1
|
Wang L, Bi S, Li Z, Liao A, Li Y, Yang L, Zhou X, Gao Y, Liu X, Zou Y, Zhang X, Shi J, Yu S, Yu Z, Guo J. Napabucasin deactivates STAT3 and promotes mitoxantrone-mediated cGAS-STING activation for hepatocellular carcinoma chemo-immunotherapy. Biomaterials 2025; 313:122766. [PMID: 39180916 DOI: 10.1016/j.biomaterials.2024.122766] [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: 05/18/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The immune resistance of tumor microenvironment (TME) causes immune checkpoint blockade therapy inefficient to hepatocellular carcinoma (HCC). Emerging strategies of using chemotherapy regimens to reverse the immune resistance provide the promise for promoting the efficiency of immune checkpoint inhibitors. The induction of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) in tumor cells evokes the adaptive immunity and remodels the immunosuppressive TME. In this study, we report that mitoxantrone (MIT, a chemotherapeutic drug) activates the cGAS-STING signaling pathway of HCC cells. We provide an approach to augment the efficacy of MIT using a signal transducer and activator of transcription 3 (STAT3) inhibitor called napabucasin (NAP). We prepare an aminoethyl anisamide (AEAA)-targeted polyethylene glycol (PEG)-modified poly (lactic-co-glycolic acid) (PLGA)-based nanocarrier for co-delivery of MIT and NAP. The resultant co-nanoformulation can elicit the cGAS-STING-based immune responses to reshape the immunoresistant TME in the mice orthotopically grafted with HCC. Consequently, the resultant co-nanoformulation can promote anti-PD-1 antibody for suppressing HCC development, generating long-term survival, and inhibiting tumor recurrence. This study reveals the potential of MIT to activate the cGAS-STING signaling pathway, and confirms the feasibility of nano co-delivery for MIT and NAP on achieving HCC chemo-immunotherapy.
Collapse
Affiliation(s)
- Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Shengnan Bi
- Department of Pharmacy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Zhuo Li
- Department of Pharmacy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Anqi Liao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xinyi Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yuqiong Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xiaobo Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Xuemei Zhang
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jia Shi
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shihan Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Cao J, Sun J, Zhang Y, Dong Z, Li M, Liu F, Younas A, Zhang N, Chen Y. An antigen/chemotherapy co-loaded DNA nanocube inserts into tumor cell plasma membrane and enhances chemo- and immunotherapy. Int J Pharm 2025; 669:125068. [PMID: 39662859 DOI: 10.1016/j.ijpharm.2024.125068] [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: 05/28/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
Breast cancer is the most frequently diagnosed female cancer. Combined chemo- and immunotherapies have been extensively explored to treat breast cancer. To improve the efficacy of the combined therapies, this study designed a hollow DNA nanocube with four cholesterol anchors (C2.2) that could be inserted into breast cancer cell plasma membrane. A model antigen-ovalbumin (OVA257-264) was further conjugated to C2.2 to construct C2.2-OVA and a model chemotherapy-Doxorubicin (DOX) was also loaded to C2.2-OVA to prepare DOX@C2.2-OVA for the combined chemo- and immunotherapy to treat breast cancer. C2.2, C2.2-OVA, and DOX@C2.2-OVA were successfully prepared and possessed square-like shape and small size. C2.2 and C2.2-OVA could be anchored in breast cancer cell plasma membrane for at least 10 h. C2.2-OVA demonstrated the significantly higher activation rates of DC cells than free OVA. Although C2.2 showed no cytotoxicity, C2.2 increased the potency of 5-FU and carboplatin to MCF-7 and 4 T1 breast cancer cells. DOX@C2.2-OVA treatment to 4 T1 tumor in vivo showed the significant reduction of tumor size and weight, and resulted in more DCs and CD8+ T cells in 4 T1 tumor. In summary, DOX@C2.2-OVA that could be inserted into tumor cell plasma membrane enhanced the combined chemo- and immunotherapy for the breast cancer treatment.
Collapse
Affiliation(s)
- Jun Cao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Junhong Sun
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China; Bayinguoleng Mongolian Autonomous Prefecture People's Hospital, Korla 841000, Xinjiang Uygur Autonomous Region, PR China
| | - Yuzhen Zhang
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Zhuolin Dong
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Mengru Li
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Fenfen Liu
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Ayesha Younas
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Nan Zhang
- Department of Pharmaceutics and Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, Henan, PR China.
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, Henan, PR China.
| |
Collapse
|
3
|
Zou T, Huang Y, Zhou Z, He S, Liu J, Chen Y, Liu H, Luo Z, Liu M, Wei H, Yu C. A minimalist multifunctional nano-prodrug for drug resistance reverse and integration with PD-L1 mAb for enhanced immunotherapy of hepatocellular carcinoma. J Nanobiotechnology 2024; 22:750. [PMID: 39627819 PMCID: PMC11613529 DOI: 10.1186/s12951-024-03027-w] [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: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/08/2024] Open
Abstract
Clinical treatment of hepatocellular carcinoma (HCC) with 5-fluorouracil (5-FU), the primary anticancer agent, remains unsatisfactory due to the glutathione (GSH)-associated drug resistance and immunosuppressive microenvironment of HCC. To develop a facile yet robust strategy to overcome 5-FU resistance for enhanced immunotherapy treatment of HCC via all dimensional GSH exhaustion, we report in this study construction of a minimalist prodrug consisting of 5-FU linked to an indoleamine-(2,3)-dioxygenase (IDO) inhibitor (IND) via a disulfide bridge, FU-SS-IND that can further self-assemble into stabilized nanoparticles, FU-SS-IND NPs. Specifically, besides the disulfide linker-induced GSH exhaustion, IND inhibits GSH biosynthesis and enhances the effector function of T cells for turning a "cold" tumor to a "hot" one, which synergistically achieving a tumor inhibition rate (TIR) of 92.5% in a 5-FU resistant mice model. Most importantly, FU-SS-IND NPs could upregulate programmed death ligand 1 (PD-L1) expression on the surface of tumor cells, which enables facile combination with immune checkpoint blockade (ICB) for a ultimate prolonged survival lifetime of 5-FU-resistant tumors-bearing mice. Overall, the minimalist bioreducible nano-prodrug developed herein demonstrates great translatable potential for efficiently reversing drug resistance and enhancing immunotherapy of HCC.
Collapse
Affiliation(s)
- Ting Zou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yun Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zongtao Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuangyan He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jia Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yalan Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hongdu Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhonghui Luo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Miaoxin Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - CuiYun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China.
| |
Collapse
|
4
|
Farzeen Z, Khan RRM, Chaudhry AR, Pervaiz M, Saeed Z, Rasheed S, Shehzad B, Adnan A, Summer M. Dostarlimab: A promising new PD-1 inhibitor for cancer immunotherapy. J Oncol Pharm Pract 2024; 30:1411-1431. [PMID: 39056234 DOI: 10.1177/10781552241265058] [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: 07/28/2024]
Abstract
OBJECTIVE Dostarlimab, a humanized monoclonal PD-1 blocking antibody, is being tested as a cancer therapy in this review. Specifically, it addresses mismatch repair failure in endometrial cancer and locally progressed rectal cancer patients. DATA SOURCES A thorough database search found Dostarlimab clinical trials and studies. Published publications and ongoing clinical trials on Dostarlimab's efficacy as a single therapy and in conjunction with other medicines across cancer types were searched. DATA SUMMARY The review recommends Dostarlimab for endometrial cancer mismatch repair failure, as supported by GARNET studies. The analysis also highlights locally advanced rectal cancer findings. In the evolving area of cancer therapy, immune checkpoint inhibitors including pembrolizumab, avelumab, atezolizumab, nivolumab, and durvalumab were discussed. CONCLUSIONS Locally advanced rectal cancer patients responded 100% to Dostarlimab. Many clinical trials, including ROSCAN, AMBER, IOLite, CITRINO, JASPER, OPAL, PRIME, PERLA, and others, are investigating Dostarlimab in combination treatment. This research sheds light on Dostarlimab's current and future possibilities, in improving cancer immunotherapy understanding.
Collapse
Affiliation(s)
- Zubaria Farzeen
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | | | - Ayoub Rashid Chaudhry
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Pervaiz
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Zohaib Saeed
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Shahzad Rasheed
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Behram Shehzad
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Ahmad Adnan
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
5
|
Ye M, Hu J, Han L, Zhang H, Xue P, Kang Y, Bai S, Xu Z. Neighboring Effect-Initiated Supramolecular Nanocomplex with Sequential Infiltration as Irreversible Apoptosis Inducer for Synergetic Chemo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402809. [PMID: 39137339 PMCID: PMC11481388 DOI: 10.1002/advs.202402809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/03/2024] [Indexed: 08/15/2024]
Abstract
Chemotherapy-based combination regimens are recommended as first-line treatment for colorectal cancer. However, multidrug resistance (MDR) and limited drug infiltration in tumor microenvironment remain critical challenges. Herein, a pH/redox dual activated supramolecular DAS@CD-OxPt (IV) nanoparticles (NPs) via host-guest molecular recognition to achieve relay drugs delivery of active oxaliplatin (OxPt (IV)) and Src inhibitor dasatinib (DAS) between tumor cells is developed. DAS@CD-OxPt (IV) NPs exhibit prolonged circulation in the blood and intra-tumoral retention. Triggered by the endo/lysosome (pH 5.0), flexible DAS@CD-OxPt (IV) NPs exhibited proton-driven in situ assembly to form nanofiber in tumor cells. Dual chemotherapeutic agents released from DAS@CD-OxPt (IV) NPs synergistically cause irreversible DNA damage by blocking p53-mediated DNA repair. Supramolecular nanofibers can further serve as the "ammunition depot" to continuously release drugs from dying cells and transport them into neighboring tumor cells, leading to domino-like cell death and enhanced immunogenicity. Furthermore, DAS@CD-OxPt (IV) NPs combined with immune checkpoint blockade (ICB) therapy strikingly suppress CT26 tumor growth and pulmonary metastasis.
Collapse
Affiliation(s)
- Mengjie Ye
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Junfeng Hu
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Linlin Han
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Hengbo Zhang
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
| | - Shuang Bai
- Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Researchthe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular SensingMinistry of EducationSchool of Materials and Energy & Chongqing Engineering Research Center, for Micro‐Nano Biomedical Materials and DevicesSouthwest UniversityChongqing400715P. R. China
- State Key Laboratory of Chemo/Biosensing and ChemometricsHunan UniversityChangsha410082P. R. China
- Yibin Academy of Southwest UniversityYibin644000China
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province College of Chemistry and Chemical EngineeringHainan Normal UniversityHaikou571158China
| |
Collapse
|
6
|
Yu H, Liu S, Yuan Z, Huang H, Yan P, Zhu W. Targeted co-delivery of rapamycin and oxaliplatin by liposomes suppresses tumor growth and metastasis of colorectal cancer. Biomed Pharmacother 2024; 178:117192. [PMID: 39098178 DOI: 10.1016/j.biopha.2024.117192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
The activation of tumor cell immunogenicity through oxaliplatin (OXP)-induced immunogenic cell death (ICD) has significant implications in cancer treatment. However, the anti-tumor effect of OXP monotherapy still has many shortcomings, and the systemic administration of OXP leads to low drug concentration at the tumor site, which is susceptible to systemic toxic side effects. In this study, a combined therapeutic strategy using folate-modified nanoliposomes co-delivered with rapamycin (Rapa) and OXP (abbreviated as FA@R/O Lps) is proposed for the treatment of colorectal cancer (CRC). Rapa and OXP can directly inhibit tumor cell proliferation and induce apoptosis. OXP induces ICD by triggering the release of danger signals, such as HMGB1, ATP, and calreticulin. FA@R/O Lps with a particle size of about 134.1±1.8 nm and a small dispersion were successfully prepared. This novel liposomal system can be used to target and increase drug accumulation in tumors. In-vivo experiments showed that FA@R/O Lps successfully inhibit CRC growth and liver metastasis, and simultaneously reduce off-target toxicity. In particular, FA@R/O Lps showed greater therapeutic effects than free Rapa/OXP and R/O Lps. Taken together, this study provides a novel combination of Rapa and OXP, and a nano-delivery system for enhanced anti-CRC efficacy. The results suggest that FA@R/O Lps could be a promising strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Shengyao Liu
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 250, Changgangdong Road, Guangzhou 510260, China
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Hanhui Huang
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
7
|
Ma X, Lin N, Hu K, Xu C, Yang Q, Feng Y, Liu P, Ding H, Xu M, Shi Q, Chen H, Xue F. An acid-activatable fluorouracil prodrug for colorectal cancer synergistic therapy. Acta Biomater 2024; 185:350-360. [PMID: 39013485 DOI: 10.1016/j.actbio.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
5-Fluorouracil has demonstrated certain efficiency in patients with colorectal cancer. However, significant side effects of use by injection are common. To address this issue defects, a reengineered 5'-deoxy-5-fluorocytidine (DFCR) based drug delivery system (POACa) is developed as a prominent tumor-selective nano-activator. Investigations demonstrate that the constructed nano-activator exhibits good biocompatibility and high therapeutic efficiency in mice with subcutaneous and orthotopic SW-480 colorectal tumors, as its activity is strictly dependent on the tumor-associated acid environment and thymidine phosphorylase. These strategies diminish the off-target toxicity and improve the specificity and sensitivity of human colorectal cancer cells to 5-Fu, obtaining potent efficiency by the combination of H2O2 mediated oxidative stress, calcium overload and 5-Fu-induced chemotherapy (the combination index is 0.11). Overall, the engineered nano-activator exhibits a high therapeutic index in vitro and in vivo. STATEMENT OF SIGNIFICANCE: In this study, we designed and prepared a pH-responsive polymer to synchronously deliver DFCR (5'-deoxy-5-fluorocytidine, a prodrug of 5-Fu), Ca2+ and H2O2. The constructed nano-activator was denoted as POACa. (1) To address the problem of premature leakage of cargo by physical embedding, our research modified the inactive prodrug DFCR through chemical bonding. (2) The activation of the prepared nano-activator was strictly dependent on the tumor-associated acid environment and thymidine phosphorylase, providing the drug delivery system with inherent safety. (3) A distinctly low combination index value (0.11) of CaO2 and DFCR indicated that POACa has a prominent tumor suppression effect by tumor calcium overload sensitized chemotherapy and H2O2 mediated cytotoxicity.
