1
|
Jin W, Zhang Y, Wang B, Kang Z, Li H, Song J, Chen Y, Xiong H, Chen J. Structural optimization and characterization of highly potent and selective STAT3 inhibitors for the treatment of triple negative breast cancer. Eur J Med Chem 2025; 287:117332. [PMID: 39938409 DOI: 10.1016/j.ejmech.2025.117332] [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: 12/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/14/2025]
Abstract
Effective targeted treatments for triple-negative breast cancer (TNBC), which has the worst prognosis among various types of breast cancer, are lacking owing to its clinical heterogeneity and malignant nature. STAT3, a key transcription factor, regulates multiple physiological functions. Aberrant activation of STAT3 plays a pivotal role in the initiation and progression of TNBC and is closely associated with a poor prognosis. Therefore, targeting STAT3 is a promising potential therapeutic approach for TNBC. In this study, we further optimized the core structure of 6f, which our research group previously identified as a STAT3 inhibitor and treatment for osteosarcoma, to identify additional potential STAT3 inhibitors for TNBC treatment. We identified WR-S-462 as a high-binding affinity inhibitor of STAT3 that effectively suppresses its phosphorylation and biological functions in vitro. Notably, WR-S-462 significantly inhibits TNBC growth and metastasis in a dose-dependent manner, providing robust evidence for its potential as a clinical intervention for TNBC.
Collapse
Affiliation(s)
- Wangrui Jin
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Yuzhu Zhang
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Baozhen Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China
| | - Zhaoyong Kang
- Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China
| | - Huachao Li
- Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), 318000, Taizhou, China
| | - Jingfeng Song
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China
| | - Yihua Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products and Yunnan College of Modern Biomedical Industry, Kunming Medical University, Yunnan, 650500, Kunming, China; Shanghai Key Laboratory of Regulatory Biology, The Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 200241, Shanghai, China.
| | - Hai Xiong
- Institute for Advanced Study, Shenzhen University, 518060, Shenzhen, China.
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, Ningxia, 750004, China; Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Ningxia, 750004, China.
| |
Collapse
|
2
|
Dou K, Lu J, Xing Y, Wang R, Won M, Kim J, Yu F, Seung Kim J. Metabolic Acidity/H 2O 2 Dual-Cascade-Activatable Molecular Imaging Platform Toward Metastatic Breast Tumor Malignancy. Angew Chem Int Ed Engl 2025; 64:e202419191. [PMID: 39511909 DOI: 10.1002/anie.202419191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
Fluorescence imaging in the second near-infrared window (NIR-II) is crucial for accurate tumor diagnosis, offering superior resolution and penetration capabilities. Current NIR-II probes are limited by either being "always on" or responding to one stimulus, leading to low signal-to-noise ratios and potential false positives. We introduced a dual-lock-controlled probe, HN-PBA, activated by both H2O2 and tumor acidic environment. This dual response ensures bright fluorescence at tumor sites, leading to higher tumor-to-normal tissue ratios (T/NT) compared to conventional "always on" probes and probes activated only by H2O2. This strategy allows precise tumor identification and removal of primary and metastatic tumors, achieving superior T/NT ratios (24.3/6.4 for orthotopic and lung metastasis, respectively). Our probe also effectively detected lung metastatic foci as small as≤0.7 mm and showed the capability for accurate lesion localization in clinical breast cancer specimens. This dual-stimuli-responsive strategy could aid future diagnostic probe design.
Collapse
Affiliation(s)
- Kun Dou
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma Ministry of Education, The First Affiliated Hospital of Hainan Medical University Hainan Medical University, Haikou, 571199, China
| | - Jiao Lu
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma Ministry of Education, The First Affiliated Hospital of Hainan Medical University Hainan Medical University, Haikou, 571199, China
| | - Yanlong Xing
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma Ministry of Education, The First Affiliated Hospital of Hainan Medical University Hainan Medical University, Haikou, 571199, China
| | - Rui Wang
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma Ministry of Education, The First Affiliated Hospital of Hainan Medical University Hainan Medical University, Haikou, 571199, China
| | - Miae Won
- Department of Chemistry, Korea University, Seoul, 02841, Korea
- R&D institute TheranoChem Incorporation, Seoul, 02856, Korea
- College of Pharmacy, Dongduk Women's University, Seoul, 02748, Korea
| | - Jungryun Kim
- Department of Chemistry, Korea University, Seoul, 02841, Korea
| | - Fabiao Yu
- Key Laboratory of Haikou Trauma, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Emergency and Trauma Ministry of Education, The First Affiliated Hospital of Hainan Medical University Hainan Medical University, Haikou, 571199, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Korea
- R&D institute TheranoChem Incorporation, Seoul, 02856, Korea
| |
Collapse
|
3
|
Yang N, Sun S, Xu J, Gong F, Lei H, Hao Y, Pei Z, Wang C, Yu Q, Nie J, Jiang N, Ni C, Cheng L. Manganese Galvanic Cells Intervene in Tumor Metabolism to Reinforce cGAS-STING Activation for Bidirectional Synergistic Hydrogen-Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414929. [PMID: 39775989 DOI: 10.1002/adma.202414929] [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: 10/01/2024] [Revised: 12/07/2024] [Indexed: 01/11/2025]
