1
|
Giurini EF, Ralph O, Pappas SG, Gupta KH. Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy. Mol Cancer Ther 2025; 24:18-32. [PMID: 39311547 DOI: 10.1158/1535-7163.mct-24-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 01/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over 5 years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on the recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.
Collapse
Affiliation(s)
- Eileena F Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Oliver Ralph
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Sam G Pappas
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Kajal H Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
2
|
Hu Q, Feng J, Qi L, Jin Y. Stromal Reprogramming Optimizes KRAS-Specific Chemotherapy Inducing Antitumor Immunity in Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:61583-61598. [PMID: 39480275 DOI: 10.1021/acsami.4c10404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a clinically challenging cancer and is often characterized with rich stroma and mutated KRAS, which determines the tumor microenvironment (TME) and therapy response. Turning immunologically "cold" PDAC into "hot" is an unmet need to improve the therapeutic outcome. Herein, we propose a programmable strategy by sequential delivery of pirfenidone (PFD) and nanoengineered KRAS specific inhibitor (AMG510) and gemcitabine (GEM) liposomes. PFD could achieve precise reduction of the extracellular matrix (ECM) by reprogramming pancreatic stellate cells (PSCs). Subsequently, targeting the KRAS-directed oncogenic signaling pathway effectively inhibited tumor proliferation and migration, which sensitized a chemotherapeutic drug and promoted immunogenic cell death (ICD). In preclinical mouse models of PDAC, PFD mediated stromal modulation enhanced the deep penetration of nanoparticles and improved their subsequent performance in tumor growth inhibition. The molecular mechanisms elucidated that the stroma intervention and KRAS signal pathway regulation reshaped the immunosuppression of PDAC and optimized cytotoxic T-cell-mediated antitumor immunity with sustained antitumor memory. Overall, our study provides a practical strategy with clinical translational promise for immunologically cold tumor PDAC treatment.
Collapse
Affiliation(s)
- Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jiayu Feng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Lulu Qi
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
3
|
Liu D, Guo L, Waasdorp C, Meijer SL, Bootsma S, Oyarce C, Bijlsma MF, van Laarhoven HWM. Hyaluronidase improves the efficacy of nab-paclitaxel after prolonged angiogenesis inhibition in preclinical models for esophagogastric cancer. Biomed Pharmacother 2024; 178:117261. [PMID: 39106708 DOI: 10.1016/j.biopha.2024.117261] [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: 05/06/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND Long-term anti-angiogenesis leads to pruned vasculature, densely deposited extracellular matrix (ECM), and consequently reduced chemotherapy delivery in esophagogastric cancer (EGC). To address this issue, we evaluated the efficacy of adding a hyaluronidase or a NO-donor to the regimen of chemotherapy and anti-angiogenic drugs. METHODS A patient-derived EGC xenograft model was developed. Grafted mice were randomly assigned to four experimental groups and one control group. The experimental groups received DC101, a murine angiogenesis inhibitor, and nab-paclitaxel (NPTX), with the addition of hyaluronidase (PEGPH20), or NO-donor (nitroglycerine, NTG), or their combination, respectively. We compared tumor growth during 17 days of treatment. We performed immunohistochemistry for ECM components hyaluronan (HA) and collagen, CD31 for endothelial cells, and γH2AX for DNA damage. The positively stained areas were quantified, and vessel diameters were measured using QuPath software. RESULTS Prolonged DC101 treatment induced deposition of HA (p<0.01) and collagen (p<0.01). HA was effectively degraded by PEGPH20 (p<0.001), but not by NTG as expected. Both PEGPH20 (p<0.05) and NTG (p<0.01) dilated vessels collapsed in response to long-term DC101 treatment. However, only PEGPH20 (rather than NTG) was found to significantly inhibit tumor growth (p<0.05) in combination with NPTX and DC101. CONCLUSIONS These findings suggest that the mechanical barrier of HA is the major reason responsible for the resistance developed during prolonged anti-angiogenesis in EGC. Incorporating PEGPH20 into the existing treatment regimen is promising to improve outcomes for patients with EGC.
Collapse
Affiliation(s)
- Dajia Liu
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| | - Lihui Guo
- Amsterdam UMC location University of Amsterdam, Department Experimental Immunology, Amsterdam Infection and Immunity Center, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Cynthia Waasdorp
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Sybren L Meijer
- Amsterdam UMC Location University of Amsterdam, Department of Pathology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Sanne Bootsma
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| | - Cesar Oyarce
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC Location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Luo C, He S, Shi F, Zhou J, Shang L. The Role of TRAIL Signaling in Cancer: Searching for New Therapeutic Strategies. BIOLOGY 2024; 13:521. [PMID: 39056714 PMCID: PMC11274015 DOI: 10.3390/biology13070521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Cancer continues to pose a significant threat to global health, with its status as a leading cause of death remaining unchallenged. Within the realm of cancer research, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) stands out as a critical player, having been identified in the 1990s as the tenth member of the TNF family. This review examines the pivotal role of TRAIL in cancer biology, focusing on its ability to induce apoptosis in malignant cells through both endogenous and exogenous pathways. We provide an in-depth analysis of TRAIL's intracellular signaling and intercellular communication, underscoring its potential as a selective anticancer agent. Additionally, the review explores TRAIL's capacity to reshape the tumor microenvironment, thereby influencing cancer progression and response to therapy. With an eye towards future developments, we discuss the prospects of harnessing TRAIL's capabilities for the creation of tailored, precision-based cancer treatments, aiming to enhance efficacy and improve patient survival rates.