Collapse
Affiliation(s)
- Xiaoqian Ma
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Nuo Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Ke Hu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Chao Xu
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China. Clinical Medical Center for Digestive Diseases, Fujian Provincial Hospital, China
| | - Qing Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Yushuo Feng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Peifei Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Haizhen Ding
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Mengjiao Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Qianqian Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Hongmin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China.
| | - Fangqin Xue
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China. Clinical Medical Center for Digestive Diseases, Fujian Provincial Hospital, China.
| |
Collapse
|
8
|
Chen Y, Dai L, Shi K, Pan M, Yuan L, Qian Z. Cabazitaxel-Loaded Thermosensitive Hydrogel System for Suppressed Orthotopic Colorectal Cancer and Liver Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404800. [PMID: 38934894 PMCID: PMC11434046 DOI: 10.1002/advs.202404800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
The treatment of colorectal cancer is always a major challenge in the field of cancer research. The number of estimated new cases of colorectal cancer worldwide in 2020 is 1 148 515, and the estimated number of deaths is 576 858, revealing that mortality accounted for approximately half of the disease incidence. The development of new drugs and strategies for colorectal cancer treatment is urgently needed. Thermosensitive injectable hydrogel PDLLA-PEG-PDLLA (PLEL) loaded with cabazitaxel (CTX) is used to explore its anti-tumor effect on mice with orthotopic colorectal cancer. CTX/PLEL is characterized by a solution state at room temperature and a hydrogel state at physiologic temperature. The excipients MPEG-PCL and PDLLA-PEG-PDLLA have good biocompatibility and biodegradability. The simple material synthesis and preparation process renders this system cost-effective and more conducive to clinical transformation. An orthotopic colorectal cancer model is established by transplantation subcutaneous tumors onto the cecum of mice. According to the results of experiments in vivo, CTX/PLEL significantly inhibits orthotopic colorectal cancer and liver metastasis in mice. The results indicate that CTX/PLEL nanoparticle preparations have high security and excellent anti-tumor effects, and have great application potential in colorectal cancer therapy.
Collapse
Affiliation(s)
- Yu Chen
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Liqun Dai
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Kun Shi
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Meng Pan
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Liping Yuan
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| | - Zhiyong Qian
- Department of BiotherapyCancer Center and State Key Laboratory of Biotherapy West China HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
9
|
Luo Z, Jiang M, Cheng N, Zhao X, Liu H, Wang S, Lin Q, Huang J, Guo X, Liu X, Shan X, Lu Y, Shi Y, Luo L, You J. Remodeling the hepatic immune microenvironment and demolishing T cell traps to enhance immunotherapy efficacy in liver metastasis. J Control Release 2024; 373:890-904. [PMID: 39067794 DOI: 10.1016/j.jconrel.2024.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Immune checkpoint inhibitors (ICIs) exhibit compromised therapeutic efficacy in many patients with advanced cancers, particularly those with liver metastases. Much of this incapability can be ascribed as an irresponsiveness resulting from the "cold" hepatic tumor microenvironment that acts as T cell "traps" for which there currently lack countermeasures. We report a novel nanomedicine that converts the hepatic immune microenvironment to a "hot" phenotype by targeting hepatic macrophage-centric T cell elimination. Using the nanomedicine, composed of KIRA6 (an endothelium reticulum stress inhibitor), α-Tocopherol nanoemulsions, and anti-PD1 antibodies, we found its potency in murine models of orthotopic colorectal tumors and hepatic metastases, restoring immune responses and enhancing anti-tumor effects. A post-treatment scrutiny of the immune microenvironment landscape in the liver reveals repolarization of immunosuppressive hepatic macrophages, upregulation of Th1-like effector CD4+ T cells, and rejuvenation of dendritic cells along with CD8+ T cells. These findings suggest adaptations of liver-centric immune milieu modulation strategies to improve the efficacy of ICIs for a variety of "cold" tumors and their liver metastases.
Collapse
Affiliation(s)
- Zhenyu Luo
- School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Ningtao Cheng
- School of Medicine, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China; Hangzhou Yuhang BoYu Intelligent Health Innovation Lab, Hangzhou, Zhejiang 311121, China.
| | - Xiaoqi Zhao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Huihui Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Qing Lin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xinyu Shan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
10
|
Ouyang B, Shan C, Shen S, Dai X, Chen Q, Su X, Cao Y, Qin X, He Y, Wang S, Xu R, Hu R, Shi L, Lu T, Yang W, Peng S, Zhang J, Wang J, Li D, Pang Z. AI-powered omics-based drug pair discovery for pyroptosis therapy targeting triple-negative breast cancer. Nat Commun 2024; 15:7560. [PMID: 39215014 PMCID: PMC11364624 DOI: 10.1038/s41467-024-51980-9] [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: 08/27/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Due to low success rates and long cycles of traditional drug development, the clinical tendency is to apply omics techniques to reveal patient-level disease characteristics and individualized responses to treatment. However, the heterogeneous form of data and uneven distribution of targets make drug discovery and precision medicine a non-trivial task. This study takes pyroptosis therapy for triple-negative breast cancer (TNBC) as a paradigm and uses data mining of a large TNBC cohort and drug databases to establish a biofactor-regulated neural network for rapidly screening and optimizing compound pyroptosis drug pairs. Subsequently, biomimetic nanococrystals are prepared using the preferred combination of mitoxantrone and gambogic acid for rational drug delivery. The unique mechanism of obtained nanococrystals regulating pyroptosis genes through ribosomal stress and triggering pyroptosis cascade immune effects are revealed in TNBC models. In this work, a target omics-based intelligent compound drug discovery framework explores an innovative drug development paradigm, which repurposes existing drugs and enables precise treatment of refractory diseases.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, P. R. China
| | - Caihua Shan
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Shun Shen
- Pharmacy Department & Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, P. R. China
| | - Xinnan Dai
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Qingwang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xiaomin Su
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Yongbin Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xifeng Qin
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ying He
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Siyu Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruizhe Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Tun Lu
- School of Computer Science, Fudan University, Shanghai, 200438, P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Shaojun Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University); Zhuhai, Guangdong, 519000, P. R. China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| | - Dongsheng Li
- Microsoft Research Asia, Shanghai, 200232, P. R. China.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| |
Collapse
|
11
|
Cao C, Li Y, Shi F, Jiang S, Li Y, Yang L, Zhou X, Gao Y, Tang F, Li H, Han S, Yu Z, Zou Y, Guo J. Nano co-delivery of doxorubicin and plumbagin achieves synergistic chemotherapy of hepatocellular carcinoma. Int J Pharm 2024; 661:124424. [PMID: 38971510 DOI: 10.1016/j.ijpharm.2024.124424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug used for hepatocellular carcinoma (HCC) treatment, but its effectiveness can be dramatically dampened by cancer cell chemoresistance. Signal transducer and activator of transcription 3 (STAT3) is implicated with drug resistance in a range of cancers (e.g., HCC), and the STAT3 inhibition can reverse the resistance of cancer cells to chemotherapeutic drugs. In the present study, a combination regimen to improve the efficiency of DOX was provided via the STAT3 blockade using plumbagin (PLB). A poly(lactic-co-glycolic acid) decorated by polyethylene glycol and aminoethyl anisamide was produced in the present study with the hope of generating the nanoparticles for co-delivery of DOX and PLB. The resulting co-formulation suppressed the STAT3 activity and achieved the synergistic chemotherapy, which led to tumor inhibition in the mice with subcutaneous DOX-resistant HCC, without causing any toxicity. The present study reveals the synergism of DOX and PLB, and demonstrates a promising combinatorial approach for treating HCC.
Collapse
Affiliation(s)
- Chenyu Cao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yifei Li
- Department of Geriatrics, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Fangzhou Shi
- Department of Geriatrics, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Shanshan Jiang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xinyi Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yuqiong Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Feiyan Tang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Huan Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Shulan Han
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
12
|
Santos JAV, Silva D, Marques MPM, Batista de Carvalho LAE. Platinum-based chemotherapy: trends in organic nanodelivery systems. NANOSCALE 2024; 16:14640-14686. [PMID: 39037425 DOI: 10.1039/d4nr01483a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Despite the investment in platinum drugs research, cisplatin, carboplatin and oxaliplatin are still the only Pt-based compounds used as first line treatments for several cancers, with a few other compounds being approved for administration in some Asian countries. However, due to the severe and worldwide impact of oncological diseases, there is an urge for improved chemotherapeutic approaches. Furthermore, the pharmaceutical application of platinum complexes is hindered by their inherent toxicity and acquired resistance. Nanodelivery systems rose as a key strategy to overcome these challenges, with recognized versatility and ability towards improving the safety, bioavailability and efficacy of the available drugs. Among the known nanocarriers, organic systems have been widely applied, taking advantage of their potential as drug vehicles. Researchers have mainly focused on the development of lipidic and polymeric carriers, including supramolecular structures, with an overall improvement of encapsulated platinum complexes. Herein, an overview of recent trends and strategies is presented, with the main focus on the encapsulation of platinum compounds into organic nanocarriers, showcasing the evolution in the design and development of these promising systems. This comprehensive review highlights formulation methods as well as characterization procedures, providing insights that may be helpful for the development of novel platinum nanocarriers aiming at future pharmaceutical applications.
