1
|
Lee J, McClure S, Weichselbaum RR, Mimee M. Designing live bacterial therapeutics for cancer. Adv Drug Deliv Rev 2025; 221:115579. [PMID: 40228606 DOI: 10.1016/j.addr.2025.115579] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Humans are home to a diverse community of bacteria, many of which form symbiotic relationships with their host. Notably, tumors can also harbor their own unique bacterial populations that can influence tumor growth and progression. These bacteria, which selectively colonize hypoxic and acidic tumor microenvironments, present a novel therapeutic strategy to combat cancer. Advancements in synthetic biology enable us to safely and efficiently program therapeutic drug production in bacteria, further enhancing their potential. This review provides a comprehensive guide to utilizing bacteria for cancer treatment. We discuss key considerations for selecting bacterial strains, emphasizing their colonization efficiency, the delicate balance between safety and anti-tumor efficacy, and the availability of tools for genetic engineering. We also delve into strategies for precise spatiotemporal control of drug delivery to minimize adverse effects and maximize therapeutic impact, exploring recent examples of engineered bacteria designed to combat tumors. Finally, we address the underlying challenges and future prospects of bacterial cancer therapy. This review underscores the versatility of bacterial therapies and outlines strategies to fully harness their potential in the fight against cancer.
Collapse
Affiliation(s)
- Jaehyun Lee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Sandra McClure
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago 60637, USA; The Ludwig Center for Metastasis Research, University of Chicago, Chicago 60637, USA
| | - Mark Mimee
- Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchoissois Family Institute, University of Chicago, Chicago, IL 60637, USA; Committee On Molecular Metabolism and Nutrition, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Zhang J, Wang X, Guo L, Xiao S, Meng D, Shang M, Sun X, Shi D, Zhao Y, Liu R, Huang S, Zeng X, Li J. Dual-responsive nanoscale ultrasound contrast agent as an oxidative stress amplifier for enhanced DNA damage in BRCA-proficient ovarian cancer. Mater Today Bio 2025; 32:101761. [PMID: 40270892 PMCID: PMC12017913 DOI: 10.1016/j.mtbio.2025.101761] [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: 01/16/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
PARP inhibitor (PARPi)-based synthetic lethal therapies have displayed limited benefits in BRCA-proficient ovarian cancer. To potentiate the application of PARPi, an ultrasound contrast agent OLA-NDs for delivery of the PARPi olaparib (OLA) was established for enhancing DNA damage by blocking DNA repair. OLA-NDs were endowed with endogenous pH- and exogenous ultrasound (US)-responsiveness to target tumors, as well as contrast-enhanced US imaging for diagnostic and therapeutic integration. OLA-NDs could upregulate NOX4 to induce oxidative stress and sensitize BRCA wild-type A2780 cells to DNA oxidative damage through the utilization of ultrasound-targeted microbubble destruction (UTMD). In addition, the strategy further increased ROS production by interfering with mitochondrial function, thereby exacerbating DNA double-strand breaks (DSBs) and inducing mitochondria-mediated apoptosis. As a consequence, the combined application of UTMD and OLA-NDs demonstrated significant antitumor effects in vitro and in vivo. This combined strategy of amplifying oxidative damage improved lethality by promoting DNA DSBs and apoptosis with reduced adverse side effects, which would provide new insight for the clinical application of PARPi in BRCA-proficient ovarian cancer.
Collapse
Affiliation(s)
- Jialu Zhang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shuting Huang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xinyu Zeng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Department of Ultrasound, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong, 266035, China
| |
Collapse
|
3
|
Wu M, Liang B, Zhang L, Wu B, Liu J. Cobalt carbonate nanorods enhance chemotherapy via neutralization of acidic tumor microenvironment and generation of carbonate radical anions for necrosis. Colloids Surf B Biointerfaces 2025; 250:114563. [PMID: 39978250 DOI: 10.1016/j.colsurfb.2025.114563] [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: 11/05/2024] [Revised: 12/26/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
One of the hallmarks of cancer is the acidic extracellular space surrounding the tumor, which is linked to metabolic reprogramming and the use of glycolysis. Additionally, the acidic tumor microenvironment (TME) establishes a physiological barrier called "ion trapping" and significantly lowers the ability of cells to absorb weak-base chemotherapy agents. Although CO32- containing agents and nanoformulations could effectively neutralize the tumor acidity, the CO32- based therapeutic effect was insufficiently investigated. Herein, we fabricated cobalt carbonate (CoCO3) nanorods as drug carriers with acidity-responsive dissociation and acidity neutralization properties for the loading of hydrophobic and weak-basic drugs, evodiamine (EVO). After effective surface modification, CoCO3-PEG-EVO could effectively accumulate in the tumor and inhibit the growth of the tumor. On the one hand, acidity neutralization of CoCO3-PEG-EVO could lead to the ion trapping overcome and cellular uptake of EVO enhancement for effective cancer cell apoptosis. On the other hand, the high level of H2O2 in the tumor and HCO3- from dissociated CoCO3-PEG-EVO could cause the generation of CO3·- through a Fenton-like reaction while not hydroxyl radical (·OH) for cancer cell necrosis. Our results thus point to a potent yet easily prepared CoCO3 nanosystem (CoCO3-PEG-EVO) to induce cancer cell death, and because of its well-defined composition and excellent biocompatibility, it may be used in clinical settings in the future.
Collapse
Affiliation(s)
- Min Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, PR China
| | - Bing Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, PR China
| | - Lu Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, PR China
| | - Benmeng Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, PR China
| | - Jingjing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, PR China; The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, PR China.
| |
Collapse
|
4
|
Chen Y, Sun B, Yu Z, Cheng Z, Chen S, Chang B. Hollow heterojunction nanocages of zeolitic imidazolate framework-8@zinc sulfide@copper sulfide for synergistic tumor therapy. J Colloid Interface Sci 2025; 695:137811. [PMID: 40344729 DOI: 10.1016/j.jcis.2025.137811] [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: 02/12/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
The tumor microenvironment (TME) poses significant challenges for achieving efficient cancer remission through monotherapy, making multimodal synergistic therapy a key strategy in tumor treatment. Here, we designed and constructed a hollow core-shell nanocage based on zeolitic imidazolate framework-8 (ZIF-8), zeolitic imidazolate framework-8@zinc sulfide@copper sulfide (ZIF-8@ZnS@CuS, denoted as ZZCS), for the combined application of chemodynamic therapy (CDT), photothermal therapy (PTT) and photocatalytic therapy (PCT). The Cu2+ ions in ZZCS imparted peroxidase (POD)-like and catalase (CAT)-like activities, enabling the generation of toxic hydroxyl radicals (OH) through POD-like catalysis to enhance CDT effect, while also reacting with overexpressed glutathione (GSH) to elevate intracellular reactive oxygen species (ROS) levels. To overcome the inhibitory effects of hypoxia on PCT in the TME, ZZCS catalyzed the production of O2 from endogenous hydrogen peroxide (H2O2) via CAT-like activity, alleviating hypoxia and enhancing the PCT effect. Furthermore, hollow core-shell heterojunction structure of ZZCS exhibited excellent near-infrared absorption and multiple light reflection effects. Under 808 nm laser irradiation, ZZCS showed a high photothermal conversion efficiency (η = 78.1 %) and generated significant amounts of ROS (OH, O2-, 1O2), enabling the synergistic elimination of tumor cells via PTT and PCT. Benefiting from the combined CDT/PTT/PCT effects, ZZCS demonstrated excellent therapeutic efficacy in vivo, nearly eradicating tumors. This study may provide a promising foundation and potential direction for advancing multimodal tumor therapy. Future studies will focus on conducting long-term investigations to systematically evaluate the efficacy of ZZCS core-shell nanocages in achieving durable tumor eradication.
Collapse
Affiliation(s)
- Yuqin Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Benjian Sun
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhouyu Yu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Si Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
5
|
Mavuluri J, Dhungana Y, Jones LL, Bhatara S, Shi H, Yang X, Lim SE, Reyes N, Chi H, Yu J, Geiger TL. GPR65 Inactivation in Tumor Cells Drives Antigen-Independent CAR T-cell Resistance via Macrophage Remodeling. Cancer Discov 2025; 15:1018-1036. [PMID: 39998425 PMCID: PMC12046320 DOI: 10.1158/2159-8290.cd-24-0841] [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: 06/10/2024] [Revised: 11/28/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
SIGNIFICANCE The study identifies GPR65 as an important determinant of B-cell acute lymphoblastic leukemia response to CAR T-cell therapy. Notably, GPR65 absence signals CAR T resistance. By emphasizing the therapeutic potential of targeting VEGFA or host macrophages, our study identifies routes to optimize CAR T-cell therapy outcomes in hematologic malignancies via tumor microenvironment manipulation.
Collapse
Affiliation(s)
- Jayadev Mavuluri
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Yogesh Dhungana
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Lindsay L. Jones
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Sheetal Bhatara
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Xu Yang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Song-Eun Lim
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Noemi Reyes
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hongbo Chi
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Terrence L. Geiger
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
6
|
Zhao N, Shi Y, Liu P, Lv C. pH-responsive carbohydrate polymer-based nanoparticles in cancer therapy. Int J Biol Macromol 2025; 306:141236. [PMID: 39978518 DOI: 10.1016/j.ijbiomac.2025.141236] [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: 01/07/2025] [Revised: 02/01/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Using the specific features of the tumor microenvironment (TME) for the development and design of novel nanomaterials can improve the capacity in tumor suppression. One of the prominent features of the TME is the mild acidic pH. Therefore, the development of pH-responsive nanoparticles can lead to the release of cargo and therapeutics at the tumor site, improving the selectivity and specificity. The materials used for the development of nanoparticles should possess a number of unique features including biocompatibility and anti-cancer activity. Hence, a special attention has been directed towards the use of carbohydrate polymers in the development of nanoparticles. The carbohydrate polymers can develop smart nanoparticles respond to the pH in TME to increase targeting ability and provide controlled drug release. Such approach is also beneficial in decreasing the side effects of systemic chemotherapy. The pH-responsive nanoparticles developed from carbohydrate polymers can be also used for the combination chemotherapy/immunotherapy/phototherapy of cancer. Furthermore, these nanoparticles demonstrate theranostic application capable of cancer diagnosis and therapy. Further attention to the large-scale production, biocompatibility and long-term safety of carbohydrate polymer-based pH-responsive nanoparticles should be directed to improve the clinical translation in the treatment of cancer patients.