Collapse
Affiliation(s)
- Cheng Luo
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Shan He
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jianhua Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| | - Li Shang
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| |
Collapse
|
5
|
Chiu H, Chau Fang A, Chen YH, Koi RX, Yu KC, Hsieh LH, Shyu YM, Amer TA, Hsueh YJ, Tsao YT, Shen YJ, Wang YM, Chen HC, Lu YJ, Huang CC, Lu TT. Mechanistic and Kinetic Insights into Cellular Uptake of Biomimetic Dinitrosyl Iron Complexes and Intracellular Delivery of NO for Activation of Cytoprotective HO-1. JACS AU 2024; 4:1550-1569. [PMID: 38665642 PMCID: PMC11040670 DOI: 10.1021/jacsau.4c00064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024]
Abstract
Dinitrosyl iron unit (DNIU), [Fe(NO)2], is a natural metallocofactor for biological storage, delivery, and metabolism of nitric oxide (NO). In the attempt to gain a biomimetic insight into the natural DNIU under biological system, in this study, synthetic dinitrosyl iron complexes (DNICs) [(NO)2Fe(μ-SCH2CH2COOH)2Fe(NO)2] (DNIC-COOH) and [(NO)2Fe(μ-SCH2CH2COOCH3)2Fe(NO)2] (DNIC-COOMe) were employed to investigate the structure-reactivity relationship of mechanism and kinetics for cellular uptake of DNICs, intracellular delivery of NO, and activation of cytoprotective heme oxygenase (HO)-1. After rapid cellular uptake of dinuclear DNIC-COOMe through a thiol-mediated pathway (tmax = 0.5 h), intracellular assembly of mononuclear DNIC [(NO)2Fe(SR)(SCys)]n-/[(NO)2Fe(SR)(SCys-protein)]n- occurred, followed by O2-induced release of free NO (tmax = 1-2 h) or direct transfer of NO to soluble guanylate cyclase, which triggered the downstream HO-1. In contrast, steady kinetics for cellular uptake of DNIC-COOH via endocytosis (tmax = 2-8 h) and for intracellular release of NO (tmax = 4-6 h) reflected on the elevated activation of cytoprotective HO-1 (∼50-150-fold change at t = 3-10 h) and on the improved survival of DNIC-COOH-primed mesenchymal stem cell (MSC)/human corneal endothelial cell (HCEC) under stressed conditions. Consequently, this study unravels the bridging thiolate ligands in dinuclear DNIC-COOH/DNIC-COOMe as a switch to control the mechanism, kinetics, and efficacy for cellular uptake of DNICs, intracellular delivery of NO, and activation of cytoprotective HO-1, which poses an implication on enhanced survival of postengrafted MSC for advancing the MSC-based regenerative medicine.
Collapse
Affiliation(s)
- Han Chiu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Anyelina Chau Fang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Yi-Hong Chen
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Ru Xin Koi
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Kai-Ching Yu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Li-Hung Hsieh
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Yueh-Ming Shyu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Tarik Abdelkareem
Mostafa Amer
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yi-Jen Hsueh
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yu-Ting Tsao
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yang-Jin Shen
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yun-Ming Wang
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Hung-Chi Chen
- Department
of Ophthalmology and Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- College
of Medicine, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chieh-Cheng Huang
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
| | - Tsai-Te Lu
- Institute
of Biomedical Engineering, National Tsing
Hua University, Hsinchu 30013 Taiwan
- Department
of Chemistry, National Tsing Hua University, Hsinchu 30013 Taiwan
- Department
of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
6
|
Xiang Y, Chen Q, Nan Y, Liu M, Xiao Z, Yang Y, Zhang J, Ying X, Long X, Wang S, Sun J, Huang Q, Ai K. Nitric Oxide‐Based Nanomedicines for Conquering TME Fortress: Say “NO” to Insufficient Tumor Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202312092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Indexed: 01/02/2025]
Abstract
AbstractAlmost all cancer treatments are significantly limited by the strong tumor microenvironment (TME) fortress formed by abnormal vasculature, dense extracellular matrix (ECM), multidrug resistance (MDR) system, and immune “cold” environment. In the huge efforts of dismantling the TME fortress, nitric oxide (NO)‐based nanomedicines are increasingly occupying a central position and have already been identified as super “strong polygonal warriors” to dismantle TME fortress for efficient cancer treatment, benefiting from NO's unique physicochemical properties and extremely fascinating biological effects. However, there is a paucity of systematic review to elaborate on the progress and fundamental mechanism of NO‐based nanomedicines in oncology from this aspect. Herein, the key characteristics of TME fortress and the potential of NO in reprogramming TME are delineated and highlighted. The evolution of NO donors and the advantages of NO‐based nanomedicines are discussed subsequently. Moreover, the latest progress of NO‐based nanomedicines for solid tumors is comprehensively reviewed, including normalizing tumor vasculature, overcoming ECM barrier, reversing MDR, and reactivating the immunosuppression TME. Lastly, the prospects, limitations, and future directions on NO‐based nanomedicines for TME manipulation are discussed to provide new insights into the construction of more applicable anticancer nanomedicines.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yayun Nan