Collapse
Affiliation(s)
- João A V Santos
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Daniela Silva
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Maria Paula M Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Luís A E Batista de Carvalho
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
13
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
14
|
Ma Y, Zhu H, Jiang X, Zhou Z, Zhou Y, Tian Y, Tu L, Lu J, Niu Y, Du L, Si Z, Fang H, Liu H, Liu Y, Chen P. Synthesis and Biological Activity of 2-Chloro-8-methoxy-5-methyl-5 H-indolo [2,3- b] Quinoline for the Treatment of Colorectal Cancer by Modulating PI3K/AKT/mTOR Pathways. ACS OMEGA 2024; 9:30698-30707. [PMID: 39035959 PMCID: PMC11256334 DOI: 10.1021/acsomega.4c03101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024]
Abstract
Developing novel drugs from natural products has proven to be a very effective strategy. Neocryptolepine was isolated from Cryptolepis sanguinolenta, a traditional endemic African herb, which exerts a wide range of biological activities such as antimalaria, antibacterial, and antitumor. 2-Chloro-8-methoxy-5-methyl-5H-indolo [2,3-b] quinoline (compound 49) was synthesized, and its cytotoxicity was assessed on pancreatic cancer PANC-1 cells, colorectal cancer HCT116 cells, liver cancer SMMC-7721 cells, and gastric cancer AGS cells in vitro. The results of the in vitro assay showed that compound 49 exerted remarkable cytotoxicity on colorectal cancer HCT116 and Caco-2 cells. The cytotoxicity of compound 49 to colorectal cancer HCT116 cells was 17 times higher than that of neocryptolepine and to human normal intestinal epithelial HIEC cells was significantly reduced. Compound 49 exhibited significant cytotoxicity against the colorectal cancer HCT116 and Caco-2 cells, with IC50 of 0.35 and 0.54 μM, respectively. The mechanism of cytotoxicity of compound 49 to colorectal cancer HCT116 and Caco-2 cells was further investigated. The results showed that compound 49 could inhibit colony formation and cell migration. Moreover, compound 49 could arrest the cell cycle at the G2/M phase, promote the production of reactive oxygen species, reduce mitochondrial membrane potential, and induce apoptosis. The results of Western blot indicated that compound 49 showed cytotoxicity on HCT116 and Caco-2 cells by modulating the PI3K/AKT/mTOR signaling pathway. In conclusion, these results suggested that compound 49 may be a potentially promising lead compound for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yunhao Ma
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Hongmei Zhu
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Xinrong Jiang
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Zhongkun Zhou
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Yong Zhou
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Yanan Tian
- Faculty
of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao, Macau 999078, China
| | - Lixue Tu
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Juan Lu
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Yuqing Niu
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Liqian Du
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Zhenzhen Si
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Hong Fang
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Huanxiang Liu
- Faculty
of Applied Sciences, Macao Polytechnic University, R. de Luís Gonzaga Gomes, Macao, Macau 999078, China
| | - Yingqian Liu
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| | - Peng Chen
- School
of Pharmacy, Lanzhou University, No. 199 Donggang West Road, Lanzhou, Gansu 730000, China
| |
Collapse
|
15
|
Sangani PS, Yazdani S, Khalili-Tanha G, Ghorbani E, Al-Hayawi IS, Fiuji H, Khazaei M, Hassanian SM, Kiani M, Ghayour-Mobarhan M, Ferns GA, Nazari E, Avan A. The therapeutic impact of programmed death - 1 in the treatment of colorectal cancer. Pathol Res Pract 2024; 259:155345. [PMID: 38805760 DOI: 10.1016/j.prp.2024.155345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/27/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024]
Abstract
Colorectal cancer (CRC) is the most common type of newly diagnosed cancer. Metastatic spread and multifactorial chemoresistance have limited the benefits of current therapies. Hence, it is imperative to identify new therapeutic agents to increase treatment efficacy. One of CRC's most promising immunotherapeutic targets is programmed death-1 (PD-1), a cell surface receptor that regulates immune responses. In this paper, we provide an overview of the therapeutic impact of PD-1 in the treatment of CRC. Cancer cells can exploit the PD-1 pathway by upregulating its programmed death-ligand 1 (PD-L1) ligand to evade immune surveillance. The binding of PD-L1 to PD-1 inhibits T cell function, leading to tumor immune escape. PD-1 inhibitors, such as pembrolizumab and nivolumab, block the PD-1/PD-L1 interaction. Clinical trials evaluating PD-1 inhibitors in advanced CRC have shown promising results. In patients with microsatellite instability-high (MSI-H) or mismatch repair-deficient (dMMR) tumors characterized by high mutation rates and increased immunogenicity, PD-1 blockade has demonstrated remarkable efficacy. As a result, pembrolizumab and nivolumab have received accelerated approval by regulatory authorities for the treatment of MSI-H/dMMR metastatic CRC. Additionally, combination approaches, such as combining PD-1 inhibitors with other immunotherapies or targeted agents, are being explored. Despite the success of PD-1 inhibitors in CRC, challenges still exist. Immune-related adverse events can occur and require close monitoring. In conclusion, PD-1 inhibitors have demonstrated significant therapeutic impact, particularly in patients with MSI-H/dMMR tumors.
Collapse
Affiliation(s)
- Pooria Salehi Sangani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soroush Yazdani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hamid Fiuji
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - MohammadAli Kiani
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Elham Nazari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq; School of Mechanical, Medical and Process Engineering, Science and Engineering Faculty, Queensland University of Technology, 2 George St, Brisbane City, QLD 4000, Australia; Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
16
|
Shao W, Yang Y, Shen W, Ren L, WenwenWang, Zhu P. Hyaluronic acid-conjugated methotrexate and 5-fluorouracil for targeted drug delivery. Int J Biol Macromol 2024; 273:132671. [PMID: 38823747 DOI: 10.1016/j.ijbiomac.2024.132671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/10/2024] [Accepted: 05/24/2024] [Indexed: 06/03/2024]
Abstract
The delivery of chemotherapeutical drugs via nanomaterials has become a focus of pharmaceutical research over several decades due to improved drug delivery to cancer cells, decreased side effects on normal tissues, and increased therapeutic efficacy. Herein, a novel hyaluronic acid-conjugated methotrexate and 5-fluorouracil nanodrug system has been developed to address the critical limitations associated with the high toxicity and side effects of methotrexate and 5-fluorouracil. Furthermore, this nanodrug system enhances the targeting capacity of drug molecules and facilitates the potential integration of multimodal drug therapies. Concomitantly, the synergistic effects of MTX with 5-fluorouracil have been shown to improve the therapeutic index of MTX while attenuating the associated toxicities of MTX. The structure and micromorphology of the novel nanodrug can be confirmed by 1HNMR, FT-IR, UV-Vis, DLS, TEM, and AFM. Due to the ability of HA to bind to CD44 receptors activated on the surface of cancer cells and its enhanced permeability and retention (EPR) effect, the novel nanodrug we designed and synthesized can effectively target cancer cells. Cell counting Kit-8 (CCK8), flow cytometry, and live-dead staining assays in vitro showed that this nanodrug system had high targeting and antitumor activity against CD44 receptors. By using drugs to act on patient-derived colorectal, liver, and breast cancer organoids, the anticancer effect of the nanodrug was identified and verified. These results showed that the nanodrug system developed in this study may have great potential as a targeted therapy for cancer.
Collapse
Affiliation(s)
- Wanfei Shao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, PR China
| | - Yanfang Yang
- Guangxi Zhuoqiang Technology Co. LTD, Nanning, Guangxi 530000, China
| | - Weidong Shen
- Department of Gastroenterology, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, China.
| | - Lei Ren
- Nanjing University of Chinese Medicine Affiliated Jiangyin Traditional Chinese Medicine Hospital, Jiangyin 214400, Jiangsu, China
| | - WenwenWang
- Nanjing University of Chinese Medicine Affiliated Jiangyin Traditional Chinese Medicine Hospital, Jiangyin 214400, Jiangsu, China
| | - Peizhi Zhu
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225002, PR China.
| |
Collapse
|
17
|
Ding J, Lu Y, Zhao X, Long S, Du J, Sun W, Fan J, Peng X. Activating Iterative Revolutions of the Cancer-Immunity Cycle in Hypoxic Tumors with a Smart Nano-Regulator. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400196. [PMID: 38734875 DOI: 10.1002/adma.202400196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The activation of sequential events in the cancer-immunity cycle (CIC) is crucial for achieving effective antitumor immunity. However, formidable challenges, such as innate and adaptive immune resistance, along with the off-target adverse effects of nonselective immunomodulators, persist. In this study, a tumor-selective nano-regulator named PNBJQ has been presented, focusing on targeting two nonredundant immune nodes: inducing immunogenic cancer cell death and abrogating immune resistance to fully activate endogenous tumor immunity. PNBJQ is obtained by encapsulating the immunomodulating agent JQ1 within a self-assembling system formed by linking a Type-I photosensitizer to polyethylene glycol through a hypoxia-sensitive azo bond. Benefiting from the Type-I photosensitive mechanism, PNBJQ triggers the immunogenic cell death of hypoxic tumors under near-infrared (NIR) light irradiation. This process resolves innate immune resistance by stimulating sufficient cytotoxic T-lymphocytes. Simultaneously, PNBJQ smartly responds to the hypoxic tumor microenvironment for precise drug delivery, adeptly addressing adaptive immune resistance by using JQ1 to downregulate programmed death ligand 1 (PD-L1) and sustaining the response of cytotoxic T lymphocytes. The activatable synergic photoimmunotherapy promotes an immune-promoting tumor microenvironment by activating an iterative revolution of the CIC, which remarkably eradicates established hypoxic tumors and suppresses distal lesions under low light dose irradiation.
Collapse
Affiliation(s)
- Junying Ding
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
| | - Yang Lu
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
| | - Xueze Zhao
- Department of Chemistry, The University of Hong Kong, Hong Kong, SAR, 999077, China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo, 315016, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo, 315016, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo, 315016, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, China
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
18
|
Liu J, Lin C, Wu M, Wang Y, Chen S, Yang T, Xie C, Kong Y, Wu W, Wang J, Ma X, Teng C. Co-delivery of indomethacin and uricase as a new strategy for inflammatory diseases associated with high uric acid. Drug Deliv Transl Res 2024; 14:1820-1838. [PMID: 38127247 DOI: 10.1007/s13346-023-01487-5] [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] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
Uric acid is the final metabolite in humans. High level of uric acid chronically induces urate deposition, aggravates kidney damage, and concomitantly causes an increase in inflammatory factors. Alleviating acute inflammation and decreasing uric acid levels are the key points in the treatment of inflammatory diseases associated with high uric acid. However, a drug delivery system that combines anti-inflammatory and uric acid reduction functions at the same time remains a challenge to be settled. Here, we designed a nanocrystal-based co-delivery platform, IND Nplex, characterized by loading of indomethacin (IND) and uricase. Compared with free IND or uricase, IND Nplex possessed a better anti-inflammatory effect by restraining the release of inflammation-related factors in vitro. In addition, pharmacokinetic and biodistribution studies revealed that IND Nplex significantly prolonged the retention time in vivo and was more concentrated in the kidney. In acute gouty arthritis model rats, IND Nplex markedly relieved ankle joint swelling and mitigated synovial inflammation. In acute kidney injury model rats, IND Nplex indicated better biocompatibility and significant amelioration of renal fibrosis. Moreover, IND Nplex showed the effect of anti-inflammatory and improved renal function via determination of inflammatory factors and biochemical markers in the serum and kidney. In conclusion, these results indicate that IND Nplex exerts anti-inflammatory activity and uric acid-lowering effect and could become a promising candidate for the treatment of uric acid-related diseases.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Chenshi Lin
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Man Wu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Yingjie Wang
- Center for Translational Imaging, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, USA
| | - Shenyu Chen
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Taiwang Yang
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Chenlu Xie
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Yue Kong
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Wenliang Wu
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Jiaping Wang
- Department of Pharmacy, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332000, China
| | - Xiaonan Ma
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 210009, China.
| | - Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
19
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
20
|
Yang G, Cao Y, Yang X, Cui T, Tan NZV, Lim YK, Fu Y, Cao X, Bhandari A, Enikeev M, Efetov S, Balaban V, He M. Advancements in nanomedicine: Precision delivery strategies for male pelvic malignancies - Spotlight on prostate and colorectal cancer. Exp Mol Pathol 2024; 137:104904. [PMID: 38788248 DOI: 10.1016/j.yexmp.2024.104904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Pelvic malignancies consistently pose significant global health challenges, adversely affecting the well-being of the male population. It is anticipated that clinicians will continue to confront these cancers in their practice. Nanomedicine offers promising strategies that revolutionize the treatment of male pelvic malignancies by providing precise delivery methods that aim to improve the efficacy of therapeutic outcomes while minimizing side effects. Nanoparticles are designed to encapsulate therapeutic agents and selectively target cancer cells. They can also be loaded with theragnostic agents, enabling multifunctional capabilities. OBJECTIVE This review aims to summarize the latest nanomedicine research into clinical applications, focusing on nanotechnology-based treatment strategies for male pelvic malignancies, encompassing chemotherapy, radiotherapy, immunotherapy, and other cutting-edge therapies. The review is structured to assist physicians, particularly those with limited knowledge of biochemistry and bioengineering, in comprehending the functionalities and applications of nanomaterials. METHODS Multiple databases, including PubMed, the National Library of Medicine, and Embase, were utilized to locate and review recently published articles on advancements in nano-drug delivery for prostate and colorectal cancers. CONCLUSION Nanomedicine possesses considerable potential in improving therapeutic outcomes and reducing adverse effects for male pelvic malignancies. Through precision delivery methods, this emerging field presents innovative treatment modalities to address these challenging diseases. Nevertheless, the majority of current studies are in the preclinical phase, with a lack of sufficient evidence to fully understand the precise mechanisms of action, absence of comprehensive pharmacotoxicity profiles, and uncertainty surrounding long-term consequences.