Abstract
The cGAS-STING pathway is pivotal in initiating antitumor immunity. However, tumor metabolism, particularly glycolysis, negatively regulates the activation of the cGAS-STING pathway. Herein, Mn galvanic cells (MnG) are prepared via liquid-phase exfoliation and in situ galvanic replacement to modulate tumor metabolism, thereby enhancing cGAS-STING activation for bidirectional synergistic H2-immunotherapy. The obtained MnG can be etched by water, enabling efficient and sustained generation of H2 gas and Mn2+. MnG not only activated and amplified the cGAS-STING pathway through the sustained release of Mn2+ but also regulated tumor glucose metabolism to inhibit the expression of three prime repair exonuclease 2 (TREX2), thereby synergistically enhancing the activation of the cGAS-STING pathway. The injection of MnG into tumors resulted in a robust immune response, thereby providing favorable support for antitumor therapy. Consequently, the combination of MnG with immune checkpoint blockade therapy resulted in significant suppression of both primary tumors and distant tumors. Furthermore, the MnG-lipiodol dispersion exhibited remarkable efficacy in combination with transarterial embolization (TAE)-gas-immunotherapy in a rabbit orthotopic liver tumor model. The present study underscores the significance of employing a metal galvanic cell strategy for enhanced immunotherapy, thereby offering a novel approach for rational design of bioactive materials to augment immunotherapeutic effectiveness.
Collapse
Affiliation(s)
- Nailin Yang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Shumin Sun
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jiachen Xu
- Department of Vascular Surgery and Interventional Radiology, The Forth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215125, China
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Chenya Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Qiao Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jihu Nie
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Nan Jiang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| |
Collapse
|
4
|
Yang J, Liu Y, Du Z, Zhou Q, Yang L, Ye Q, Pan J, Zou W, Chen C, Jin B. Antitumor activity of niclosamide-mediated oxidative stress against acute lymphoblastic leukemia. Carcinogenesis 2024; 45:940-952. [PMID: 38820079 DOI: 10.1093/carcin/bgae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 05/13/2024] [Accepted: 05/30/2024] [Indexed: 06/02/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a heterogeneous clonal disease originated from B- or T-cell lymphoid precursor cells. ALL is often refractory or relapses after treatment. Novel treatments are anxiously needed in order to achieve a better response and prolonged overall survival in ALL patients. In the present study, we aimed at examining the antitumor effect of niclosamide on ALL. We investigated the effects of niclosamide on the proliferation and apoptosis in vitro, the growth of ALL cells in xenografted NOD-Prkdcem26Cd52 il2rgem26Cd22 /Nju (NCG) mice. The results showed that niclosamide treatment potently inhibited the growth of ALL cells and induced apoptosis via elevating the levels of reactive oxygen species and activating TP53. These findings suggest that niclosamide may be a promisingly potential agent for ALL therapy.
Collapse
Affiliation(s)
- Jing Yang
- Division of Hematology/Oncology, Department of Pediatrics, 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yong Liu
- Division of Hematology/Oncology, Department of Pediatrics, 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zefan Du
- Division of Hematology/Oncology, Department of Pediatrics, 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qin Zhou
- Division of Hematology/Oncology, Department of Pediatrics, 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Luo Yang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 South Xianlie Road, Guangzhou 510060, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qianyun Ye
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 South Xianlie Road, Guangzhou 510060, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 South Xianlie Road, Guangzhou 510060, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Waiyi Zou
- Department of Hematology, The First Affiliated Hospital, 58 Zhongshan Road II, Guangzhou 510080, Sun Yat-sen University, Guangzhou, China
| | - Chun Chen
- Division of Hematology/Oncology, Department of Pediatrics, 628 Zhenyuan Road, Xinhu Street, Guangming District, Shenzhen, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Bei Jin
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, 54 South Xianlie Road, Guangzhou 510060, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Nethi SK, Kothadiya S, White BM, Rachagani S, Bardhan R, Mallapragada SK. Polyanhydride Copolymer-Based Niclosamide Nanoparticles for Inhibiting Triple-Negative Breast Cancer: Metabolic Responses and Synergism with Paclitaxel. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70362-70377. [PMID: 39666980 DOI: 10.1021/acsami.4c17961] [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: 12/14/2024]
Abstract