- Geriatric Medical Center People's Hospital of Ningxia Hui Autonomous Region Yinchuan Ningxia 750002 P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yuqi Yang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Jinping Zhang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Xingyu Long
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Jian Sun
- College of Pharmacy Xinjiang Medical University Urumqi 830017 P. R. China
| | - Qiong Huang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and Treatment Ministry of Education Xiangya Hospital Central South University Changsha 410078 P. R. China
| |
Collapse
|
7
|
Mu HY, Lin CM, Chu LA, Lin YH, Li J, Liu CY, Huang HC, Cheng SL, Lee TY, Lee HM, Chen HM, Tsai YJ, Chen Y, Huang JH. Ex Vivo Evaluation of Combination Immunotherapy Using Tumor-Microenvironment-on-Chip. Adv Healthc Mater 2024; 13:e2302268. [PMID: 37748773 DOI: 10.1002/adhm.202302268] [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: 07/17/2023] [Revised: 09/03/2023] [Indexed: 09/27/2023]
Abstract
Combination immunotherapy has emerged as a promising strategy to address the challenges associated with immune checkpoint inhibitor (ICI) therapy in breast cancer. The efficacy of combination immunotherapy hinges upon the intricate and dynamic nature of the tumor microenvironment (TME), characterized by cellular heterogeneity and molecular gradients. However, current methodologies for drug screening often fail to accurately replicate these complex conditions, resulting in limited predictive capacity for treatment outcomes. Here, a tumor-microenvironment-on-chip (TMoC), integrating a circulation system and ex vivo tissue culture with physiological oxygen and nutrient gradients, is described. This platform enables spatial infiltration of cytotoxic CD8+ T cells and their targeted attack on the tumor, while preserving the high complexity and heterogeneity of the TME. The TMoC is employed to assess the synergistic effect of five targeted therapy drugs and five chemotherapy drugs in combination with immunotherapy, demonstrating strong concordance between chip and animal model responses. The TMoC holds significant potential for advancing drug development and guiding clinical decision-making, as it offers valuable insights into the complex dynamics of the TME.
Collapse
Affiliation(s)
- Hsuan-Yu Mu
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Chiao-Min Lin
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
- Brain Research Center, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Ya-Hui Lin
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Ji Li
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Chao-Yu Liu
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Hsi-Chien Huang
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Sheng-Liang Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Tsung-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Hsin Mei Lee
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Hsin-Min Chen
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Yun-Jen Tsai
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| | - Jen-Huang Huang
- Department of Chemical Engineering, National Tsing Hua University, 101, Sec. 2, Kuang-Fu Rd., Hsinchu, 30013, Taiwan
| |
Collapse
|
8
|
Chakkera M, Foote JB, Farran B, Nagaraju GP. Breaking the stromal barrier in pancreatic cancer: Advances and challenges. Biochim Biophys Acta Rev Cancer 2024; 1879:189065. [PMID: 38160899 DOI: 10.1016/j.bbcan.2023.189065] [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: 08/04/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Pancreatic cancer (PC) remains a leading cause of mortality worldwide due to the absence of early detection methods and the low success rates of traditional therapeutic strategies. Drug resistance in PC is driven by its desmoplastic stroma, which creates a barrier that shields cancer niches and prevents the penetration of drugs. The PC stroma comprises heterogeneous cellular populations and non-cellular components involved in aberrant ECM deposition, immunosuppression, and drug resistance. These components can influence PC development through intricate and complex crosstalk with the PC cells. Understanding how stromal components and cells interact with and influence the invasiveness and refractoriness of PC cells is thus a prerequisite for developing successful stroma-modulating strategies capable of remodeling the PC stroma to alleviate drug resistance and enhance therapeutic outcomes. In this review, we explore how non-cellular and cellular stromal components, including cancer-associated fibroblasts and tumor-associated macrophages, contribute to the immunosuppressive and tumor-promoting effects of the stroma. We also examine the signaling pathways underlying their activation, tumorigenic effects, and interactions with PC cells. Finally, we discuss recent pre-clinical and clinical work aimed at developing and testing novel stroma-modulating agents to alleviate drug resistance and improve therapeutic outcomes in PC.
