1
|
Abdullah KM, Sharma G, Singh AP, Siddiqui JA. Nanomedicine in Cancer Therapeutics: Current Perspectives from Bench to Bedside. Mol Cancer 2025; 24:169. [PMID: 40490771 DOI: 10.1186/s12943-025-02368-w] [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: 03/26/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Cancer is among the leading causes of death worldwide, with projections indicating that it will claim 35 million lives by the year 2050. Conventional therapies, such as chemotherapy and immune modulation, have reduced cancer mortality to some extent; however, they have limited efficacy due to their broad mode of action, often resulting in cytotoxic effects on normal cells along with the malignant tissues, ultimately limiting their overall optimal therapeutic efficacy outcomes.Rapid advances in nanotechnology and an evolving understanding of cancer mechanisms have propelled the development of a diverse array of nanocarriers to vanquish the hurdles in achieving sophisticated drug delivery with reduced off-target toxicity. Nanoformulations can deliver the anti-cancer agents precisely to the tumor cell by integrating a multitarget approach that allows for tissue-, cell-, or organelle-specific delivery and internalization. Despite the immense interest and unmatched advancements in modern oncology equipped with nanomedicines, only a few nanoformulations have successfully translated into clinical settings. A major reason behind this shortcoming is the lack of a rationale design incorporating smart, responsive targeting features, leading to a compromised therapeutic window due to inefficient internalization or erroneous intracellular localization with unsuccessful payload release. This review aims to summarize the recent perspective of nanomedicine and its translation to clinical practice, with a particular focus on the evolution of strategies used in tumor targeting from traditional EPR-based passive mechanisms to advanced active and multi-stage approaches. We highlight the coupling of organelle-specific and stimuli-responsive nanocarriers, discuss the potential of biomimetic and cell-mediated delivery systems, and also shed light on technologies such as microfluidics, tumor-on-chip models, and AI-assisted synthesis. Finally, this review explores translational hurdles ranging from biological and manufacturing challenges to regulatory bottlenecks and outlines how innovative modeling systems and engineering solutions can bridge the gap from bench to bedside in cancer nanotherapeutics.
Collapse
Affiliation(s)
- K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Ajay P Singh
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jawed A Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
2
|
Al-Shehaby N, Elshoky HA, Zidan M, Salaheldin TA, Gaber MH, Ali MA, El-Sayed NM. In vitro localization of modified zinc oxide nanoparticles showing selective anticancer effects against colorectal carcinoma using biophysical techniques. Sci Rep 2025; 15:16811. [PMID: 40369004 PMCID: PMC12078601 DOI: 10.1038/s41598-025-00434-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
In recent decades, despite advancements in conventional cancer therapies, their serious side effects on both healthy and tumor cells remain a major concern. Aiming to address indiscriminate drug distribution, unwanted toxicity, and high chemotherapy doses, this study explores the targeted delivery of zinc oxide nanoparticles (ZnO NPs). ZnO NPs were synthesized and coated with bovine serum albumin (BSA) and tetraethoxysilane (TEOS) to control cellular uptake and enhance anticancer activity. Characterized by UV-visible spectroscopy, DLS, FTIR, XRD, and TEM, ZnO, ZnOB, and ZnOT particles displayed sizes of 140 ± 13.6 nm, 342 ± 8.4 nm, and 145 ± 23.8 nm, respectively, with ZnOT showing a positive charge of + 19.3 ± 4.16 mV, enhancing stability and cellular interaction. Cytotoxicity assays revealed ZnO's potent anticancer effect in Caco-2 cells with an IC50 of 219 µg/ml, while ZnOB and ZnOT showed moderate toxicity (IC50 values of 308 µg/ml and 235 µg/ml). HepG2 cells maintained viability close to 100%, highlighting ZnO NPs' selectivity for Caco-2 cells. Flow cytometry and confocal microscopy indicated differential uptake, with ZnOB showing the highest uptake in Caco-2 cells after 24 h at 37 °C, increasing fluorescence intensity by over 80% compared to ZnO. ZnOT notably increased late apoptotic cells by 65% in Caco-2 lines and caused a 40% rise in G2/M phase arrest. Mitochondrial function assays showed that ZnO reduced mitochondrial membrane potential by over 30%, indicating stress induction. These results support the potential of ZnO-based nanoparticles in colorectal cancer treatment, offering selective cytotoxicity, enhanced cellular uptake, and clear apoptotic activity, making them a promising alternative to conventional chemotherapy.
Collapse
Affiliation(s)
- Nouran Al-Shehaby
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
| | - Hisham A Elshoky
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt.
- Nanotechnology and Advanced Materials Central Lab, Agricultural Research Center, P.O. 588 Orman, 9 Elgamaa St., Giza, 12619, Egypt.
- Regional Center for Food and Feed, Agricultural Research Center, Giza, 12619, Egypt.
| | - Mona Zidan
- Immunology research program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
| | - Taher A Salaheldin
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- SUNY Schenectady Community College, New York, USA
| | - Mohamed H Gaber
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Maha A Ali
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Nayera M El-Sayed
- Physics Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
3
|
Elblová P, Anthi J, Liu M, Lunova M, Jirsa M, Stephanopoulos N, Lunov O. DNA Nanostructures for Rational Regulation of Cellular Organelles. JACS AU 2025; 5:1591-1616. [PMID: 40313805 PMCID: PMC12042030 DOI: 10.1021/jacsau.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
DNA nanotechnology has revolutionized materials science and biomedicine by enabling precise manipulation of matter at the nanoscale. DNA nanostructures (DNs) in particular represent a promising frontier for targeted therapeutics. Engineered DNs offer unprecedented molecular programmability, biocompatibility, and structural versatility, making them ideal candidates for advanced drug delivery, organelle regulation, and cellular function modulation. This Perspective explores the emerging role of DNs in modulating cellular behavior through organelle-targeted interventions. We highlight current advances in nuclear, mitochondrial, and lysosomal targeting, showcasing applications ranging from gene delivery to cancer therapeutics. For instance, DNs have enabled precision mitochondrial disruption in cancer cells, lysosomal pH modulation to enhance gene silencing, and nuclear delivery of gene-editing templates. While DNs hold immense promise for advancing nanomedicine, outstanding challenges include optimizing biological interactions and addressing safety concerns. This Perspective highlights the current potential of DNs for rational control of targeted organelles, which could lead to novel therapeutic strategies and advancement of precision nanomedicines in the future.
Collapse
Affiliation(s)
- Petra Elblová
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Faculty
of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16 Prague 2, Czech Republic
| | - Judita Anthi
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Minghui Liu
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Mariia Lunova
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Milan Jirsa
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Nicholas Stephanopoulos
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Oleg Lunov
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
| |
Collapse
|
4
|
Malhotra A, Dehghankelishadi P, Kaur I, Marshall M, Rudd D, Wojnilowicz M, Nowell CJ, Fulcher AJ, Esser L, Tong WY, Cifuentes A, Wagstaff KM, Voelcker NH. Triple-Negative Breast Cancer Aptamer-Targeting Porous Silicon Nanocarrier. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5955-5969. [PMID: 39804251 DOI: 10.1021/acsami.4c18453] [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: 01/31/2025]
Abstract
Common treatment approaches for triple-negative breast cancer (TNBC) are associated with severe side effects due to the unfavorable biodistribution profile of potent chemotherapeutics. Here, we explored the potential of TNBC-targeting aptamer-decorated porous silicon nanoparticles (pSiNPs) as targeted nanocarriers for TNBC. A "salt-aging" strategy was employed to fabricate a TNBC-targeting aptamer functionalized pSiNP that was highly colloidally stable. Doxorubicin (Dox) was efficiently loaded into nanoparticles (179 ± 5 μg/mg of pSiNP) and experienced pH-dependent release kinetics. Further experiments highlighted that clathrin-mediated endocytosis was the primary route that aptamer-pSiNP conjugates take to enter the endolysosomal compartment of the MCF10Ca1h TNBC cells. A time-interval colocalization study shows the accumulation of an aptamer-decorated pSiNP conjugate in the lysosomes of TNBC cells, unlike for antibody-decorated pSiNPs, leading to particle-induced lysosomal swelling and membrane destabilization. Dox-loaded aptamer-pSiNPs efficiently reduced the viability of the TNBC cells (11.8 ± 1.5%) compared to nontargeted nanoparticles (58.2 ± 8.8%) while the developed system showed a low level of toxicity in healthy cells, both in vitro and in vivo. These findings have laid the foundation for further investigating the potential of aptamer-pSiNP conjugates as a targeted treatment strategy in preclinical TNBC models.
Collapse
Affiliation(s)
- Ankit Malhotra
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Pouya Dehghankelishadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Ishdeep Kaur
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Morgan Marshall
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3168, Australia
| | - David Rudd
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Marcin Wojnilowicz
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria 3168, Australia
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Alex J Fulcher
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3168, Australia
| | - Lars Esser
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria 3168, Australia
| | - Wing Yin Tong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Anna Cifuentes
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Kylie M Wagstaff
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3168, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Clayton, Victoria 3168, Australia
- Materials Science and Engineering, Monash University, 14 Alliance Lane, Clayton, Victoria 3168, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| |
Collapse
|
5
|
Yuan F, Lerman LO. Targeted therapeutic strategies for the kidney. Expert Opin Ther Targets 2024; 28:979-989. [PMID: 39491501 PMCID: PMC11617265 DOI: 10.1080/14728222.2024.2421756] [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: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Kidney diseases impose a significant burden with high incidence and mortality rates. Current treatment options for kidney diseases are limited, necessitating urgent development of novel and effective therapeutic strategies to delay or reverse disease progression. Targeted therapies for the kidney hold promise in significantly enhancing treatment outcomes, offering hope to patients afflicted with renal disorders. AREAS COVERED This review summarized advances in kidney-targeted therapies including genes, peptides and proteins, cell-based, nanoparticles, and localized delivery routes. We also explored the potential clinical applications, prospects, and challenges of targeted therapies for renal disorders. EXPERT OPINION Advances in targeted therapies for renal conditions have enhanced therapeutic outcomes. Clinical application of kidney-targeted therapies is currently limited by renal structure and the scarcity of robust biomarkers. Bridging the gap from basic and pre-clinical research targeting the kidney to achieving clinical translation remains a formidable challenge.
