621). Conclusions TACE treatment combined with TKI plus ICI regime resulted in longer OS than treatment with TKI plus ICI alone for patients with unresectable HCC.
Collapse
Affiliation(s)
- Yue Hu
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tao Pan
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Cai
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Quan-Sheng He
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Bao Zheng
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming-Sheng Huang
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zai-Bo Jiang
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun-Wei Chen
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun Wu
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
You H, Yuan D, Li Q, Zhang N, Kong D, Yu T, Liu X, Liu X, Zhou R, Kong F, Zheng K, Tang R. Hepatitis B virus X protein increases LASP1 SUMOylation to stabilize HER2 and facilitate hepatocarcinogenesis. Int J Biol Macromol 2023; 226:996-1009. [PMID: 36473530 DOI: 10.1016/j.ijbiomac.2022.11.312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 07/05/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The hepatitis B virus (HBV) X protein (HBX), a viral macromolecule, plays a vital role in the development of HBV-related hepatocellular carcinoma (HCC). Increased expression of HER2 is linked to HBV infection, and HBX is responsible for HER2 upregulation in HCC. Nevertheless, the underlying molecular mechanisms are not yet fully understood. In the study, we discovered that HBX promoted HER2 expression to facilitate the sensitization of the insulin signaling pathway and enhance the growth and migration of HCC cells. Mechanistically, the viral protein enhanced the stability of HER2 by preventing its ubiquitination-mediated proteasomal degradation through LASP1, which could bind to HER2. Furthermore, increased SUMOylation of LASP1 contributed to the upregulation of HER2 and the interaction of LASP1 with HER2. In addition, RANBP2 and RANGAP1 were found to interact with LASP1 and promote SUMOylation of LASP1 to upregulate HER2 expression in HBX-associated hepatoma cells. In summary, our work provides a novel insight into hepatocarcinogenesis mediated by HBX and estimates the detailed mechanisms related to the increase in HER2 regulated by the viral protein, which might help provide a theoretical basis for identifying novel targets for HBV-positive HCC treatment.
Collapse
Affiliation(s)
- Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongchen Yuan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ning Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Delong Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tong Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rui Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China; National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China; National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Xiao J, Liu Z, Wang J, Zhang S, Zhang Y. Identification of cuprotosis-mediated subtypes, the development of a prognosis model, and influence immune microenvironment in hepatocellular carcinoma. Front Oncol 2022; 12:941211. [PMID: 36110946 PMCID: PMC9468823 DOI: 10.3389/fonc.2022.941211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/05/2022] [Indexed: 12/25/2022] Open
Abstract
Purpose Cuprotosis is a newly discovered form of non-apoptotic regulated cell death and is characterized by copper-dependent and associated with mitochondrial respiration. However, the prognostic significance and function of cuprotosis-related genes (CRGs) in hepatocellular carcinoma (HCC) are unknown. This study aims to develop cuprotosis-mediated patterns-related gene (CMPRG) prediction models for the prognosis of patients with HCC, exploring the functional underlying the CRGs on the influence of tumor microenvironment (TME) features. Experimental design This study obtained transcriptome profiling and the corresponding clinical information from the TCGA and GEO databases. Besides, the Cox regression model with LASSO was implemented to build a multi-gene signature, which was then validated in an internal validation set and two external validation sets through Kaplan-Meier, DCA, and ROC analyses. Results According to the LASSO analysis, we screened out a cuprotosis-mediated pattern 5-gene combination (including PBK; MMP1; GNAZ; GPC1 and AKR1D1). A nomogram was constructed for the presentation of the final model. The ROC curve assessed the model’s predictive ability, which resulted in an area under the curve (AUC) values ranging from 0.604 to 0.787 underwent internal and two external validation sets. Meanwhile, the risk score divided the patients into two groups of high and low risk, and the survival rate of high-risk patients was significantly lower than that of low-risk patients (P<0.01). The risk score could be an independent prognostic factor in the multifactorial Cox regression analysis (P<0.01). Functional analysis revealed that immune status, mutational loads, and drug sensitivity differed between the two risk groups. Conclusions In summary, we identified three cuprotosis-mediated patterns in HCC. And CMPRGs are a promising candidate biomarker for HCC early detection, owing to their strong performance in predicting HCC prognosis and therapy. Quantifying cuprotosis-mediated patterns in individual samples may help improve the understanding of multiomic characteristics and guide the development of targeted therapy for HCC.