The heterogeneity of tumors and the lack of effective therapies have resulted in triple-negative breast cancer (TNBC) exhibiting the least favorable outcomes among breast cancer subtypes. TNBC is characterized by its aggressive nature, often leading to high rates of relapse, metastasis, and mortality. Niclosamide (Nic), an Food and Drug Administration-approved anthelmintic drug, has been repurposed for cancer treatment; however, its application for TNBC is hindered by significant challenges, including strong hydrophobicity, poor aqueous solubility, and low bioavailability. This study aimed to develop Nic nanoparticles (Nic NPs) using biodegradable and biocompatible polyanhydride copolymers to enhance Nic's bioavailability and therapeutic efficacy. Nic NPs effectively inhibited migration, proliferation, and clonogenicity in both murine and human TNBC cells, inducing apoptosis and suppressing STAT3 signaling. For the first time, we utilized Raman spectroscopy and Seahorse extracellular flux assays to demonstrate the metabolic responses of TNBC cells to Nic NPs, revealing significant metabolic alterations, including the inhibition of mitochondrial respiration and glycolysis. Additionally, this study is the first to explore the combination therapy of repurposed Nic with the approved chemotherapeutic agent paclitaxel in the 4T1 TNBC immunocompetent mouse model. The combination of Nic NPs and paclitaxel significantly reduced tumor growth without adversely affecting the body weight of tumor-bearing mice. In summary, these findings suggest that Nic NPs could serve as a promising component in combination therapies for the effective treatment of TNBC.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| | - Siddhant Kothadiya
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
| | - Brianna M White
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri 65211, United States
- Roy Blunt NextGen Precision Health, University of Missouri, Columbia, Missouri 65211, United States
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa50011, United States
- Nanovaccine Institute, Iowa State University, Ames, Iowa50011, United States
| |
Collapse
|
6
|
Kim JH, Lee J, Lee KW, Xiong H, Li M, Kim JS. Trapped in Cells: A Selective Accumulation Approach for Type-I Photodynamic Ablation of Cancer Stem-like Cells. JACS AU 2024; 4:3657-3667. [PMID: 39328753 PMCID: PMC11423316 DOI: 10.1021/jacsau.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024]
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme responsible for converting aldehyde functional groups into carboxylate metabolites. Elevated ALDH activity is a characteristic feature of cancer stem-like cells (CSCs). As a novel approach to target the CSC trait of overexpressing ALDH, we aimed to utilize ALDH activity for the selective accumulation of a photosensitizer in ALDHHigh CSCs. A novel ALDH substrate photosensitizer, SCHO, with thionylated coumarin and N-ethyl-4-(aminomethyl)benzaldehyde was developed to achieve this goal. Our study demonstrated the efficient metabolism of the aldehyde unit of SCHO into carboxylate, leading to its accumulation in ALDHHigh MDA-MB-231 cells. Importantly, we established the selectivity of SCHO as an ALDHHigh cell photosensitizer as it is not a substrate for ABC transporters. SCHO-based photodynamic therapy triggers apoptosis and pyroptosis in MDA-MB-231 cells and further reduces the characteristics of CSCs. Our study presents a novel strategy to target CSCs by exploiting their cellular metabolism to enhance photosensitizer accumulation, highlighting the potential of photodynamic therapy as a powerful tool for eliminating ALDHHigh CSCs.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- Department
of Chemical and Systems Biology, Chem-H
and Stanford Cancer Institute, Stanford School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Jieun Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Kyung-Woo Lee
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Hao Xiong
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
7
|
An S, Chun J, Lee J, Kim YS, Noh M, Ko H. Unraveling Stereochemical Structure-Activity Relationships of Sesquiterpene Lactones for Inhibitory Effects on STAT3 Activation. Biomol Ther (Seoul) 2024; 32:627-634. [PMID: 39091020 PMCID: PMC11392665 DOI: 10.4062/biomolther.2023.210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/15/2024] [Accepted: 02/23/2024] [Indexed: 08/04/2024] Open
Abstract
Sesquiterpene lactones, a class of natural compounds abundant in the Asteraceae family, have gained attention owing to their diverse biological activities, and particularly their anti-proliferative effects on human cancer cells. In this study, we systematically investigated the structure-activity relationship of ten sesquiterpene lactones with the aim of elucidating the structural determinants for the STAT3 inhibition governing their anti-proliferative effects. Our findings revealed a significant correlation between the STAT3 inhibitory activity and the anti-proliferative effects of sesquiterpene lactones in MDA-MB-231 breast cancer cell lines. Among the compounds tested, alantolactone and isoalantolactone emerged as the most potent STAT3 inhibitors, highlighting their potential as candidates for anticancer drug development. Through protein-ligand docking studies, we revealed the structural basis of STAT3 inhibition by sesquiterpene lactones, emphasizing the critical role of hydrogen-bonding interactions with key residues, including Arg609, Ser611, Glu612, and Ser613, in the SH2 domain of STAT3. Furthermore, our conformational analysis revealed the decisive role of the torsion angle within the geometry-optimized structures of sesquiterpene lactones in their STAT3 inhibitory activity (R=0.80, p<0.01). These findings not only provide preclinical evidence for sesquiterpene lactones as promising phytomedicines against diseases associated with abnormal STAT3 activation, but also highlight the importance of stereochemical aspects in their activity.