Collapse
Affiliation(s)
- Fei Yuan
- Division of Nephrology and Hypertension, Mayo Clinic; Rochester, MN, USA
- Department of Urology, National Children’s Medical Center, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic; Rochester, MN, USA
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
6
|
Li Y, Li XM, Wei LS, Ye JF. Advancements in mitochondrial-targeted nanotherapeutics: overcoming biological obstacles and optimizing drug delivery. Front Immunol 2024; 15:1451989. [PMID: 39483479 PMCID: PMC11524880 DOI: 10.3389/fimmu.2024.1451989] [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: 06/20/2024] [Accepted: 09/19/2024] [Indexed: 11/03/2024] Open
Abstract
In recent decades, nanotechnology has significantly advanced drug delivery systems, particularly in targeting subcellular organelles, thus opening new avenues for disease treatment. Mitochondria, critical for cellular energy and health, when dysfunctional, contribute to cancer, neurodegenerative diseases, and metabolic disorders. This has propelled the development of nanomedicines aimed at precise mitochondrial targeting to modulate their function, marking a research hotspot. This review delves into the recent advancements in mitochondrial-targeted nanotherapeutics, with a comprehensive focus on targeting strategies, nanocarrier designs, and their therapeutic applications. It emphasizes nanotechnology's role in enhancing drug delivery by overcoming biological barriers and optimizing drug design for specific mitochondrial targeting. Strategies exploiting mitochondrial membrane potential differences and specific targeting ligands improve the delivery and mitochondrial accumulation of nanomedicines. The use of diverse nanocarriers, including liposomes, polymer nanoparticles, and inorganic nanoparticles, tailored for effective mitochondrial targeting, shows promise in anti-tumor and neurodegenerative treatments. The review addresses the challenges and future directions in mitochondrial targeting nanotherapy, highlighting the need for precision, reduced toxicity, and clinical validation. Mitochondrial targeting nanotherapy stands at the forefront of therapeutic strategies, offering innovative treatment perspectives. Ongoing innovation and research are crucial for developing more precise and effective treatment modalities.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, China
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiao-meng Li
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Li-si Wei
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Adu-Amankwaah F, Februarie C, Nyambo K, Maarman G, Tshililo N, Mabasa L, Mavumengwana V, Baatjies L. Cytotoxic properties, glycolytic effects and high-resolution respirometry mitochondrial activities of Eriocephalus racemosus against MDA-MB 231 triple-negative breast cancer. BMC Complement Med Ther 2024; 24:332. [PMID: 39256791 PMCID: PMC11389270 DOI: 10.1186/s12906-024-04615-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) represents a significant global health crisis due to its resistance to conventional therapies and lack of specific molecular targets. This study explored the potential of Eriocephalus racemosus (E. racemosus) as an alternative treatment for TNBC. The cytotoxic properties and high-resolution respirometry mitochondrial activities of E. racemosus against the MDA-MB 231 TNBC cell line were evaluated. METHODS Hexane solvent and bioactive fraction extractions of E. racemosus were performed, while mass spectrometry-based metabolite profiling was used to identify the phytochemical constituents of the extracts. The extracts were further tested against MDA-MB 231 TNBC cells to determine their cytotoxicity. The mode of cell death was determined using flow cytometry. The activities of caspases 3, 8, and 9 were assessed using a multiplex activity assay kit. Glycolytic activity and High-resolution respirometry measurements of mitochondrial function in the MDA-MB 231 cell line were conducted using the Seahorse XFp and Oroboros O2K. RESULTS Metabolite profiling of E. racemosus plant crude extracts identified the presence of coumarins, flavonoids, sesquiterpenoids, triterpenoids, and unknown compounds. The extracts demonstrated promising cytotoxic activities, with a half maximal inhibitory concentration (IC50) of 12.84 µg/mL for the crude hexane extract and 15.49 µg/mL for the bioactive fraction. Further, the crude hexane and bioactive fraction extracts induced apoptosis in the MDA-MB-231 TNBC cells, like the reference drug cisplatin (17.44%, 17.26% and 20.25%, respectively) compared to untreated cells. Caspase 3 activities confirmed the induction of apoptosis in both cisplatin and the plant crude extracts, while caspase 8 and 9 activities confirmed the activation of both the intrinsic and extrinsic apoptosis pathways. Increased levels of glycolytic activity were observed in the hexane crude extract. High-resolution respiratory measurements showed elevated mitochondrial activities in all mitochondrial states except for complex-IV activity. CONCLUSION These findings support further exploration of E. racemosus as a potential therapeutic agent for TNBC, offering a promising avenue for the development of targeted treatments with minimal adverse effects.
Collapse
Affiliation(s)
- Francis Adu-Amankwaah
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Februarie
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Kudakwashe Nyambo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerald Maarman
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health Science, CARMA: Centre for Cardio-Metabolic Research in Africa, Stellenbosch University, Cape Town, 8000, South Africa
| | - Ndivhuwo Tshililo
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lawrence Mabasa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Vuyo Mavumengwana
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa
| | - Lucinda Baatjies
- South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- Biomedical Research and Innovation Platform (BRIP), Medical Research Council, Tygerberg, Cape Town, South Africa.
| |
Collapse
|
8
|
Zhang Q, Xia Y, Wang L, Wang Y, Bao Y, Zhao GS. Targeted anti-angiogenesis therapy for advanced osteosarcoma. Front Oncol 2024; 14:1413213. [PMID: 39252946 PMCID: PMC11381227 DOI: 10.3389/fonc.2024.1413213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
To date, despite extensive research, the prognosis of advanced osteosarcoma has not improved significantly. Thus, patients experience a reduced survival rate, suggesting that a reevaluation of current treatment strategies is required. Recently, in addition to routine surgery, chemotherapy and radiotherapy, researchers have explored more effective and safer treatments, including targeted therapy, immunotherapy, anti-angiogenesis therapy, metabolic targets therapy, and nanomedicine therapy. The tumorigenesis and development of osteosarcoma is closely related to angiogenesis. Thus, anti-angiogenesis therapy is crucial to treat osteosarcoma; however, recent clinical trials found that it has insufficient efficacy. To solve this problem, the causes of treatment failure and improve treatment strategies should be investigated. This review focuses on summarizing the pathophysiological mechanisms of angiogenesis in osteosarcoma and recent advances in anti-angiogenesis treatment of osteosarcoma. We also discuss some clinical studies, with the aim of providing new ideas to improve treatment strategies for osteosarcoma and the prognosis of patients.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Pain and Rehabilitation, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuxuan Xia
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - LiYuan Wang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Wang
- Department of Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Sheng Zhao
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Harisa GI, Faris TM, Sherif AY, Alzhrani RF, Alanazi SA, Kohaf NA, Alanazi FK. Coding Therapeutic Nucleic Acids from Recombinant Proteins to Next-Generation Vaccines: Current Uses, Limitations, and Future Horizons. Mol Biotechnol 2024; 66:1853-1871. [PMID: 37578574 DOI: 10.1007/s12033-023-00821-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023]
Abstract
This study aims to highlight the potential use of cTNAs in therapeutic applications. The COVID-19 pandemic has led to significant use of coding therapeutic nucleic acids (cTNAs) in terms of DNA and mRNA in the development of vaccines. The use of cTNAs resulted in a paradigm shift in the therapeutic field. However, the injection of DNA or mRNA into the human body transforms cells into biological factories to produce the necessary proteins. Despite the success of cTNAs in the production of corona vaccines, they have several limitations such as instability, inability to cross biomembranes, immunogenicity, and the possibility of integration into the human genome. The chemical modification and utilization of smart drug delivery cargoes resolve cTNAs therapeutic problems. The success of cTNAs in corona vaccine production provides perspective for the eradication of influenza viruses, Zika virus, HIV, respiratory syncytial virus, Ebola virus, malaria, and future pandemics by quick vaccine design. Moreover, the progress cTNAs technology is promising for the development of therapy for genetic disease, cancer therapy, and currently incurable diseases.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia.
- Department of Biochemistry and Molecular Biology, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Tarek M Faris
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Abdelrahman Y Sherif
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
| | - Riyad F Alzhrani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
- Nanobiotechnology Research Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Alanazi
- Pharmaceutical Care Services, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science Collage of Pharmacy, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Neveen A Kohaf
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11651, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
10
|
Alsharif ST, Gardouh AM, Mandour MF, Alaqais ZM, Alharbi LK, Almarwani MJ, Mokhtar HI, Hisham FA, Abdellah MM, Mohamed GM, Shorog EM, Almaeen AH, Atteia HH, Zaitone SA. Antitumor activity and targeting p53-PUMA mRNA expression by 5-flurouracil PLGA-lipid polymeric nanoparticles in mouse mammary carcinomas: comparison to free 5-flurouracil. Toxicol Mech Methods 2024; 34:385-397. [PMID: 38083807 DOI: 10.1080/15376516.2023.2294083] [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: 04/24/2023] [Accepted: 12/02/2023] [Indexed: 01/10/2024]
Abstract
Polymeric poly (lactic-co-glycolic acid) (PLGA)-lipid hybrid nanoparticles (PNPs)-based therapy are powerful carriers for various therapeutic agents. This study was conducted to evaluate the chemotherapeutic potential of free 5-flurouracil (5FU) and synthetized 5FU-PNPs and impact on p53-dependent apoptosis in mammary carcinomas (MCs) grown in mice. Breast cancer cells were injected in Swiss albino female mice and 2 bilateral masses of MC were confirmed after one week. Mice were distributed to five experimental groups; Group 1: MC control group. Groups 2 and 3: MC + free 5FU [5 or 10 mg per kg] groups. Groups 4 and 5: synthetized MC+ 5FU-PNPs [5 or 10 mg per kg] groups. Medications were administered orally, twice weekly for 3 weeks. Then, tumors were dissected, and sections were stained with hematoxylin-eosin (HE) while the other MC was used for measuring of cell death and inflammatory markers. Treatment with 5FU-PNPs suppressed the MC masses and pathologic scores based on HE-staining. Similarly, greater proapoptotic activity was recorded in 5FU-PNPs groups compared to free 5FU groups as shown by significant upregulation in tumoral p53 immunostaining. The current results encourage the utility of PNPs for improving the antitumor effect of 5FU. The chemotherapeutic potential was mediated through enhancement of tumoral p53-mediated p53 up-regulated modulator of apoptosis (PUMA) genes. Additional studies are warranted for testing the antitumor activity of this preparation in other mouse models of breast cancer.