Collapse
Affiliation(s)
- Nanxi Zhao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yang Shi
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Pai Liu
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Chengzhou Lv
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Trejo-Solís C, Escamilla-Ramírez Á, Gómez-Manzo S, Castillo-Rodriguez RA, Palomares-Alonso F, Castillo-Pérez C, Jiménez-Farfán D, Sánchez-García A, Gallardo-Pérez JC. The pentose phosphate pathway (PPP) in the glioma metabolism: A potent enhancer of malignancy. Biochimie 2025; 232:117-126. [PMID: 39894336 DOI: 10.1016/j.biochi.2025.01.013] [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: 06/20/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
The glioma hallmark includes reprogramming metabolism to support biosynthetic and bioenergetic demands, as well as to maintain their redox equilibrium. It has been suggested that the pentose phosphate pathway (PPP) and glycolysis are directly involved in the dynamics and regulation of glioma cell proliferation and migration. The PPP is implicated in cellular redox homeostasis and the modulation of signaling pathways, which play a fundamental role in the progression of tumors to malignant grades, metastasis, and drug resistance. Several studies have shown that in glioblastoma cells, the activity, expression, and metabolic flux of some PPP enzymes increase, leading to heightened activity of the pathway. This generates higher levels of DNA, lipids, cholesterol, and amino acids, favoring rapid cell proliferation. Due to the crucial role played by the PPP in the development of glioma cells, enzymes from this pathway have been proposed as potential therapeutic targets. This review summarizes and highlights the role that the PPP plays in glioma cells and focuses on the key functions of the enzymes and metabolites generated by this pathway, as well as the regulation of the PPP. The studies described in this article enrich the understanding of the PPP as a therapeutic tool in the search for pharmacological targets for the development of a new generation of drugs to treat glioma.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico.
| | | | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, CDMX, 04530, Mexico.
| | - Rosa Angélica Castillo-Rodriguez
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, 62790, Xochitepec, Mexico.
| | - Francisca Palomares-Alonso
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico
| | - Carlos Castillo-Pérez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico.
| | - Dolores Jiménez-Farfán
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico.
| | - Aurora Sánchez-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología, 14080, Ciudad de Mexico, Mexico; Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico.
| |
Collapse
|
8
|
Tran RL, Li T, de la Cerda J, Schuler FW, Khaled AS, Pudakalakatti S, Bhattacharya PK, Sinharay S, Pagel MD. Potentiation of immune checkpoint blockade with a pH-sensitizer as monitored in two pre-clinical tumor models with acidoCEST MRI. Br J Cancer 2025; 132:744-753. [PMID: 39994445 PMCID: PMC11997056 DOI: 10.1038/s41416-025-02962-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Tumor acidosis causes resistance to immune checkpoint blockade (ICB). We hypothesized that a "pH-sensitizer" can increase tumor extracellular pH (pHe) and improve tumor control following ICB. We also hypothesized that pHe measured with acidoCEST MRI can predict improved tumor control with ICB. METHODS We tested the effects of pH-sensitizers on proton efflux rate (PER), cytotoxicity, T cell activation, tumor immunogenicity, tumor growth and survival using 4T1 and B16-F10 tumor cells. We measured in vivo tumor pHe of 4T1 and B16-F10 models with acidoCEST MRI. RESULTS Among the pH-sensitizers tested, someprazole caused the greatest reduction in PER without exhibiting cytotoxicity or reducing T cell activation. Esomeprazole improved 4T1 tumor control with ICB administered one day after the pH-sensitizer. Tumor pHe positively correlated with TCF-1 + CD4 effector and CD8 T cell intratumoral frequencies and predicted improved 4T1 tumor control with ICB. For comparison, esomeprazole had a mild effect on B16-F10 tumor pHe, and worsened tumor control with ICB and increased intratumoral myeloid and dendritic cell (DC) frequencies. CONCLUSIONS A pH-sensitizer can improve tumor control with ICB, and acidoCEST MRI can be used to measure pHe and predict tumor control, but only in the 4T1 model and not the B16-F10 model.
Collapse
Affiliation(s)
- Renee L Tran
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Tianzhe Li
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jorge de la Cerda
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - F William Schuler
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | - Alia S Khaled
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sanhita Sinharay
- Centre for Biosystems Science & Engineering, Indian Institute of Science, Bangalore, India
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Kim S, Jeong DI, Karmakar M, Huh JW, Hong EH, Kim DJ, Ko HJ, Cho HJ, Lee KB. Multifunctional Bioactive Dual-Layered Nanofibrous Matrix for Effective Breast Cancer Therapy and Enhanced Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500717. [PMID: 40296483 DOI: 10.1002/smll.202500717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/15/2025] [Indexed: 04/30/2025]
Abstract
Surgical resection is the primary treatment for triple-negative breast cancer (TNBC). Post-operative complications, including tumor recurrence and bacterial infections, hinder complete remission and long-term recovery. To address these challenges, a multifunctional bioactive dual-layered nanofibrous matrix (MBDL-NanoMat) featuring adaptive shape control, excellent wound adherence, tunable drug release profiles, and superior biocompatibility for post-surgical applications is developed. The MBDL-NanoMat comprises a hydrophilic (HyPhil) layer and a hydrophobic (HyPhob) layer, offering distinct functionalities. The HyPhil layer, electrospun with gelatin and copper peroxide nanoparticles (Cu NPs), rapidly releases Cu NPs to induce anticancer effects through chemodynamic therapy (CDT), ferroptosis, and cuproptosis along with antibacterial action. Near-infrared laser irradiation enhances therapeutic efficacy through photothermal therapy (PTT). The HyPhob layer ensures prolonged therapeutic effects by releasing therapeutic molecules, such as rapamycin, enabling sustained chemotherapy (CT) and antibacterial activity. This synergistic therapeutic system integrates multiple mechanisms-CT, CDT, PTT, ferroptosis, and cuproptosis-suppressing tumor recurrence and accelerating wound healing. Preclinical results demonstrated enhanced angiogenesis, collagen deposition, and dermal regeneration without systemic safety. In short, the MBDL-NanoMat platform offers a unique advantage in post-surgical TNBC care by simultaneously targeting tumor recurrence and facilitating wound healing. Further investigation of this platform can significantly improve oncological and regenerative medicine strategies.
Collapse
Affiliation(s)
- Sungyun Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Da In Jeong
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Mrinmoy Karmakar
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Ji-Won Huh
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Eun-Hye Hong
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Dae-Joon Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Hyun-Jong Cho
- Department of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
- Department of Regulatory Science Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Institute of Regulatory Innovation through Science (IRIS), Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
10
|
Xu W, Li Y, Zeng Z, Guo G. Crosstalk of lactate metabolism-related subtypes, establishment of a prognostic signature and immune infiltration characteristics in colon adenocarcinoma. Sci Rep 2025; 15:14599. [PMID: 40287503 PMCID: PMC12033353 DOI: 10.1038/s41598-025-98735-0] [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: 07/17/2024] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Colon adenocarcinoma (COAD) is a common malignant tumor of digestive tract and lactate metabolism has been linked to tumor development and progression. In this study, we sought to investigate the influence of lactate metabolism-related genes (LRGs) prognosis. We also aimed to identify distinct LRG-related clusters and develop a risk signature for assessing patient prognosis, immunological characteristics, and response to therapy. We analyzed data from The Cancer Genome Atlas (TCGA) to reveal the expression and mutational features of LRGs in COAD patients. In the integrated TCGA and GSE39582 cohort, consensus clustering analysis was employed to classify patients into two distinct LRG-related clusters. Using differentially expressed genes (DRGs) from these two clusters, we established a LRG-related gene cluster and prognostic signature which was used to classify patients into high-risk and low-risk groups. An validation cohort was used to validate the predictive ability of risk signature and expression of 6 candidate LRGs was confirmed through quantitative real-time PCR (qRT-PCR). Nomograms were created to visually represent the clinical value of LRG-related signature. Furthermore, we extensively examined differences in immune cell infiltration, tumor mutational load (TMB), microsatellite instability (MSI) and drug sensitivity between two risk groups. Analysis of the integrated TCGA and GSE39582 cohorts revealed two distinct LRG-related clusters and gene clusters with significant differences in overall survival (OS) and tumor microenvironment. We developed a LRG-related signature comprising 6 candidate LRGs that reliably predicted OS and qRT-PCR validation confirmed the expression of LRGs. Based on the median risk score, patients were divided into low-risk and high-risk groups, with low-risk group showing better survival. Furthermore, patients in high-risk group were more sensitive to chemotherapy and associated with higher TMB, higher proportion of MSI-H. Our study provides a valuable method for guiding clinical management and personalized treatment of COAD patients, which offers new insights into individualized treatment strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Wenwei Xu
- The Department of Gastrointestinal Surgery, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, China
| | - Yongjian Li
- The Department of Gastrointestinal Surgery, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, China
| | - Zhaoshang Zeng
- The Department of Gastrointestinal Surgery, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, China
| | - Guanjun Guo
- The Department of Gastrointestinal Surgery, Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Foshan, 528000, Guangdong, China.
| |
Collapse
|
11
|
Saha S, Tandon R, Sanku J, Kumari A, Shukla R, Srivastava N. siRNA-based Therapeutics in Hormone-driven Cancers: Advancements and benefits over conventional treatments. Int J Pharm 2025; 674:125463. [PMID: 40081431 DOI: 10.1016/j.ijpharm.2025.125463] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Hormone-related cancers, also known as hormone-sensitive or hormone-dependent cancers, rely on hormones such as estrogen, testosterone, and progesterone for growth. These malignancies, including breast, pituitary, thyroid, ovarian, uterine, cervical, and prostate cancers, often exhibit accelerated progression in response to hormonal signaling. Small interfering RNA (siRNA) has emerged as a groundbreaking gene suppression therapy since the FDA approval of its first product in 2018. With over 200 ongoing clinical trials, siRNA is being actively explored as a targeted treatment for hormone-related cancers. Its ability to silence specific oncogenes offers significant advantages over conventional therapies, which are often associated with toxicity, resistance, and non-specific targeting. However, challenges in siRNA delivery remain a major barrier to its clinical translation, limiting its ability to reach target cells effectively. This review evaluates the potential of siRNA in hormone-related cancers, addressing the shortcomings of traditional treatments while examining novel strategies to enhance siRNA delivery and overcome tumor microenvironment obstacles. Notably, no existing literature comprehensively consolidates siRNA-based therapies for these cancers, emphasizing the importance of this manuscript in bridging current knowledge gaps and advancing the translational application of siRNA therapeutics.