Collapse
Affiliation(s)
- Seungchan An
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaemoo Chun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Joohee Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yeong Shik Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyejin Ko
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| |
Collapse
|
8
|
Liang Q, Zhang S, Liu J, Zhou X, Syamimi Ariffin N, Wei J, Shi C, Ma X, Zhang Y, Huang R. Discovery of novel 1,8-naphthalimide piperazinamide based benzenesulfonamides derivatives as potent carbonic anhydrase IX inhibitors and ferroptosis inducers for the treatment of triple-negative breast cancer. Bioorg Chem 2024; 150:107596. [PMID: 38941699 DOI: 10.1016/j.bioorg.2024.107596] [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: 04/07/2024] [Revised: 06/16/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
A novel series of 1,8-naphthalimide piperazinamide based benzenesulfonamides derivatives were designed and synthesized as carbonic anhydrase IX (CA IX) inhibitors and ferroptosis inducers for the treatment of triple-negative breast cancer (TNBC). The representative compound 9o exhibited more potent inhibitory activity and selective against CA IX over off-target CA II, compared with positive control SLC-0111. Molecular docking study was also performed to gain insights into the binding interactions of 9o in the binding pocket of CAIX. Moreover, compound 9o exhibited superior antitumor activities against breast cancer cells under hypoxia than that of normoxia conditions. Mechanism studies revealed that compound 9o could act as DNA intercalator and effectively suppressed cell migration, arrested the cell cycle at G1/S phase and induced apoptosis in MDA-MB-231 cells, while inducing ferroptosis accompanied by the dissipation of MMP and the elevation intracellular levels of ROS. Notably, in vivo studies demonstrated that 9o effectively inhibited tumor growth and metastasis in a highly metastatic murine breast cancer 4 T1 xenograft model. Taken together, this study suggests that compound 9o represents a potent and selective CA IX inhibitor and ferroptosis inducer for the treatment of TNBC.
Collapse
Affiliation(s)
- Qiaoling Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Shi Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Jiajia Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xiaoqun Zhou
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, 42300 Bandar Puncak Alam, Selangor
| | - Nur Syamimi Ariffin
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, 42300 Bandar Puncak Alam, Selangor
| | - Jianhua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Chengyi Shi
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Xianli Ma
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| | - Rizhen Huang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
9
|
Chen W, Wang Z, Hong G, Du J, Song F, Peng X. Self-assembly-integrated tumor targeting and electron transfer programming towards boosting tumor type I photodynamic therapy. Chem Sci 2024; 15:10945-10953. [PMID: 39027272 PMCID: PMC11253188 DOI: 10.1039/d4sc03008g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Type I photodynamic therapy (PDT) is attracting increasing interest as an effective solution to the poor prognosis of patients with hypoxic tumors. The development of functional type I photosensitizers is limited by a lack of feasible strategies to systematically modulate electron transfer (ET) in photosensitization. Herein, we present an easily accessible approach for the preparation of nanophotosensitizers with self-assembly-integrated tumor-targeting and ET programming towards boosting tumor type I PDT. Specifically, a dual functional amphiphile PS-02 was designed with a ligand (6-NS) that had the ability to not only target tumor cell marker carbonic anhydrase IX (CAIX) but also regulate the ET process for type I PDT. The amphiphile PS-02 tended to self-assemble into PS-02 nanoparticles (NPs), which exhibited a local "ET-cage effect" due to the electron-deficient nature of 6-NS. It is noteworthy that when PS-02 NPs selectively targeted the tumor cells, the CAIX binding enabled the uncaging of the inhibited ET process owing to the electron-rich characteristic of CAIX. Therefore, PS-02 NPs integrated tumor targeting and CAIX activation towards boosting type I PDT. As a proof of concept, the improved PDT performance of PS-02 NPs was demonstrated with tumor cells under hypoxic conditions and solid tumor tissue in mouse in vivo experiments. This work provides a practical paradigm to develop versatile type I PDT nano-photosensitizers by simply manipulating ET and easy self-assembling.
Collapse
Affiliation(s)
- Wenlong Chen
- Shenzhen Research Institute of Shandong University, A301 Virtual University Park in South District of Shenzhen 518057 P. R. China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Zehui Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Gaobo Hong
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Fengling Song
- Shenzhen Research Institute of Shandong University, A301 Virtual University Park in South District of Shenzhen 518057 P. R. China
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology Dalian 116024 P. R. China
| |
Collapse
|
10
|
Wang Y, Cheng X, Yin N, Wang M, Qin G, Tang J, Zhang Y, Xu Q. A Hypochlorite-Activated Theranostic Prodrug for Selective Imaging of High Myeloperoxidase Expression Acute Myeloid Leukemia Cells and Drug Release. Anal Chem 2024. [PMID: 39012074 DOI: 10.1021/acs.analchem.4c00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Acute myeloid leukemia (AML) is a fatal hematologic disease. Diagnosis and proper treatment are important for prognosis. High myeloperoxidase (MPO) expression AML cells are characterized with high levels of hypochlorite (ClO-). In this study, we report a ClO--activated theranostic agent, FNC, for AML therapy. FNC responds to ClO- specifically in high MPO expression AML cells, resulting in bright fluorescence and chlorambucil release. FNC can be used to quickly distinguish high MPO expression AML cells from other cells, including low MPO expression leukemia and activated inflammatory cells. FNC exhibits selective toxicity to highly MPO expression AML cells and can efficiently inhibit tumor growth. Meanwhile, FNC can be used to indicate differentiation through the detection of ClO-.