Collapse
Affiliation(s)
- Sara T Alsharif
- PharmD program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Ahmed M Gardouh
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Jadara University, Irbid, Jordan
| | - Mohamed F Mandour
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Zood M Alaqais
- PharmD program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Lama K Alharbi
- PharmD program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Maha J Almarwani
- PharmD program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Hatem I Mokhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia, Egypt
| | - Fatma Azzahraa Hisham
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Mahmoud Abdellah
- Department of Pathology, Faculty of Medicine, Galala University, Suez, Egypt
- Department of Pathology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Ghena M Mohamed
- Nutrition and Food Science Department, College of Home Economics, Tabuk University, Tabuk, Saudi Arabia
| | - Eman M Shorog
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | | | - Hebatallah H Atteia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
11
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
12
|
Janero DR. Current strategic trends in drug discovery: the present as prologue. Expert Opin Drug Discov 2024; 19:147-159. [PMID: 37936504 DOI: 10.1080/17460441.2023.2275640] [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: 08/21/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Escalating costs and inherent uncertainties associated with drug discovery invite initiatives to improve its efficiency and de-risk campaigns for inventing better therapeutics. One such initiative involves recognizing and exploiting current approaches in therapeutics invention with molecular mechanisms of action that hold promise for designing and targeting new chemical entities as drugs. AREAS COVERED This perspective considers the current contextual framework around three drug-discovery approaches and evaluates their potential to help identify new targets/modalities in small-molecule molecular pharmacology: diversifying ligand-directed phenotypes for G protein-coupled receptor (GPCR) pharmacotherapeutic signaling; developing therapeutic-protein degraders and stabilizers for proximity-inducing pharmacology; and mining organelle biology for druggable therapeutic targets. EXPERT OPINION The contemporary drug-discovery approaches examined appear generalizable and versatile to have applications in therapeutics invention beyond those case studies discussed herein. Accordingly, they may be considered strategic trends worthy of note in advancing the field toward novel ways of addressing pharmacotherapeutically unmet medical needs.
Collapse
Affiliation(s)
- David R Janero
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, and Health Sciences Entrepreneurs, Northeastern University, Boston, MA, USA
| |
Collapse
|
13
|
Šlachtová V, Chovanec M, Rahm M, Vrabel M. Bioorthogonal Chemistry in Cellular Organelles. Top Curr Chem (Cham) 2023; 382:2. [PMID: 38103067 PMCID: PMC10725395 DOI: 10.1007/s41061-023-00446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/12/2023] [Indexed: 12/17/2023]
Abstract
While bioorthogonal reactions are routinely employed in living cells and organisms, their application within individual organelles remains limited. In this review, we highlight diverse examples of bioorthogonal reactions used to investigate the roles of biomolecules and biological processes as well as advanced imaging techniques within cellular organelles. These innovations hold great promise for therapeutic interventions in personalized medicine and precision therapies. We also address existing challenges related to the selectivity and trafficking of subcellular dynamics. Organelle-targeted bioorthogonal reactions have the potential to significantly advance our understanding of cellular organization and function, provide new pathways for basic research and clinical applications, and shape the direction of cell biology and medical research.
Collapse
Affiliation(s)
- Veronika Šlachtová
- Department of Bioorganic and Medicinal Chemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10, Prague 6, Czech Republic
| | - Marek Chovanec
- Department of Bioorganic and Medicinal Chemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10, Prague 6, Czech Republic
- University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| | - Michal Rahm
- Department of Bioorganic and Medicinal Chemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10, Prague 6, Czech Republic
- University of Chemistry and Technology, Technická 5, 166 28, Prague 6, Czech Republic
| | - Milan Vrabel
- Department of Bioorganic and Medicinal Chemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 166 10, Prague 6, Czech Republic.
| |
Collapse
|
14
|
Pozza MD, Mesdom P, Abdullrahman A, Prieto Otoya TD, Arnoux P, Frochot C, Niogret G, Saubaméa B, Burckel P, Hall JP, Hollenstein M, Cardin CJ, Gasser G. Increasing the π-Expansive Ligands in Ruthenium(II) Polypyridyl Complexes: Synthesis, Characterization, and Biological Evaluation for Photodynamic Therapy Applications. Inorg Chem 2023; 62:18510-18523. [PMID: 37913550 DOI: 10.1021/acs.inorgchem.3c02606] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Lack of selectivity is one of the main issues with currently used chemotherapies, causing damage not only to altered cells but also to healthy cells. Over the last decades, photodynamic therapy (PDT) has increased as a promising therapeutic tool due to its potential to treat diseases like cancer or bacterial infections with a high spatiotemporal control. Ruthenium(II) polypyridyl compounds are gaining attention for their application as photosensitizers (PSs) since they are generally nontoxic in dark conditions, while they show remarkable toxicity after light irradiation. In this work, four Ru(II) polypyridyl compounds with sterically expansive ligands were studied as PDT agents. The Ru(II) complexes were synthesized using an alternative route to those described in the literature, which resulted in an improvement of the synthesis yields. Solid-state structures of compounds [Ru(DIP)2phen]Cl2 and [Ru(dppz)2phen](PF6)2 have also been obtained. It is well-known that compound [Ru(dppz)(phen)2]Cl2 binds to DNA by intercalation. Therefore, we used [Ru(dppz)2phen]Cl2 as a model for DNA interaction studies, showing that it stabilized two different sequences of duplex DNA. Most of the synthesized Ru(II) derivatives showed very promising singlet oxygen quantum yields, together with noteworthy photocytotoxic properties against two different cancer cell lines, with IC50 in the micro- or even nanomolar range (0.06-7 μM). Confocal microscopy studies showed that [Ru(DIP)2phen]Cl2 and [Ru(DIP)2TAP]Cl2 accumulate preferentially in mitochondria, while no mitochondrial internalization was observed for the other compounds. Although [Ru(dppn)2phen](PF6)2 did not accumulate in mitochondria, it interestingly triggered an impairment in mitochondrial respiration after light irradiation. Among others, [Ru(dppn)2phen](PF6)2 stands out for its very good IC50 values, correlated with a very high singlet oxygen quantum yield and mitochondrial respiration disruption.
Collapse
Affiliation(s)
- Maria Dalla Pozza
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| | - Pierre Mesdom
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| | - Ahmad Abdullrahman
- Department of Pharmacy, Chemistry and Pharmacy Building, University of Reading, Whiteknights Campus, Reading, Berkshire RG6 6AD, U.K
| | | | | | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, Nancy F-54000, France
| | - Germain Niogret
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Departement of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Paris 75015, France
| | - Bruno Saubaméa
- Université Paris Cité, INSERM, CNRS, P-MIM, Plateforme d'Imagerie Cellulaire et Moléculaire (PICMO), Paris F-75006, France
| | - Pierre Burckel
- Université de Paris, Institut de physique du globe de Paris, CNRS, Paris F-75005, France
| | - James P Hall
- Department of Pharmacy, Chemistry and Pharmacy Building, University of Reading, Whiteknights Campus, Reading, Berkshire RG6 6AD, U.K
| | - Marcel Hollenstein
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Departement of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, Paris 75015, France
| | - Christine J Cardin
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6 6AD, U.K
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health, Paris 75005, France
| |
Collapse
|
15
|
Žmudová Z, Šanderová Z, Liegertová M, Vinopal S, Herma R, Sušický L, Müllerová M, Strašák T, Malý J. Biodistribution and toxicity assessment of methoxyphenyl phosphonium carbosilane dendrimers in 2D and 3D cell cultures of human cancer cells and zebrafish embryos. Sci Rep 2023; 13:15477. [PMID: 37726330 PMCID: PMC10509138 DOI: 10.1038/s41598-023-42850-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
The consideration of human and environmental exposure to dendrimers, including cytotoxicity, acute toxicity, and cell and tissue accumulation, is essential due to their significant potential for various biomedical applications. This study aimed to evaluate the biodistribution and toxicity of a novel methoxyphenyl phosphonium carbosilane dendrimer, a potential mitochondria-targeting vector for cancer therapeutics, in 2D and 3D cancer cell cultures and zebrafish embryos. We assessed its cytotoxicity (via MTT, ATP, and Spheroid growth inhibition assays) and cellular biodistribution. The dendrimer cytotoxicity was higher in cancer cells, likely due to its specific targeting to the mitochondrial compartment. In vivo studies using zebrafish demonstrated dendrimer distribution within the vascular and gastrointestinal systems, indicating a biodistribution profile that may be beneficial for systemic therapeutic delivery strategies. The methoxyphenyl phosphonium carbosilane dendrimer shows promise for applications in cancer cell delivery, but additional studies are required to confirm these findings using alternative labelling methods and more physiologically relevant models. Our results contribute to the growing body of evidence supporting the potential of carbosilane dendrimers as vectors for cancer therapeutics.
Collapse
Affiliation(s)
- Zuzana Žmudová
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| | - Zuzana Šanderová
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| | - Michaela Liegertová
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic.
| | - Stanislav Vinopal
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| | - Regina Herma
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| | - Luděk Sušický
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| | - Monika Müllerová
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
- Institute of Chemical Process Fundamentals of the CAS, Prague, Czech Republic
| | - Tomáš Strašák
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
- Institute of Chemical Process Fundamentals of the CAS, Prague, Czech Republic
| | - Jan Malý
- CENAB, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Ústí nad Labem, Czech Republic
| |
Collapse
|
16
|
Jana B, Jin S, Go EM, Cho Y, Kim D, Kim S, Kwak SK, Ryu JH. Intra-Lysosomal Peptide Assembly for the High Selectivity Index against Cancer. J Am Chem Soc 2023; 145:18414-18431. [PMID: 37525328 DOI: 10.1021/jacs.3c04467] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Lysosomes remain powerful organelles and important targets for cancer therapy because cancer cell proliferation is greatly dependent on effective lysosomal function. Recent studies have shown that lysosomal membrane permeabilization induces cell death and is an effective way to treat cancer by bypassing the classical caspase-dependent apoptotic pathway. However, most lysosome-targeted anticancer drugs have very low selectivity for cancer cells. Here, we show intra-lysosomal self-assembly of a peptide amphiphile as a powerful technique to overcome this problem. We designed a peptide amphiphile that localizes in the cancer lysosome and undergoes cathepsin B enzyme-instructed supramolecular assembly. This localized assembly induces lysosomal swelling, membrane permeabilization, and damage to the lysosome, which eventually causes caspase-independent apoptotic death of cancer cells without conventional chemotherapeutic drugs. It has specific anticancer effects and is effective against drug-resistant cancers. Moreover, this peptide amphiphile exhibits high tumor targeting when attached to a tumor-targeting ligand and causes significant inhibition of tumor growth both in cancer and drug-resistant cancer xenograft models.