Collapse
Affiliation(s)
- Sayani Saha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Jhansi Sanku
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Anchala Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India.
| |
Collapse
|
12
|
Kim EY, Abides J, Keller CR, Martinez SR, Li W. Tumor Microenvironment Lactate: Is It a Cancer Progression Marker, Immunosuppressant, and Therapeutic Target? Molecules 2025; 30:1763. [PMID: 40333742 PMCID: PMC12029365 DOI: 10.3390/molecules30081763] [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: 03/10/2025] [Revised: 04/12/2025] [Accepted: 04/12/2025] [Indexed: 05/09/2025] Open
Abstract
The "Warburg effect" is a term coined a century ago for the preferential use of glycolysis over aerobic respiration in tumor cells for energy production, even under aerobic conditions. Although this is a less efficient mechanism of generating energy from glucose, aerobic glycolysis, in addition to the canonical anaerobic glycolysis, is an effective means of lactate production. The abundant waste product, lactate, yielded by the dual glycolysis in a tumor, has been discovered to be a major biomolecule that drives cancer progression. Lactate is a metabolic energy source that, via cell membrane lactate transporters, shuttles in and out of cancer cells as well as cancer cell-associated stromal cells and immune cells within the tumor microenvironment (TME). Additionally, lactate serves as a pH tuner, signaling ligand and transducer, epigenetic and gene transcription regulator, TME modifier, immune suppressor, chemoresistance modulator, and prognostic marker. With such broad functionalities, the production-consumption-reproduction of TME lactate fuels tumor growth and dissemination. Here, we elaborate on the lactate sources that contribute to the pool of lactate in the TME, the functions of TME lactate, the influence of the TME lactate on immune cell function and local tissue immunity, and anticancer therapeutic approaches adopting lactate manipulations and their efficacies. By scrutinizing these properties of the TME lactate and others that have been well addressed in the field, it is expected that a better weighing of the influence of the TME lactate on cancer development, progression, prognosis, and therapeutic efficacy can be achieved.
Collapse
Affiliation(s)
- Eugene Y. Kim
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Joyce Abides
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Chandler R. Keller
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| | - Steve R. Martinez
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Providence Regional Cancer Partnership, Providence Regional Medical Center, Everett, WA 98201, USA
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (E.Y.K.); (J.A.); (C.R.K.)
| |
Collapse
|
13
|
Ibe A, Nakanishi M, Higashimoto K, Muragaki Y, Ehata S. An acidic microenvironment promotes lymphatic metastasis of melanoma by Thy-1 in endothelial cells and integrin αvβ3 in tumor cells. Discov Oncol 2025; 16:498. [PMID: 40205271 PMCID: PMC11981969 DOI: 10.1007/s12672-025-02276-x] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Melanoma tissues exhibit an acidic microenvironment compared with that of surrounding normal tissues. However, the effects of acidic conditions on the lymphatic metastasis, a crucial prognostic factor for patients with melanoma, are unclear. In the present study, we aimed to investigate the role of the acidic microenvironment in the function of lymphatic endothelial cells. We first conducted gene expression profiling using human dermal lymphatic endothelial cells (HDLECs) treated with low pH media. Based on these results, we focused on Thy-1/CD90, whose expression increased in a time-dependent manner in HDLECs under acidic conditions. Immunohistochemical analysis of primary tumor tissues in a mouse melanoma model revealed an increased expression of Thy-1 in lymphatic endothelial cells. The expression of integrin αvβ3, a receptor for Thy-1, was also up-regulated in melanoma cells under acidic conditions. The adhesion of HDLECs to melanoma cells was accelerated under acidic conditions, which was reduced by Thy-1 knockdown in HDLECs. Furthermore, lymphatic metastasis was significantly attenuated in a mouse melanoma metastasis model when inoculated with integrin αv-silenced melanoma cells. These results suggest that acid-induced Thy-1 in lymphatic endothelial cells, as well as integrin αvβ3 in melanoma cells, may promote their mutual cellular adhesion, contributing to lymphatic metastasis.
Collapse
Affiliation(s)
- Akiya Ibe
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Masako Nakanishi
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Kurumi Higashimoto
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Yasuteru Muragaki
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Shogo Ehata
- Department of Pathology, School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| |
Collapse
|
14
|
Parbin NS, Banik B. Copper peroxide incorporated BSA-NPs: a pH-responsive, self-supplying source of reactive oxygen species for cancer cell destruction via polarization of macrophages to the M1 phenotype. Chem Commun (Camb) 2025; 61:5605-5608. [PMID: 40105257 DOI: 10.1039/d5cc00216h] [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: 03/20/2025]
Abstract
Herein, we produced self-assembled CuPer-BSA nanoparticles (NPs) formed as a result of incorporation of copper peroxide into bovine serum albumin (BSA) and serving as a self-supplying source of Cu2+ and H2O2 at biologically relevant acidic pH, and in these conditions activating a Fenton-type reaction to generate hydroxyl radicals (˙OH). ROS-induced polarization of RAW 264.7 macrophages to the M1 phenotype resulted in cellular secretions that showed significant anti-cancer efficacy against HeLa cells.
Collapse
Affiliation(s)
- Nursaima Sultana Parbin
- Department of Chemistry, Cotton University, Panbazar, Guwahati-781001, Assam, India.
- Department of Chemistry, Gauhati University, Gopinath Bordoloi Nagar, Guwahati-781014, Assam, India
| | - Bhabatosh Banik
- Department of Chemistry, Cotton University, Panbazar, Guwahati-781001, Assam, India.
| |
Collapse
|
15
|
Liu S, Liu C, He Y, Li J. Benign non-immune cells in tumor microenvironment. Front Immunol 2025; 16:1561577. [PMID: 40248695 PMCID: PMC12003390 DOI: 10.3389/fimmu.2025.1561577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
The tumor microenvironment (TME) is a highly complex and continuous evolving ecosystem, consisting of a diverse array of cellular and non-cellular components. Among these, benign non-immune cells, including cancer-associated fibroblasts (CAFs), adipocytes, endothelial cells (ECs), pericytes (PCs), Schwann cells (SCs) and others, are crucial factors for tumor development. Benign non-immune cells within the TME interact with both tumor cells and immune cells. These interactions contribute to tumor progression through both direct contact and indirect communication. Numerous studies have highlighted the role that benign non-immune cells exert on tumor progression and potential tumor-promoting mechanisms via multiple signaling pathways and factors. However, these benign non-immune cells may play different roles across cancer types. Therefore, it is important to understand the potential roles of benign non-immune cells within the TME based on tumor heterogeneity. A deep understanding allows us to develop novel cancer therapies by targeting these cells. In this review, we will introduce several types of benign non-immune cells that exert on different cancer types according to tumor heterogeneity and their roles in the TME.
Collapse
Affiliation(s)
- Shaowen Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chunhui Liu
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Li
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Department of Molecular Pathology, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
16
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [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: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
17
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Gao F, Shah R, Xin G, Wang R. Metabolic Dialogue Shapes Immune Response in the Tumor Microenvironment. Eur J Immunol 2025; 55:e202451102. [PMID: 40223597 PMCID: PMC11995254 DOI: 10.1002/eji.202451102] [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: 10/24/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025]
Abstract
The fate of immune cells is fundamentally linked to their metabolic program, which is also influenced by the metabolic landscape of their environment. The tumor microenvironment represents a unique system for intercellular metabolic interactions, where tumor-derived metabolites suppress effector CD8+ T cells and promote tumor-promoting macrophages, reinforcing an immune-suppressive niche. This review will discuss recent advancements in metabolism research, exploring the interplay between various metabolites and their effects on immune cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Fengxia Gao
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Rushil Shah
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| | - Gang Xin
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Ruoning Wang
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
19
|
Zhang G, Zhang X, Yue K, Zhong W. Mechanistic study of enhanced drug release in mixed pH-responsive peptide-loaded liposomes. J Biomol Struct Dyn 2025:1-15. [PMID: 40126078 DOI: 10.1080/07391102.2025.2481581] [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: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Liposomes serving as nanocarriers offer significant advantages in drug delivery for tumor treatment. There still exists challenges in controlling drug release by disintegrating the liposome membrane for the improvement of therapeutic efficiency. In this paper, a novel method involving the mixture of short peptides with pH-responsive characteristics into the cargo has been introduced. This approach facilitates the release of doxorubicin (DOX) in the acidic tumor tissue environment. The efficacy of this improvement was elucidated through molecular dynamics simulations and experiments. Liposomes incorporating a 1:1 ratio of peptides-DOX exhibited pronounced pH sensitivity and an enhanced drug release profile. The underlying mechanism is attributed to the peptides entering tumor tissues and undergoing protonation in acidic conditions, which increases the hydrophilicity of the peptide-DOX clusters and the internal surface tension of the liposomes. This alteration disrupts the balance between the inner and outer surface tensions of the nanocarrier, causing the liposomes to structurally disintegrate and thus enhancing drug release. The results from both thermodynamic analysis results and experimental data confirm the augmented drug release efficiency of this method, offering valuable theoretical insights for nanoparticle design and determining the optimal mixing ratio for therapeutic applications.
Collapse
Affiliation(s)
- Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Xilong Zhang
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| |
Collapse
|
20
|
Zhang L, Feng Q, Zheng C, Li Y, Ge X, Jin T, Hu G, Tan Z, Wang J, Xu J, Jiang L, Wang D, Ying Z, Zhao X, Cheng K, Li Q, Ge M. Antigen-Targeting Inserted Nanomicelles Guide Pre-Existing Immunity to Kill Head and Neck Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410629. [PMID: 40091501 DOI: 10.1002/advs.202410629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/02/2025] [Indexed: 03/19/2025]
Abstract
A significant challenge in cancer therapy is the identification of suitable targets that are specifically and uniformly expressed across heterogeneous tumors. The efficacy of pre-existing antiviral immunity in tumor treatment is limited by the absence of corresponding targets. This study develops a novel platform of antigen-targeted inserted nanomicelles, preS1 (an antigen of hepatitis B virus)-pHLIP nanomicelles, in which tumor-targeting nanomicelles release antigens that label tumor tissue for pre-existing immunity-mediated lysis in situ. In animal models of head and neck cancers, including head and neck squamous cell carcinoma and anaplastic thyroid cancer, preS1-pHLIP nanomicelles effectively inhibited tumor growth, recurrence, and metastasis in animals pre-immunized with preS1. This therapeutic effect is associated with an increase in the proportion of preS1-specific B cells and activated tumor-specific T cells within the tumor microenvironment. Overall, this work has engineered a nanomicelle that can disguise tumor cells as viruses and achieve tumor killing through the pre-existing antiviral immune response. This strategy presents a novel approach for treating tumors with ambiguous therapeutic target profiles.