Collapse
Affiliation(s)
- Yuting Wang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Xiaoyu Cheng
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Nan Yin
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Mingxiu Wang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
- School of Public Health, Shanxi Medical University, Taiyuan 030001, P. R. China
| | - Guixin Qin
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Jiali Tang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| | - Qingling Xu
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 101408, P. R. China
| |
Collapse
|
11
|
Failli M, Demir S, Del Río-Álvarez Á, Carrillo-Reixach J, Royo L, Domingo-Sàbat M, Childs M, Maibach R, Alaggio R, Czauderna P, Morland B, Branchereau S, Cairo S, Kappler R, Armengol C, di Bernardo D. Computational drug prediction in hepatoblastoma by integrating pan-cancer transcriptomics with pharmacological response. Hepatology 2024; 80:55-68. [PMID: 37729391 PMCID: PMC11185924 DOI: 10.1097/hep.0000000000000601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/11/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND AND AIMS Hepatoblastoma (HB) is the predominant form of pediatric liver cancer, though it remains exceptionally rare. While treatment outcomes for children with HB have improved, patients with advanced tumors face limited therapeutic choices. Additionally, survivors often suffer from long-term adverse effects due to treatment, including ototoxicity, cardiotoxicity, delayed growth, and secondary tumors. Consequently, there is a pressing need to identify new and effective therapeutic strategies for patients with HB. Computational methods to predict drug sensitivity from a tumor's transcriptome have been successfully applied for some common adult malignancies, but specific efforts in pediatric cancers are lacking because of the paucity of data. APPROACH AND RESULTS In this study, we used DrugSense to assess drug efficacy in patients with HB, particularly those with the aggressive C2 subtype associated with poor clinical outcomes. Our method relied on publicly available collections of pan-cancer transcriptional profiles and drug responses across 36 tumor types and 495 compounds. The drugs predicted to be most effective were experimentally validated using patient-derived xenograft models of HB grown in vitro and in vivo. We thus identified 2 cyclin-dependent kinase 9 inhibitors, alvocidib and dinaciclib as potent HB growth inhibitors for the high-risk C2 molecular subtype. We also found that in a cohort of 46 patients with HB, high cyclin-dependent kinase 9 tumor expression was significantly associated with poor prognosis. CONCLUSIONS Our work proves the usefulness of computational methods trained on pan-cancer data sets to reposition drugs in rare pediatric cancers such as HB, and to help clinicians in choosing the best treatment options for their patients.
Collapse
Affiliation(s)
- Mario Failli
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples “Federico II”, Naples, Italy
| | - Salih Demir
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Germany
| | - Álvaro Del Río-Álvarez
- Childhood Liver Oncology Group (c-LOG), Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | - Juan Carrillo-Reixach
- Childhood Liver Oncology Group (c-LOG), Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
- Nottingham Clinical Trials Unit, Nottingham, United Kingdom
| | - Laura Royo
- Childhood Liver Oncology Group (c-LOG), Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | - Montserrat Domingo-Sàbat
- Childhood Liver Oncology Group (c-LOG), Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
| | | | - Rudolf Maibach
- International Breast Cancer Study Group Coordinating Center, Bern, Switzerland
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Piotr Czauderna
- Department of Surgery and Urology for Children and Adolescents, Medical University of Gdansk, Gdansk, Poland
| | - Bruce Morland
- Department of Oncology, Birmingham Women’s and Children’s Hospital, Birmingham, United Kingdom
| | | | - Stefano Cairo
- XenTech, Evry, France
- Champions Oncology, Rockville, Maryland, USA
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Germany
| | - Carolina Armengol
- Childhood Liver Oncology Group (c-LOG), Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Catalonia, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Madrid, Spain
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
12
|
Kumar S, Chatterjee N, Misra SK. Suitably Incorporated Hydrophobic, Redox-Active Drug in Poly Lactic Acid-Graphene Nanoplatelet Composite Generates 3D-Printed Medicinal Patch for Electrostimulatory Therapeutics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:11858-11872. [PMID: 38801374 DOI: 10.1021/acs.langmuir.3c03338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Polymer carbon composites have been reported for improved mechanical, thermal and electrical properties to provide reduced side effect by 3D printing personalized biomedical drug delivery devices. But control on homogeneity in loading and release of dopants like carbon allotropes and drugs, respectively, in the bulk and on the surface has always been a challenge. Herein, we are reporting a methodological cascade to achieve a model, customizable, 3D printed, homogeneously layered and electrically stimulatory, PLA-Graphene nanoplatelet (hl-PLGR) based drug delivery device, called 3D-est-MediPatch. The medicinal patch has been prepared by 3D-printing a Nic-hl-PLGR composite obtained by incorporating a redox active model drug, niclosamide (Nic) in hl-PLGR. The composite of Nic-hl-PLGR was characterized in three sequentially complex forms─composite film, hot melt extruded (HME) filament, and 3D printed (3DP) patches to understand the effect of filament extrusion and 3D-printing processes on Nic-hl-PLGR composite and overall drug incorporation efficiency and control. The incorporation of graphene was found to improve the homogeneity of the drug, and the hot melt extrusion improved the dispersion of drug and graphene fillers in the composite. The electroresponsive drug release from the Nic-hl-PLGR composite was found to be controllably accelerated compared to the drug release by diffusion, in simulated buffer condition. The released drug concentration was found to reach within the IC50 range for malignant melanoma cell (A375) and showed in vitro selectively, with reduced effects in noncancerous, fibroblast cells (NIH3T3). Further, the feasibility of application for this system was assessed in generating personalized 3D-est-MediPatch for skin, liver and spleen tissues in ex-vivo scenario. It showed excellent feasibility and efficacy of the 3D-est-MediPatch in controlled and personalized release of drugs during electrostimulation. Thus, a model platform, 3D-est-MediPatch, could be achieved by suitably incorporating a hydrophobic, redox-active drug (niclosamide) in poly lactic acid-graphene nanoplatelet composite for electrostimulatory therapeutics with reduced side effects.