Collapse
Affiliation(s)
- Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seongeon Jin
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Eun Min Go
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Yumi Cho
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sang Kyu Kwak
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
17
|
Binnebose AM, Mullis AS, Haughney SL, Narasimhan B, Bellaire BH. Nanotherapeutic delivery of antibiotic cocktail enhances intra-macrophage killing of Mycobacterium marinum. FRONTIERS IN ANTIBIOTICS 2023; 2:1162941. [PMID: 39816663 PMCID: PMC11732124 DOI: 10.3389/frabi.2023.1162941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/26/2023] [Indexed: 01/18/2025]
Abstract
Mycobacterium marinum is a waterborne pathogen responsible for tuberculosis-like infections in cold-blooded animals and is an opportunistic pathogen in humans. M. marinum is the closest genetic relative of the Mycobacterium tuberculosis complex and is a reliable surrogate for drug susceptibility testing. We synthesized and evaluated two nanoparticle (NP) formulations for compatibility with rifampicin, isoniazid, pyrazinamide, and ethambutol (PIRE), the front-line antimycobacterial drugs used in combination against active tuberculosis infections. Improved in vitro antimicrobial activity was observed with encapsulated rifampicin alone or in a cocktail of drugs formulated through co-encapsulation in amphiphilic polyanhydride NPs. Broth antimicrobial testing revealed that the encapsulation of PIRE in NP resulted in a significant increase in antimicrobial activity, with the benefit over soluble formulations at biologically relevant concentrations ranging from >10 to >3,000 fold. M. marinum-infected human macrophages treated with NP-PIRE were cleared of viable bacteria in 48 h following a single treatment, representing a >4 log reduction in colony-forming units and a >2,000-fold increase in antimicrobial activity. The amphiphilic polyanhydride nanoparticles demonstrated the ability to co-encapsulate PIRE antibiotics and enhance their antimicrobial activity against M. marinum in infected macrophages in culture and in vitro. These data suggest that polyanhydride nanoparticles are a promising nanotherapeutic for combatting Mycobacterium infections through improved intracellular targeting of encapsulated antibiotics.
Collapse
Affiliation(s)
- Andrea M. Binnebose
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
- Department of Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
- Cargill Animal Nutrition, Elk River, MN, United States
| | - Adam S. Mullis
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Shannon L. Haughney
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Bryan H. Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
- Department of Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Wei Y, Xia X, Li H, Gao H. Influence factors on and potential strategies to amplify receptor-mediated nanodrug delivery across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1713-1730. [PMID: 37542516 DOI: 10.1080/17425247.2023.2245332] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/22/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION A major challenge in treating central nervous system (CNS) disorders is to achieve adequate drug delivery across the blood-brain barrier (BBB). Receptor-mediated nanodrug delivery as a Trojan horse strategy has become an exciting approach. However, these nanodrugs do not accumulate significantly in the brain parenchyma, which greatly limits the therapeutic effect of drugs. Amplifying the efficiency of receptor-mediated nanodrug delivery across the BBB becomes the holy grail in the treatment of CNS disorders. AREAS COVERED In this review, we tend to establish links between dynamic BBB and receptor-mediated nanodrug delivery, starting with the delivery processes across the BBB, describing factors affecting nanodrug delivery efficiency, and summarizing potential strategies that may amplify delivery efficiency. EXPERT OPINION Receptor-mediated nanodrug delivery is a common approach to significantly enhance the efficiency of brain-targeting delivery. As BBB is constantly undergoing changes, it is essential to investigate the impact of diseases on the effectiveness of brain-targeting nanodrug delivery. More critically, there are several barriers to achieving brain-targeting nanodrug delivery in the five stages of receptor-mediated transcytosis (RMT), and the impacts can be conflicting, requiring intricate balance. Further studies are also needed to investigate the material toxicity of nanodrugs to address the issue of clinical translation.
Collapse
Affiliation(s)
- Ya Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu, P. R. China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
19
|
Keller T, Trinks N, Brand J, Trippmacher S, Stahlhut P, Albrecht K, Papastavrou G, Koepsell H, Sauer M, Groll J. Design of Nanohydrogels for Targeted Intracellular Drug Transport to the Trans-Golgi Network. Adv Healthc Mater 2023; 12:e2201794. [PMID: 36739269 PMCID: PMC11469190 DOI: 10.1002/adhm.202201794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/27/2023] [Indexed: 02/06/2023]
Abstract
Nanohydrogels combine advantages of hydrogels and nanoparticles. In particular, they represent promising drug delivery systems. Nanogel synthesis by oxidative condensation of polyglycidol prepolymers, that are modified with thiol groups, results in crosslinking by disulfide bonds. Hereby, biomolecules like the antidiabetic peptide RS1-reg, derived from the regulatory protein RS1 of the Na+ -D-glucose cotransporter SGLT1, can be covalently bound by cysteine residues to the nanogel in a hydrophilic, stabilizing environment. After oral uptake, the acid-stable nanogels protect their loading during gastric passage from proteolytic degradation. Under alkaline conditions in small intestine the nanohydrogels become mucoadhesive, pass the intestinal mucosa and are taken up into small intestinal enterocytes by endocytosis. Using Caco-2 cells as a model for small intestinal enterocytes, by confocal laser scanning microscopy and structured illumination microscopy, the colocalization of fluorescent-labeled RS1-reg with markers of endosomes, lysosomes, and trans-Golgi-network after uptake with polyglycidol-based nanogels formed by precipitation polymerization is demonstrated. This indicates that RS1-reg follows the endosomal pathway. In the following, the design of bespoken nanohydrogels for specific targeting of RS1-reg to its site of action at the trans-Golgi network is described that might also represent a way of targeted transport for other drugs to their targets at the Golgi apparatus.
Collapse
Affiliation(s)
- Thorsten Keller
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Nora Trinks
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jessica Brand
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Steffen Trippmacher
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Krystyna Albrecht
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Georg Papastavrou
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Hermann Koepsell
- Institute of Anatomy and Cell BiologyUniversity of WürzburgKoellikerstraße 697070WürzburgGermany
| | - Markus Sauer
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| |
Collapse
|
20
|
Kim WK, Choi W, Deshar B, Kang S, Kim J. Golgi Stress Response: New Insights into the Pathogenesis and Therapeutic Targets of Human Diseases. Mol Cells 2023; 46:191-199. [PMID: 36574967 PMCID: PMC10086555 DOI: 10.14348/molcells.2023.2152] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 12/29/2022] Open
Abstract
The Golgi apparatus modifies and transports secretory and membrane proteins. In some instances, the production of secretory and membrane proteins exceeds the capacity of the Golgi apparatus, including vesicle trafficking and the post-translational modification of macromolecules. These proteins are not modified or delivered appropriately due to insufficiency in the Golgi function. These conditions disturb Golgi homeostasis and induce a cellular condition known as Golgi stress, causing cells to activate the 'Golgi stress response,' which is a homeostatic process to increase the capacity of the Golgi based on cellular requirements. Since the Golgi functions are diverse, several response pathways involving TFE3, HSP47, CREB3, proteoglycan, mucin, MAPK/ETS, and PERK regulate the capacity of each Golgi function separately. Understanding the Golgi stress response is crucial for revealing the mechanisms underlying Golgi dynamics and its effect on human health because many signaling molecules are related to diseases, ranging from viral infections to fatal neurodegenerative diseases. Therefore, it is valuable to summarize and investigate the mechanisms underlying Golgi stress response in disease pathogenesis, as they may contribute to developing novel therapeutic strategies. In this review, we investigate the perturbations and stress signaling of the Golgi, as well as the therapeutic potentials of new strategies for treating Golgi stress-associated diseases.
Collapse
Affiliation(s)
- Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Wooseon Choi
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Barsha Deshar
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Shinwon Kang
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G, Canada
| | - Jiyoon Kim
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
21
|
Youden B, Jiang R, Carrier AJ, Servos MR, Zhang X. A Nanomedicine Structure-Activity Framework for Research, Development, and Regulation of Future Cancer Therapies. ACS NANO 2022; 16:17497-17551. [PMID: 36322785 DOI: 10.1021/acsnano.2c06337] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Despite their clinical success in drug delivery applications, the potential of theranostic nanomedicines is hampered by mechanistic uncertainty and a lack of science-informed regulatory guidance. Both the therapeutic efficacy and the toxicity of nanoformulations are tightly controlled by the complex interplay of the nanoparticle's physicochemical properties and the individual patient/tumor biology; however, it can be difficult to correlate such information with observed outcomes. Additionally, as nanomedicine research attempts to gradually move away from large-scale animal testing, the need for computer-assisted solutions for evaluation will increase. Such models will depend on a clear understanding of structure-activity relationships. This review provides a comprehensive overview of the field of cancer nanomedicine and provides a knowledge framework and foundational interaction maps that can facilitate future research, assessments, and regulation. By forming three complementary maps profiling nanobio interactions and pathways at different levels of biological complexity, a clear picture of a nanoparticle's journey through the body and the therapeutic and adverse consequences of each potential interaction are presented.
Collapse
Affiliation(s)
- Brian Youden
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Runqing Jiang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, Ontario N2G 1G3, Canada
| | - Andrew J Carrier
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Xu Zhang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| |
Collapse
|
22
|
Moreno-Echeverri AM, Susnik E, Vanhecke D, Taladriz-Blanco P, Balog S, Petri-Fink A, Rothen-Rutishauser B. Pitfalls in methods to study colocalization of nanoparticles in mouse macrophage lysosomes. J Nanobiotechnology 2022; 20:464. [PMID: 36309696 PMCID: PMC9618187 DOI: 10.1186/s12951-022-01670-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background In the field of nanoscience there is an increasing interest to follow dynamics of nanoparticles (NP) in cells with an emphasis on endo-lysosomal pathways and long-term NP fate. During our research on this topic, we encountered several pitfalls, which can bias the experimental outcome. We address some of these pitfalls and suggest possible solutions. The accuracy of fluorescence microscopy methods has an important role in obtaining insights into NP interactions with lysosomes at the single cell level including quantification of NP uptake in a specific cell type. Methods Here we use J774A.1 cells as a model for professional phagocytes. We expose them to fluorescently-labelled amorphous silica NP with different sizes and quantify the colocalization of fluorescently-labelled NP with lysosomes over time. We focus on confocal laser scanning microscopy (CLSM) to obtain 3D spatial information and follow live cell imaging to study NP colocalization with lysosomes. Results We evaluate different experimental parameters that can bias the colocalization coefficients (i.e., Pearson’s and Manders’), such as the interference of phenol red in the cell culture medium with the fluorescence intensity and image post-processing (effect of spatial resolution, optical slice thickness, pixel saturation and bit depth). Additionally, we determine the correlation coefficients for NP entering the lysosomes under four different experimental set-ups. First, we found out that not only Pearson’s, but also Manders’ correlation coefficient should be considered in lysosome-NP colocalization studies; second, there is a difference in NP colocalization when using NP of different sizes and fluorescence dyes and last, the correlation coefficients might change depending on live-cell and fixed-cell imaging set-up. Conclusions The results summarize detailed steps and recommendations for the experimental design, staining, sample preparation and imaging to improve the reproducibility of colocalization studies between the NP and lysosomes. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01670-9.