Collapse
Affiliation(s)
- Lizhuo Zhang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| | - Qingqing Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, 310006, China
| | - Chuanming Zheng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| | - Yuanqiang Li
- Zhejiang Cancer Hospital, Hangzhou, 310005, China
| | - Xinyang Ge
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Tiefeng Jin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, China
| | - Gaofeng Hu
- Zhejiang Cancer Hospital, Hangzhou, 310005, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| | - Jiafeng Wang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
- Department of Thyroid and Breast Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, 551700, China
| | - Jiajie Xu
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| | - Liehao Jiang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| | - Dan Wang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | | | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou, 310005, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Zhejiang Provincial Clinical Research Center for Head & Neck Cancer, Hangzhou, 310014, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, 310014, China
| |
Collapse
|
21
|
Kondo A, Nakamura A. Overcoming cross-reactivity of antibodies against human lactate dehydrogenase. J Immunol Methods 2025; 538:113821. [PMID: 39900137 DOI: 10.1016/j.jim.2025.113821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/19/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025]
Abstract
Lactate dehydrogenase subunit A (LD-A) plays an important role in cancer regulation and therapy. We attempted to develop an enzyme-linked immune-solvent assay (ELISA) for LD-A in human serum. However, commercial antibodies against LD-A exhibited cross-reactivity with an unknown protein. The unknown protein was purified and characterized by protein sequencing and Western blotting. In addition, we attempted to prepare a specific antibody for the ELISA using partially synthesized peptides of LD-A as immunogens. The epitope position in LD-A was carefully selected based on bioinformatics analysis. Peptide sequencer elucidated a ten amino acid sequence of the purified protein at the N-terminal. A BLAST search revealed that this sequence perfectly matched that at the N-terminus of the IgG heavy chain (H-chain). Furthermore, we demonstrated that twelve commercially available antibodies targeting LD-A or LD-B (subunit B) primarily cross-reacted with IgG or its H-chain, with only one specific antibody for each subunit. As the specific antibody against LD-A is no longer commercially accessible, we successfully produced two kinds of specific antibodies using partially synthesized LD-A peptides as immunogens. In conclusion, we have successfully produced specific antibodies against LD-A. Moreover, our findings underscore the utility of bioinformatics tools for determining the optimal positions of immunizing peptides.
Collapse
Affiliation(s)
- Akira Kondo
- Department of Clinical Laboratory Science, Tenri University, Nara, Japan.
| | - Ayumu Nakamura
- Department of Clinical Laboratory Science, Tenri University, Nara, Japan
| |
Collapse
|
22
|
Li Y, Fu B, Jiang W. Emerging Roles of Nanozyme in Tumor Metabolism Regulation: Mechanisms, Applications, and Future Directions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11552-11577. [PMID: 39936939 DOI: 10.1021/acsami.4c20417] [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: 02/13/2025]
Abstract
Nanozymes, nanomaterials with intrinsic enzyme activity, have garnered significant attention in recent years due to their catalytic abilities comparable to natural enzymes, cost-effectiveness, high catalytic activities, and stability against environmental fluctuations. As functional analogs of natural enzymes, nanozymes participate in various critical metabolic processes, including glucose metabolism, lactate metabolism, and the maintenance of redox homeostasis, all of which are essential for normal cellular functions. However, disruptions in these metabolic pathways frequently promote tumorigenesis and progression, making them potential therapeutic targets. While several therapies targeting tumor metabolism are currently in clinical or preclinical stages, their efficacy requires further enhancement. Consequently, nanozymes that target tumor metabolism are regarded as a promising therapeutic strategy. Despite extensive studies investigating the application of nanozymes in tumor metabolism, relevant reviews are relatively scarce. This article first introduces the physicochemical properties and biological behaviors of nanozymes. Subsequently, we analyze the role of nanozymes in tumor metabolism and explore their potential applications in tumor therapy. In conclusion, this review aims to foster innovative research in related fields and advance the development of nanozyme-based strategies for cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Yikai Li
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, Jilin 130000, China
| | - Bowen Fu
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, Jilin 130000, China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| |
Collapse
|
23
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [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: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
24
|
Zhang Y, Li Y, Fang B, Du Y, Peng P. Framework Nucleic Acids: Innovative Tools for Cellular Sensing and Therapeutics. Chembiochem 2025; 26:e202400810. [PMID: 39653648 DOI: 10.1002/cbic.202400810] [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: 09/29/2024] [Revised: 12/07/2024] [Indexed: 12/18/2024]
Abstract
As emerging biomaterials, framework nucleic acids (FNAs) have recently demonstrated great potential in the biomedical field due to their high programmability, biocompatibility, unique structural diversity, and precise molecular design capabilities. This review focuses on the applications of FNAs in cellular sensing and disease treatment. First, we systematically introduce the applications of FNAs in cellular sensing, including their precise recognition and response to the extracellular tumor microenvironment, cell membrane proteins, and intracellular biomarkers. Subsequently, we review the potential of FNAs in disease treatment, covering their applications and development in drug delivery, regulation of cell behavior, and immunomodulation. We also discuss the limitations and potential role of FNAs in personalized medicine, precision diagnostics, and advanced therapies. The broad application of FNAs is expected to drive significant breakthroughs in future biomedical technological innovations and clinical translation.
Collapse
Affiliation(s)
- Yihan Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yuting Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Bowen Fang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yi Du
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Pai Peng
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| |
Collapse
|
25
|
Cortini M, Ilieva E, Massari S, Bettini G, Avnet S, Baldini N. Uncovering the protective role of lipid droplet accumulation against acid-induced oxidative stress and cell death in osteosarcoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167576. [PMID: 39561857 DOI: 10.1016/j.bbadis.2024.167576] [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: 04/29/2024] [Revised: 10/14/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Extracellular acidosis stemming from altered tumor metabolism promotes cancer progression by enabling tumor cell adaptation to the hostile microenvironment. In osteosarcoma, we have previously shown that acidosis increases tumor cell survival alongside substantial lipid droplet accumulation. In this study, we explored the role of lipid droplet formation in mitigating cellular stress induced by extracellular acidosis in osteosarcoma cells, thereby enhancing tumor survival during progression. Specifically, we examined how lipid droplets shield against reactive oxygen species induced by extracellular acidosis. We demonstrated that lipid droplet biogenesis is critical for acid-exposed tumor cell survival, as it starts shortly after acid exposure (24 h) and inversely correlates with ROS levels (DCFH-DA assay), lipid peroxidation (Bodipy assay), and the antioxidant response, as also revealed by NRF2 transcript. Additionally, extracellular metabolites, such as lactate, and interaction with mesenchymal stromal cells within the tumor microenvironment intensify lipid droplet build-up in osteosarcoma cells. Critically, upon targeting two key proteins implicated in LD formation - PLIN2 and DGAT1 - cell viability significantly declined while ROS production escalated. In summary, our findings underscore the vital reliance of acid-exposed tumor cells on lipid droplet formation to scavenge oxidative stress. We conclude that the rewiring of lipid metabolism driven by microenvironmental cues is of paramount importance for the survival of metabolically altered osteosarcoma cells in acidic condition. Overall, we suggest that targeting key members of lipid droplet biogenesis may eradicate more aggressive and resistant tumor cells, uncovering potential new treatment strategies for osteosarcoma.
Collapse
Affiliation(s)
- Margherita Cortini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Elizabeta Ilieva
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Stefania Massari
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy
| | - Giuliano Bettini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, 40100 Ozzano dell'Emilia, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy.
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, Università di Bologna, 40127 Bologna, Italy; Biomedical Science, Technology and Nanobiotechnology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| |
Collapse
|
26
|
Ma R, Zhang Y, Ji H, Fu H, Gu S, Su Q, Lin Y, Deng Q, Xue W, Yang Y. Glucose oxidase-driven self-accelerating drug release nanosystem based on metal-phenolic networks orchestrates tumor chemotherapy and ferroptosis-based therapy. Int J Biol Macromol 2025; 290:139103. [PMID: 39716697 DOI: 10.1016/j.ijbiomac.2024.139103] [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: 11/01/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
Nanocarriers responding to tumor microenvironment have been extensively exploited to improve the antitumor outcome of chemotherapeutic drugs. However, selectively and completely releasing drugs within the tumor remains a challenge, thereby limiting the therapeutic effect of drug delivery nanosystem. To tackle this challenge, a metal-phenolic networks (MPNs)-based nanosystem (F-MGD) showing the capability of self-accelerating drug release was originally fabricated in this study. Glucose oxidase (GOx) encapsulated in F-MGD could conduct the glucose transformation in tumor to cause the oxygen consumption and the production of gluconic acid and H2O2. Therefore, F-MGD with acid and hypoxia sensitivities thoroughly disintegrated under the aggravated acidity and hypoxia to achieve a more complete drug release. Besides, the product of H2O2 was readily decomposed into hydroxyl radicals via the iron ion-mediated Fenton reaction, which markedly augmented the oxidative stress in tumor cells and promoted ferroptosis. The results of both in vitro and in vivo assays demonstrated the significant antitumor efficacy of F-MGD. Collectively, this study proposes a strategy to expedite drug release in tumor and improve the tumor treatment effect by combining ferroptosis-based therapy and chemotherapy.
Collapse
Affiliation(s)
- Rongying Ma
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yufei Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hongting Ji
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Huiling Fu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Shuzhen Gu
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qianhong Su
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yumian Lin
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Qingchun Deng
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Yang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
27
|
Greenshields-Watson A, Vavourakis O, Spoendlin FC, Cagiada M, Deane CM. Challenges and compromises: Predicting unbound antibody structures with deep learning. Curr Opin Struct Biol 2025; 90:102983. [PMID: 39862761 DOI: 10.1016/j.sbi.2025.102983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Therapeutic antibodies are manufactured, stored and administered in the free state; this makes understanding the unbound form key to designing and improving development pipelines. Prediction of unbound antibodies is challenging, specifically modelling of the CDRH3 loop, where inaccuracies are potentially worse due to a bias in structural data towards antibody-antigen complexes. This class imbalance provides a challenge for deep learning models trained on this data, potentially limiting generalisation to unbound forms. Here we discuss the importance of unbound structures in antibody development pipelines. We explore how the latest generation of structure predictors can provide new insights and assess how conformational heterogeneity may influence binding kinetics. We hypothesise that generative models may address some of these issues. While prediction of antibodies in complex is essential, we should not ignore the need for progress in modelling the unbound form.