Collapse
Affiliation(s)
- Mohana Chakkera
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
9
|
Shah VM, Rizvi S, Smith A, Tsuda M, Krieger M, Pelz C, MacPherson K, Eng J, Chin K, Munks MW, Daniel CJ, Al-Fatease A, Yardimci GG, Langer EM, Brody JR, Sheppard BC, Alani AWG, Sears RC. Micelle-Formulated Juglone Effectively Targets Pancreatic Cancer and Remodels the Tumor Microenvironment. Pharmaceutics 2023; 15:2651. [PMID: 38139993 PMCID: PMC10747591 DOI: 10.3390/pharmaceutics15122651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 12/24/2023] Open
Abstract
Pancreatic cancer remains a formidable challenge due to limited treatment options and its aggressive nature. In recent years, the naturally occurring anticancer compound juglone has emerged as a potential therapeutic candidate, showing promising results in inhibiting tumor growth and inducing cancer cell apoptosis. However, concerns over its toxicity have hampered juglone's clinical application. To address this issue, we have explored the use of polymeric micelles as a delivery system for juglone in pancreatic cancer treatment. These micelles, formulated using Poloxamer 407 and D-α-Tocopherol polyethylene glycol 1000 succinate, offer an innovative solution to enhance juglone's therapeutic potential while minimizing toxicity. In-vitro studies have demonstrated that micelle-formulated juglone (JM) effectively decreases proliferation and migration and increases apoptosis in pancreatic cancer cell lines. Importantly, in-vivo, JM exhibited no toxicity, allowing for increased dosing frequency compared to free drug administration. In mice, JM significantly reduced tumor growth in subcutaneous xenograft and orthotopic pancreatic cancer models. Beyond its direct antitumor effects, JM treatment also influenced the tumor microenvironment. In immunocompetent mice, JM increased immune cell infiltration and decreased stromal deposition and activation markers, suggesting an immunomodulatory role. To understand JM's mechanism of action, we conducted RNA sequencing and subsequent differential expression analysis on tumors that were treated with JM. The administration of JM treatment reduced the expression levels of the oncogenic protein MYC, thereby emphasizing its potential as a focused, therapeutic intervention. In conclusion, the polymeric micelles-mediated delivery of juglone holds excellent promise in pancreatic cancer therapy. This approach offers improved drug delivery, reduced toxicity, and enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Vidhi M. Shah
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
| | - Syed Rizvi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 South Moody Avenue, Portland, OR 97201, USA
| | - Alexander Smith
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
| | - Motoyuki Tsuda
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Madeline Krieger
- Cancer Early Detection Advanced Research Center, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carl Pelz
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Kevin MacPherson
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Jenny Eng
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Koei Chin
- Cancer Early Detection Advanced Research Center, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael W. Munks
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
| | - Colin J. Daniel
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Adel Al-Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Guraiger, Abha 62529, Saudi Arabia
| | - Galip Gürkan Yardimci
- Cancer Early Detection Advanced Research Center, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Jonathan R. Brody
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Surgery, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Brett C. Sheppard
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
- Department of Surgery, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
| | - Adam WG. Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 South Moody Avenue, Portland, OR 97201, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Rosalie C. Sears
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA; (V.M.S.)
- Department of Molecular and Medical Genetics, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| |
Collapse
|
10
|
Ciflik KB, Dogan HT, Dogan E, Ciflik BO, Kocer B, Karaoglanoglu N, Yekeler E. Can tumour budding change the future of pulmonary adenocarcinoma? Pathol Res Pract 2023; 250:154808. [PMID: 37748210 DOI: 10.1016/j.prp.2023.154808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND Pulmonary adenocarcinoma shows different prognosis even in the same pathological subtype and stage. In this study, it is aimed to investigate the relationship between tumour budding and known prognostic values and clinicopathological features in pulmonary adenocarcinoma. METHODS In this study, there have been 77 patients diagnosed with primary pulmonary adenocarcinoma. In the evaluation process, the number of budding between 0 and 4 is accepted as low budding (Bd1), the number of budding between 5 and 9 is considered as medium budding (Bd2), and the number of budding above 10 is considered as high budding (Bd3). RESULTS According to the findings of the study, it can be seen that there is a statistical difference between tumour budding and stromal fibrosis (p < 0.001). The presence of pleural invasion, lymph vascular invasion and perineural invasion in patients with Bd3 is found to be statistically higher than the patients with Bd1 (p = 0.048) (p = 0.041) (p = 0.029). CONCLUSIONS Tumour budding has been associated with pleural invasion, lymph vascular invasion, perineural invasion, and stromal fibrosis. This study is the first to show the relationship between tumour budding and stromal fibrosis in pulmonary adenocarcinomas. The role of tumour budding in lung cancers remains to be clarified.
Collapse
Affiliation(s)
| | - Hayriye Tatli Dogan
- Department of Pathology, Ankara Yildirim Beyazit University, Ankara, Turkey.
| | - Elif Dogan
- Department of Pathology, Ankara City Hospital, Ankara, Turkey.
| | | | - Bulent Kocer
- Department of Thoracic Surgery, Ankara City Hospital, Ankara, Turkey.
| | | | - Erdal Yekeler
- Department of Thoracic Surgery, Ankara City Hospital, Ankara, Turkey.