Collapse
|
23
|
Wang J, Zhang Z, Zhang R, Du H, Zhou T, Wang F. "Willow Branch" DNA Self-Assembly for Cancer Dual-Target and Proliferation Inhibition. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:11778-11786. [PMID: 36102591 DOI: 10.1021/acs.langmuir.2c01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
DNA nanotechnology is beginning to yield unique advantages in the area of drug delivery. For the dual-targeting and proliferation suppression of cancer cells, a "willow branch" DNA assembly based on rolling circle amplification (RCA) was built. Three single-stranded DNAs, including antibody modified cDNAs, aptamer cDNAs, and simple cDNAs, were employed in the DNA self-assembly, along with the RCA scaffolds (every 63 bases is a repeat unit). "Willow branch" DNA (WB DNA) assembly successfully linked multiple antibodies and aptamers together to achieve dual targeting of cancer cells. Binding of CD44 antibodies and S2.2 aptamers to receptors on the cell membrane inhibits both pathways, β-catenin signaling and nuclear factor-kappa B-specific transcription activity, through feedback regulation. Results demonstrated that WB DNA assembly could effectively exert multivalency clustering cell-surface receptors, modulating signal pathways and inhibiting proliferation. This study proposes a new approach for cancer dual-target and proliferation inhibition by clustering multivalent receptors.
Collapse
Affiliation(s)
- Jiawei Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Zhiqing Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ruyan Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Huan Du
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ting Zhou
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Fang Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
24
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
25
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
26
|
Yang Z, Zhang Z, Zhao Y, Ye Q, Li X, Meng L, Long J, Zhang S, Zhang L. Organelle Interaction and Drug Discovery: Towards Correlative Nanoscopy and Molecular Dynamics Simulation. Front Pharmacol 2022; 13:935898. [PMID: 35795548 PMCID: PMC9251060 DOI: 10.3389/fphar.2022.935898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
The inter-organelle interactions, including the cytomembrane, endoplasmic reticulum, mitochondrion, lysosome, dictyosome, and nucleus, play the important roles in maintaining the normal function and homeostasis of cells. Organelle dysfunction can lead to a range of diseases (e.g., Alzheimer's disease (AD), Parkinson's disease (PD), and cancer), and provide a new perspective for drug discovery. With the development of imaging techniques and functional fluorescent probes, a variety of algorithms and strategies have been developed for the ever-improving estimation of subcellular structures, organelle interaction, and organelle-related drug discovery with accounting for the dynamic structures of organelles, such as the nanoscopy technology and molecular dynamics (MD) simulations. Accordingly, this work summarizes a series of state-of-the-art examples of the recent progress in this rapidly changing field and uncovering the drug screening based on the structures and interactions of organelles. Finally, we propose the future outlook for exciting applications of organelle-related drug discovery, with the cooperation of nanoscopy and MD simulations.
Collapse
Affiliation(s)
- Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Zichen Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| | - Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| | - Qiushi Ye
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| | - Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| | - Lingjie Meng
- School of Chemistry, Xi’an Jiaotong University, Xi’an, China
- Instrumental Analysis Center, Xi’an Jiaotong University, Xi’an, China
| | - Jiangang Long
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
27
|
Sun S, Yang Y, Niu H, Luo M, Wu ZS. Design and application of DNA nanostructures for organelle-targeted delivery of anticancer drugs. Expert Opin Drug Deliv 2022; 19:707-723. [PMID: 35618266 DOI: 10.1080/17425247.2022.2083603] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION DNA nanostructures targeting organelles are of great significance for the early diagnosis and precise therapy of human cancers. This review is expected to promote the development of DNA nanostructure-based cancer treatment with organelle-level precision in the future. AREAS COVERED In this review, we introduce the different principles for targeting organelles, summarize the progresses in the development of organelle-targeting DNA nanostructures, highlight their advantages and applications in disease treatment, and discuss current challenges and future prospects. EXPERT OPINION Accurate targeting is a basic problem for effective cancer treatment. However, current DNA nanostructures cannot meet the actual needs. Targeting specific organelles is expected to further improve the therapeutic effect and overcome tumor cell resistance, thereby holding great practical significance for tumor treatment in the clinic. With the deepening of the research on the molecular mechanism of disease development, especially on tumorigenesis and tumor progression, and increasing understanding of the behavior of biological materials in living cells, more versatile DNA nanostructures will be constructed to target subcellular organelles for drug delivery, essentially promoting the early diagnosis of cancers, classification, precise therapy and the estimation of prognosis in the future.
Collapse
Affiliation(s)
- Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China.,Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Huimin Niu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China.,Fujian Key Laboratory of Aptamers Technology, The 900th Hospital of Joint Logistics Support Force, Fuzhou 350025, China
| | - Mengxue Luo
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| |
Collapse
|
28
|
The Promise of Nanotechnology in Personalized Medicine. J Pers Med 2022; 12:jpm12050673. [PMID: 35629095 PMCID: PMC9142986 DOI: 10.3390/jpm12050673] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Both personalized medicine and nanomedicine are new to medical practice. Nanomedicine is an application of the advances of nanotechnology in medicine and is being integrated into diagnostic and therapeutic tools to manage an array of medical conditions. On the other hand, personalized medicine, which is also referred to as precision medicine, is a novel concept that aims to individualize/customize therapeutic management based on the personal attributes of the patient to overcome blanket treatment that is only efficient in a subset of patients, leaving others with either ineffective treatment or treatment that results in significant toxicity. Novel nanomedicines have been employed in the treatment of several diseases, which can be adapted to each patient-specific case according to their genetic profiles. In this review, we discuss both areas and the intersection between the two emerging scientific domains. The review focuses on the current situation in personalized medicine, the advantages that can be offered by nanomedicine to personalized medicine, and the application of nanoconstructs in the diagnosis of genetic variability that can identify the right drug for the right patient. Finally, we touch upon the challenges in both fields towards the translation of nano-personalized medicine.
Collapse
|
29
|
Liu F, Danylchuk DI, Andreiuk B, Klymchenko AS. Dynamic covalent chemistry in live cells for organelle targeting and enhanced photodynamic action. Chem Sci 2022; 13:3652-3660. [PMID: 35432899 PMCID: PMC8966643 DOI: 10.1039/d1sc04770a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/03/2022] [Indexed: 12/22/2022] Open
Abstract
Organelle-specific targeting enables increasing the therapeutic index of drugs and localizing probes for better visualization of cellular processes. Current targeting strategies require conjugation of a molecule of interest with organelle-targeting ligands. Here, we propose a concept of dynamic covalent targeting of organelles where the molecule is conjugated with its ligand directly inside live cells through a dynamic covalent bond. For this purpose, we prepared a series of organelle-targeting ligands with a hydrazide residue for reacting with dyes and drugs bearing a ketone group. We show that dynamic hydrazone bond can be formed between these hydrazide ligands and a ketone-functionalized Nile Red dye (NRK) in situ in model lipid membranes or nanoemulsion droplets. Fluorescence imaging in live cells reveals that the targeting hydrazide ligands can induce preferential localization of NRK dye and an anti-cancer drug doxorubicin in plasma membranes, mitochondria and lipid droplets. Thus, with help of the dynamic covalent targeting, it becomes possible to direct a given bioactive molecule to any desired organelle inside the cell without its initial functionalization by the targeting ligand. Localizing the same NRK dye in different organelles by the hydrazide ligands is found to affect drastically its photodynamic activity, with the most pronounced phototoxic effects in mitochondria and plasma membranes. The capacity of this approach to tune biological activity of molecules can improve efficacy of drugs and help to understand better their intracellular mechanisms.
Collapse
Affiliation(s)
- Fei Liu
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, ITI Chimie des Systèmes Complexes, Université de Strasbourg 74 Route du Rhin 67401 Illkirch France
| | - Dmytro I Danylchuk
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, ITI Chimie des Systèmes Complexes, Université de Strasbourg 74 Route du Rhin 67401 Illkirch France
| | - Bohdan Andreiuk
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, ITI Chimie des Systèmes Complexes, Université de Strasbourg 74 Route du Rhin 67401 Illkirch France
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, ITI Chimie des Systèmes Complexes, Université de Strasbourg 74 Route du Rhin 67401 Illkirch France
| |
Collapse
|
30
|
Yoshinaga N, Numata K. Rational Designs at the Forefront of Mitochondria-Targeted Gene Delivery: Recent Progress and Future Perspectives. ACS Biomater Sci Eng 2022; 8:348-359. [PMID: 34979085 DOI: 10.1021/acsbiomaterials.1c01114] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mitochondria play an essential role in cellular metabolism and generate energy in cells. To support these functions, several proteins are encoded in the mitochondrial DNA (mtDNA). The mutation of mtDNA causes mitochondrial dysfunction and ultimately results in a variety of inherited diseases. To date, gene delivery systems targeting mitochondria have been developed to ameliorate mtDNA mutations. However, applications of these strategies in mitochondrial gene therapy are still being explored and optimized. Thus, from this perspective, we herein highlight recent mitochondria-targeting strategies for gene therapy and discuss future directions for effective mitochondria-targeted gene delivery.
Collapse
Affiliation(s)
- Naoto Yoshinaga
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Keiji Numata
- Biomacromolecule Research Team, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan.,Department of Material Chemistry, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Roy B, Mengji R, Roy S, Pal B, Jana A, Singh NDP. NIR-Responsive Lysosomotropic Phototrigger: An "AIE + ESIPT" Active Naphthalene-Based Single-Component Photoresponsive Nanocarrier with Two-Photon Uncaging and Real-Time Monitoring Ability. ACS APPLIED MATERIALS & INTERFACES 2022; 14:4862-4870. [PMID: 35049266 DOI: 10.1021/acsami.1c19022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In recent times, organelle-targeted drug delivery systems have gained tremendous attention due to the site-specific delivery of active drug molecules, resulting in enhanced bioefficacy. In this context, a phototriggered drug delivery system (DDS) for releasing an active molecule is superior, as it provides spatial and temporal control over the release. So far, a near-infrared (NIR) light-responsive organelle-targeted DDS has not yet been developed. Hence, we introduced a two-photon NIR light-responsive lysosome-targeted "AIE + ESIPT" active single-component DDS based on the naphthalene chromophore. The two-photon absorption cross section of our DDS is 142 GM at 850 nm. The DDS was converted into pure organic nanoparticles for biological applications. Our nano-DDS is capable of selective targeting, AIE luminogenic imaging, and drug release within the lysosome. In vitro studies using cancerous cell lines showed that our single-component photoresponsive nanocarrier exhibited enhanced cytotoxicity and real-time monitoring ability of drug release.
Collapse
Affiliation(s)
- Biswajit Roy
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Rakesh Mengji
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Samrat Roy
- Department of Physics, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Bipul Pal
- Department of Physics, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal 741246, India
| | - Avijit Jana
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology Hyderabad, Uppal Road, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - N D Pradeep Singh
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
32
|
Cerrato CP, Langel Ü. An update on cell-penetrating peptides with intracellular organelle targeting. Expert Opin Drug Deliv 2022; 19:133-146. [PMID: 35086398 DOI: 10.1080/17425247.2022.2034784] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Cell-penetrating peptide (CPP) technologies represent an important strategy to address drug delivery to specific intracellular compartments by covalent conjugation to targeting sequences, potentially enabling strategies to combat most diseases. AREAS COVERED This updated review article provides an overview of current intracellular organelle targeting by CPP. The targeting strategies of CPP and CPP/cargo complexes to specific cells or intracellular organelles are summarized, and the review provides an update on the current data for their pharmacological and therapeutical applications. EXPERT OPINION Targeted drug delivery is moving from the level of tissue or specific pathogenic cell to the level of specific organelle that is the target of the drug, an important aspect in drug design and development. Organelle-targeted drug delivery results in improved efficacy, ability to control mode of action, reduction of undesired toxicities and side effects, and possibility to overcome drug resistance mechanisms.