Collapse
Affiliation(s)
- Guodong Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Te Cui
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yuen Kai Lim
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Fu
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinren Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aanchal Bhandari
- HBT Medical College and Dr. R N Cooper Municipal General Hospital, Mumbai, India
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Sergey Efetov
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir Balaban
- Clinic of Coloproctology and Minimally Invasive Surgery, Sechenov University, Moscow, Russia
| | - Mingze He
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| |
Collapse
|
21
|
Hong Z, Li Y, Chen M, Chen X, Deng X, Wu Y, Wang C, Qiu C. Protosappanin B enhances the chemosensitivity of 5-fluorouracil in colon adenocarcinoma by regulating the LINC00612/microRNA-590-3p/Golgi phosphoprotein 3 axis. Discov Oncol 2024; 15:193. [PMID: 38806777 PMCID: PMC11133243 DOI: 10.1007/s12672-024-01036-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND 5-fluorouracil (5-FU) is conventionally used in chemotherapy for colon adenocarcinomas. Acquired resistance of 5-FU remains a clinical challenge in colon cancer, and efforts to develop targeted agents to reduce resistance have not yielded success. Protosappanin B (PSB), the main component of Lignum Sappan extract, is known to exhibit anti-tumor effects. However, whether and how PSB could improve 5-FU resistance in colon cancer have not yet been established. In this study, we aimed to explore the effects and underlying mechanisms of PSB in 5-FU-induced chemoresistance in colon adenocarcinoma. METHODS Forty-seven paired colon cancer tissue samples from patients who received 5-FU chemotherapy were collected as clinical samples. Two 5-FU resistant colon cancer cell lines were established for in vitro experiments. Reverse transcription-quantitative PCR (RT-qPCR) was performed to determine the mRNA and microRNA (miRNA) expression levels in colon adenocarcinoma tissues and cell lines. Cell Counting Kit-8 (CCK-8) and flow cytometry assays were performed to evaluate cell proliferation and apoptosis, respectively. RESULTS LINC00612 was highly expressed in colon adenocarcinoma samples and 5-FU resistant colon cancer cells. LINC00612 knockdown enhances 5-FU chemosensitivity in 5-FU resistant cells. Notably, PSB treatment attenuated LINC00612 expression in 5-FU resistant colon adenocarcinoma cells. Moreover, PSB treatment reversed the increase in LINC00612-induced 5-FU resistance. Mechanistically, LINC00612 specifically bound to miR-590-3p, which promoted 5-FU resistance in colon adenocarcinoma cells and attenuated the inhibitory effect of LINC00612 on GOLPH3 expression. CONCLUSION PSB attenuates 5-FU chemoresistance in colon adenocarcinoma by regulating the LINC00612/miRNA-590-3p/GOLPH3 axis.
Collapse
Affiliation(s)
- Zhongshi Hong
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Yachen Li
- Medical Department, The Second Affiliated Hospital of Fujian Medical University, No.34 Zhongshan North Road, Quanzhou, 362000, Fujian, China
| | - Mingliang Chen
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Xiaojing Chen
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Xian Deng
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Yuze Wu
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China
| | - Chunxiao Wang
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China.
| | - Chengzhi Qiu
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, No.34, Zhongshan North Road, Quanzhou, Fujian, 362000, China.
| |
Collapse
|
22
|
Lang X, Wang X, Han M, Guo Y. Nanoparticle-Mediated Synergistic Chemoimmunotherapy for Cancer Treatment. Int J Nanomedicine 2024; 19:4533-4568. [PMID: 38799699 PMCID: PMC11127654 DOI: 10.2147/ijn.s455213] [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: 12/16/2023] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Until now, there has been a lack of effective strategies for cancer treatment. Immunotherapy has high potential in treating several cancers but its efficacy is limited as a monotherapy. Chemoimmunotherapy (CIT) holds promise to be widely used in cancer treatment. Therefore, identifying their involvement and potential synergy in CIT approaches is decisive. Nano-based drug delivery systems (NDDSs) are ideal delivery systems because they can simultaneously target immune cells and cancer cells, promoting drug accumulation, and reducing the toxicity of the drug. In this review, we first introduce five current immunotherapies, including immune checkpoint blocking (ICB), adoptive cell transfer therapy (ACT), cancer vaccines, oncolytic virus therapy (OVT) and cytokine therapy. Subsequently, the immunomodulatory effects of chemotherapy by inducing immunogenic cell death (ICD), promoting tumor killer cell infiltration, down-regulating immunosuppressive cells, and inhibiting immune checkpoints have been described. Finally, the NDDSs-mediated collaborative drug delivery systems have been introduced in detail, and the development of NDDSs-mediated CIT nanoparticles has been prospected.
Collapse
Affiliation(s)
- Xiaoxue Lang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, People’s Republic of China
| |
Collapse
|
23
|
Chen J, Chen K, Xue S, Cheng X, Qi Y, Wang H, Li W, Cheng G, Xiong Y, Mu C, Gu M. Integration of caveolin-mediated cytosolic delivery and enzyme-responsive releasing of squalenoyl nanoparticles enhance the anti-cancer efficacy of chidamide in pancreatic cancer. Int J Pharm 2024; 655:124072. [PMID: 38561133 DOI: 10.1016/j.ijpharm.2024.124072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/29/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
We explored the potential of overcoming the dense interstitial barrier in pancreatic cancer treatment by enhancing the uptake of hydrophilic chemotherapeutic drugs. In this study, we synthesized the squalenoyl-chidamide prodrug (SQ-CHI), linking lipophilic squalene (SQ) with the hydrophilic antitumor drug chidamide (CHI) through a trypsin-responsive bond. Self-assembled nanoparticles with sigma receptor-bound aminoethyl anisamide (AEAA) modification, forming AEAA-PEG-SQ-CHI NPs (A-C NPs, size 116.6 ± 0.4 nm), and reference nanoparticles without AEAA modification, forming mPEG-SQ-CHI NPs (M-C NPs, size 88.3 ± 0.3 nm), were prepared. A-C NPs exhibited significantly higher in vitro CHI release (74.7 %) in 0.5 % trypsin medium compared to release (20.2 %) in medium without trypsin. In vitro cell uptake assays revealed 3.6 and 2.3times higher permeation of A-C NPs into tumorspheres of PSN-1/HPSC or CFPAC-1/HPSC, respectively, compared to M-C NPs. Following intraperitoneal administration to subcutaneous tumor-bearing nude mice, the A-C NPs group demonstrated significant anti-pancreatic cancer efficacy, inducing cancer cell apoptosis and inhibiting proliferation in vivo. Mechanistic studies revealed that AEAA surface modification on nanoparticles promoted intracellular uptake through caveolin-mediated endocytosis. This nanoparticle system presents a novel therapeutic approach for pancreatic cancer treatment, offering a delivery strategy to enhance efficacy through improved tumor permeation, trypsin-responsive drug release, and specific cell surface receptor-mediated intracellular uptake.
Collapse
Affiliation(s)
- Junyan Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Kaidi Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Shuai Xue
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou 313000, Zhejiang, China.
| | - Yuwei Qi
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Hangjie Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Wei Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Guilin Cheng
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Yang Xiong
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Chaofeng Mu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Mancang Gu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| |
Collapse
|
24
|
Wang Y, Wang L, Li T, Ouyang M, Xiong H, Zhou D. Bimetallic nanoparticles as cascade sensitizing amplifiers for low-dose and robust cancer radio-immunotherapy. Acta Pharm Sin B 2024; 14:1787-1800. [PMID: 38572091 PMCID: PMC10985033 DOI: 10.1016/j.apsb.2023.11.028] [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: 08/31/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 04/05/2024] Open
Abstract
Radiotherapy (RT) is one of the most feasible and routinely used therapeutic modalities for treating malignant tumors. In particular, immune responses triggered by RT, known as radio-immunotherapy, can partially inhibit the growth of distantly spreading tumors and recurrent tumors. However, the safety and efficacy of radio-immunotherapy is impeded by the radio-resistance and poor immunogenicity of tumor. Herein, we report oxaliplatin (IV)-iron bimetallic nanoparticles (OXA/Fe NPs) as cascade sensitizing amplifiers for low-dose and robust radio-immunotherapy. The OXA/Fe NPs exhibit tumor-specific accumulation and activation of OXA (II) and Fe2+ in response to the reductive and acidic microenvironment within tumor cells. The cascade reactions of the released metallic drugs can sensitize RT by inducing DNA damage, increasing ROS and O2 levels, and amplifying the immunogenic cell death (ICD) effect after RT to facilitate potent immune activation. As a result, OXA/Fe NPs-based low-dose RT triggered a robust immune response and inhibited the distant and metastatic tumors effectively by a strong abscopal effect. Moreover, a long-term immunological memory effect to protect mice from tumor rechallenging is observed. Overall, the bimetallic NPs-based cascade sensitizing amplifier system offers an efficient radio-immunotherapy regimen that addresses the key challenges.
Collapse
Affiliation(s)
- Yupeng Wang
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lina Wang
- Testing and Analysis Center, Hebei Normal University, Shijiazhuang 050024, China
| | - Tao Li
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Min Ouyang
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hejian Xiong
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Dongfang Zhou
- Department of Ultrasonic Diagnosis, Zhujiang Hospital, Key Laboratory of Mental Health of the Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
25
|
Cao W, Zhang X, Li R, Li Z, Lu A, Yu F, Sun L, Wang J, Wang Z, He H. Lipid core-shell nanoparticles co-deliver FOLFOX regimen and siPD-L1 for synergistic targeted cancer treatment. J Control Release 2024; 368:52-65. [PMID: 38368946 DOI: 10.1016/j.jconrel.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
FOLFOX regimen, composed of folinic acid, 5-fluorouracil (5-FU) and oxaliplatin (OXP), has been used as clinical standard therapeutic regimen in treatments of colorectal cancer (CRC) and esophageal squamous cell carcinoma (ESCC). To further improve its therapeutic outcomes, FOLFOX was combined with anti-PD-1 antibody to form an advanced chemo-immune combination strategy, which has been proven more efficient in controlling cancer progression and prolonging patients' survival in various clinical trials. However, bad tumor accumulation, relative high toxicity, numerous treatment cycles with high fees and low compliance as well as drug resistance seriously limit the prognosis of FOLFOX regimen. The "all-in-one" formulations, which could precisely delivery multidrug regimen into tumor sites and cells, showed a promising application prospect for targeted drug delivery as well as reducing side effects. However, the design and preparation of the "all-in-one" formulation with high drug encapsulation efficiencies for all drugs was still challenging. Herein, a lipid core-shell nanoparticle codelivery platform was designed for simultaneous encapsulation of variant FOLFOX composed of miriplatin (MiPt), 5-Fluoro-2'-deoxyuridine 5'-monophosphate (FdUMP), calcium folinate (CF) and PD-L1 siRNA (siPD-L1) with high efficiencies, and their synergistic anti-tumor mechanisms were studied, respectively. MiPt, a precursor of OXP, was validated capable of inducing efficient immunogenic cell death (ICD) in this work. Additionally, ICD-mediated release of damage associated molecular patterns functionalized synergistically with PD-L1 silence by siPD-L1 to overcome chemoresistance, reverse suppressive tumor microenvironment and recruit more CD8+ T cells. FdUMP, as the intracellular active form of 5-FU, could induce large amounts of reactive oxygen species to enhance the ICD. CF worked as the sensitizer of FdUMP. The enhanced long-term anti-tumor effect of the prepared "all-in-one" formulation compared to free drug regimen and other controls, was verified in heterotopic CRC mice models and ESCC mice models, providing new thoughts for researchers and showing a promising prospect of translation into clinical applications.