Collapse
Affiliation(s)
- Jingjing Xiao
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhenhua Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jinlong Wang
- Department of Critical Care Medicine, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Shuaimin Zhang
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yi Zhang
- Department of Hepatobiliary Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
6
|
Zhang Y, Pang S, Sun B, Zhang M, Jiao X, Lai L, Qian Y, Yang N, Yang W. ELOVLs Predict Distinct Prognosis Value and Immunotherapy Efficacy In Patients With Hepatocellular Carcinoma. Front Oncol 2022; 12:884066. [PMID: 35912257 PMCID: PMC9334671 DOI: 10.3389/fonc.2022.884066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a primary malignancy of the liver with high prevalence worldwide and poor prognosis. It has been verified that elongation of very-long-chain fatty acids gene family (ELOVLs), a group of genes that responsible for elongation of saturated and polyunsaturated fatty acids, participate in the pathogenesis and development of multiplex disease including cancers. However, the functions and prognosis of ELOVLs in HCC are still indistinguishable. Methods First, we searched the mRNA expression and survival data of ELOVLs in patients with HCC via the data of The Cancer Genome Atlas (TCGA). The prognosis value of ELOVLs on HCC was assessed by Kaplan–Meier plotter and Cox regression analysis. reverse transcription quantitative- polymerase chain reaction (RT-qPCR), Western blot (WB), and immunohistochemistry were applied to assess the specific mRNA and protein expression of ELOVLs in HCC clinical specimens of our cohort. Then, the functional enrichment of ELOVL1 especially the pathways relating to the immune was conducted utilizing the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) analysis. Additionally, TIMER, CIBERSOR, and tumor immune dysfunction and exclusion (TIDE) were employed to evaluate the relationship between ELOVL1 and immune responses. Last, the correlation of ELOVL1 with genome heterogeneity [microsatellite instability (MSI), tumor mutational burden (TMB), mutant-allele tumor heterogeneity (MATH), homologous recombination deficiency (HRD), purity, ploidy, loss of heterozygosity (LOH), and neoantigens] and mutational landscape were also evaluated basing on the date in TCGA. Results Significant expression alteration was observed in ELOVLs family at the pan-cancer level. In liver cancer, ELOVL1 and ELOVL3 were strongly associated with poor prognosis of HCC by survival analysis and differential expression analysis. Immunohistochemistry microarray, WB, and RT-qPCR confirmed that ELOVL1 but not ELOVL3 played an important role in HCC. Mechanistically, functional network analysis revealed that ELOVL1 might be involved in the immune response. ELOVL1 could affect immune cell infiltration and immune checkpoint markers such as PD-1 and CTLA4 in HCC. Meanwhile, high expression of ELOVL1 would be insensitive to immunotherapy. Correlation analysis of immunotherapy markers showed that ELOVL1 has been associated with MSI, TMB, and oncogene mutations such as TP53. Conclusion ELOVLs play distinct prognostic value in HCC. ELOVL1 could predict the poor prognosis and might be a potential indicator of immunotherapy efficacy in HCC patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shujie Pang
- Department V of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Minbo Zhang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ning Yang
- Department V of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Ning Yang, ; Wenzhuo Yang,
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ning Yang, ; Wenzhuo Yang,
| |
Collapse
|
7
|