Collapse
Affiliation(s)
| | - Ülo Langel
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.,Laboratory of Molecular Biotechnology, Institute of Technology, University of Tartu, Tartu, Estonia
| |
Collapse
|
33
|
Lv S, Sylvestre M, Prossnitz AN, Yang LF, Pun SH. Design of Polymeric Carriers for Intracellular Peptide Delivery in Oncology Applications. Chem Rev 2021; 121:11653-11698. [PMID: 33566580 DOI: 10.1021/acs.chemrev.0c00963] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent decades, peptides, which can possess high potency, excellent selectivity, and low toxicity, have emerged as promising therapeutics for cancer applications. Combined with an improved understanding of tumor biology and immuno-oncology, peptides have demonstrated robust antitumor efficacy in preclinical tumor models. However, the translation of peptides with intracellular targets into clinical therapies has been severely hindered by limitations in their intrinsic structure, such as low systemic stability, rapid clearance, and poor membrane permeability, that impede intracellular delivery. In this Review, we summarize recent advances in polymer-mediated intracellular delivery of peptides for cancer therapy, including both therapeutic peptides and peptide antigens. We highlight strategies to engineer polymeric materials to increase peptide delivery efficiency, especially cytosolic delivery, which plays a crucial role in potentiating peptide-based therapies. Finally, we discuss future opportunities for peptides in cancer treatment, with an emphasis on the design of polymer nanocarriers for optimized peptide delivery.
Collapse
Affiliation(s)
| | | | - Alexander N Prossnitz
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | | | | |
Collapse
|
34
|
Aydin F, Durumeric AEP, da Hora GCA, Nguyen JDM, Oh MI, Swanson JMJ. Improving the accuracy and convergence of drug permeation simulations via machine-learned collective variables. J Chem Phys 2021; 155:045101. [PMID: 34340389 DOI: 10.1063/5.0055489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Understanding the permeation of biomolecules through cellular membranes is critical for many biotechnological applications, including targeted drug delivery, pathogen detection, and the development of new antibiotics. To this end, computer simulations are routinely used to probe the underlying mechanisms of membrane permeation. Despite great progress and continued development, permeation simulations of realistic systems (e.g., more complex drug molecules or biologics through heterogeneous membranes) remain extremely challenging if not intractable. In this work, we combine molecular dynamics simulations with transition-tempered metadynamics and techniques from the variational approach to conformational dynamics to study the permeation mechanism of a drug molecule, trimethoprim, through a multicomponent membrane. We show that collective variables (CVs) obtained from an unsupervised machine learning algorithm called time-structure based Independent Component Analysis (tICA) improve performance and substantially accelerate convergence of permeation potential of mean force (PMF) calculations. The addition of cholesterol to the lipid bilayer is shown to increase both the width and height of the free energy barrier due to a condensing effect (lower area per lipid) and increase bilayer thickness. Additionally, the tICA CVs reveal a subtle effect of cholesterol increasing the resistance to permeation in the lipid head group region, which is not observed when canonical CVs are used. We conclude that the use of tICA CVs can enable more efficient PMF calculations with additional insight into the permeation mechanism.
Collapse
Affiliation(s)
- Fikret Aydin
- Quantum Simulation Group, Lawrence Livermore National Laboratory, Livermore, California 94550, USA
| | | | - Gabriel C A da Hora
- Department of Chemistry, University of Utah, Salt Lake City, Utah 84112-0850, USA
| | - John D M Nguyen
- Department of Chemistry, University of Utah, Salt Lake City, Utah 84112-0850, USA
| | - Myong In Oh
- Department of Chemistry, University of Utah, Salt Lake City, Utah 84112-0850, USA
| | - Jessica M J Swanson
- Department of Chemistry, University of Utah, Salt Lake City, Utah 84112-0850, USA
| |
Collapse
|
35
|
Tremi I, Spyratou E, Souli M, Efstathopoulos EP, Makropoulou M, Georgakilas AG, Sihver L. Requirements for Designing an Effective Metallic Nanoparticle (NP)-Boosted Radiation Therapy (RT). Cancers (Basel) 2021; 13:cancers13133185. [PMID: 34202342 PMCID: PMC8269428 DOI: 10.3390/cancers13133185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent advances in nanotechnology gave rise to trials with various types of metallic nanoparticles (NPs) to enhance the radiosensitization of cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy. This work reviews the physical and chemical mechanisms leading to the enhancement of ionizing radiation’s detrimental effects on cells and tissues, as well as the plethora of experimental procedures to study these effects of the so-called “NPs’ radiosensitization”. The paper presents the need to a better understanding of all the phases of actions before applying metallic-based NPs in clinical practice to improve the effect of IR therapy. More physical and biological experiments especially in vivo must be performed and simulation Monte Carlo or mathematical codes based on more accurate models for all phases must be developed. Abstract Many different tumor-targeted strategies are under development worldwide to limit the side effects and improve the effectiveness of cancer therapies. One promising method is to enhance the radiosensitization of the cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy using metallic nanoparticles (NPs). Radiotherapy with MV photons is more commonly available and applied in cancer clinics than high LET particle radiotherapy, so the addition of high-Z NPs has the potential to further increase the efficacy of photon radiotherapy in terms of NP radiosensitization. Generally, when using X-rays, mainly the inner electron shells are ionized, which creates cascades of both low and high energy Auger electrons. When using high LET particles, mainly the outer shells are ionized, which give electrons with lower energies than when using X-rays. The amount of the produced low energy electrons is higher when exposing NPs to heavy charged particles than when exposing them to X-rays. Since ions traverse the material along tracks, and therefore give rise to a much more inhomogeneous dose distributions than X-rays, there might be a need to introduce a higher number of NPs when using ions compared to when using X-rays to create enough primary and secondary electrons to get the desired dose escalations. This raises the questions of toxicity. This paper provides a review of the fundamental processes controlling the outcome of metallic NP-boosted photon beam and ion beam radiation therapy and presents some experimental procedures to study the biological effects of NPs’ radiosensitization. The overview shows the need for more systematic studies of the behavior of NPs when exposed to different kinds of ionizing radiation before applying metallic-based NPs in clinical practice to improve the effect of IR therapy.
Collapse
Affiliation(s)
- Ioanna Tremi
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Maria Souli
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Mersini Makropoulou
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Correspondence: (A.G.G.); (L.S.)
| | - Lembit Sihver
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Correspondence: (A.G.G.); (L.S.)
| |
Collapse
|
36
|
Lee JH, Shin HJ, Kim YD, Lim DK. Real-time surface-enhanced Raman scattering-based live cell monitoring of the changes in mitochondrial membrane potential. NANOSCALE ADVANCES 2021; 3:3470-3480. [PMID: 36133723 PMCID: PMC9418680 DOI: 10.1039/d0na01076f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/07/2021] [Indexed: 06/13/2023]
Abstract
Obtaining molecular information on cells in real time has been a critical challenge in studying the interaction between molecules of interest and intracellular components. Fluorescence-based methods have long served as excellent tools to study such important interactions. In this paper, we introduce a Raman scattering-based method as a promising platform to achieve the real-time monitoring of subtle molecular changes occurring within cells. We found that the Raman scattering-based method enabled monitoring changes in the mitochondrial membrane potential at the single-cell level in rheumatoid arthritis synovial fibroblasts induced by tumor necrosis factor-alpha (TNF-α) protein, various chemicals (MgCl2, FCCP, and sodium pyruvate), and a non-chemical stimulus (i.e., light). The triphenylphosphine-modified gold nanoparticles were selectively localized in the mitochondria and showed the characteristic Raman spectrum of cytochrome C and other Raman spectra of molecular components inside the cell. The surface-enhanced Raman spectrum originating from mitochondria was sensitively changed over time when mitochondrial depolarization was induced by the addition of TNF-α, or chemicals known to induce mitochondrial depolarization. The Raman-based signal changes were well matched with results of the conventional fluorescence-based analysis. However, in contrast to the conventional approach, the Raman-based method enables monitoring such changes in real time and provides detailed molecular information in terms of the interaction of molecules. Therefore, these results highlight the possibility of surface-enhanced Raman scattering-based live cell analysis for future proteomics or drug-screening applications.
Collapse
Affiliation(s)
- Ji Hye Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul South Korea
| | - Hyeon Jeong Shin
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul South Korea
| | - Yong Duk Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul South Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul South Korea
| |
Collapse
|
37
|
Hu Q, He C, Lu Z, Xu L, Fu Z. Mitochondria and Endoplastic Reticulum Targeting Strategy for Enhanced Phototherapy. ACS APPLIED BIO MATERIALS 2021; 4:3015-3026. [PMID: 35014389 DOI: 10.1021/acsabm.1c00155] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To ensure improved efficacy and minimized toxicity of therapeutic molecules, it is generally accepted that specifically delivering them to the subcellular site of their action will be attractive. Phototherapy has received considerable attention because of its noninvasiveness, high temporal-spatial resolution, and minimal drug resistance. As important functional organelles in cells, mitochondria and endoplasmic reticulum (ER) participate in fundamental cellular processes, which make them much more sensitive to reactive oxygen species (ROS) and hyperthermia. Thus, mitochondria- or ER-targeted phototherapy will be rational strategies for synergetic cancer therapy. In this review, we focus on the latest advances in molecules and nanomaterials currently used for mitochondria- and ER-targeted phototherapy.
Collapse
Affiliation(s)
- Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Chao He
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhuoting Lu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liwang Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
38
|
El-Kahky D, Attia M, Easa SM, Awad NM, Helmy EA. Interactive Effects of Biosynthesized Nanocomposites and Their Antimicrobial and Cytotoxic Potentials. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:903. [PMID: 33916082 PMCID: PMC8067103 DOI: 10.3390/nano11040903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/14/2022]
Abstract
The present study investigated the biosynthesis of silver (AgNPs), zinc oxide (ZnONPs) and titanium dioxide (TiO2NPs) nanoparticles using Aspergillusoryzae, Aspergillusterreus and Fusariumoxysporum. Nanocomposites (NCs) were successfully synthesized by mixing nanoparticles using a Sonic Vibra-Cell VC/VCX processor. A number of analytical techniques were used to characterize the synthesized biological metal nanoparticles. Several experiments tested biologically synthesized metal nanoparticles and nanocomposites against two types of human pathogenic bacteria, including Gram-positive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA), and Gram-negative Escherichia coli and Pseudomonasaeruginosa. Additionally, the antitumor activity in HCT-116 cells (colonic carcinoma) was also evaluated. Significant antimicrobial effects of various synthesized forms of nanoparticles and nanocomposites against E. coli and P. aeruginosa bacteria were detected. Various synthesized biogenic forms of nanoparticles and nanocomposite (9.0 to 29 mm in diameter) had high antibacterial activity and high antitumor activity against HCT-116 cells (colonic carcinoma) with IC50 values of 0.7-100 µg/mL. Biosynthesized NPs are considered an alternative to large-scale biosynthesized metallic nanoparticles and nanocomposites, are simple and cost effective, and provide stable nanomaterials.