Collapse
Affiliation(s)
- Alexander Greenshields-Watson
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom.
| | - Odysseas Vavourakis
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| | - Fabian C Spoendlin
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| | - Matteo Cagiada
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom; Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, United Kingdom
| |
Collapse
|
28
|
Tang S, Li Y, Fang Y, Tu M, Wu S, Cen Y, Xu J. Simultaneously delivery of functional gallium ions and hydrogen sulfide to endow potentiated treatment efficacy in chemo- and PARPi-resistant ovarian cancer. J Nanobiotechnology 2025; 23:73. [PMID: 39893477 PMCID: PMC11786553 DOI: 10.1186/s12951-025-03167-7] [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: 11/29/2024] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
Limited therapeutic options are available for patients with platinum-resistant ovarian cancer (OC). Herein, we developed gallium sulfide-embedded bovine serum albumin nanoformulations (Ga2S3-BSA NMs) with a size of ~ 11 nm via a self-assembly approach. As the nanoformulations degraded in an acidic cancer microenvironment, Ga3+ and H2S gas were simultaneously released to exert their combined anticancer effects. In A2780-CIS and SKOV3-CIS platinum-resistant OC cells, Ga3+ and H2S released from Ga2S3-BSA NMs synergistically enhanced DNA damage, which arrested the cell cycle at S and G2/M phases and suppressed cell proliferation. Meanwhile, Ga2S3-BSA NMs significantly inhibited NF-κB signaling and Bcl2 protein expression, leading to cell apoptosis. Furthermore, Ga2S3-BSA NMs increased cellular lipid peroxidation and triggered ferroptosis. RNA-seq analysis further clarified the comprehensive antitumor mechanisms of Ga2S3-BSA NMs. More importantly, the therapeutic efficacy of Ga2S3-BSA NMs and their ability to enhance the sensitivity to carboplatin and fluzoparib with negligible toxicity were further confirmed in a platinum-resistant OC animal model. Altogether, our results demonstrated a potentially safe and practical strategy by using Ga2S3-BSA NMs to combat drug resistance in platinum-resistant OC.
Collapse
Affiliation(s)
- Sangsang Tang
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yangyang Li
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yifeng Fang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Mengyan Tu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Shenglong Wu
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yixuan Cen
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, Zhejiang, 310020, China.
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, #1 Xueshi Road, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
29
|
Quaresima B, Scicchitano S, Faniello MC, Mesuraca M. Role of solute carrier transporters in ovarian cancer (Review). Int J Mol Med 2025; 55:24. [PMID: 39611477 PMCID: PMC11637498 DOI: 10.3892/ijmm.2024.5465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/11/2024] [Indexed: 11/30/2024] Open
Abstract
Solute carrier (SLC) transporters are involved in various biological processes associated with metabolic reprogramming and cancer, supporting the increased requirement of nutrients and energy. Over the past decade, there have been significant advancements in understanding the expression and function of SLCs in ovarian cancer (OC). This gynecological condition has a high mortality rate and limited treatment options; thus, early diagnosis remains a target clinically. OC exhibits complexity and heterogeneity, resulting in different clinical characteristics, resistance to chemotherapy drugs and poor prognosis. Additionally, SLCs have a different expression pattern between healthy and tumor tissue, and consequently, their inhibition or activation could modify signaling pathways involved in the tumor growth process, such as cell proliferation, apoptosis and drug accumulation. The present review aims to consolidate current data to provide a comprehensive understanding of the potential importance of SLCs in OC. Additionally, it seeks to offer guidance for further research on utilizing SLCs as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Barbara Quaresima
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, I-88100 Catanzaro, Italy
| | - Stefania Scicchitano
- Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, I-88100 Catanzaro, Italy
| | | | - Maria Mesuraca
- Correspondence to: Dr Maria Mesuraca or Dr Barbara Quaresima, Department of Experimental and Clinical Medicine, 'Magna Graecia' University of Catanzaro, Viale Europa, I-88100 Catanzaro, Italy, E-mail: , E-mail:
| |
Collapse
|
30
|
Liu L, Fu S, Gu H, Li Y, Zhu G, Ai H, Li W. Platinum(IV)-Backboned Polymer Prodrug-Functionalized Manganese Oxide Nanoparticles for Enhanced Lung Cancer Chemoimmunotherapy via Amplifying Stimulator of Interferon Genes Activation. ACS NANO 2025; 19:2726-2741. [PMID: 39772457 DOI: 10.1021/acsnano.4c15115] [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: 01/11/2025]
Abstract
The stimulator of interferon genes (STING) pathway exhibits great potential in remodeling the immunosuppressive tumor microenvironment and initiating antitumor immunity. However, how to effectively activate STING and avoid undesired toxicity after systemic administration remains challenging. Herein, platinum(IV)-backboned polymer prodrug-coated manganese oxide nanoparticles (DHP/MnO2NP) with pH/redox dual responsive properties are developed to precisely release cisplatin and Mn2+ in the tumor microenvironment and synergistically amplify STING activation. In vitro, we demonstrate that DHP/MnO2NP can effectively induce tumor cell DNA damage and leak into the cytoplasm, cooperating with Mn2+ to promote STING activation and significantly upregulate the expression of proinflammatory cytokines. Additionally, DHP/MnO2NP can selectively release cisplatin and Mn2+ to mediate tumor killing while reducing toxicity to normal cells. In vivo, DHP/MnO2NP exerted increased therapeutic efficacy by inducing STING activation and initiating robust antitumor immunity. Specifically, DHP/MnO2NP effectively skewed tumor-associated macrophages toward a proinflammatory phenotype and upregulated the expression of proinflammatory cytokines in tumors by up to 99-fold relative to the control. And the infiltration of CD8+ T cells was also significantly increased. When STING signaling was blocked, the antitumor effects and immunostimulatory efficacy of DHP/MnO2NP were significantly inhibited. Moreover, DHP/MnO2NP possess the advantage of enhanced tumor homing and retention, resulting in stronger and longer-lasting anticancer effects. Overall, DHP/MnO2NP provide a potential platform for potentiating cancer chemoimmunotherapy and hold promise for precision treatment.
Collapse
Affiliation(s)
- Li Liu
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Shengxiang Fu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, P. R. China
| | - Haojie Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P. R. China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Guonian Zhu
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, P. R. China
| | - Weimin Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Chengdu 610041, P. R. China
| |
Collapse
|
31
|
Cerioli N, Bououdina W, Mereu A, Natsaridis E, Salsetta J, Cova A, Lupoli G, D’Angelo E, Rivoltini L, Figdor CG, Huber V, Tagit O. Reprogramming the melanoma and immunosuppressive myeloid cells with esomeprazole-loaded PLGA nanoparticles. iScience 2025; 28:111559. [PMID: 39839438 PMCID: PMC11750290 DOI: 10.1016/j.isci.2024.111559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/04/2024] [Accepted: 12/05/2024] [Indexed: 01/23/2025] Open
Abstract
Proton pump inhibitors have been explored for potentiating cancer therapies via reverting the tumor acidity and promoting the activation of anti-tumor immune responses. To regulate the intracellular pH of melanoma and immunosuppressive myeloid cells, we developed poly(L-lactide-co-glycolide) nanoparticles loaded with esomeprazole (ESO-NPs). The effect of ESO-NPs on melanoma cells was observed as alkalinization and reduction of melanin content accompanied by a decrease of microphthalmia-associated transcription factor (MITF), poliovirus receptor (PVR), and programmed death ligand 1 (PD-L1) immune checkpoint expression. ESO-NP treatment of melanoma-patient-derived and in vitro-induced myeloid-derived suppressor cells (MDSCs) reduced the expression of immunosuppression-associated molecules PD-L1, CD206, and CD163 on patient-derived myeloid cells while inducing the expression of co-stimulatory molecule CD86 and HLA-DR in the in vitro model. Our findings suggest that reprogramming the intracellular pH of melanoma and immune-suppression-associated myeloid cells with ESO-NPs can modulate the expression of proteins involved in resistance to cancer therapy and immunosuppression, thus potentially improving the response to immunotherapies.
Collapse
Affiliation(s)
- Nicola Cerioli
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Wissem Bououdina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alessandro Mereu
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Evangelos Natsaridis
- Group of Biointerfaces, Institute of Chemistry and Bioanalytics, School of Life Sciences FHNW, Muttenz, Switzerland
| | - Jeannette Salsetta
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Agata Cova
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianpiero Lupoli
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa D’Angelo
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Licia Rivoltini
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carl G. Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Veronica Huber
- Translational Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Oya Tagit
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Group of Biointerfaces, Institute of Chemistry and Bioanalytics, School of Life Sciences FHNW, Muttenz, Switzerland
| |
Collapse
|
32
|
Sabit H, Arneth B, Pawlik TM, Abdel-Ghany S, Ghazy A, Abdelazeem RM, Alqosaibi A, Al-Dhuayan IS, Almulhim J, Alrabiah NA, Hashash A. Leveraging Single-Cell Multi-Omics to Decode Tumor Microenvironment Diversity and Therapeutic Resistance. Pharmaceuticals (Basel) 2025; 18:75. [PMID: 39861138 PMCID: PMC11768313 DOI: 10.3390/ph18010075] [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: 10/31/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Recent developments in single-cell multi-omics technologies have provided the ability to identify diverse cell types and decipher key components of the tumor microenvironment (TME), leading to important advancements toward a much deeper understanding of how tumor microenvironment heterogeneity contributes to cancer progression and therapeutic resistance. These technologies are able to integrate data from molecular genomic, transcriptomic, proteomics, and metabolomics studies of cells at a single-cell resolution scale that give rise to the full cellular and molecular complexity in the TME. Understanding the complex and sometimes reciprocal relationships among cancer cells, CAFs, immune cells, and ECs has led to novel insights into their immense heterogeneity in functions, which can have important consequences on tumor behavior. In-depth studies have uncovered immune evasion mechanisms, including the exhaustion of T cells and metabolic reprogramming in response to hypoxia from cancer cells. Single-cell multi-omics also revealed resistance mechanisms, such as stromal cell-secreted factors and physical barriers in the extracellular matrix. Future studies examining specific metabolic pathways and targeting approaches to reduce the heterogeneity in the TME will likely lead to better outcomes with immunotherapies, drug delivery, etc., for cancer treatments. Future studies will incorporate multi-omics data, spatial relationships in tumor micro-environments, and their translation into personalized cancer therapies. This review emphasizes how single-cell multi-omics can provide insights into the cellular and molecular heterogeneity of the TME, revealing immune evasion mechanisms, metabolic reprogramming, and stromal cell influences. These insights aim to guide the development of personalized and targeted cancer therapies, highlighting the role of TME diversity in shaping tumor behavior and treatment outcomes.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza 3237101, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldingerstr. 1, 35043 Marburg, Germany
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH 43210, USA
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza 3237101, Egypt
| | - Aysha Ghazy
- Department of Agricultural Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza 3237101, Egypt
| | - Rawan M. Abdelazeem
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza 3237101, Egypt
| | - Amany Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Ibtesam S. Al-Dhuayan
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Jawaher Almulhim
- Department of Biological Sciences, King Faisal University, Alahsa 31982, Saudi Arabia
| | - Noof A. Alrabiah
- Department of Biological Sciences, King Faisal University, Alahsa 31982, Saudi Arabia
| | - Ahmed Hashash
- Department of Biomedicine, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
33
|
Kim TM, Subba SH, Hwang YK, Kim SG, Park J, Jin EJ, Park SY. Electrical and fluorescence in situ monitoring of tumor microenvironment-based pH-responsive polymer dot coated surface. Talanta 2025; 281:126840. [PMID: 39265419 DOI: 10.1016/j.talanta.2024.126840] [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: 06/19/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
A boronate-ester structure forming a pH-responsive polymer dot (Plu-PD) coated biosensor between carbonized-sp2 rich dopamine-alginate [PD(Alg)] and boronic acid-grafted Pluronic (BA-Pluronic) was developed for the electrochemical and fluorescence detection of cancer cells. The reduced fluorescence (FL) resulting from fluorescence resonance energy transfer (FRET) mediated by π-π interactions within Plu-PD was successfully reinvigorated through the specific cleavage of the boronate-ester bond, triggered by the acidic conditions prevailing in the cancer microenvironment. The anomalous variations in extracellular pH levels observed in cancer (pH ∼6.8), as opposed to the normal cellular pH range of approximately 7.4, serve as robust indicators for discerning cancer cells from their healthy counterparts. Moreover, the Plu-PD coated surface demonstrated remarkable adaptability in modulating its surface structure, concurrently exhibiting tunable electroconductivity under reduced pH conditions, thereby imparting selective responsiveness to cancer cells. The pH-modulated conductivity change was validated by a reduction in resistance from 211 ± 9.7 kΩ at pH 7.4 to 73.9 ± 9.4 kΩ and 61.5 ± 11.5 kΩ at pH 6.8 and 6.0, respectively. The controllable electrochemical characteristics were corroborated through in vitro treatment of cancer cells (HeLa, B16F10, and SNU-C2A) via LED experiments and wireless output analysis. In contrast, identical treatments yielded a limited response in normal cell line (CHO-K1). Notably, the Plu-PD coated surface can be seamlessly integrated with a wireless system to facilitate real-time monitoring of the sensing performance in the presence of cancer and normal cells, enabling rapid and accurate cancer diagnosis using a smartphone.