| |
Collapse
|
11
|
Duan QY, Zhu YX, Jia HR, Wang SH, Wu FG. Nanogels: Synthesis, properties, and recent biomedical applications. PROGRESS IN MATERIALS SCIENCE 2023; 139:101167. [DOI: 10.1016/j.pmatsci.2023.101167] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Liao CJ, Tseng YT, Cheng YA, Dayao LA, Iffland-Mühlhaus L, Gee LB, Ribson RD, Chan TS, Apfel UP, Lu TT. Ligand Control of Dinitrosyl Iron Complexes for Selective Superoxide-Mediated Nitric Oxide Monooxygenation and Superoxide-Dioxygen Interconversion. J Am Chem Soc 2023; 145:20389-20402. [PMID: 37683125 DOI: 10.1021/jacs.3c05577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Through nitrosylation of [Fe-S] proteins, or the chelatable iron pool, a dinitrosyl iron unit (DNIU) [Fe(NO)2] embedded in the form of low-molecular-weight/protein-bound dinitrosyl iron complexes (DNICs) was discovered as a metallocofactor assembled under inflammatory conditions with elevated levels of nitric oxide (NO) and superoxide (O2-). In an attempt to gain biomimetic insights into the unexplored transformations of the DNIU under inflammation, we investigated the reactivity toward O2- by a series of DNICs [(NO)2Fe(μ-MePyr)2Fe(NO)2] (1) and [(NO)2Fe(μ-SEt)2Fe(NO)2] (3). During the superoxide-induced conversion of DNIC 1 into DNIC [(K-18-crown-6-ether)2(NO2)][Fe(μ-MePyr)4(μ-O)2(Fe(NO)2)4] (2-K-crown) and a [Fe3+(MePyr)x(NO2)y(O)z]n adduct, stoichiometric NO monooxygenation yielding NO2- occurs without the transient formation of peroxynitrite-derived •OH/•NO2 species. To study the isoelectronic reaction of O2(g) and one-electron-reduced DNIC 1, a DNIC featuring an electronically localized {Fe(NO)2}9-{Fe(NO)2}10 electronic structure, [K-18-crown-6-ether][(NO)2Fe(μ-MePyr)2Fe(NO)2] (1-red), was successfully synthesized and characterized. Oxygenation of DNIC 1-red leads to the similar assembly of DNIC 2-K-crown, of which the electronic structure is best described as paramagnetic with weak antiferromagnetic coupling among the four S = 1/2 {FeIII(NO-)2}9 units and S = 5/2 Fe3+ center. In contrast to DNICs 1 and 1-red, DNICs 3 and [K-18-crown-6-ether][(NO)2Fe(μ-SEt)2Fe(NO)2] (3-red) display a reversible equilibrium of "3 + O2- ⇋ 3-red + O2(g)", which is ascribed to the covalent [Fe(μ-SEt)2Fe] core and redox-active [Fe(NO)2] unit. Based on this study, the supporting/bridging ligands in dinuclear DNIC 1/3 (or 1-red/3-red) control the selective monooxygenation of NO and redox interconversion between O2- and O2 during reaction with O2- (or O2).
Collapse
Affiliation(s)
- Cheng-Jhe Liao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Ting Tseng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-An Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Loise Ann Dayao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Linda Iffland-Mühlhaus
- Department of Chemistry and Biochemistry, Inorganic Chemistry I, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Leland B Gee
- LCLS, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Ryan D Ribson
- LCLS, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Ting-Shan Chan
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Ulf-Peter Apfel
- Department of Chemistry and Biochemistry, Inorganic Chemistry I, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Electrosynthesis, Fraunhofer UMSICHT, 46047 Oberhausen, Germany
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
13
|
Chuang WH, Chou YT, Chen YH, Kuo TH, Liaw WF, Lu TT, Kao CF, Wang YM. Neuroprotective Effect of NO-Delivery Dinitrosyl Iron Complexes (DNICs) on Amyloid Pathology in the Alzheimer's Disease Cell Model. ACS Chem Neurosci 2023; 14:2922-2934. [PMID: 37533298 DOI: 10.1021/acschemneuro.3c00348] [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: 08/04/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment, memory loss, and behavioral deficits. β-amyloid1-42 (Aβ1-42) aggregation is a significant cause of the pathogenesis in AD. Despite the numerous types of research, the current treatment efficacy remains insufficient. Hence, a novel therapeutic strategy is required. Nitric oxide (NO) is a multifunctional gaseous molecule. NO displays a neuroprotective role in the central nervous system by inhibiting the Aβ aggregation and rescuing memory and learning deficit through the NO signaling pathway. Targeting the NO pathway might be a therapeutic option; however, NO has a limited half-life under the biological system. To address this issue, a biomimetic dinitrosyl iron complex [(NO)2Fe(μ-SCH2CH2COOH)2Fe(NO)2] (DNIC-COOH) that could stably deliver NO was explored in the current study. To determine whether DNIC-COOH exerts anti-AD efficacy, DNIC-COOH was added to neuron-like cells and primary cortical neurons along with Aβ1-42. This study found that DNIC-COOH protected neuronal cells from Aβ-induced cytotoxicity, potentiated neuronal functions, and facilitated Aβ1-42 degradation through the NO-sGC-cGMP-AKT-GSK3β-CREB/MMP-9 pathway.