Collapse
Affiliation(s)
- Dina El-Kahky
- Microbiology Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt;
| | - Magdy Attia
- Agricultural Microbiology Department, National Research Centre, 33 El-Bohouth Street, (Former El-Tahrir Street) Dokki, Giza 12622, Egypt; (M.A.); (N.M.A.)
| | - Saadia M. Easa
- Microbiology Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt;
| | - Nemat M. Awad
- Agricultural Microbiology Department, National Research Centre, 33 El-Bohouth Street, (Former El-Tahrir Street) Dokki, Giza 12622, Egypt; (M.A.); (N.M.A.)
| | - Eman A. Helmy
- Microbiology Department, The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11651, Egypt;
| |
Collapse
|
39
|
Rajagopal P, Jayandharan GR, Krishnan UM. Evaluation of the Anticancer Activity of pH-Sensitive Polyketal Nanoparticles for Acute Myeloid Leukemia. Mol Pharm 2021; 18:2015-2031. [PMID: 33780253 DOI: 10.1021/acs.molpharmaceut.0c01243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polyketals are a class of acid-responsive polymers that have been relatively less explored for drug delivery applications compared to polyesters. The degradation of these polymers is accelerated in an acidic medium and does not result in acidic byproducts. Their biocompatibility depends on the diol used for the synthesis. The present work aims to synthesize, characterize, and fabricate nanospheres of an aliphatic polyketal for delivery of the nucleotide analogue cytarabine toward the treatment of acute myeloid leukemia (AML). The internalization mechanism of the nanospheres was probed, and its implication on the nuclear localization and escape from the endo-lysosomal compartments were studied. The drug-loaded polyketal nanoparticles reduced the cell viability to a greater extent compared with the free drug. The effect of the drug-loaded polyketal nanoparticles on the differential gene expression of leukemic cells was investigated for the first time to understand their therapeutic implications. It was found that treatment with drug-loaded polyketal nanoparticles downregulated AML-specific genes involved in cell proliferation and recurrence compared to the free drug. The protein expression studies were performed for selected genes obtained from gene expression analysis. Biodistribution studies showed that the poly(cyclohexane-1,4-diyl acetone dimethylene ketal) (PCADK) nanoparticles exhibit prolonged circulation time. Overall, our results suggest that polyketal-based delivery of cytarabine represents a more effective alternative strategy for AML therapy.
Collapse
Affiliation(s)
- Pratheppa Rajagopal
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India.,The Mehta Family Centre for Engineering In Medicine, Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University Thanjavur 613401, India.,School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur 613401, India.,School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613401, India
| |
Collapse
|
40
|
Liu J, Ding G, Chen S, Xue C, Chen M, Wu X, Yuan Q, Zheng J, Yang R. Multifunctional Programmable DNA Nanotrain for Activatable Hypoxia Imaging and Mitochondrion-Targeted Enhanced Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9681-9690. [PMID: 33606499 DOI: 10.1021/acsami.0c21681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Programmable DNA-based nanostructures (e.g., nanotrains, nanoflowers, and DNA dendrimers) provide new approaches for safe and effective biological imaging and tumor therapy. However, few studies have reported that DNA-based nanostructures respond to the hypoxic microenvironment for activatable imaging and organelle-targeted tumor therapy. Herein, we innovatively report an azoreductase-responsive, mitochondrion-targeted multifunctional programmable DNA nanotrain for activatable hypoxia imaging and enhanced efficacy of photodynamic therapy (PDT). Cyanine structural dye (Cy3) and black hole quencher 2 (BHQ2), which were employed as a fluorescent mitochondrion-targeted molecule and azoreductase-responsive element, respectively, covalently attached to the DNA hairpin monomers. The extended guanine (G)-rich sequence at the end of the DNA hairpin monomer served as a nanocarrier for the photosensitizer 5,10,15,20-tetrakis(4-N-methylpyridiniumyl) porphyrin (TMPyP4). Upon initiation between the DNA hairpin monomer and initiation probe, the fluorescence of Cy3 and the singlet oxygen (1O2) generation of TMPyP4 in the programmable nanotrain were effectively quenched by BHQ2 through the fluorescence resonance energy transfer (FRET) process. Once the programmable nanotrain entered cancer cells, the azo bond in BHQ2 will be reduced to amino groups by the high expression of azoreductase under hypoxia conditions; then, the fluorescence of Cy3 and the 1O2 generation of TMPyP4 will significantly be restored. Furthermore, due to the mitochondrion-targeting characteristic endowed by Cy3, the TMPyP4-loaded nanotrain would accumulate in the mitochondria of cancer cells and then demonstrate enhanced PDT efficacy under light irradiation. We expect that this programmable DNA nanotrain-based multifunctional nanoplatform could be effectively used for activatable imaging and high performance of PDT in hypoxia-related biomedical field.
Collapse
Affiliation(s)
- Jin Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Institute of Chemical Biology and Nanomedicine (ICBN), Hunan University, Changsha 410082, China
| | - Ge Ding
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Health Science Center, School of Biomedical Engineering, Shenzhen University, Shenzhen 518055, China
| | - Shiya Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Institute of Chemical Biology and Nanomedicine (ICBN), Hunan University, Changsha 410082, China
| | - Caoye Xue
- Hunan Institute of Sports Science, Changsha 410003, China
| | - Mian Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Health Science Center, School of Biomedical Engineering, Shenzhen University, Shenzhen 518055, China
| | - Xu Wu
- NHC Key Laboratory of Carcinogenesis and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha 410083, China
| | - Quan Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Institute of Chemical Biology and Nanomedicine (ICBN), Hunan University, Changsha 410082, China
| | - Jing Zheng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Institute of Chemical Biology and Nanomedicine (ICBN), Hunan University, Changsha 410082, China
| | - Ronghua Yang
- School of Chemistry and Biological Engineering, Changsha University of Science and Technology, Changsha 410004, China
| |
Collapse
|
41
|
Andraos C, Gulumian M. Intracellular and extracellular targets as mechanisms of cancer therapy by nanomaterials in relation to their physicochemical properties. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1680. [PMID: 33111484 PMCID: PMC7988657 DOI: 10.1002/wnan.1680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022]
Abstract
Cancer nanomedicine has evolved in recent years and is only expected to increase due to the ease with which nanomaterials (NMs) may be manipulated to the advantage of the cancer patient. The success of nanomedicine is dependent on the cell death mechanism, which in turn is dependent on the organelle initially targeted. The success of cancer nanomedicine is also dependent on other cellular mechanisms such as the induction of autophagy dysfunction, manipulation of the tumor microenvironment (TME) and secretome or induction of host immune responses. Current cancer phototherapies for example, photothermal- or photodynamic therapies as well as radio enhancement also form a major part of cancer nanomedicine. In general, cancer nanomedicine may be grouped into those NMs exhibiting inherent anti-cancer properties that is, self-therapeutic NMs (Group 1), NMs leading to localization of phototherapies or radio-enhancement (Group 2), and NMs as nanocarriers in the absence or presence of external radiation (Group 3). The recent advances of these three groups, together with their advantages and disadvantages as well as their cellular mechanisms and ultimate outcomes are summarized in this review. By exploiting these different intracellular mechanisms involved in initiating cell death pathways, it is possible to synthesize NMs that may have the desirable characteristics to maximize their efficacy in cancer therapy. Therefore, a summary of these important physicochemical characteristics is also presented that need to be considered for optimal cancer cell targeting and initiation of mechanisms that will lead to cancerous cell death. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Charlene Andraos
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
| | - Mary Gulumian
- Toxicology DepartmentNational Institute for Occupational HealthJohannesburgSouth Africa
- Haematology and Molecular Medicine DepartmentUniversity of the WitwatersrandJohannesburgSouth Africa
- Water Research Group, Unit for Environmental Sciences and ManagementNorth West UniversityPotchefstroomSouth Africa
| |
Collapse
|
42
|
Kim BJ, Fang Y, He H, Xu B. Trypsin-Instructed Self-Assembly on Endoplasmic Reticulum for Selectively Inhibiting Cancer Cells: Dedicated to Professor George M. Whitesides on the occasion of his 80th birthday. Adv Healthc Mater 2021; 10:e2000416. [PMID: 32342647 PMCID: PMC7725443 DOI: 10.1002/adhm.202000416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Selectively targeting the endoplasmic reticulum (ER) of cancer cells, though promising a new strategy for cancer therapy, remains underdeveloped. Enzyme-instructed self-assembly (EISA) is emerging as a promising approach for selectively targeting ER of cancer cells. This work reports an easily accessible branched peptide that consists of a D-tetrapeptide backbone and a branch with the sequence of KYDKKKKDG (K: lysine; Y: tyrosine; D: aspratic acid; G: glycine), being an EISA substrate of typsin-1 (PRSS1), selectively inhibits cancer cells. Depending on the type of cells, the level of PRSS1 expression dictates the cytotoxicity of the branched peptide. Moreover, immunostaining and fluorescent imaging reveal that PRSS1 overexpresses on the ER of a high-grade serous ovarian cancer cell line (OVSAHO). The overexpression of PRSS1 renders the branched peptide to exhibit high selectivity against OVSAHO by the in situ formation of the peptide assemblies on the ER of OVSAHO cells, which causes ER stress and eventual cell death. This work, illustrating trypsin-guided EISA for inhibiting cancer cells by enzymatic reaction on ER for the first time, offers a new way to target the subcellular organelles of cancer cells for potential cancer therapy.