Collapse
Affiliation(s)
- Weiran Cao
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xue Zhang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Rui Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zijie Li
- Department of Immuno-oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fei Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Lu Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Zhiyu Wang
- Department of Immuno-oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, International Joint Laboratory of Ocular Diseases, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
26
|
Zhang JJ, Xu QJ, Zhang Y, Zhou Q, Lv R, Chen Z, He W. Recent advances in nanocarriers for clinical platinum(II) anticancer drugs. Coord Chem Rev 2024; 505:215676. [DOI: 10.1016/j.ccr.2024.215676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
27
|
Yu Z, Huang L, Guo J. Anti-stromal nanotherapeutics for hepatocellular carcinoma. J Control Release 2024; 367:500-514. [PMID: 38278367 DOI: 10.1016/j.jconrel.2024.01.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Hepatocellular carcinoma (HCC), the most commonly diagnosed primary liver cancer, has become a leading cause of cancer-related death worldwide. Accumulating evidence confirms that the stromal constituents within the tumor microenvironment (TME) exacerbate HCC malignancy and set the barriers to current anti-HCC treatments. Recent developments of nano drug delivery system (NDDS) have facilitated the application of stroma-targeting therapeutics, disrupting the stromal TME in HCC. This review discusses the stromal activities in HCC development and therapy resistance. In addition, it addresses the delivery challenges of NDDS for stroma-targeting therapeutics (termed anti-stromal nanotherapeutics in this review), and provides recent advances in anti-stromal nanotherapeutics for safe, effective, and specific HCC therapy.
Collapse
Affiliation(s)
- Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
28
|
Mohammadian S, Avan A, Khazaei M, Maghami P. The advancing of polymeric core-shell ZnO nanocomposites containing 5-fluorouracil for improving anticancer activity in colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:899-911. [PMID: 37530786 DOI: 10.1007/s00210-023-02643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
The study investigated the use of 5-fluorouracil-loaded ZnO nanocomposites (5-FU/Gd-ZnO NCs) as a potential treatment for cancer. 5-FU is a commonly used drug for cancer treatment but has undesirable side effects. The materials were characterized using various techniques, including PXRD, FTIR, FESEM, TEM, DLS, £-potential, and AFM. The data showed that the nanocomposites had a plate-like agglomeration with particle diameters ranging from 317.6 to 120.1 nm. The IC50 value of 5-FU-ZnO, which inhibits cell growth, was found to be 1.85 ppm. The effects of 5-FU-ZnO on inflammatory markers were also examined. While 5-FU increased the levels of TNF-a and IL-1b, the nanocomposites were able to reduce these levels. Additionally, the 5-FU/Gd-ZnO-NCs group showed an increase in thiol levels and a decrease in catalase and superoxide dismutase levels. Flow cytometry results showed that 5-FU, ZnO-NCs, and 5-FU/Gd-ZnO-NCs did not have any additive or synergistic effects on the suppression or eradication of cancer cells. In vivo, experiments showed that the 5-FU/Gd-ZnO NCs had similar necrotic characteristics and reduced fibrosis and collagen deposition compared to the free medication. The nanocomposites also exhibited higher antioxidative activity and lower inflammatory responses compared to the 5-FU group. It was shown that 5-FU/Gd-ZnO-NCs successfully inhibit cell proliferation. The in vivo results were comparable to those obtained with free 5-FU, suggesting the potential of these nanocomposites as therapeutic agents.
Collapse
Affiliation(s)
- Samaneh Mohammadian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Maghami
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
29
|
Chen J, Hu S, Sun M, Shi J, Zhang H, Yu H, Yang Z. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur J Pharm Sci 2024; 193:106688. [PMID: 38171420 DOI: 10.1016/j.ejps.2023.106688] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/23/2023] [Accepted: 12/31/2023] [Indexed: 01/05/2024]
Abstract
The limitations of conventional cancer treatment are driving the emergence and development of nanomedicines. Research in liposomal nanomedicine for cancer therapy is rapidly increasing, opening up new horizons for cancer treatment. Liposomal nanomedicine, which focuses on targeted drug delivery to improve the therapeutic effect of cancer while reducing damage to normal tissues and cells, has great potential in the field of cancer therapy. This review aims to clarify the advantages of liposomal delivery systems in cancer therapy. We describe the recent understanding of spatiotemporal fate of liposomes in the organism after different routes of drug administration. Meanwhile, various types of liposome-based drug delivery systems that exert their respective advantages in cancer therapy while reducing side effects were discussed. Moreover, the combination of liposomal agents with other therapies (such as photodynamic therapy and photothermal therapy) has demonstrated enhanced tumor-targeting efficiency and therapeutic efficacy. Finally, the opportunities and challenges faced by the field of liposome nanoformulations for entering the clinical treatment of cancer are highlighted.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life Sciences, Jilin University, Changchun, China
| | - Siyuan Hu
- School of Life Sciences, Jilin University, Changchun, China
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun, China
| | - Jianan Shi
- School of Life Sciences, Jilin University, Changchun, China
| | - Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Hongmei Yu
- China-Japan Union Hospital, Jilin University, Changchun, China.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
30
|
Zhou X, Li D, Xia S, Ma X, Li R, Mu Y, Liu Z, Zhang L, Zhou Q, Zhuo W, Ding K, Lin A, Liu W, Liu X, Zhou T. RNA-based modulation of macrophage-mediated efferocytosis potentiates antitumor immunity in colorectal cancer. J Control Release 2024; 366:128-141. [PMID: 38104775 DOI: 10.1016/j.jconrel.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Tumor-associated macrophages play pivotal roles in tumor progression and metastasis. Macrophage-mediated clearance of apoptotic cells (efferocytosis) supports inflammation resolution, contributing to immune evasion in colorectal cancers. To reverse this immunosuppressive process, we propose a readily translatable RNA therapy to selectively inhibit macrophage-mediated efferocytosis in tumor microenvironment. A clinically approved lipid nanoparticle platform (LNP) is employed to encapsulate siRNA for the phagocytic receptor MerTK (siMerTK), enabling selective MerTK inhibition in the diseased organ. Decreased MerTK expression in tumor-associated macrophages results in apoptotic cell accumulation and immune activation in tumor microenvironment, leading to suppressed tumor growth and better survival in both liver and peritoneal metastasis models of colorectal cancers. siMerTK delivery combined with PD-1 blockade further produces enhanced antimetastatic efficacy with reactivated intratumoral immune milieu. Collectively, LNP-based siMerTK delivery combined with immune checkpoint therapy may present a feasible modality for metastatic colorectal cancer therapy.
Collapse
Affiliation(s)
- Xuefei Zhou
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Dezhi Li
- Department of Oncology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Shenglong Xia
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Xixi Ma
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China
| | - Rong Li
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yongli Mu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zimo Liu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lu Zhang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Quan Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Zhuo
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Liu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiangrui Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| | - Tianhua Zhou
- Cancer Center, Zhejiang University, Hangzhou 310058, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China.
| |
Collapse
|
31
|
Zhou BY, Liu H, Pu YY, Wang LF, Sun YK, Yin HH, Lu D, Ye X, Hu XY, Wang X, Han H, Xia HS, Zhao CK, Xu HX. Quantitative analysis of pre-treatment dynamic contrast-enhanced ultrasound for assessing the response of colorectal liver metastases to chemotherapy plus targeted therapy: a dual-institutional study. Abdom Radiol (NY) 2024; 49:414-424. [PMID: 37853236 DOI: 10.1007/s00261-023-04055-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVES To investigate the clinical value of pre-treatment quantitative contrast-enhanced ultrasound (CEUS) in assessing the response of colorectal liver metastases (CRLM) to chemotherapy plus targeted therapy. METHODS This study retrospectively enrolled 50 CRLM patients from the Zhongshan Hospital, Fudan University as the training cohort and 14 patients from Shanghai Tenth People's Hospital as the testing cohort. Patients underwent the CEUS examination before receiving chemotherapy (CAPOX, FOLFOX, FOLFIRI, or FOLFOXIRI) plus targeted therapy (Bevacizumab or Cetuximab). The therapy response was determined according to Response Evaluation Criteria in Solid Tumors version 1.1 based on pre-treatment CT and 3-month follow-up CT after therapy. Dynamic analysis was performed by VueBox® software. Time-intensity curves with quantitative perfusion parameters were obtained. In the training cohort, univariable and multivariable logistic regression analyses were used to develop the predictive model of therapy response. The predictive performance of the developed model was validated in the testing cohort. RESULTS After the logistic regression analyses, the peak enhancement (PE) (odds ratio = 1.640; 95% confidence intervals [CI] 1.022-2.633) and time to peak (TTP) (odds ratio = 0.495; 95% CI 0.246-0.996) were determined as independent predictive factors. PE and TTP generated from VueBox® were not affected by ultrasound instruments and contrast agent dosage in therapy response evaluation (P > 0.05). The logistic regression model achieved satisfactory prediction performance (area under the curve: 0.923 in the training cohort and 0.854 in the testing cohort). CONCLUSION CEUS with dynamic quantitative perfusion analysis, which presents high consistency, has potential practical value in predicting the response of CRLM to chemotherapy plus targeted therapy.
Collapse
Affiliation(s)
- Bo-Yang Zhou
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Hui Liu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
| | - Yin-Ying Pu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Li-Fan Wang
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Yi-Kang Sun
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Hao-Hao Yin
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Dan Lu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Xing Ye
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Xin-Yuan Hu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Xi Wang
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Hong Han
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
| | - Han-Sheng Xia
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China.
| | - Chong-Ke Zhao
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China.