Collapse
Affiliation(s)
- Wen-Han Chuang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ting Chou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ting-Han Kuo
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biomedical Science and Environmental Biology, Department of Dentistry, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
14
|
Li Y, Yang KD, Duan HY, Du YN, Ye JF. Phage-based peptides for pancreatic cancer diagnosis and treatment: alternative approach. Front Microbiol 2023; 14:1231503. [PMID: 37601380 PMCID: PMC10433397 DOI: 10.3389/fmicb.2023.1231503] [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: 05/30/2023] [Accepted: 07/06/2023] [Indexed: 08/22/2023] Open
Abstract
Pancreatic cancer is a devastating disease with a high mortality rate and a lack of effective therapies. The challenges associated with early detection and the highly aggressive nature of pancreatic cancer have limited treatment options, underscoring the urgent need for better disease-modifying therapies. Peptide-based biotherapeutics have become an attractive area of research due to their favorable properties such as high selectivity and affinity, chemical modifiability, good tissue permeability, and easy metabolism and excretion. Phage display, a powerful technique for identifying peptides with high affinity and specificity for their target molecules, has emerged as a key tool in the discovery of peptide-based drugs. Phage display technology involves the use of bacteriophages to express peptide libraries, which are then screened against a target of interest to identify peptides with desired properties. This approach has shown great promise in cancer diagnosis and treatment, with potential applications in targeting cancer cells and developing new therapies. In this comprehensive review, we provide an overview of the basic biology of phage vectors, the principles of phage library construction, and various methods for binding affinity assessment. We then describe the applications of phage display in pancreatic cancer therapy, targeted drug delivery, and early detection. Despite its promising potential, there are still challenges to be addressed, such as optimizing the selection process and improving the pharmacokinetic properties of phage-based drugs. Nevertheless, phage display represents a promising approach for the development of novel targeted therapies in pancreatic cancer and other tumors.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Ya-nan Du
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
15
|
Franchi M, Karamanos KA, Cappadone C, Calonghi N, Greco N, Franchi L, Onisto M, Masola V. Colorectal Cancer Cell Invasion and Functional Properties Depend on Peri-Tumoral Extracellular Matrix. Biomedicines 2023; 11:1788. [PMID: 37509428 PMCID: PMC10376217 DOI: 10.3390/biomedicines11071788] [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: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
We investigated how the extracellular matrix (ECM) affects LoVo colorectal cancer cells behavior during a spatiotemporal invasion. Epithelial-to-mesenchymal transition (EMT) markers, matrix-degrading enzymes, and morphological phenotypes expressed by LoVo-S (doxorubicin-sensitive) and higher aggressive LoVo-R (doxorubicin-resistant) were evaluated in cells cultured for 3 and 24 h on Millipore filters covered by Matrigel, mimicking the basement membrane, or type I Collagen reproducing a desmoplastic lamina propria. EMT and invasiveness were investigated with RT-qPCR, Western blot, and scanning electron microscopy. As time went by, most gene expressions decreased, but in type I Collagen samples, a strong reduction and high increase in MMP-2 expression in LoVo-S and -R cells occurred, respectively. These data were confirmed by the development of an epithelial morphological phenotype in LoVo-S and invading phenotypes with invadopodia in LoVo-R cells as well as by protein-level analysis. We suggest that the duration of culturing and type of substrate influence the morphological phenotype and aggressiveness of both these cell types differently. In particular, the type I collagen meshwork, consisting of large fibrils confining inter fibrillar micropores, affects the two cell types differently. It attenuates drug-sensitive LoVo-S cell aggressiveness but improves a proteolytic invasion in drug-resistant LoVo-R cells as time goes by. Experimental studies on CRC cells should examine the peri-tumoral ECM components, as well as the dynamic physical conditions of TME, which affect the behavior and aggressiveness of both drug-sensitive and drug-resistant LoVo cells differently.
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, 47900 Rimini, Italy
| | | | - Concettina Cappadone
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Nicola Greco
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy
| | - Leonardo Franchi
- Department of Medicine, University of Bologna, 40126 Bologna, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy
| | - Valentina Masola
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy
| |
Collapse
|
16
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
17
|
Nanotechnology for next-generation cancer immunotherapy: State of the art and future perspectives. J Control Release 2023; 356:14-25. [PMID: 36805873 DOI: 10.1016/j.jconrel.2023.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Over the past decade, immunotherapy aiming to activate an effective antitumor immune response has ushered in a new era of cancer treatment. However, the efficacy of cancer immunotherapy is limited by low response rates and high systemic toxicity. Nanotechnology is an encouraging platform for the development of next-generation cancer immunotherapy to effectively treat advanced cancer. Nanotechnology-enabled immunotherapy has remarkable advantages, ranging from the increased bioavailability and stability of immunotherapeutic agents to the enhanced activation of immune cells and favorable safety profiles. Nanotechnology-enabled immunotherapy can target solid tumors through reprogramming or stimulating immune cells (i.e., nanovaccines); modulating the immunosuppressive tumor microenvironment; or targeting tumor cells and altering their responses to immune cells to generate effective antitumor immunity. In this Oration, I introduce the advanced strategies currently being pursued by our laboratory and other groups to improve the therapeutic efficacy of cancer immunotherapy and discuss the potential challenges and future directions.
Collapse
|
18
|
Yang HZ, Guo Y, Pu L, Yu XQ, Zhang J. Fluorescent Self-Reporting Lipid Nanoparticles for Nitric Oxide/Gene Co-Delivery and Combination Therapy. Mol Pharm 2023; 20:1404-1414. [PMID: 36594589 DOI: 10.1021/acs.molpharmaceut.2c00973] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The combination cancer therapy of nitric oxide (NO) with gene therapy is a promising method for tumor treatment. However, efficient co-delivery of gas and therapeutic genes to tumor cells remains a challenge. Herein, we designed a nano-sized ultraviolet (UV) light-responsive cationic lipid vector DPNO(Zn). Fluorescence spectroscopy and confocal imaging experiments revealed that DPNO(Zn) lipid nanoparticles (LNPs) could rapidly release NO under low-power UV light irradiation. Moreover, the fluorescence turn-on might take place along with the release of NO, indicating the self-reporting ability. Gene delivery experiments showed that DPNO(Zn) LNPs had good gene transfection ability, making such materials a good candidate for gas/gene combination therapy. In vitro antitumor assay demonstrated that the co-delivery system was more effective in inhibiting tumor cell proliferation than individual NO or pTrail treatment. Studies on the mechanism of tumor cell apoptosis induced by NO/pTrail co-delivery showed that NO could not only effectively increase the accumulation of p53 protein in tumor cells, thereby promoting the activation of caspase-3, but also induce mitochondrial damage. On the other hand, the Trail protein expressed by pTrail gene could enhance the degree of NO-induced caspase-3 activation, indicating the synergistic effect. These results proved that DPNO(Zn) LNP may serve as a multifunctional nanocarrier for potential tumor therapy.