Collapse
Affiliation(s)
- Tae Min Kim
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Sunu Hangma Subba
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Yoon Kil Hwang
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Seul Gi Kim
- Department of Green Bio Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Junyoung Park
- Department of Biological Sciences, College of Health Sciences, Wonkwang University, Jeonbuk, 54538, Iksan, Republic of Korea
| | - Eun-Jung Jin
- Department of Biological Sciences, College of Health Sciences, Wonkwang University, Jeonbuk, 54538, Iksan, Republic of Korea
| | - Sung Young Park
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Green Bio Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
34
|
Wu X, Ma L, Zhang Y, Liu S, Cheng L, You C, Dong Z. Application progress of nanomaterials in the treatment of prostate cancer. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:1-12. [PMID: 39187009 DOI: 10.1016/j.pharma.2024.08.009] [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: 07/12/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Prostate cancer is one of the most common malignant tumors in men, which seriously threatens the survival and quality of life of patients. At present, there are serious limitations in the treatment of prostate cancer, such as drug tolerance, drug resistance and easy recurrence. Sonodynamic therapy and chemodynamic therapy are two emerging tumor treatment methods, which activate specific drugs or sonosensitizers through sound waves or chemicals to produce reactive oxygen species and kill tumor cells. Nanomaterials are a kind of nanoscale materials with many excellent physical properties such as high targeting, drug release regulation and therapeutic monitoring. Sonodynamic therapy and chemodynamic therapy combined with the application of nanomaterials can improve the therapeutic effect of prostate cancer, reduce side effects and enhance tumor immune response. This article reviews the application progress of nanomaterials in the treatment of prostate cancer, especially the mechanism, advantages and challenges of nanomaterials in sonodynamic therapy and chemodynamic therapy, which provides new ideas and prospects for research in this field.
Collapse
Affiliation(s)
- Xuewu Wu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Longtu Ma
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Yang Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, China
| | - Shuai Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Long Cheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Chengyu You
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China
| | - Zhilong Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Gansu 730030, China.
| |
Collapse
|
35
|
Ren X, Deng S, Xiang L, Gu H, Tang Y, Wang Y, Tong S, Peng L, Liu D. Positive associations between dietary potential acid load and renal cancer incidence and mortality: Results from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening trial. Public Health 2025; 238:229-238. [PMID: 39689651 DOI: 10.1016/j.puhe.2024.12.004] [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: 09/11/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVES To explore the relationships between dietary potential acid load and renal cancer (RC) incidence and mortality. STUDY DESIGN A prospective cohort study involving 97,166 U S. adults aged 55-74 years. METHODS Data utilized in this study were drawn from the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO). Dietary potential acid load was assessed by dietary acid load (DAL), potential renal acid load (PRAL) and net endogenous acid production (NEAP) scores using a validated Diet History Questionnaire (DHQ). Cox proportional hazards regression was used to estimate hazard ratios (HRs) and 95 % confidence intervals (CIs) for RC incidence and mortality with adjustment for potential confounders. Restricted cubic spline (RCS) plots were utilized to reveal whether there were nonlinear associations between the DAL, PRAL, and NEAP scores and RC incidence and mortality. RESULTS During a follow-up period of 859,907 and 1,467,573 person-years, 423 RC cases and 221 mortality cases were documented. DAL, PRAL, and NEAP scores were positively associated with RC incidence (DAL: HR 1.41; 95% CI 1.05, 1.90; P-trend = 0.010; PRAL: HR 1.45; 95% CI 1.10, 1.91; P-trend = 0.010; NEAP: HR 1.39; 95% CI 1.04, 1.85; P-trend = 0.029). Similar associations were observed between DAL, PRAL, and NEAP scores and RC mortality. The RCS plots demonstrated a nonlinear association between DAL score and RC risk, while a linear association was observed between DAL score and RC mortality. Similar linear associations were found between PRAL and NEAP scores and RC incidence and mortality. CONCLUSION A higher dietary potential acid load was associated with higher risk of RC incidence and mortality in American adults.
Collapse
Affiliation(s)
- Xiaorui Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shijiang Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhao Tang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiwen Tong
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Dengliang Liu
- Department of Gastrointestinal Surgery, Chongqing Jiulongpo People's Hospital, Chongqing, China; Department of General Surgery, Xipeng Town Health Center of Jiulongpo District, Chongqing, China.
| |
Collapse
|
36
|
Peng T, Shao X, Song W, Xu W, Xiong W, He Y, Ding Y, Huang Y. Intratumoral lactic acid neutralization strategy for boosting chemoimmunotherapy using liposomal sodium bicarbonate. Sci Bull (Beijing) 2024; 69:3936-3948. [PMID: 39547906 DOI: 10.1016/j.scib.2024.08.042] [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: 01/09/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 11/17/2024]
Abstract
Glycolysis-related lactic acid overproduction creates an "ion-trapping" barrier and immunosuppressive tumor microenvironment that compromise effective intratumoral drug delivery and therapy. Therefore, normalization of tumor microenvironment via lactic acid neutralization can be a promising avenue for overcoming this therapeutic hurdle. In this study, the flexible liposomes loaded with sodium bicarbonate (NaHCO3@Flip) were used as a nano-adjuvant to boost chemoimmunotherapy. Their effects on assisting DOXIL and anti-programmed cell death protein 1 (PD-1) therapy were investigated. NaHCO3@Flip achieved deep tumor penetration, with the ability to neutralize lactic acid and normalize the acidic tumor microenvironment. NaHCO3@Flip is biosafe and can enhance cellular uptake efficiency of doxorubicin (DOX) by overcoming the ion-trapping barrier and amplify immunogenic cell death induced by DOX. The combination therapy of liposomal DOX and NaHCO3@Flip demonstrated enhanced inhibition of tumor growth. NaHCO3@Flip can also synergize with PD-1 antibody therapy. NaHCO3@Flip has the potential to serve as a therapeutic adjuvant for boosting chemoimmunotherapy by overcoming the ion-trapping effect and normalizing the tumor microenvironment.
Collapse
Affiliation(s)
- Taoxing Peng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinyue Shao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqin Song
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weihua Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wei Xiong
- Zhongshan Institute for Drug Discovery, Shanghai Institutes of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yihao He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; Zhongshan Institute for Drug Discovery, Shanghai Institutes of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| |
Collapse
|
37
|
Sekar S, Srikanth S, Mukherjee AG, Gopalakrishnan AV, Wanjari UR, Vellingiri B, Renu K, Madhyastha H. Biogenesis and functional implications of extracellular vesicles in cancer metastasis. Clin Transl Oncol 2024:10.1007/s12094-024-03815-8. [PMID: 39704958 DOI: 10.1007/s12094-024-03815-8] [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: 06/21/2024] [Accepted: 11/23/2024] [Indexed: 12/21/2024]
Abstract
Extracellular vesicles (EVs) play a crucial role in the complex process of cancer metastasis by facilitating cellular communication and influencing the microenvironment to promote the spread and establishment of cancer cells in distant locations. This paper explores the process of EV biogenesis, explaining their various sources that range from endosomal compartments to plasma membrane shedding. It also discusses the complex mechanisms that control the sorting of cargo within EVs, determining their chemical makeup. We investigate the several functions of EVs in promoting the spread of cancer to other parts of the body. These functions include influencing the immune system, creating environments that support the formation of metastases before they occur, and aiding in the transformation of cells from an epithelial to a mesenchymal state. Moreover, we explore the practical consequences of EV cargo, such as nucleic acids, proteins, and lipids, in influencing the spread of cancer cells, from the beginning of invasion to the creation of secondary tumor sites. Examining recent progress in the field of EV-based diagnostics and treatments, we explore the potential of EVs as highly promising biomarkers for predicting the course of cancer and as targets for therapeutic intervention. This review aims to provide a complete understanding of the biology of EVs in the context of cancer metastasis. By unravelling the nuances of EV biology, it seeks to pave the way for new tactics in cancer detection, treatment, and management.