Minami Y, Takaki H, Yamakado K, Kudo M. How Compatible Are Immune Checkpoint Inhibitors and Thermal Ablation for Liver Metastases? Cancers (Basel) 2022; 14:cancers14092206. [PMID: 35565338 PMCID: PMC9103121 DOI: 10.3390/cancers14092206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although immune checkpoint inhibitors (ICIs) have achieved great progression in cancer treatment, the efficacy of ICI monotherapy is still limited. Meanwhile, the negative efficacy of thermal ablation for liver metastases is the high rate of local tumor progression. Since thermal ablation-induced inflammation and increases in tumor antigens have been suggested to promote the cancer-immunity cycle, thermal ablation and ICI can boost the immune response against cancer cells as one of the positive synergy effects. The findings of preclinical and clinical research have provided supportive evidence for the combination of ICIs with thermal ablation reversing T-cell exhaustion and demonstrating synergy. However, the clinical feasibility of immune response activation by combination therapy with ICI monotherapy and thermal ablation appears to be limited, it may be not very common phenomena. Abstract Cancer immunotherapy, which reactivates the weakened immune cells of cancer patients, has achieved great success, and several immune checkpoint inhibitors (ICIs) are now available in clinical practice. Despite promising clinical outcomes, favorable responses are only observed in a fraction of patients, and resistance mechanisms, including the absence of tumor antigens, have been reported. Thermal ablation involves the induction of irreversible damage to cancer cells by localized heat and may result in the release of tumor antigens. The combination of immunotherapy and thermal ablation is an emerging therapeutic option with enhanced efficacy. Since thermal ablation-induced inflammation and increases in tumor antigens have been suggested to promote the cancer-immunity cycle, the combination of immuno-oncology (IO) therapy and thermal ablation may be mutually beneficial. In preclinical and clinical studies, the combination of ICI and thermal ablation significantly inhibited tumor growth, and synergistic antitumor effects appeared to prolong the survival of patients with secondary liver cancer. However, evidence for the efficacy of ICI monotherapy combined with thermal ablation is currently insufficient. Therefore, the clinical feasibility of immune response activation by ICI monotherapy combined with thermal ablation may be limited, and thermal ablation may be more compatible with dual ICIs (the IO–IO combination) to induce strong immune responses.
Collapse
Affiliation(s)
- Yasunori Minami
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi Osaka-Sayama, Osaka 589-8511, Japan;
- Correspondence: ; Tel.: +81-72-366-0221 (ext. 3525); Fax: +81-72-367-2880
| | - Haruyuki Takaki
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa Nishinomiya, Nishinomiya 663-8501, Japan; (H.T.); (K.Y.)
| | - Koichiro Yamakado
- Department of Radiology, Hyogo College of Medicine, 1-1 Mukogawa Nishinomiya, Nishinomiya 663-8501, Japan; (H.T.); (K.Y.)
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-Higashi Osaka-Sayama, Osaka 589-8511, Japan;
| |
Collapse
|
8
|
Vène E, Jarnouen K, Ribault C, Vlach M, Verres Y, Bourgeois M, Lepareur N, Cammas-Marion S, Loyer P. Circumsporozoite Protein of Plasmodium berghei- and George Baker Virus A-Derived Peptides Trigger Efficient Cell Internalization of Bioconjugates and Functionalized Poly(ethylene glycol)-b-poly(benzyl malate)-Based Nanoparticles in Human Hepatoma Cells. Pharmaceutics 2022; 14:pharmaceutics14040804. [PMID: 35456637 PMCID: PMC9028075 DOI: 10.3390/pharmaceutics14040804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
In order to identify the peptides, selected from the literature, that exhibit the strongest tropism towards human hepatoma cells, cell uptake assays were performed using biotinylated synthetic peptides bound to fluorescent streptavidin or engrafted onto nanoparticles (NPs), prepared from biotin-poly(ethylene glycol)-block-poly(benzyl malate) (Biot-PEG-b-PMLABe) via streptavidin bridging. Two peptides, derived from the circumsporozoite protein of Plasmodium berghei- (CPB) and George Baker (GB) Virus A (GBVA10-9), strongly enhanced the endocytosis of both streptavidin conjugates and NPs in hepatoma cells, compared to primary human hepatocytes and non-hepatic cells. Unexpectedly, the uptake of CPB- and GBVA10-9 functionalized PEG-b-PMLABe-based NPs by hepatoma cells involved, at least in part, the peptide binding to apolipoproteins, which would promote NP’s interactions with cell membrane receptors of HDL particles. In addition, CPB and GBVA10-9 peptide–streptavidin conjugates favored the uptake by hepatoma cells over that of the human macrophages, known to strongly internalize nanoparticles by phagocytosis. These two peptides are promising candidate ligands for targeting hepatocellular carcinomas.