Collapse
Affiliation(s)
- Hui-Zhen Yang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| | - Yu Guo
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| | - Lin Pu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.,Department of Chemistry, University of Virginia, McCormick Rd, Charlottesville, Virginia 22904, United States
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China.,Asymmetric Synthesis and Chiral Technology Key Laboratory of Sichuan Province, Department of Chemistry, Xihua University, Chengdu 610039, P. R. China
| | - Ji Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
19
|
Yagolovich AV, Gasparian ME, Dolgikh DA. Recent Advances in the Development of Nanodelivery Systems Targeting the TRAIL Death Receptor Pathway. Pharmaceutics 2023; 15:pharmaceutics15020515. [PMID: 36839837 PMCID: PMC9961178 DOI: 10.3390/pharmaceutics15020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
The TRAIL (TNF-related apoptosis-inducing ligand) apoptotic pathway is extensively exploited in the development of targeted antitumor therapy due to TRAIL specificity towards its cognate receptors, namely death receptors DR4 and DR5. Although therapies targeting the TRAIL pathway have encountered many obstacles in attempts at clinical implementation for cancer treatment, the unique features of the TRAIL signaling pathway continue to attract the attention of researchers. Special attention is paid to the design of novel nanoscaled delivery systems, primarily aimed at increasing the valency of the ligand for improved death receptor clustering that enhances apoptotic signaling. Optionally, complex nanoformulations can allow the encapsulation of several therapeutic molecules for a combined synergistic effect, for example, chemotherapeutic agents or photosensitizers. Scaffolds for the developed nanodelivery systems are fabricated by a wide range of conventional clinically approved materials and innovative ones, including metals, carbon, lipids, polymers, nanogels, protein nanocages, virus-based nanoparticles, dendrimers, DNA origami nanostructures, and their complex combinations. Most nanotherapeutics targeting the TRAIL pathway are aimed at tumor therapy and theranostics. However, given the wide spectrum of action of TRAIL due to its natural role in immune system homeostasis, other therapeutic areas are also involved, such as liver fibrosis, rheumatoid arthritis, Alzheimer's disease, and inflammatory diseases caused by bacterial infections. This review summarizes the recent innovative developments in the design of nanodelivery systems modified with TRAIL pathway-targeting ligands.
Collapse
Affiliation(s)
- Anne V. Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Correspondence:
| | - Marine E. Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Dmitry A. Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
20
|
Lee TY, Lu HH, Cheng HT, Huang HC, Tsai YJ, Chang IH, Tu CP, Chung CW, Lu TT, Peng CH, Chen Y. Delivery of nitric oxide with a pH-responsive nanocarrier for the treatment of renal fibrosis. J Control Release 2023; 354:417-428. [PMID: 36627025 DOI: 10.1016/j.jconrel.2022.12.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023]
Abstract
Fibrosis is an excessive accumulation of extracellular matrix (ECM) that may cause severe organ dysfunction. Nitric oxide (NO), a multifunctional gaseous signaling molecule, may inhibit fibrosis, and delivery of NO may serve as a potential antifibrotic strategy. However, major limitations in the application of NO to treat fibrotic diseases include its nonspecificity, short half-life and low availability in fibrotic tissue. Herein, we aimed to develop a stimuli-responsive drug carrier to deliver NO to halt kidney fibrosis. We manufactured a nanoparticle (NP) composed of pH-sensitive poly[2-(diisopropylamino)ethyl methacrylate (PDPA) polymers to encapsulate a NO donor, a dinitrosyl iron complex (DNIC; [Fe2(μ-SEt)2(NO)4]). The NPs were stable at physiological pH 7.4 but disintegrated at pH 4.0-6.0. The NPs showed significant cytotoxicity to cultured human myofibroblasts and were able to inhibit the activation of myofibroblasts, as indicated by a lower expression level of α-smooth muscle actin and the synthesis of a major ECM component, collagen I, in cultured human myofibroblasts. When given to mice treated with unilateral ureteral ligation/obstruction (UUO) to induce kidney fibrosis, these NPs remained in blood at a stable concentration for as long as 24 h and might enter the fibrotic kidneys to suppress myofibroblast activation and collagen I production, leading to a 70% reduction in the fibrotic area. In summary, our strategy to assemble a NO donor, the iron nitrosyl complex DNIC, into pH-responsive NPs proves effective in treating renal fibrosis and warrants further investigation for its therapeutic potential.