Collapse
Affiliation(s)
- Sandarbh Kumar
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, 208016, India
| | - Niranjan Chatterjee
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, 208016, India
| | - Santosh Kumar Misra
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, 208016, India
- Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, 208016, India
| |
Collapse
|
13
|
Lu R, Hong J, Fu T, Zhu Y, Tong R, Ai D, Wang S, Huang Q, Chen C, Zhang Z, Zhang R, Guo H, Li B. Loss of OVOL2 in Triple-Negative Breast Cancer Promotes Fatty Acid Oxidation Fueling Stemness Characteristics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308945. [PMID: 38627980 PMCID: PMC11199980 DOI: 10.1002/advs.202308945] [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: 11/21/2023] [Revised: 03/10/2024] [Indexed: 06/27/2024]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer, has a poor prognosis and lacks effective treatment strategies. Here, the study discovered that TNBC shows a decreased expression of epithelial transcription factor ovo-like 2 (OVOL2). The loss of OVOL2 promotes fatty acid oxidation (FAO), providing additional energy and NADPH to sustain stemness characteristics, including sphere-forming capacity and tumor initiation. Mechanistically, OVOL2 not only suppressed STAT3 phosphorylation by directly inhibiting JAK transcription but also recruited histone deacetylase 1 (HDAC1) to STAT3, thereby reducing the transcriptional activation of downstream genes carnitine palmitoyltransferase1 (CPT1A and CPT1B). PyVT-Ovol2 knockout mice develop a higher number of primary breast tumors with accelerated growth and increased lung-metastases. Furthermore, treatment with FAO inhibitors effectively reduces stemness characteristics of tumor cells, breast tumor initiation, and metastasis, especially in OVOL2-deficient breast tumors. The findings suggest that targeting JAK/STAT3 pathway and FAO is a promising therapeutic strategy for OVOL2-deficient TNBC.
Collapse
Affiliation(s)
- Ruipeng Lu
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Jingjing Hong
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Tong Fu
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Yu Zhu
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Ruiqi Tong
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Di Ai
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Shuai Wang
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Qingsong Huang
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Ceshi Chen
- Academy of Biomedical EngineeringKunming Medical UniversityKunming650500China
- The Third Affiliated HospitalKunming Medical UniversityKunming650118China
| | - Zhiming Zhang
- Department of Breast SurgeryThe First Affiliated Hospital of Xiamen UniversityXiamen361009China
| | - Rui Zhang
- Xiamen Cell Therapy Research CenterThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361003China
| | - Huiling Guo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| | - Boan Li
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen UniversityXiamen361104China
| |
Collapse
|
14
|
Yao SY, Ying AK, Jiang ZT, Cheng YQ, Geng WC, Hu XY, Cai K, Guo DS. Single Molecular Nanomedicines Based on Macrocyclic Carrier-Drug Conjugates for Concentration-Independent Encapsulation and Precise Activation of Drugs. J Am Chem Soc 2024; 146:14203-14212. [PMID: 38733560 DOI: 10.1021/jacs.4c03238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Nanomedicines often rely on noncovalent self-assembly and encapsulation for drug loading and delivery. However, challenges such as reproducibility issues due to the multicomponent nature, off-target activation caused by premature drug release, and complex pharmacokinetics arising from assembly dissociation have hindered their clinical translation. In this study, we introduce an innovative design concept termed single molecular nanomedicine (SMNM) based on macrocyclic carrier-drug conjugates. Through the covalent linkage of two chemotherapy drugs to a hypoxia-cleavable macrocyclic carrier, azocalix[4]arene, we obtained two self-included complexes to serve as SMNMs. The intramolecular inclusion feature of the SMNMs has not only demonstrated comprehensive shielding and protection for the drugs but also effectively prevented off-target drug leakage, thereby significantly reducing their side effects and enhancing their antitumor therapeutic efficacy. Additionally, the attributes of being a single component and molecularly dispersed confer advantages such as ease of preparation and good reproducibility for SMNMs, which is desirable for clinical applications.