| | - Hui-Xiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| |
Collapse
|
32
|
Wang H, Xiang D, Lu X, Fang L, Cui C, Shi Q, Yang X. Human serum albumin-bound paclitaxel nanoparticle inhibits cervical carcinoma cell proliferation and oxidative damage through CYP3A4 and CYP2C8. Heliyon 2024; 10:e24460. [PMID: 38347900 PMCID: PMC10859773 DOI: 10.1016/j.heliyon.2024.e24460] [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: 10/17/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 02/15/2024] Open
Abstract
Background Cervical cancer (CC) is currently the most common malignant tumour in the female reproductive tract, and paclitaxel (PTX) is a commonly used chemotherapeutic agent, but tumour cell resistance will seriously affect the therapeutic efficacy of PTX. Nanoparticle human serum albumin-bound paclitaxel (Nano-HSA-PTX) is a novel drug delivery modality that may have superior effects to PTX alone. Objective To clarify the effect of Nano-HSA-PTX on cervical carcinoma (CC) cells and the underlying mechanisms. Methods After the preparation of Nano-HSA-PTX, its morphology was observed by electron transmission microscope (TEM), and its entrapment efficiency (EE%) and drug loading rate (DL%) were detected. Nano-HSA-PTX was compared with conventional PTX for drug metabolism. Additionally, CC HeLa and SiHa cells were purchased and divided into three groups to treat with Nano-HSA-PTX, PTX and normal saline, respectively. MTT, cell cloning, Transwell and cell scratch assays were carried out to determine cell proliferation, invasion and migration, flow cytometry and Western blotting were performed to detect apoptosis rate and apoptosis-related protein expression, and PCR was conducted to quantify oxidative damage indicators. Further, CYP3A4 and CYP2C8 expression patterns in CC cells (HeLa and SiHa) and human normal cervical epithelia (End1/E6E7) and the changes of their levels under the intervention of Nano-HSA-PTX were measured. Subsequently, C57BL/6mice were purchased for subcutaneous tumorigenesis experiment to observe the impact of Nano-HSA-PTX on tumor growth. Results Under TEM, Nano-HSA-PTX was complete and arranged compactly, with a stable structure and markedly higher EE% and DL% than PTX (P < 0.05). Under Nano-HSA-PTX intervention, the proliferation, invasion, migration and oxidative damage of HeLa and SiHa were significantly decreased compared with the control and PTX groups, while the apoptosis was increased (P < 0.05). Besides, elevated CYP3A4 and CYP2C8 levels were observed in CC cells, which were inhibited by Nano-HSA-PTX and PTX (P < 0.05). Finally, tumorigenesis experiments in nude mice revealed that Nano-HSA-PTX could inhibit tumor growth. Conclusion Compared with PTX, Nano-HSA-PTX has a superior effect of inhibiting CC activity. And this mechanism of action was carried out by inhibiting the expression of CYP3A4 and CYP2C8.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), 214015, Wuxi, Jiangsu, China
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Dajun Xiang
- Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), 214015, Wuxi, Jiangsu, China
| | - Xianyi Lu
- Department of Obstetrics and Gynecology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), 214015, Wuxi, Jiangsu, China
| | - Ling Fang
- Department of Dermatology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), Wuxi, Jiangsu, 214105, China
| | - Chengjun Cui
- Department of Pathology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), Wuxi, Jiangsu, 214105, China
| | - Qifeng Shi
- Department of Pathology, Wuxi Xishan People's Hospital, (Wuxi Branch of Zhongda Hospital Southeast University), Wuxi, Jiangsu, 214105, China
| | - Xiaojun Yang
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
33
|
Zu M, Ma Y, Zhang J, Sun J, Shahbazi MA, Pan G, Reis RL, Kundu SC, Liu J, Xiao B. An Oral Nanomedicine Elicits In Situ Vaccination Effect against Colorectal Cancer. ACS NANO 2024; 18:3651-3668. [PMID: 38241481 DOI: 10.1021/acsnano.3c11436] [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: 01/21/2024]
Abstract
Oral administration is the most preferred approach for treating colon diseases, and in situ vaccination has emerged as a promising cancer therapeutic strategy. However, the lack of effective drug delivery platforms hampered the application of in situ vaccination strategy in oral treatment of colorectal cancer (CRC). Here, we construct an oral core-shell nanomedicine by preparing a silk fibroin-based dual sonosensitizer (chlorin e6, Ce6)- and immunoadjuvant (imiquimod, R837)-loaded nanoparticle as the core, with its surface coated with plant-extracted lipids and pluronic F127 (p127). The resultant nanomedicines (Ce6/R837@Lp127NPs) maintain stability during their passage through the gastrointestinal tract and exert improved locomotor activities under ultrasound irradiation, achieving efficient colonic mucus infiltration and specific tumor penetration. Thereafter, Ce6/R837@Lp127NPs induce immunogenic death of colorectal tumor cells by sonodynamic treatment, and the generated neoantigens in the presence of R837 serve as a potent in situ vaccine. By integrating with immune checkpoint blockades, the combined treatment modality inhibits orthotopic tumors, eradicates distant tumors, and modulates intestinal microbiota. As the first oral in situ vaccination, this work spotlights a robust oral nanoplatform for producing a personalized vaccine against CRC.
Collapse
Affiliation(s)
- Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Ya Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Jun Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7LD, U.K
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco 4805-017, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga 4800-058, Guimarães, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco 4805-017, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga 4800-058, Guimarães, Portugal
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
34
|
Li M, Liu Y, Liu Y, Lin J, Ding L, Wu S, Gong J. Fabrication of targeted and pH responsive lysozyme-hyaluronan nanoparticles for 5-fluorouracil and curcumin co-delivery in colorectal cancer therapy. Int J Biol Macromol 2024; 254:127836. [PMID: 37931859 DOI: 10.1016/j.ijbiomac.2023.127836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Green nanotechnology is considered a promising method to construct functional materials with significant anticancer activity, while overcoming the shortcomings of traditional synthesis process complexity and high organic solvents consumption. Thus, in this study, we report for the first time the rational design and green synthesis of functionalized 5-fluorouracil and curcumin co-loaded lysozyme-hyaluronan composite colloidal nanoparticles (5-Fu/Cur@LHNPs) for better targeted colorectal cancer therapy with minimized side effects. The functionalized 5-Fu/Cur@LHNPs exhibit stabilized particle size (126.1 nm) with excellent homogeneity (PDI = 0.1), favorable colloidal stabilities, and excellent re-dispersibility. In vitro cell experiments illustrate that the cellular uptake of 5-Fu/Cur@LHNPs was significantly improved and further promoted a higher apoptosis ratio of HCT-116 cells. Compared with the control group, the 5-Fu/Cur@LHNPs formulation group achieved effective inhibition (60.1 %) of colorectal tumor growth. The alcohol-free self-assembly method to construct 5-Fu/Cur@LHNPs is simple and safe for a translational chemotherapy drug, also to promote more robust delivery systems for treating colorectal cancer.
Collapse
Affiliation(s)
- Maolin Li
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, PR China
| | - Yin Liu
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, PR China
| | - Yanbo Liu
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, PR China
| | - Jiawei Lin
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, PR China
| | - Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, PR China.
| | - Songgu Wu
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, PR China; Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China.
| | - Junbo Gong
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300072, PR China; Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, PR China
| |
Collapse
|
35
|
Bagheri M, Zandieh MA, Daryab M, Samaei SS, Gholami S, Rahmanian P, Dezfulian S, Eary M, Rezaee A, Rajabi R, Khorrami R, Salimimoghadam S, Hu P, Rashidi M, Ardakan AK, Ertas YN, Hushmandi K. Nanostructures for site-specific delivery of oxaliplatin cancer therapy: Versatile nanoplatforms in synergistic cancer therapy. Transl Oncol 2024; 39:101838. [PMID: 38016356 DOI: 10.1016/j.tranon.2023.101838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
As a clinically approved treatment strategy, chemotherapy-mediated tumor suppression has been compromised, and in spite of introducing various kinds of anticancer drugs, cancer eradication with chemotherapy is still impossible. Chemotherapy drugs have been beneficial in improving the prognosis of cancer patients, but after resistance emerged, their potential disappeared. Oxaliplatin (OXA) efficacy in tumor suppression has been compromised by resistance. Due to the dysregulation of pathways and mechanisms in OXA resistance, it is suggested to develop novel strategies for overcoming drug resistance. The targeted delivery of OXA by nanostructures is described here. The targeted delivery of OXA in cancer can be mediated by polymeric, metal, lipid and carbon nanostructures. The advantageous of these nanocarriers is that they enhance the accumulation of OXA in tumor and promote its cytotoxicity. Moreover, (nano)platforms mediate the co-delivery of OXA with drugs and genes in synergistic cancer therapy, overcoming OXA resistance and improving insights in cancer patient treatment in the future. Moreover, smart nanostructures, including pH-, redox-, light-, and thermo-sensitive nanostructures, have been designed for OXA delivery and cancer therapy. The application of nanoparticle-mediated phototherapy can increase OXA's potential in cancer suppression. All of these subjects and their clinical implications are discussed in the current review.
Collapse
Affiliation(s)
- Mohsen Bagheri
- Radiology Resident, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Babol Branch, Islamic Azad University, Babol, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Eary
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
36
|
Wang J, Li L, Xu ZP. Enhancing Cancer Chemo-Immunotherapy: Innovative Approaches for Overcoming Immunosuppression by Functional Nanomaterials. SMALL METHODS 2024; 8:e2301005. [PMID: 37743260 DOI: 10.1002/smtd.202301005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/05/2023] [Indexed: 09/26/2023]
Abstract
Chemotherapy is a critical modality in cancer therapy to combat malignant cell proliferation by directly attacking cancer cells and inducing immunogenic cell death, serving as a vital component of multi-modal treatment strategies for enhanced therapeutic outcomes. However, chemotherapy may inadvertently contribute to the immunosuppression of the tumor microenvironment (TME), inducing the suppression of antitumor immune responses, which can ultimately affect therapeutic efficacy. Chemo-immunotherapy, combining chemotherapy and immunotherapy in cancer treatment, has emerged as a ground-breaking approach to target and eliminate malignant tumors and revolutionize the treatment landscape, offering promising, durable responses for various malignancies. Notably, functional nanomaterials have substantially contributed to chemo-immunotherapy by co-delivering chemo-immunotherapeutic agents and modulating TME. In this review, recent advancements in chemo-immunotherapy are thus summarized to enhance treatment effectiveness, achieved by reversing the immunosuppressive TME (ITME) through the exploitation of immunotherapeutic drugs, or immunoregulatory nanomaterials. The effects of two-way immunomodulation and the causes of immunoaugmentation and suppression during chemotherapy are illustrated. The current strategies of chemo-immunotherapy to surmount the ITME and the functional materials to target and regulate the ITME are discussed and compared. The perspective on tumor immunosuppression reversal strategy is finally proposed.
Collapse
Affiliation(s)
- Jingjing Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
37
|
Becceneri AB, Fuzer AM, Lopes AC, da Silva PB, Plutin AM, Batista AA, Chorilli M, Cominetti MR. Nanoencapsulation of Ruthenium Complex Ru(ThySMet): A Strategy to Improve Selective Cytotoxicity against Breast Tumor Cells in 2D and 3D Culture Models. Curr Drug Discov Technol 2024; 21:e060623217687. [PMID: 37282638 DOI: 10.2174/1570163820666230606110457] [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: 03/21/2023] [Revised: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND Ruthenium complexes have shown promise in treating many cancers, including breast cancer. Previous studies of our group have demonstrated the potential of the trans- [Ru(PPh3)2(N,N-dimethylN'-thiophenylthioureato-k2O,S)(bipy)]PF6 complex, the Ru(ThySMet), in the treatment of breast tumor cancers, both in 2D and 3D culture systems. Additionally, this complex presented low toxicity when tested in vivo. AIMS Improve the Ru(ThySMet) activity by incorporating the complex into a microemulsion (ME) and testing its in vitro effects. METHODS The ME-incorporated Ru(ThySMet) complex, Ru(ThySMet)ME, was tested for its biological effects in two- (2D) and three-dimensional (3D) cultures using different types of breast cells, MDAMB- 231, MCF-10A, 4T1.13ch5T1, HMT-3522 and Balb/C 3T3 fibroblasts. RESULTS An increased selective cytotoxicity of the Ru(ThySMet)ME for tumor cells was found in 2D cell culture, compared with the original complex. This novel compound also changed the shape of tumor cells and inhibited cell migration with more specificity. Additional 3D cell culture tests using the non-neoplastic S1 and the triple-negative invasive T4-2 breast cells have shown that Ru(ThySMet)ME presented increased selective cytotoxicity for tumor cells compared with the 2D results. The morphology assay performed in 3D also revealed its ability to reduce the size of the 3D structures and increase the circularity in T4-2 cells. CONCLUSION These results demonstrate that the Ru(ThySMet)ME is a promising strategy to increase its solubility, delivery, and bioaccumulation in target breast tumors.
Collapse
Affiliation(s)
- Amanda Blanque Becceneri
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luís, Km 235, São Carlos, São Paulo, 13565-905, Brazil
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, Vila Monte Alegre, Ribeirão Preto, SP, 14040-903, Brazil
| | - Angelina Maria Fuzer
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luís, Km 235, São Carlos, São Paulo, 13565-905, Brazil
| | - Ana Carolina Lopes
- School of Pharmaceutical Sciences, São Paulo State University, Rodovia Araraquara-Jau, km. 1, Araraquara, São Paulo, 14800-903, Brazil
| | - Patrícia Bento da Silva
- School of Pharmaceutical Sciences, São Paulo State University, Rodovia Araraquara-Jau, km. 1, Araraquara, São Paulo, 14800-903, Brazil
| | - Ana Maria Plutin
- Facultad de Química, Universidad de la Habana. Zapata s/n entre G y Carlitos Aguirre, Habana, 10400, Cuba
| | - Alzir Azevedo Batista
- Department of Chemistry, Federal University of São Carlos. Rod. Washington Luís, Km 235, São Carlos, São Paulo, 13565-905, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University, Rodovia Araraquara-Jau, km. 1, Araraquara, São Paulo, 14800-903, Brazil
| | - Márcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luís, Km 235, São Carlos, São Paulo, 13565-905, Brazil
| |
Collapse
|
38
|
Wang J, Ma J, Tai Z, Li L, Zhang T, Cheng T, Yu J, Zhu Q, Bao L, Chen Z. Nanocarrier-Mediated Immunogenic Cell Death for Melanoma Treatment. Int J Nanomedicine 2023; 18:7149-7172. [PMID: 38059000 PMCID: PMC10697015 DOI: 10.2147/ijn.s434582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
Melanoma, a highly aggressive skin tumor, exhibits notable features including heterogeneity, a high mutational load, and innate immune escape. Despite advancements in melanoma treatment, current immunotherapies fail to fully exploit the immune system's maximum potential. Activating immunogenic cell death (ICD) holds promise in enhancing tumor cell immunogenicity, stimulating immune amplification response, improving drug sensitivity, and eliminating tumors. Nanotechnology-enabled ICD has emerged as a compelling therapeutic strategy for augmenting cancer immunotherapy. Nanoparticles possess versatile attributes, such as prolonged blood circulation, stability, and tumor-targeting capabilities, rendering them ideal for drug delivery. In this review, we elucidate the mechanisms underlying ICD induction and associated therapeutic strategies. Additionally, we provide a concise overview of the immune stress response associated with ICD and explore the potential synergistic benefits of combining ICD induction methods with the utilization of nanocarriers.