Collapse
Affiliation(s)
- Tsung-Ying Lee
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hung-Hsun Lu
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hui-Teng Cheng
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Zhu Bei City 302, Taiwan
| | - Hsi-Chien Huang
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yun-Jen Tsai
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - I-Hsiang Chang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chao-Peng Tu
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chieh-Wei Chung
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chi-How Peng
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Yunching Chen
- Institute of Biomedical Engineering, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
21
|
Targeting TRAIL Death Receptors in Triple-Negative Breast Cancers: Challenges and Strategies for Cancer Therapy. Cells 2022; 11:cells11233717. [PMID: 36496977 PMCID: PMC9739296 DOI: 10.3390/cells11233717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor necrosis factor (TNF) superfamily member TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis in cancer cells via death receptor (DR) activation with little toxicity to normal cells or tissues. The selectivity for activating apoptosis in cancer cells confers an ideal therapeutic characteristic to TRAIL, which has led to the development and clinical testing of many DR agonists. However, TRAIL/DR targeting therapies have been widely ineffective in clinical trials of various malignancies for reasons that remain poorly understood. Triple negative breast cancer (TNBC) has the worst prognosis among breast cancers. Targeting the TRAIL DR pathway has shown notable efficacy in a subset of TNBC in preclinical models but again has not shown appreciable activity in clinical trials. In this review, we will discuss the signaling components and mechanisms governing TRAIL pathway activation and clinical trial findings discussed with a focus on TNBC. Challenges and potential solutions for using DR agonists in the clinic are also discussed, including consideration of the pharmacokinetic and pharmacodynamic properties of DR agonists, patient selection by predictive biomarkers, and potential combination therapies. Moreover, recent findings on the impact of TRAIL treatment on the immune response, as well as novel strategies to address those challenges, are discussed.
Collapse
|
22
|
The microbiota and aging microenvironment in pancreatic cancer: Cell origin and fate. Biochim Biophys Acta Rev Cancer 2022; 1877:188826. [DOI: 10.1016/j.bbcan.2022.188826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/30/2022]
|
23
|
Gan J, Sun L, Guan C, Ren T, Zhang Q, Pan S, Zhang Q, Chen H. Preparation and Properties of Salecan-Soy Protein Isolate Composite Hydrogel Induced by Thermal Treatment and Transglutaminase. Int J Mol Sci 2022; 23:9383. [PMID: 36012648 PMCID: PMC9409434 DOI: 10.3390/ijms23169383] [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: 07/13/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 11/18/2022] Open
Abstract
Salecan (Sal) is a novel marine microbial polysaccharide. In the present research, Sal and soy protein isolate (SPI) were adopted to fabricate Sal-SPI composite hydrogel based on a stepwise process (thermal treatment and transglutaminase induction). The effect of Sal concentration on morphology, texture properties, and the microstructure of the hydrogel was evaluated. As Sal concentration varied from 0.4 to 0.6 wt%, hydrogel elasticity increased from 0.49 to 0.85 mm. Furthermore, the internal network structure of Sal-SPI composite hydrogel also became denser and more uniform as Sal concentration increased. Rheological studies showed that Sal-SPI elastic hydrogel formed under the gelation process. Additionally, FTIR and XRD results demonstrated that hydrogen bonds formed between Sal and SPI molecules, inferring the formation of the interpenetrating network structure. This research supplied a green and simple method to fabricate Sal-SPI double network hydrogels.
Collapse
Affiliation(s)
- Jing Gan
- College of Life Science, Yantai University, No. 30 Qingquan Road, Laishan Strict, Yantai 264000, China
| | - Lirong Sun
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Chenxia Guan
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Teng Ren
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Qinling Zhang
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Shihui Pan
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Qian Zhang
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| | - Hao Chen
- Marine College, Shandong University, No. 180 Wenhua West Road, Gao Strict, Weihai 264209, China
| |
Collapse
|
24
|
Ma X, Li SJ, Liu Y, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Bioengineered nanogels for cancer immunotherapy. Chem Soc Rev 2022; 51:5136-5174. [PMID: 35666131 DOI: 10.1039/d2cs00247g] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed increasingly rapid advances in nanocarrier-based biomedicine aimed at improving treatment paradigms for cancer. Nanogels serve as multipurpose and constructed vectors formed via intramolecular cross-linking to generate drug delivery systems, which is attributed predominantly to their satisfactory biocompatibility, bio-responsiveness, high stability, and low toxicity. Recently, immunotherapy has experienced unprecedented growth and has become the preferred strategy for cancer treatment, and mainly involves the mobilisation of the immune system and an enhanced anti-tumour immunity of the tumour microenvironment. Despite the inspiring success, immunotherapeutic strategies are limited due to the low response rates and immune-related adverse events. Like other nanomedicines, nanogels are comparably limited by lower focal enrichment rates upon introduction into the organism via injection. Because nanogels are three-dimensional cross-linked aqueous materials that exhibit similar properties to natural tissues and are structurally stable, they can comfortably cope with shear forces and serum proteins in the bloodstream, and the longer circulation life increases the chance of nanogel accumulation in the tumour, conferring deep tumour penetration. The large specific surface area can reduce or eliminate off-target effects by introducing stimuli-responsive functional groups, allowing multiple physical and chemical modifications for specific purposes to improve targeting to specific immune cell subpopulations or immune organs, increasing the bioavailability of the drug, and conferring a low immune-related adverse events on nanogel therapies. The slow release upon reaching the tumour site facilitates long-term awakening of the host's immune system, ultimately achieving enhanced therapeutic effects. As an effective candidate for cancer immunotherapy, nanogel-based immunotherapy has been widely used. In this review, we mainly summarize the recent advances of nanogel-based immunotherapy to deliver immunomodulatory small molecule drugs, antibodies, genes and cytokines, to target antigen presenting cells, form cancer vaccines, and enable chimeric antigen receptor (CAR)-T cell therapy. Future challenges as well as expected and feasible prospects for clinical treatment are also highlighted.
Collapse
Affiliation(s)
- Xianbin Ma
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Shu-Jin Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Yuantong Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Tian Zhang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Peng Xue
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Yuejun Kang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine, Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Zhigang Xu
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy & Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China.
| |
Collapse
|