Collapse
Affiliation(s)
- Shun-Yu Yao
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - An-Kang Ying
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ze-Tao Jiang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yuan-Qiu Cheng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Kang Cai
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi 844000, China
| |
Collapse
|
15
|
Chen MM, Li Y, Zhu Y, Geng WC, Chen FY, Li JJ, Wang ZH, Hu XY, Tang Q, Yu Y, Sun T, Guo DS. Supramolecular 3 in 1: A Lubrication and Co-Delivery System for Synergistic Advanced Osteoarthritis Therapy. ACS NANO 2024; 18:13117-13129. [PMID: 38727027 DOI: 10.1021/acsnano.4c01939] [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: 05/22/2024]
Abstract
The complexity, heterogeneity, and drug resistance of diseases necessitate a shift in therapeutic paradigms from monotherapy to combination therapy, which could augment treatment efficiency. Effective treatment of advanced osteoarthritis (OA) requires addressing three key factors contributing to its deterioration: chronic joint inflammation, lubrication dysfunction, and cartilage-tissue degradation. Herein, we present a supramolecular nanomedicine of multifunctionality via molecular recognition and self-assembly. The employed macrocyclic carrier, zwitterion-modified cavitand (CV-2), not only accurately loads various drugs but also functions as a therapeutic agent with lubricating properties for the treatment of OA. Kartogenin (KGN), a drug for articular cartilage regeneration and protection, and flurbiprofen (FP), an anti-inflammatory agent, were coloaded onto CV-2 assembly, forming a supramolecular nanomedicine KGN&FP@CV-2. The three-in-one combination therapy of KGN&FP@CV-2 addresses the three pathological features for treating OA collectively, and thus provides long-term therapeutic benefits for OA through sustained drug release and intrinsic lubrication in vivo. The multifunctional integration of macrocyclic delivery and therapeutics provides a simple, flexible, and universal platform for the synergistic treatment of diseases involving multiple drugs.
Collapse
Affiliation(s)
- Meng-Meng Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yuqiao Li
- Spine Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Yujie Zhu
- Center for Supramolecular Chemistry & Catalysis and Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Wen-Chao Geng
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Fang-Yuan Chen
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Juan-Juan Li
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ze-Han Wang
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xin-Yue Hu
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
| | - Qiong Tang
- Department of Respiratory, Tianjin Union Medical Center, Tianjin 300121, China
| | - Yang Yu
- Center for Supramolecular Chemistry & Catalysis and Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Tianwei Sun
- Spine Surgery, Tianjin Union Medical Center, Tianjin 300121, China
| | - Dong-Sheng Guo
- College of Chemistry, State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin 300071, China
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, College of Chemistry and Environmental Sciences, Kashi University, Kashi 844000, China
| |
Collapse
|
16
|
Liu L, Lei H, Hou G, Zhang L, Chen Y, Lu Y, Pei Z, Ge J, Wu J, Zhou J, Cheng L. Gas-Amplified Metalloimmunotherapy with Dual Activation of Pyroptosis and the STING Pathway for Remodeling the Immunosuppressive Cervical Cancer Microenvironment. ACS NANO 2024; 18:12830-12844. [PMID: 38709246 DOI: 10.1021/acsnano.4c00017] [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: 05/07/2024]
Abstract
The immunosuppressive microenvironment of cervical cancer significantly hampers the effectiveness of immunotherapy. Herein, PEGylated manganese-doped calcium sulfide nanoparticles (MCSP) were developed to effectively enhance the antitumor immune response of the cervical cancer through gas-amplified metalloimmunotherapy with dual activation of pyroptosis and STING pathway. The bioactive MCSP exhibited the ability to rapidly release Ca2+, Mn2+, and H2S in response to the tumor microenvironment. H2S disrupted the calcium buffer system of cancer cells by interfering with the oxidative phosphorylation pathway, leading to calcium overload-triggered pyroptosis. On the other hand, H2S-mediated mitochondrial dysfunction further promoted the release of mitochondrial DNA (mtDNA), enhancing the activation effect of Mn2+ on the cGAS-STING signaling axis and thereby activating immunosuppressed dendritic cells. The released H2S acted as an important synergist between Mn2+ and Ca2+ by modulating dual signaling mechanisms to bridge innate and adaptive immune responses. The combination of MCSP NPs and PD-1 immunotherapy achieved synergistic antitumor effects and effectively inhibited tumor growth. This study reveals the potential collaboration between H2S gas therapy and metalloimmunotherapy and provides an idea for the design of nanoimmunomodulators for rational regulation of the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Lin Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Guanghui Hou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Youdong Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Yujie Lu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jun Ge
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jie Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| |
Collapse
|
17
|
Supuran CT. Drug interactions of carbonic anhydrase inhibitors and activators. Expert Opin Drug Metab Toxicol 2024; 20:143-155. [PMID: 38450431 DOI: 10.1080/17425255.2024.2328152] [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: 01/22/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
INTRODUCTION Carbonic anhydrases (CAs, EC 4.2.1.1) have been established drug targets for decades, with their inhibitors and activators possessing relevant pharmacological activity and applications in various fields. At least 11 sulfonamides/sulfamates are clinically used as diuretics, antiglaucoma, antiepileptic, or antiobesity agents and one derivative, SLC-0111, is in clinical trials as antitumor/antimetastatic agent. The activators were less investigated with no clinically used agent. AREAS COVERED Drug interactions between CA inhibitors/activators and various other agents are reviewed in publications from the period March 2020 - January 2024. EXPERT OPINION Drug interactions involving these agents revealed several interesting findings. Acetazolamide plus loop diuretics is highy effective in acute decompensated heart failure, whereas ocular diseases such as X-linked retinoschisis and macular edema were treated by acetazolamide plus bevacizumab or topical NSAIDs. Potent anti-infective effects of acetazolamide and other CAIs, alone or in combination with other agents were demonstrated for the management of Neisseria gonorrhoea, vancomycin resistant enterococci, Acanthamoeba castellanii, Trichinella spiralis, and Cryptococcus neoformans infections. Topiramate, in combination with phentermine is incresingly used for the management of obesity, whereas zonisamide plus levodopa is highly effective for Parkinson's disease. Acetazolamide, methazolamide, ethoxzolamide, and SLC-0111 showed synergistic antitumor/antimetastatic action in combination with many other antitumor drugs.