Collapse
Affiliation(s)
- Elise Vène
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU Rennes, F-35033 Rennes, France
| | - Kathleen Jarnouen
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Catherine Ribault
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- INRAE, Institut AGRO, PEGASE UMR 1348, F-35590 Saint-Gilles, France
| | - Yann Verres
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Mickaël Bourgeois
- CRCINA, Inserm, CNRS, Université de Nantes, F-44000 Nantes, France;
- ARRONAX Cyclotron, F-44817 Saint Herblain, France
| | - Nicolas Lepareur
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Comprehensive Cancer Center Eugène Marquis, F-35000 Rennes, France
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| | - Sandrine Cammas-Marion
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Institut des Sciences Chimiques de Rennes (ISCR), Ecole Nationale Supérieure de Chimie de Rennes, CNRS UMR 6226, University of Rennes, F-35042 Rennes, France
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Correspondence: (N.L.); (S.C.-M.); (P.L.)
| |
Collapse
|
9
|
Chinnadurai R, Porter AP, Patel M, Lipat AJ, Forsberg MH, Rajan D, Hematti P, Capitini CM, Bruker C. Hepatocellular Carcinoma Cells Are Protected From Immunolysis by Mesenchymal Stromal Cells Through Indoleamine 2,3 Dioxygenase. Front Cell Dev Biol 2021; 9:715905. [PMID: 34869307 PMCID: PMC8633446 DOI: 10.3389/fcell.2021.715905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
B7 family proteins serve as checkpoint molecules that protect tumors from T cell mediated lysis. Tryptophan degrading enzymes indoleamine 2,3 dioxygenase (IDO) and tryptophan 2,3 dioxygenase (TDO) also induce T cell immune tolerance. However, little is known about the relative contribution of B7 molecules, tryptophan degrading enzymes, as well as the impact of tumor and stromal cell interactions to the development of immunosuppressive tumor microenvironment. To investigate such interactions, we used a tripartite model of human hepatocellular carcinoma cell line (HepG2) and mesenchymal stromal cells (MSCs) co-cultured with peripheral blood mononuclear cells (PBMCs). Co-culture of HepG2 cells and activated PBMCs demonstrate that HepG2 cells undergo PBMC mediated cytolysis, despite constitutive expression of B7-H3 and upregulation of PD-L1 by IFNγ. Knockdown of B7-H3, PD-L1 or IDO does not modulate PBMC mediated lysis of HepG2 cells. However, TNFα preactivation enhances lysis of HepG2 cells, and blocking of TNFα production from PBMCs protects HepG2 cells. On the other hand, MSCs protect HepG2 cells from PBMC mediated lysis, even in the presence of TNFα. Further investigation showed that MSC mediated protection is associated with the unique secretome profile of upregulated and downregulated cytokines and chemokines. IFNγ activated MSCs are superior to TNFα activated or control MSCs in protecting HepG2 cells. Blockade of IFNγ driven IDO activity completely abolishes the ability of MSCs to protect HepG2 cells from cytolysis by PBMCs. These results suggest that inhibition of IFNγ activation of IDO induction in stromal cells, combined with usage of TNFα, could be a novel immunotherapeutic strategy to induce regression of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Amanda Paige Porter
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Mihir Patel
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Ariel Joy Lipat
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Mathews H Forsberg
- Department of Pediatrics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Devi Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Peiman Hematti
- Department of Medicine, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Christian M Capitini
- Department of Pediatrics, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Charles Bruker
- Department of Pathology, Memorial Health University Medical Center, Savannah, GA, United States
| |
Collapse
|
10
|
Heinrich S, Craig AJ, Ma L, Heinrich B, Greten TF, Wang XW. Understanding tumour cell heterogeneity and its implication for immunotherapy in liver cancer using single-cell analysis. J Hepatol 2021; 74:700-715. [PMID: 33271159 DOI: 10.1016/j.jhep.2020.11.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Over the last decade, precision medicine and immunotherapeutic approaches have become increasingly popular in oncology. Early clinical trials reported promising results, but response rates in phase III clinical trials have been suboptimal. Knowledge gained from subsequent translational studies indicates the importance of targeting the tumour microenvironment to overcome resistance to immunotherapy. In this era of precision medicine, it is crucial to consider inter- as well as intratumoural heterogeneity. Single-cell analysis is a cutting-edge technology that enables us to better define the tumour cell community and to identify potential targets for immunotherapy or combination treatments. This review focuses on single-cell analysis in the context of immunotherapy in liver cancer, including the rationale behind studying hepatocellular carcinoma biology at a single-cell level. Single-cell technologies have the potential to revolutionise our understanding of resistance mechanisms and to guide drug discovery efforts, leading to further advances in personalised medicine.