Collapse
Affiliation(s)
- Jiandong Wang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Department of Pharmacy, Third Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Jinyuan Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| | - Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| | - Tingrui Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| | - Tingting Cheng
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Department of Pharmacy, Third Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Junxia Yu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Department of Pharmacy, Third Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People’s Republic of China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| | - Leilei Bao
- Department of Pharmacy, Third Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, People’s Republic of China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, 200443, People’s Republic of China
| |
Collapse
|
39
|
Dai X, Du Y, Li Y, Yan F. Nanomaterials-based precision sonodynamic therapy enhancing immune checkpoint blockade: A promising strategy targeting solid tumor. Mater Today Bio 2023; 23:100796. [PMID: 37766898 PMCID: PMC10520454 DOI: 10.1016/j.mtbio.2023.100796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Burgeoning is an evolution from conventional photodynamic therapy (PDT). Thus, sonodynamic therapy (SDT) regulated by nanoparticles (NPs) possesses multiple advantages, including stronger penetration ability, better biological safety, and not reactive oxygen species (ROS)-dependent tumor-killing effect. However, the limitation to tumor inhibition instead of shrinkage and the incapability of eliminating metastatic tumors hinder the clinical potential for SDT. Fortunately, immune checkpoint blockade (ICB) can revive immunological function and induce a long-term immune memory against tumor rechallenges. Hence, synergizing NPs-based SDT with ICB can provide a promising therapeutic outcome for solid tumors. Herein, we briefly reviewed the progress in NPs-based SDT and ICB therapy. We highlighted the synergistic anti-tumor mechanisms and summarized the representative preclinical trials on SDT-assisted immunotherapy. Compared to other reviews, we provided comprehensive and unique perspectives on the innovative sonosensitizers in each trial. Moreover, we also discussed the current challenges and future corresponding solutions.
Collapse
Affiliation(s)
- Xinlun Dai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Yangyang Du
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yumei Li
- Department of Pediatric Intensive Care Unit, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
40
|
Sun Y, Lian T, Huang Q, Chang Y, Li Y, Guo X, Kong W, Yang Y, Zhang K, Wang P, Wang X. Nanomedicine-mediated regulated cell death in cancer immunotherapy. J Control Release 2023; 364:174-194. [PMID: 37871752 DOI: 10.1016/j.jconrel.2023.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Immunotherapy has attracted widespread attention in cancer treatment and has achieved considerable success in the clinical treatment of some tumors, but it has a low response rate in most tumors. To achieve sufficient activation of the immune response, significant efforts using nanotechnology have been made to enhance cancer immune response. In recent years, the induction of various regulated cell death (RCD) has emerged as a potential antitumor immuno-strategy, including processes related to apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis. In particular, damage-associated molecular patterns (DAMPs) released from the damaged membrane of dying cells act as in situ adjuvants to trigger antigen-specific immune responses by the exposure of an increased antigenicity. Thus, RCD-based immunotherapy offers a new approach for enhancing cancer treatment efficacy. Furthermore, incorporation with multimodal auxiliary therapies in cell death-based immunotherapy can trigger stronger immune responses, resulting in more efficient therapeutic outcome. This review discusses different RCD modalities and summarizes recent nanotechnology-mediated RCDs in cancer immunotherapy.
Collapse
Affiliation(s)
- Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; The Xi'an key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Ting Lian
- Research Center for Prevention and Treatment of Respiratory Disease, School of Clinical Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Qichao Huang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yawei Chang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaoyu Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Weirong Kong
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yifang Yang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
41
|
Xiang J, Liu K, Xu H, Zhao Z, Piao Y, Shao S, Tang J, Shen Y, Zhou Z. Dual Synergistic Tumor-Specific Polymeric Nanoparticles for Efficient Chemo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301216. [PMID: 37551065 PMCID: PMC10582463 DOI: 10.1002/advs.202301216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/02/2023] [Indexed: 08/09/2023]
Abstract
Chemo-immunotherapy has made significant progress in cancer treatment. However, the cancer cell self-defense mechanisms, including cell cycle checkpoint and programmed cell death-ligand 1 (PD-L1) upregulation, have greatly hindered the therapeutic efficacy. Herein, norcantharidin (NCTD)-platinum (Pt) codelivery nanoparticles (NC-NP) with tumor-sensitive release profiles are designed to overcome the self-defense mechanisms via synergistic chemo-immunotherapy. NC-NP remains stable under normal physiological conditions but quickly releases 1,2-diaminocyclohexane-platinum(II) (DACHPt, a parent drug of oxaliplatin) and NCTD in response to the tumor acidity. NCTD inhibits protein phosphatase 2A (PP2A) activity to relieve cell cycle arrest and downregulates the tumor PD-L1 expression to disrupt the programmed cell death-1 (PD-1)/PD-L1 interaction, synergistically enhancing Pt-based chemotherapy and immunogenic cell death-induced immunotherapy. As a result, NC-NP exhibits potent synergistic cytotoxicity and promotes T cell recruitment to generate robust antitumor immune responses. The dual synergism exhibits potent antitumor activity against orthotopic 4T1 tumors, providing a promising chemo-immunotherapy paradigm for cancer treatment.
Collapse
Affiliation(s)
- Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Kexin Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Hongxia Xu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Zhihao Zhao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| |
Collapse
|
42
|
Hu S, Xia K, Huang X, Zhao Y, Zhang Q, Huang D, Xu W, Chen Z, Wang C, Zhang Z. Multifunctional CaCO 3@Cur@QTX125@HA nanoparticles for effectively inhibiting growth of colorectal cancer cells. J Nanobiotechnology 2023; 21:353. [PMID: 37773145 PMCID: PMC10543835 DOI: 10.1186/s12951-023-02104-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths in humans, and effective treatments are still needed in clinical practice. Despite significant developments in anticancer drugs and inhibitors, their poor stability, water solubility, and cellular membrane permeability limit their therapeutic efficacy. To address these issues, multifunctional CaCO3 nanoparticles loaded with Curcumin (Cur) and protein deacetylase (HDAC) inhibitor QTX125, and coated with hyaluronic acid (HA) (CaCO3@Cur@QTX125@HA), were prepared through a one-step gas diffusion strategy. Dynamic light scattering (DLS), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) showed that CaCO3@Cur@QTX125@HA nanoparticles have uniform spherical morphology and elemental distribution, with diameters around 450 nm and a Zeta potential of - 8.11 mV. The controlled release of Cur from the nanoparticles was observed over time periods of 48 h. Cellular uptake showed that CaCO3@Cur@QTX125@HA nanoparticles were efficiently taken up by cancer cells and significantly inhibited their growth. Importantly, CaCO3@Cur@QTX125@HA nanoparticles showed specific inhibitory effects on CRC cell growth. Encouragingly, CaCO3@Cur@QTX125@HA nanoparticles successfully internalized into CRC patient-derived organoid (PDO) models and induced apoptosis of tumor cells. The multifunctional CaCO3@Cur@QTX125@HA nanoparticles hold promise for the treatment of CRC.
Collapse
Affiliation(s)
- Shengyun Hu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kunkun Xia
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ye Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingqing Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dongdong Huang
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China
| | - Weiyi Xu
- Department of Dermatology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Zhengju Chen
- Pooling Medical Research Institutes of 100Biotech, Beijing, 100006, China.
| | - Chenfei Wang
- Department of Dermatology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Zhiyong Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
43
|
Akbar MU, Khattak S, Khan MI, Saddozai UAK, Ali N, AlAsmari AF, Zaheer M, Badar M. A pH-responsive bi-MIL-88B MOF coated with folic acid-conjugated chitosan as a promising nanocarrier for targeted drug delivery of 5-Fluorouracil. Front Pharmacol 2023; 14:1265440. [PMID: 37745070 PMCID: PMC10517339 DOI: 10.3389/fphar.2023.1265440] [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: 07/22/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer has remained one of the leading causes of death worldwide, with a lack of effective treatment. The intrinsic shortcomings of conventional therapeutics regarding tumor specificity and non-specific toxicity prompt us to look for alternative therapeutics to mitigate these limitations. In this regard, we developed multifunctional bimetallic (FeCo) bi-MIL-88B-FC MOFs modified with folic acid-conjugated chitosan (FC) as drug delivery systems (DDS) for targeted delivery of 5-Fluorouracil (5-FU). The bi-MIL-88B nanocarriers were characterized through various techniques, including powder X-ray diffraction, scanning electron microscopy, energy-dispersive X-ray, thermogravimetric analysis, and Fourier transform infrared spectroscopy. Interestingly, 5-FU@bi-MIL-88B-FC showed slower release of 5-FU due to a gated effect phenomenon endowed by FC surface coating compared to un-modified 5-FU@bi-MIL-88B. The pH-responsive drug release was observed, with 58% of the loaded 5-FU released in cancer cells mimicking pH (5.2) compared to only 24.9% released under physiological pH (5.4). The in vitro cytotoxicity and cellular internalization experiments revealed the superiority of 5-FU@bi-MIL-88B-FC as a highly potent targeted DDS against folate receptor (FR) positive SW480 cancer cells. Moreover, due to the presence of Fe and Co in the structure, bi-MIL-88B exhibited peroxidase-like activity for chemodynamic therapy. Based on the results, 5-FU@bi-MIL-88B-FC could serve as promising candidate for smart DDS by sustained drug release and selective targeting.
Collapse
Affiliation(s)
- Muhammad Usman Akbar
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| | - Saadullah Khattak
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Malik Ihsanullah Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Umair Ali Khan Saddozai
- Department of Preventive Medicine, Institute of Bioinformatics, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Zaheer
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Muhammad Badar
- Gomal Center of Biochemistry and Biotechnology, Gomal University, Dera Ismail Khan, Pakistan
| |
Collapse
|
44
|
Zhong T, Yu J, Pan Y, Zhang N, Qi Y, Huang Y. Recent Advances of Platinum-Based Anticancer Complexes in Combinational Multimodal Therapy. Adv Healthc Mater 2023; 12:e2300253. [PMID: 37097737 DOI: 10.1002/adhm.202300253] [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: 01/24/2023] [Revised: 03/19/2023] [Indexed: 04/26/2023]
Abstract
Platinum drugs with manifest therapeutic effects are widely used, but their systemic toxicity and the drug resistance acquired by cancer cells limit their clinical applications. Thus, the exploration on appropriate methods and strategies to overcome the limitations of traditional platinum drugs becomes extremely necessary. Combination therapy of platinum drugs can inhibit tumor growth and metastasis in an additive or synergistic manner, and can potentially reduce the systemic toxicity of platinum drugs and overcome platinum-resistance. This review summarizes the various modalities and current progress in platinum-based combination therapy. The synthetic strategies and therapeutic effects of some platinum-based anticancer complexes in the combination of platinum drugs with gene editing, ROS-based therapy, thermal therapy, immunotherapy, biological modelling, photoactivation, supramolecular self-assembly and imaging modality are briefly described. Their potential challenges and prospects are also discussed. It is hoped that this review will inspire researchers to have more ideas for the future development of highly effective platinum-based anti-cancer complexes.