Collapse
Affiliation(s)
- Sneha Sekar
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sandhya Srikanth
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab, 151401, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 600077, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| |
Collapse
|
38
|
Jia W, Czabanka M, Broggini T. Cell blebbing novel therapeutic possibilities to counter metastasis. Clin Exp Metastasis 2024; 41:817-828. [PMID: 39222238 PMCID: PMC11607095 DOI: 10.1007/s10585-024-10308-z] [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: 05/23/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Cells constantly reshape there plasma membrane and cytoskeleton during physiological and pathological processes (Hagmann et al. in J Cell Biochem 73:488-499, 1999). Cell blebbing, the formation of bulges or protrusions on the cell membrane, is related to mechanical stress, changes in intracellular pressure, chemical signals, or genetic anomalies. These membrane bulges interfere with the force balance of actin filaments, microtubules, and intermediate filaments, the basic components of the cytoskeleton (Charras in J Microsc 231:466-478, 2008). In the past, these blebs with circular structures were considered apoptotic markers (Blaser et al. in Dev Cell 11:613-627, 2006). Cell blebbing activates phagocytes and promotes the rapid removal of intrinsic compartments. However, recent studies have revealed that blebbing is associated with dynamic cell reorganization and alters the movement of cells in-vivo and in-vitro (Charras and Paluch in Nat Rev Mol Cell Biol 9:730-736, 2008). During tumor progression, blebbing promotes invasion of cancer cells into blood, and lymphatic vessels, facilitating tumor progression and metastasis (Weems et al. in Nature 615:517-525, 2023). Blebbing is a dominant feature of tumor cells generally absent in normal cells. Restricting tumor blebbing reduces anoikis resistance (survival in suspension) (Weems et al. in Nature 615:517-525, 2023). Hence, therapeutic intervention with targeting blebbing could be highly selective for proliferating pro-metastatic tumor cells, providing a novel therapeutic pathway for tumor metastasis with minimal side effects. Here, we review the association between cell blebbing and tumor cells, to uncover new research directions and strategies for metastatic cancer therapy. Finaly, we aim to identify the druggable targets of metastatic cancer in relation to cell blebbing.
Collapse
Affiliation(s)
- Weiyi Jia
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Broggini
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
39
|
Ge M, Jiang F, Lin H. Nanocatalytic medicine enabled next-generation therapeutics for bacterial infections. Mater Today Bio 2024; 29:101255. [PMID: 39381264 PMCID: PMC11459013 DOI: 10.1016/j.mtbio.2024.101255] [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: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
The rapid rise of antibiotic-resistant strains and the persistence of biofilm-associated infections have significantly challenged global public health. Unfortunately, current clinical high-dose antibiotic regimens and combination therapies often fail to completely eradicate these infections, which can lead to adverse side effects and further drug resistance. Amidst this challenge, however, the burgeoning development in nanotechnology and nanomaterials brings hopes. This review provides a comprehensive summary of recent advancements in nanomaterials for treating bacterial infections. Firstly, the research progress of catalytic therapies in the field of antimicrobials is comprehensively discussed. Thereafter, we systematically discuss the strategies of nanomaterials for anti-bacterial infection therapies, including endogenous response catalytic therapy, exogenous stimulation catalytic therapy, and catalytic immunotherapy, in order to elucidate the mechanism of nanocatalytic anti-infections. Based on the current state of the art, we conclude with insights on the remaining challenges and future prospects in this rapidly emerging field.
Collapse
Affiliation(s)
- Min Ge
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Feng Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Han Lin
- Shanghai Institute of Ceramics Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai, 200050, China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, China
| |
Collapse
|
40
|
Zheng G, Shi J, Li Q, Jin X, Fang Y, Zhang Z, Cao Q, Zhu L, Shen J. BAP1 inactivation promotes lactate production by leveraging the subcellular localization of LDHA in melanoma. Cell Death Discov 2024; 10:483. [PMID: 39587076 PMCID: PMC11589756 DOI: 10.1038/s41420-024-02250-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024] Open
Abstract
BRCA1-associated protein 1 (BAP1) acts as a tumor suppressor and can affect the cell cycle, tumor immunity, and cellular metabolism through multiple pathways. In melanoma, BAP1 mutations promote tumor cell glycolysis, leading to increased lactate production. The tumor microenvironment with high lactate levels is often associated with immunosuppression and tumor progression. The inhibitory effect of BAP1 on glycolysis has been found in a variety of tumors, but the specific mechanism by which BAP1 inhibits lactate production still needs to be elucidated. In this study, we show that BAP1 can interact directly with lactate dehydrogenase (LDHA), causing LDHA to accumulate in the nucleus. Conversely, BAP1 deletion leads to the accumulation of LDHA in the cytoplasm, catalyzing the production of lactate from pyruvate that results in increased lactate levels inside and outside the cell. By elucidating the interaction between BAP1 and LDHA and the subsequent effects on lactate production in melanoma cells, this work provides insights into the mechanism of BAP1-mediated metabolic regulation. Furthermore, it may provide novel directions for the clinical treatment of BAP1-mutant melanoma.
Collapse
Affiliation(s)
- Guopei Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qian Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoliang Jin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Yan Fang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhe Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Lili Zhu
- Songjiang Research Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
- Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
41
|
Oliveira I, Rodrigues-Santos P, Ferreira L, Pires das Neves R. Synthetic and biological nanoparticles for cancer immunotherapy. Biomater Sci 2024; 12:5933-5960. [PMID: 39441658 DOI: 10.1039/d4bm00995a] [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: 10/25/2024]
Abstract
Cancer is becoming the main public health problem globally. Conventional chemotherapy approaches are slowly being replaced or complemented by new therapies that avoid the loss of healthy tissue, limit off-targets, and eradicate cancer cells. Immunotherapy is nowadays an important strategy for cancer treatment, that uses the host's anti-tumor response by activating the immune system and increasing the effector cell number, while, minimizing cancer's immune-suppressor mechanisms. Its efficacy is still limited by poor therapeutic targeting, low immunogenicity, antigen presentation deficiency, impaired T-cell trafficking and infiltration, heterogeneous microenvironment, multiple immune checkpoints and unwanted side effects, which could benefit from improved delivery systems, able to release immunotherapeutic agents to tumor microenvironment and immune cells. Nanoparticles (NPs) for immunotherapy (Nano-IT), have a huge potential to solve these limitations. Natural and/or synthetic, targeted and/or stimuli-responsive nanoparticles can be used to deliver immunotherapeutic agents in their native conformations to the site of interest to enhance their antitumor activity. They can also be used as co-adjuvants that enhance the activity of IT effector cells. These nanoparticles can be engineered in the natural context of cell-derived extracellular vesicles (EVs) or exosomes or can be fully synthetic. In this review, a detailed SWOT analysis is done through the comparison of engineered-synthetic and naturaly-derived nanoparticles in terms of their current and future use in cancer immunotherapy.
Collapse
Affiliation(s)
- Inês Oliveira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Paulo Rodrigues-Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- Center for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
42
|
Yang S, Lin M, Hao S, Ye H, Zhang X. Current hotspots and trends in cancer metabolic reprogramming: a scientometric analysis. Front Immunol 2024; 15:1497461. [PMID: 39588377 PMCID: PMC11586341 DOI: 10.3389/fimmu.2024.1497461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 11/27/2024] Open
Abstract
Background Metabolic reprogramming (MR) in cancer (CA) has been a focus of intense research in the recent two decades. This phenomenon has attracted great interest because it offers potential targets for cancer therapy. To capture the intellectual landscape of this field, we conducted a bibliometric analysis to assess the scientific output, major contributors, and trends in the MR/CA research. Methods We performed a systematic search using the Web of Science to retrieve articles published on MR of cancer from 2006 until 2023. The bibliometric tools such as Biblioshiny, VOSviewer, and Microsoft Excel were used to identify the most prolific authors, institutions, citation patterns, and keywords. We also used co-citation analysis to map the conceptual structure of the field and identify influential publications. Furthermore, we examined the literature by analyzing publication years, citations, and research impact factors. Results A total of 4,465 publications about MR/CA were retrieved. Publications on MR/CA increased rapidly from 2006 to 2023. Frontiers in Oncology published the most papers, while Cell Metabolism had the most citations. Highly cited papers were mainly published in Cancer Cell, Nature, Cell, Science and Cell Metabolism. China and the United States led the way in publications and contributed the most to MR/CA research. The University of Texas System, Chinese Academy of Sciences, and Fudan University were the most productive institutions. The profitable authors were Deberardinis Ralph J and Chiarugi Paola. The current topics included MR in tumorigenesis and progression of CA, MR of tumor cells and tumor microenvironment, the effect of MR on the CA treatment, the underlying mechanisms of MR (such as gene regulation, epigenetics, extracellular vesicles, and gut microbiota), and the modulation of MR. Some topics such as tumor microenvironment, lipid MR, circular RNA, long noncoding RNA, exosome, prognostic model, and immunotherapy may be the focus of MR/CA research in the next few years. Conclusion This study evaluated the global scientific output in the field of MR/CA research, analyzing its quantitative characteristics. It identified some significant and distinguished papers and compiled information regarding the current status and evolving trends of MR/CA research.
Collapse
Affiliation(s)
- Shanshan Yang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Miaomiao Lin
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Shaodong Hao
- Spleen and Stomach Disease Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| | - Xuezhi Zhang
- Traditional Chinese Medicine and Integrative Medicine Department, Peking University First Hospital, Beijing, China
| |
Collapse
|
43
|
Combs JE, Murray AB, Lomelino CL, Mboge MY, Mietzsch M, Horenstein NA, Frost SC, McKenna R, Becker HM. Disruption of the Physical Interaction Between Carbonic Anhydrase IX and the Monocarboxylate Transporter 4 Impacts Lactate Transport in Breast Cancer Cells. Int J Mol Sci 2024; 25:11994. [PMID: 39596062 PMCID: PMC11593560 DOI: 10.3390/ijms252211994] [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: 10/10/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
It has been previously established that breast cancer cells exhibit high expression of the monocarboxylate (lactate) transporters (MCT1 and/or MCT4) and carbonic anhydrase IX (CAIX) and form a functional metabolon for proton-coupled lactate export, thereby stabilizing intracellular pH. CD147 is the MCT accessory protein that facilitates the creation of the MCT/CAIX complex. This study describes how the small molecule Beta-Galactose 2C (BGal2C) blocks the physical and functional interaction between CAIX and either MCT1 or MCT4 in Xenopus oocytes, which reduces the rate of proton and lactate flux with an IC50 of ~90 nM. This value is similar to the Ki for inhibition of CAIX activity. Furthermore, it is shown that BGal2C blocks hypoxia-induced lactate transport in MDA-MB-231 and MCF-7 breast cancer cells, both of which express CAIX. As in oocytes, BGal2C interferes with the physical interaction between CAIX and MCTs in both cell types. Finally, X-ray crystallographic studies highlight unique interactions between BGal2C and a CAIX-mimic that are not observed within the CAII active site and which may underlie the strong specificity of BGal2C for CAIX. These studies demonstrate the utility of a novel sulfonamide in interfering with elevated proton and lactate flux, a hallmark of many solid tumors.