Collapse
Affiliation(s)
- Sophia Heinrich
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Amanda J Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Bernd Heinrich
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Tim F Greten
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA; Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Xin W Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, USA; Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, USA.
| |
Collapse
|
11
|
Silva L, Egea J, Villanueva L, Ruiz M, Llopiz D, Repáraz D, Aparicio B, Lasarte-Cia A, Lasarte JJ, Ruiz de Galarreta M, Lujambio A, Sangro B, Sarobe P. Cold-Inducible RNA Binding Protein as a Vaccination Platform to Enhance Immunotherapeutic Responses Against Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12113397. [PMID: 33207844 PMCID: PMC7696968 DOI: 10.3390/cancers12113397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Therapies based on immune checkpoint inhibitors (ICPI) have yielded promising albeit limited results in patients with hepatocellular carcinoma (HCC). Vaccines have been proposed as combination partners to enhance response rates to ICPI. Thus, we analyzed the combined effect of a vaccine based on the TLR4 ligand cold-inducible RNA binding protein (CIRP) plus ICPI. Mice were immunized with vaccines containing ovalbumin linked to CIRP (OVA-CIRP), with or without ICPI, and antigen-specific responses and therapeutic efficacy were tested in subcutaneous and orthotopic mouse models of liver cancer. OVA-CIRP elicited polyepitopic T-cell responses, which were further enhanced when combined with ICPI (anti-PD-1 and anti-CTLA-4). Combination of OVA-CIRP with ICPI enhanced ICPI-induced therapeutic responses when tested in subcutaneous and intrahepatic B16-OVA tumors, as well as in the orthotopic PM299L HCC model. This effect was associated with higher OVA-specific T-cell responses in the periphery, although many tumor-infiltrating lymphocytes still displayed an exhausted phenotype. Finally, a new vaccine containing human glypican-3 linked to CIRP (GPC3-CIRP) induced clear responses in humanized HLA-A2.01 transgenic mice, which increased upon combination with ICPI. Therefore, CIRP-based vaccines may generate anti-tumor immunity to enhance ICPI efficacy in HCC, although blockade of additional checkpoint molecules and immunosuppressive targets should be also considered.
Collapse
Affiliation(s)
- Leyre Silva
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - Josune Egea
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - Lorea Villanueva
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
| | - Marta Ruiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - Diana Llopiz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - David Repáraz
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - Belén Aparicio
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
| | - Aritz Lasarte-Cia
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
| | - Juan José Lasarte
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
| | - Marina Ruiz de Galarreta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.R.d.G.); (A.L.)
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.R.d.G.); (A.L.)
| | - Bruno Sangro
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
- Liver Unit, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Pablo Sarobe
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, 31008 Pamplona, Spain; (L.S.); (J.E.); (L.V.); (M.R.); (D.L.); (D.R.); (B.A.); (A.L.-C.); (J.J.L.)
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas CIBEREHD, 31008 Pamplona, Spain
- Correspondence: ; Tel.: +34-948-194700 (ext. 813009)
| |
Collapse
|