Collapse
Affiliation(s)
- Claudiu T Supuran
- Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, FI, Italy
| |
Collapse
|
18
|
Kim S, Park JM, Park S, Jung E, Ko D, Park M, Seo J, Nam KD, Kang YK, Lee K, Farrand L, Kim YJ, Kim JY, Seo JH. Suppression of TNBC metastasis by doxazosin, a novel dual inhibitor of c-MET/EGFR. J Exp Clin Cancer Res 2023; 42:292. [PMID: 37924112 PMCID: PMC10625208 DOI: 10.1186/s13046-023-02866-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by aggressive growth and a high propensity for recurrence and metastasis. Simultaneous overexpression of c-MET and EGFR in TNBC is associated with worse clinicopathological features and unfavorable outcomes. Although the development of new c-MET inhibitors and the emergence of 3rd-generation EGFR inhibitors represent promising treatment options, the high costs involved limit the accessibility of these drugs. In the present study, we sought to investigate the therapeutic potential of doxazosin (DOXA), a generic drug for benign prostate hyperplasia, in targeting TNBC. METHODS The effect of DOXA on TNBC cell lines in vitro was evaluated in terms of cell viability, apoptosis, c-MET/EGFR signaling pathway, molecular docking studies and impact on cancer stem cell (CSC)-like properties. An in vivo metastatic model with CSCs was used to evaluate the efficacy of DOXA. RESULTS DOXA exhibits notable anti-proliferative effects on TNBC cells by inducing apoptosis via caspase activation. Molecular docking studies revealed the direct interaction of DOXA with the tyrosine kinase domains of c-MET and EGFR. Consequently, DOXA disrupts important survival pathways including AKT, MEK/ERK, and JAK/STAT3, while suppressing CSC-like characteristics including CD44high/CD24low subpopulations, aldehyde dehydrogenase 1 (ALDH1) activity and formation of mammospheres. DOXA administration was found to suppress tumor growth, intra- and peri-tumoral angiogenesis and distant metastasis in an orthotopic allograft model with CSC-enriched populations. Furthermore, no toxic effects of DOXA were observed in hepatic or renal function. CONCLUSIONS Our findings highlight the potential of DOXA as a therapeutic option for metastatic TNBC, warranting further investigation.
Collapse
Affiliation(s)
- Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Yong Koo Kang
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Kyoungmin Lee
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
19
|
Supuran CT. Targeting carbonic anhydrases for the management of hypoxic metastatic tumors. Expert Opin Ther Pat 2023; 33:701-720. [PMID: 37545058 DOI: 10.1080/13543776.2023.2245971] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/08/2023]
Abstract
INTRODUCTION Several isoforms of the metalloenzyme carbonic anhydrase (CA, EC 4.2.1.1) are connected with tumorigenesis. Hypoxic tumors overexpress CA IX and XII as a consequence of HIF activation cascade, being involved in pH regulation, metabolism, and metastases formation. Other isoforms (CA I, II, III, IV) were also reported to be present in some tumors. AREAS COVERED Some CA isoforms are biomarkers for disease progression or response to therapy. Inhibitors, antibodies, and other procedures for targeting these enzymes for the treatment of tumors/metastases are discussed. Sulfonamides and coumarins represent the most investigated classes of inhibitors, but carboxylates, selenium, and tellurium-containing inhibitors were also investigated. Hybrid drugs of CA inhibitors with other antitumor agents for multitargeted therapy were reported. EXPERT OPINION Targeting CAs present in solid or hematological tumors with selective, targeted inhibitors is a validated approach, which has been consolidated in the last years. A host of new preclinical data and several clinical trials of antibodies and small-molecule inhibitors are ongoing, which connected with the large number of new chemotypes/procedures discovered to be effective, may lead to a breakthrough in this therapeutic area. The scientific/patent literature has been searched for on PubMed, ScienceDirect, Espacenet, and PatentGuru, from 2018 to 2023.
Collapse
Affiliation(s)
- Claudiu T Supuran
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Sesto Fiorentino, Firenze, Italy
| |
Collapse
|