Collapse
Affiliation(s)
- Tianyuan Zhong
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Jie Yu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Ning Zhang
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics, Harbin, 150000, China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
45
|
Liu Q, Li J, Chang J, Guo Y, Wen D. The characteristics and medical applications of antler stem cells. Stem Cell Res Ther 2023; 14:225. [PMID: 37649124 PMCID: PMC10468909 DOI: 10.1186/s13287-023-03456-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023] Open
Abstract
Antlers are the only fully regenerable mammalian appendages whose annual renewal is initiated by antler stem cells (ASCs), defined as a specialized type of mesenchymal stem cells (MSCs) with embryonic stem cell properties. ASCs possess the same biological features as MSCs, including the capacity for self-renewal and multidirectional differentiation, immunomodulatory functions, and the maintenance of stem cell characteristics after multiple passages. Several preclinical studies have shown that ASCs exhibit promising potential in wound healing, bone repair, osteoarthritis, anti-tissue fibrosis, anti-aging, and hair regeneration. Medical applications based on ASCs and ASC-derived molecules provide a new source of stem cells and therapeutic modalities for regenerative medicine. This review begins with a brief description of antler regeneration and the role of ASCs. Then, the properties and advantages of ASCs are described. Finally, medical research advances regarding ASCs are summarized, and the prospects and challenges of ASCs are highlighted.
Collapse
Affiliation(s)
- Qi Liu
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiannan Li
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinghui Chang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Guo
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Dacheng Wen
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
46
|
Pang Y, Chen X, Xu B, Zhang Y, Liang S, Hu J, Liu R, Luo X, Wang Y. Engineered multitargeting exosomes carrying miR-323a-3p for CRC therapy. Int J Biol Macromol 2023; 247:125794. [PMID: 37442504 DOI: 10.1016/j.ijbiomac.2023.125794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/15/2023]
Abstract
Colorectal cancer (CRC) is in the forefront of malignancies for its high incidence and mortality. 5-Fluorouracil (5-FU) is one of the most widely used effective drugs for the treatment of CRC. However, there is an urgent need in reducing its systemic side effects and chemoresistance, in order to make 5-FU-based chemotherapy more effective in the treatment of CRC. In this study, engineered CRC cells were established to overexpress miR-323a-3p, which was a tumor suppressor that targeted both EGFR and TYMS. Then miR-323a-3p-loaded exosomes (miR-Exo) were obtained with suitable methods of collection and purification. We found that miR-Exo significantly inhibited CRC cell proliferation and induced apoptosis by the way of targeting EGFR directly in the cells, which eventually led to desirable tumor regression in the cell derived xenograft (CDX) and patient derived xenograft (PDX) tumor mice models. Moreover, we discovered that miR-323a-3p released from miR-Exo directly inhibited the upregulation of thymidylate synthase (TYMS) induced by 5-FU-resistence in CRC cells, resulting in the revival of tumor cytotoxicity from 5-FU. MiR-Exo could effectively induce the CRC cell apoptosis by targeting EGFR and TYMS, and enhance the therapeutic effects of 5-FU on CRC. Our work demonstrates the potency of miR-Exo for advanced CRC biotherapy.
Collapse
Affiliation(s)
- Yechun Pang
- Department of General Surgery, Pudong New Area People's Hospital, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingshi Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiying Xu
- Department of General Surgery, Pudong New Area People's Hospital, China
| | - Yuanzhou Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunshun Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingying Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoying Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yunfeng Wang
- Department of General Surgery, Pudong New Area People's Hospital, China.
| |
Collapse
|
47
|
Wang C, He Y, He Y, Liang W, Zhou C, Wu M, Meng Z, Li W, Cao J. Prognostic and biological function value of OSBPL3 in colorectal cancer analyzed by multi-omic data analysis. BMC Gastroenterol 2023; 23:270. [PMID: 37550605 PMCID: PMC10408063 DOI: 10.1186/s12876-023-02824-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/17/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignancies in the world. This study proposes to reveal prognostic biomarkers for the prognosis and treatment of CRC patients. METHODS Differential analysis of OSBPL3 was performed in pan-cancer, and the correlation between clinical stage and OSBPL3 was analyzed. Multiple omics analysis was used to compare the relationship between survival of patients and copy number variation, single nucleotide variant, and methylation status. Survival differences between high and low OSBPL3 expression groups were analyzed. Differentially expressed genes (DEGs) between high and low OSBPL3 expression groups were obtained, and functional enrichment analysis was implemented. Correlations between immune cells and OSBPL3 was analyzed. Drug sensitivity between the two OSBPL3 expression groups was compared. Moreover, the expression of OSBPL3 was verified by immunohistochemistry and real-time quantitative PCR. RESULTS OSBPL3 was differentially expressed in 13 tumors and had some correlations with T and N stages. OSBPL3 expression was regulated by methylation and higher OSBPL3 expression was associated with poorer prognosis in CRC. 128 DEGs were obtained and they were mainly involved in signaling receptor activator activity, aspartate and glutamate metabolism. T cell gamma delta and T cell follicular helper were significantly different in the high and low OSBPL3 expression groups. Moreover, OSBPL3 showed negative correlations with multiple drugs. OSBPL3 was significantly upregulated in CRC samples compared to normal samples. CONCLUSIONS A comprehensive analysis demonstrated that OSBPL3 had potential prognostic value, and guiding significance for CRC chemotherapeutic.
Collapse
Affiliation(s)
- Chengxing Wang
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Yu He
- National Drug Clinical Trial Institution, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Weijun Liang
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Chaorong Zhou
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Meimei Wu
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Zijie Meng
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, 529000, Guangdong, China
| | - Wanglin Li
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China.
| | - Jie Cao
- The First Affiliated Hospital, Jinan University, Guangzhou, 529000, Guangdong, China.
| |
Collapse
|
48
|
Chu D, Chen J, Liu X, Liao A, Song X, Li Y, Yang L, Chen Z, Yu Z, Guo J. A tetramethylpyrazine-loaded hyaluronic acid-based hydrogel modulates macrophage polarization for promoting wound recovery in diabetic mice. Int J Biol Macromol 2023; 245:125495. [PMID: 37353128 DOI: 10.1016/j.ijbiomac.2023.125495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/15/2023] [Accepted: 06/18/2023] [Indexed: 06/25/2023]
Abstract
The failure of wound healing often causes lower limb disability and amputation of diabetic patients. Current strategies for diabetic wound management often fail to achieve the expected outcomes, and emerging alternatives are urgently needed. Recent advances in the identification of active compounds from traditional herbal medicines provide promising therapeutics for tissue repair and regeneration. In this study, the pro-healing effects of tetramethylpyrazine (TMP, a natural alkaloid found in Ligusticum chuanxiong Hort) for diabetic wounds were for the first time demonstrated. The cutaneous healing was mainly achieved by TMP-mediated macrophage polarization from pro-inflammatory to pro-healing phenotype. In addition, the topical administration of TMP was facilitated by the hyaluronic acid (HA) hydrogel for promoting the full-thickness wounds in the experimental diabetic mice. Consequently, TMP-loaded HA hydrogel (TMP-HA) profoundly accelerated the wound closure in comparison with TMP-loaded INTRASITE Gel (it is a commercial hydrogel), which was evident with the inflammation mitigation, the angiogenesis enhancement, and the collagen deposition. Our work reveals the macrophage-modulatory function of TMP for diabetic wound healing and demonstrates great potential of TMP-HA for clinical application.
Collapse
Affiliation(s)
- Di Chu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Junjun Chen
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xingmei Liu
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Anqi Liao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xiaohuan Song
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Zheng Chen
- Key Laboratory of High-Performance Plastics, Ministry of Education, National & Local Joint Engineering Laboratory for Synthesis Technology of High-Performance Polymer, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
49
|
Xia J, Dai QL, He S, Jia HJ, Liu XG, Hua H, Zhou M, Wang X. Artesunate alleviates 5-fluorouracil-induced intestinal damage by suppressing cellular senescence and enhances its antitumor activity. Discov Oncol 2023; 14:139. [PMID: 37498338 PMCID: PMC10374509 DOI: 10.1007/s12672-023-00747-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most prevalent diagnosed malignancies and one of the leading causes of cancer-related deaths worldwide. 5-Fluorouracil (5-FU) and its combination regimen are commonly used as primary chemotherapeutic agents for advanced CRC. Intestinal mucositis is one of the most frequent side effects of 5-FU. Artesunate (Arte) is derived from the wormwood plant Artemisia annua. Arte is not only effective against malaria but also diabetes, atherosclerosis, inflammation, and other conditions. The mechanism by which 5-FU damages the intestinal tract is unclear, and there is no standard treatment for diarrhea caused by 5-FU. Therefore, it is critical to discover novel and promising therapeutic drugs for 5-FU side effect treatment. METHODS The morphology and expression of genes and proteins associated with the aging of HUVECs, HIECs, and intestinal tissues were compared to the those of the control group. The cell lines and tissues were evaluated by SA-β-Gal staining, Western blotting, and RT‒qPCR. HIEC and HCT116 cell viability was assessed in vitro by a CCK-8 assay and in vivo by a subcutaneous tumor mouse assay. Tumor cell proliferation and apoptosis was evaluated by immunohistochemistry. RESULTS Here, we report that Arte alleviates the adverse side effects caused by 5-FU in intestinal tissue, and that 5-FU-induced intestinal damage is associated with drug-induced chemical inflammation and an increase in the proportion of senescent cells. Arte decreases the ratio of SA-β-Gal-positive cells and downregulated the expression of aging-related proteins (p53, p16) and aging-related genes (p53, p21). Mechanistically, Arte relieves intestinal injury by inhibiting mTOR expression, which is associated with the regulation of aging. Moreover, Arte suppresses the p38MAPK and NF-κB signaling pathways, which are related to inflammation regulation. In addition, the combined therapy of Arte plus 5-FU significantly decreases cancer cell viability in vitro. Arte and 5-FU synergistically reduce the growth of colorectal cancer (CRC) xenografts in vivo. CONCLUSIONS Overall, our findings point to the crucial treatment effect of Arte on inflammation, intestinal cell senescence, and CRC cell proliferation and offer a new option for CRC treatment.
Collapse
Affiliation(s)
- Jing Xia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Qian Long Dai
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Siyue He
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Hui-Jie Jia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Xian-Guo Liu
- Department of Oncology, The Affiliated Chengdu 363 Hospital of Southwest Medical University, No. 108, Daosangshu Street, Chengdu, 610041, China
| | - Hui Hua
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Min Zhou
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China.
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China.
| |
Collapse
|
50
|
Han S, Bao X, Zou Y, Wang L, Li Y, Yang L, Liao A, Zhang X, Jiang X, Liang D, Dai Y, Zheng QC, Yu Z, Guo J. d-lactate modulates M2 tumor-associated macrophages and remodels immunosuppressive tumor microenvironment for hepatocellular carcinoma. SCIENCE ADVANCES 2023; 9:eadg2697. [PMID: 37467325 PMCID: PMC10355835 DOI: 10.1126/sciadv.adg2697] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
The polarization of tumor-associated macrophages (TAMs) from M2 to M1 phenotype demonstrates great potential for remodeling the immunosuppressive tumor microenvironment (TME) of hepatocellular carcinoma (HCC). d-lactate (DL; a gut microbiome metabolite) acts as an endogenous immunomodulatory agent that enhances Kupffer cells for clearance of pathogens. In this study, the potential of DL for transformation of M2 TAMs to M1 was confirmed, and the mechanisms underlying such polarization were mainly due to the modulation of phosphatidylinositol 3-kinase/protein kinase B pathway. A poly(lactide-co-glycolide) nanoparticle (NP) was used to load DL, and the DL-loaded NP was modified with HCC membrane and M2 macrophage-binding peptide (M2pep), forming a nanoformulation (DL@NP-M-M2pep). DL@NP-M-M2pep transformed M2 TAMs to M1 and remodeled the immunosuppressive TME in HCC mice, promoting the efficacy of anti-CD47 antibody for long-term animal survival. These findings reveal a potential TAM modulatory function of DL and provide a combinatorial strategy for HCC immunotherapy.
Collapse
Affiliation(s)
- Shulan Han
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xueying Bao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Lingzhi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yutong Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Leilei Yang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Anqi Liao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Xuemei Zhang
- Department of Hepatopathy, Shuguang Hospital, affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Di Liang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Qing-Chuan Zheng
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun 130012, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital, affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|