Collapse
Affiliation(s)
- Jacob E. Combs
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Akilah B. Murray
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Carrie L. Lomelino
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Mam Y. Mboge
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | | | - Susan C. Frost
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32611, USA (M.M.); (S.C.F.)
| | - Holger M. Becker
- Institute of Physiological Chemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
44
|
Perez LM, Venugopal SV, Martin AS, Freedland SJ, Di Vizio D, Freeman MR. Mechanisms governing lineage plasticity and metabolic reprogramming in cancer. Trends Cancer 2024; 10:1009-1022. [PMID: 39218770 DOI: 10.1016/j.trecan.2024.08.001] [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: 04/19/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Dynamic alterations in cellular phenotypes during cancer progression are attributed to a phenomenon known as 'lineage plasticity'. This process is associated with therapeutic resistance and involves concurrent shifts in metabolic states that facilitate adaptation to various stressors inherent in malignant growth. Certain metabolites also serve as synthetic reservoirs for chromatin modification, thus linking metabolic states with epigenetic regulation. There remains a critical need to understand the mechanisms that converge on lineage plasticity and metabolic reprogramming to prevent the emergence of lethal disease. This review attempts to offer an overview of our current understanding of the interplay between metabolic reprogramming and lineage plasticity in the context of cancer, highlighting the intersecting drivers of cancer hallmarks, with an emphasis on solid tumors.
Collapse
Affiliation(s)
- Lillian M Perez
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Smrruthi V Venugopal
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anna St Martin
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephen J Freedland
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dolores Di Vizio
- Department of Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael R Freeman
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
45
|
Pujana-Vaquerizo M, Bozal-Basterra L, Carracedo A. Metabolic adaptations in prostate cancer. Br J Cancer 2024; 131:1250-1262. [PMID: 38969865 PMCID: PMC11473656 DOI: 10.1038/s41416-024-02762-z] [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: 03/13/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 07/07/2024] Open
Abstract
Prostate cancer is one of the most commonly diagnosed cancers in men and is a major cause of cancer-related deaths worldwide. Among the molecular processes that contribute to this disease, the weight of metabolism has been placed under the limelight in recent years. Tumours exhibit metabolic adaptations to comply with their biosynthetic needs. However, metabolites also play an important role in supporting cell survival in challenging environments or remodelling the tumour microenvironment, thus being recognized as a hallmark in cancer. Prostate cancer is uniquely driven by androgen receptor signalling, and this knowledge has also influenced the paths of cancer metabolism research. This review provides a comprehensive perspective on the metabolic adaptations that support prostate cancer progression beyond androgen signalling, with a particular focus on tumour cell intrinsic and extrinsic pathways.
Collapse
Affiliation(s)
- Mikel Pujana-Vaquerizo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Laura Bozal-Basterra
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
- Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), 28029, Madrid, Spain.
- Traslational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, Baracaldo, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
46
|
Ali LS, Attia YAM, Mourad S, Halawa EM, Abd Elghaffar NH, Shokry S, Attia OM, Makram M, Wadan AHS, Negm WA, Elekhnawy E. The missing link between cancer stem cells and immunotherapy. Curr Med Res Opin 2024; 40:1963-1984. [PMID: 39316769 DOI: 10.1080/03007995.2024.2407963] [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: 06/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Cancer stem cells (CSCs) are cancer cells that can self-renew and give rise to tumors. The multipotency of CSCs enables the generation of diverse cancer cell types and their potential for differentiation and resilience against chemotherapy and radiation. Additionally, specific biomarkers have been identified for them, such as CD24, CD34, CD44, CD47, CD90, and CD133. The CSC model suggests that a subset of CSCs within tumors is responsible for tumor growth. The tumor microenvironment (TME), including fibroblasts, immune cells, adipocytes, endothelial cells, neuroendocrine (NE) cells, extracellular matrix (ECM), and extracellular vesicles, has a part in shielding CSCs from the host immune response as well as protecting them against anticancer drugs. The regulation of cancer stem cell plasticity by cancer-associated fibroblasts (CAFs) occurs through specific signaling pathways that differ among various types of cancer, utilizing the IGF-II/IGF1R, FAK, and c-Met/FRA1/HEY1 signaling pathways. Due to the intricate dynamics of CSC proliferation, controlling their growth necessitates innovative approaches and much more research. Our current review speculates an outline of how the TME safeguards stem cells, their interaction with CSCs, and the involvement of the immune and inflammatory systems in CSC differentiation and maintenance. Several technologies have the ability to identify CSCs; however, each approach has limitations. We discuss how these methods can aid in recognizing CSCs in several cancer types, comprising brain, breast, liver, stomach, and colon cancer. Furthermore, we explore different immunotherapeutic strategies targeting CSCs, including stimulating cancer-specific T cells, modifying immunosuppressive TMEs, and antibody-mediated therapy targeting CSC markers.
Collapse
Affiliation(s)
- Lobna Safwat Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | | | - Sohaila Mourad
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Esraa M Halawa
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Seham Shokry
- Faculty of Science, Tanta University, Tanta, Egypt
| | - Omar M Attia
- Faculty of Medicine, Cairo University, Giza, Egypt
| | - Maha Makram
- Faculty of Science, Zagazig University, Zagazig, Egypt
| | | | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
47
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024; 25:5080-5112. [PMID: 39415049 PMCID: PMC11549407 DOI: 10.1038/s44319-024-00288-2] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
48
|
Wu M, Mao L, Zhai X, Liu J, Wang J, Li L, Duan J, Wang J, Lin S, Li J, Yu S. Microenvironmental alkalization promotes the therapeutic effects of MSLN-CAR-T cells. J Immunother Cancer 2024; 12:e009510. [PMID: 39433427 PMCID: PMC11499857 DOI: 10.1136/jitc-2024-009510] [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: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high invasion, prone metastasis, frequent recurrence and poor prognosis. Unfortunately, the curative effects of current clinical therapies, including surgery, radiotherapy, chemotherapy and immunotherapy, are still limited in patients with TNBC. In this study, we showed that the heterogeneous expression at the protein level and subcellular location of mesothelin (MSLN), a potential target for chimeric antigen receptor-T (CAR-T) cell therapy in TNBC, which is caused by acidification of the tumor microenvironment, may be the main obstacle to therapeutic efficacy. Alkalization culture or sodium bicarbonate administration significantly promoted the membrane expression of MSLN and enhanced the killing efficiency of MSLN-CAR-T cells both in vitro and in vivo, and the same results were also obtained in other cancers with high MSLN expression, such as pancreatic and ovarian cancers. Moreover, mechanistic exploration revealed that the attenuation of autophagy-lysosome function caused by microenvironmental alkalization inhibited the degradation of MSLN. Hence, alkalization of the microenvironment improves the consistency and high expression of the target antigen MSLN and constitutes a routine method for treating diverse solid cancers via MSLN-CAR-T cells.
Collapse
Affiliation(s)
- Min Wu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Ling Mao
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Xuejia Zhai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jie Liu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Junhan Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Langhong Li
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jiangjie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Shuang Lin
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jianjun Li
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Shicang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| |
Collapse
|
49
|
Zhou Y, Zou P, Chen X, Chen P, Shi M, Lang J, Chen M. Overcoming Barriers in Photodynamic Therapy Harnessing Nanogenerators Strategies. Int J Biol Sci 2024; 20:5673-5694. [PMID: 39494340 PMCID: PMC11528466 DOI: 10.7150/ijbs.100317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024] Open
Abstract
Photodynamic therapy (PDT) represents a targeted approach for cancer treatment that employs light and photosensitizers (PSs) to induce the generation of reactive oxygen species (ROS). However, PDT faces obstacles including insufficient PS localization, limited light penetration, and treatment resistance. A potential solution lies in nanogenerators (NGs), which function as self-powered systems capable of generating electrical energy. Recent progress in piezoelectric and triboelectric NGs showcases promising applications in cancer research and drug delivery. Integration of NGs with PDT holds the promise of enhancing treatment efficacy by ensuring sustained PS illumination, enabling direct electrical control of cancer cells, and facilitating improved drug administration. This comprehensive review aims to augment our comprehension of PDT principles, explore associated challenges, and underscore the transformative capacity of NGs in conjunction with PDT. By harnessing NG technology alongside PDT, significant advancement in cancer treatment can be realized. Herein, we present the principal findings and conclusions of this study, offering valuable insights into the integration of NGs to overcome barriers in PDT.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Abdominal Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Pingjin Zou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Xingmin Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Ping Chen
- Department of Abdominal Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Min Shi
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jinyi Lang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Meihua Chen
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| |
Collapse
|
50
|
Saxon E, Ali T, Peng X. Hydrogen peroxide responsive theranostics for cancer-selective activation of DNA alkylators and real-time fluorescence monitoring in living cells. Eur J Med Chem 2024; 276:116695. [PMID: 39047609 DOI: 10.1016/j.ejmech.2024.116695] [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: 02/29/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Triple negative breast cancer (TNBC) is a notoriously difficult disease to treat, and many of the existing TNBC chemotherapeutics lack tumor selectivity and the capability for simultaneously visualizing and monitoring their own activity in the biological context. However, TNBC cells have been known to generate high levels of reactive oxygen species (ROS), such as hydrogen peroxide (H2O2). To this end, three novel small molecule theranostics 1a, 1c, and 2 consisting of both H2O2-responsive nitrogen mustard prodrug and profluorophore character have been designed, synthesized, and evaluated as targeted cancer therapeutics and bioimaging agents. The three theranostics comprise of boronate esters that deactivate nitrogen mustard functional groups and fluorophores but allow their selective activation through H2O2-specific oxidative deboronation for the release of the active drug and fluorophore. The three theranostics demonstrated H2O2-inducible DNA-alkylating capability and fluorescence turn-on properties in addition to selective anticancer activity. They are particularly effective in killing TNBC MDA-MB-468 cells with high H2O2 level while safe to normal epithelial MCF-10A cell. The conjugated boron-masked fluorophores in 1c and 2 are highly responsive towards H2O2, which enabled tracking of the theranostics in living cellular mitochondria and nucleus organelles. The three theranostics 1a, 1c, and 2 are capable of both selective release of the active drug to take effect in H2O2-rich cancer sites and simultaneously monitoring its activity. This single molecule system is of utmost importance to understand the function, efficacy, and mechanism of the H2O2-activated prodrugs and theranostics within the living recipient.
Collapse
Affiliation(s)
- Eron Saxon
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53211, USA
| | - Taufeeque Ali
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53211, USA
| | - Xiaohua Peng
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer St., Milwaukee, WI, 53211, USA.
| |
Collapse
|