Collapse
Affiliation(s)
- Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Yu Fang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| |
Collapse
|
43
|
Zeng J, Shirihai OS, Grinstaff MW. Modulating lysosomal pH: a molecular and nanoscale materials design perspective. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2020; 2:25-37. [PMID: 33403369 PMCID: PMC7781074 DOI: 10.36069/jols/20201204] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lysosomes, membrane-bound organelles, play important roles in cellular processes including endocytosis, phagocytosis, and autophagy. Lysosomes maintain cellular homeostasis by generating a highly acidic environment of pH 4.5 - 5.0 and by housing hydrolytic enzymes that degrade engulfed biomolecules. Impairment of lysosomal function, especially in its acidification, is a driving force in the pathogenesis of diseases including neurodegeneration, cancer, metabolic disorders, and infectious diseases. Therefore, lysosomal pH is an attractive and targetable site for therapeutic intervention. Currently, there is a dearth of strategies or materials available to specifically modulate lysosomal acidification. This review focuses on the key aspects of how lysosomal pH is implicated in various diseases and discusses design strategies and molecular or nanoscale agents for lysosomal pH modulation, with the ultimate goal of developing novel therapeutic solutions.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06511
| | - Orian S Shirihai
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90045
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
- Department of Chemistry, Boston University, Boston, MA 02215
| |
Collapse
|
44
|
Nair JB, Joseph MM, Arya JS, Sreedevi P, Sujai PT, Maiti KK. Elucidating a Thermoresponsive Multimodal Photo-Chemotherapeutic Nanodelivery Vehicle to Overcome the Barriers of Doxorubicin Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:43365-43379. [PMID: 32880178 DOI: 10.1021/acsami.0c08762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In an attempt to circumvent the major pitfalls associated with conventional chemotherapy including drug resistance and off-target toxicity, we have adopted a strategy to simultaneously target both mitochondrial DNA (Mt-DNA) and nuclear DNA (n-DNA) with the aid of a targeted theranostic nanodelivery vehicle (TTNDV). Herein, folic acid-anchored p-sulfo-calix[4]arene (SC4)-capped hollow gold nanoparticles (HGNPs) were meticulously loaded with antineoplastic doxorubicin (Dox) and its mitochondrion-targeted analogue, Mt-Dox, in a pretuned ratio (1:100) for sustained thermoresponsive release of cargo. This therapeutic strategy was enabled to eradicate both n-DNA and Mt-DNA leaving no space to develop drug resistance. The SC4-capped HGNPs (HGNPSC4) were experimented for the first time as a photothermal (PTT) agent with 61.6% photothermal conversion efficiency, and they generated tunable localized heat more efficiently than bare HGNPs. Moreover, the cavity of SC4 facilitated the formation of an inclusion complex with folic acid to target the folate receptor expressing cancer cells and imparted enhanced biocompatibility. The as-synthesized TTNDV was demonstrated to be an ideal substrate for surface-enhanced Raman scattering (SERS) to monitor the molecular-level therapeutic progression in cells and a spheroidal model. A significant reduction in the tumor mass with a marked survival benefit was achieved in syngraft murine models through this synergistic photo-chemotherapy. Collectively, this multifunctional nanoplatform offers a robust approach to treat cancer without any scope of generating Dox resistance and off-target toxicity.
Collapse
Affiliation(s)
- Jyothi B Nair
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manu M Joseph
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
| | - Jayadev S Arya
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Padincharapad Sreedevi
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
| | - Palasseri T Sujai
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
45
|
Purushothaman B, Lee J, Hong S, Song JM. Multifunctional TPP-PEG-biotin self-assembled nanoparticle drug delivery-based combination therapeutic approach for co-targeting of GRP78 and lysosome. J Nanobiotechnology 2020; 18:102. [PMID: 32690101 PMCID: PMC7372800 DOI: 10.1186/s12951-020-00661-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Background In this study, a multifunctional tetraphenylporphyrin (TPP) conjugated polyethylene glycol with biotin (TPP-PEG-biotin) as a photo-dynamic therapy (PDT) material encapsulating a ruthenium complex 1 (Ru-1) was fabricated as self-assembled nanoparticle (Ru-1@TPP-PEG-biotin SAN) to co-target glucose-regulated protein 78 (GRP78) and the lysosome as a new anti-cancer therapeutic strategy. Results The MTT assay results reveals the enhanced anticancer activity of the Ru-1@TPP-PEG-biotin SANs due to the co-targeting of the GRP78 and lysosome. The Ru-1@TPP-PEG-biotin reduced level of GRP78 and lysosomal ceramide that contributed to the stability of the lysosomal membrane. The endoplasmic reticulum (ER) stress concomitant with the inhibition of GRP78 was clearly monitored by the phosphorylation of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), and inositol-requiring enzyme 1 α (IRE1α) kinases to indicate the activation of the unfolded protein response (UPR) signaling using immunofluorescence assay. On the other hand, the degradation of the lysosome was observed through PDT action by the Ru-1@TPP-PEG-biotin SAN treatment. This was confirmed by the co-localization assay showing the disappearance of cathepsin D and lysosomal-associated membrane protein 1 (LAMP1) in the lysosome. Conclusions Considering lysosome-mediated autophagy is an effective cancer cell survival mechanism, the degradation of the lysosome along with GRP78 inhibition by the Ru-1@TPP-PEG-biotin SAN combination therapy is suggested as a new co-targeting cancer treatment.![]()
Collapse
Affiliation(s)
| | - Jeongmin Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Sera Hong
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Joon Myong Song
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
46
|
Parra-Nieto J, Del Cid MAG, de Cárcer IA, Baeza A. Inorganic Porous Nanoparticles for Drug Delivery in Antitumoral Therapy. Biotechnol J 2020; 16:e2000150. [PMID: 32476279 DOI: 10.1002/biot.202000150] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/21/2020] [Indexed: 12/26/2022]
Abstract
The use of nanoparticles in oncology to deliver chemotherapeutic agents has received considerable attention in the last decades due to their tendency to be passively accumulated in solid tumors. Besides this remarkable property, the surface of these nanocarriers can be decorated with targeting moieties capable to recognize malignant cells which lead to selective nanoparticle uptake mainly in the diseased cells, without affecting the healthy ones. Among the different nanocarriers which have been developed with this purpose, inorganic porous nanomaterials constitute some of the most interesting due to their unique properties such as excellent cargo capacity, high biocompatibility and chemical, thermal and mechanical robustness, among others. Additionally, these materials can be engineered to present an exquisite control in the drug release behavior placing stimuli-responsive pore-blockers or sensitive hybrid coats on their surface. Herein, the recent advances developed in the use of porous inorganic nanomedicines will be described in order to provide an overview of their huge potential in the look out of an efficient and safe therapy against this complex disease. Porous inorganic nanoparticles have been designed to be accumulated in tumoral tissues; once there to recognize the target cell and finally, to release their payload in a controlled manner.
Collapse
Affiliation(s)
- Jorge Parra-Nieto
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - María Amor García Del Cid
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Iñigo Aguirre de Cárcer
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Alejandro Baeza
- Dpto. Materiales y Producción Aeroespacial, ETSI Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| |
Collapse
|
47
|
A novel mitochondrial targeted hybrid peptide modified HPMA copolymers for breast cancer metastasis suppression. J Control Release 2020; 325:38-51. [PMID: 32598957 DOI: 10.1016/j.jconrel.2020.06.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/27/2022]
Abstract
Primary tumor metastasis remains to be a tough obstacle for clinical breast cancer treatment. Since evidences have shown that mitochondria play a crucial role in tumor metastasis, we designed a mitochondrial targeted drug delivery system (P-D-R8MTS) based on N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers to simultaneously inhibit breast cancer progression and metastasis. A novel mitochondrial targeted hybrid peptide R8MTS, which consists of a cell penetrating peptide octaarginine (R8) and a mitochondrial targeting sequence ALD5MTS, was used as targeting ligand and attached to doxorubicin (DOX) as model drug (DOX-R8MTS). After entering into the tumor cells, DOX-R8MTS was pH-responsibly released from HPMA copolymer backbone in acidic lysosome and efficiently targeted to mitochondria, resulting in enhanced reactive oxygen species (ROS) generation and apoptosis initiation. By destroying mitochondria, P-D-R8MTS not only inhibited cell proliferation but also suppressed migration and invasion of breast cancer 4T1 and MDA-MB-231 cells in vitro. Moreover, P-D-R8MTS exhibited superior inhibition of tumor growth and showed no apparent lung metastatic nodules on 4T1-bearing mice in vivo, which was partially via down-regulation of typical proteins associated with tumor metastasis and invasion: matrix metalloproteinases-2 (MMP-2), vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β). Collectively, our work provided a prospectively potential strategy for metastatic cancer treatment through mitochondrial targeted drug delivery.
Collapse
|
48
|
Tsubone TM, Zhang Z, Goyal R, Santacruz C, Martins WK, Kohn J, Baptista MS. Porphyrin-Loaded TyroSpheres for the Intracellular Delivery of Drugs and Photoinduced Oxidant Species. Mol Pharm 2020; 17:2911-2924. [DOI: 10.1021/acs.molpharmaceut.0c00338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tayana Mazin Tsubone
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-900, Brazil
| | - Zheng Zhang
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854-8009, United States
| | - Ritu Goyal
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854-8009, United States
| | - Carolina Santacruz
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-900, Brazil
| | | | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854-8009, United States
| | - Mauricio S. Baptista
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
49
|
Bulumulla C, Kularatne RN, Catchpole T, Takacs A, Christie A, Gilfoyle A, Nguyen TD, Stefan MC, Csaky KG. Investigating the Effect of Esterification on Retinal Pigment Epithelial Uptake Using Rhodamine B Derivatives. Transl Vis Sci Technol 2020; 9:18. [PMID: 32821515 PMCID: PMC7409196 DOI: 10.1167/tvst.9.6.18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/12/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose This study investigated the effects of esterification and increased lipophilicity on cellular penetration, accumulation and retention in ARPE-19-nic cells using ester functionalized rhodamine B dyes. Methods Rhodamine B was esterified to generate four dyes with increasing lipophilicity. Cellular uptake, retention and mitochondrial localization were investigated in vitro using ARPE-19-nic cells using direct intracellular and extracellular and mitochondrial fluorescence quantitation, confocal and high-resolution live cell imaging and co-localization with Mito-GFP. Results Cellular penetrance, mitochondrial accumulation, and retention of the esterified dyes were increased in ARPE-19-nic cells compared with the nonesterified parent dye by direct fluorescence quantitation. Imaging demonstrated intracellular accumulation was confined to mitochondria as confirmed by colocalization with Mito-GFP. Conclusions Esterification is an effective way to increase lipophilicity of a dye to improve cellular penetration of chemical entities. These observations may be key to improving retinal drug delivery for retinal pigment epithelium–based diseases. Translational Relevance Understanding the intracellular distribution of drugs into retinal pigment epithelium cells is a critical component for identifying potential therapies for retinal pigment epithelium–based diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mihaela C Stefan
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA.,Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Karl G Csaky
- Retina Foundation of the Southwest, Dallas, TX, USA
| |
Collapse
|
50
|
Schaumann EN, Tian B. Biological Interfaces, Modulation, and Sensing with Inorganic Nano-Bioelectronic Materials. SMALL METHODS 2020; 4:1900868. [PMID: 34295965 PMCID: PMC8294120 DOI: 10.1002/smtd.201900868] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/16/2020] [Indexed: 05/30/2023]
Abstract
The last several years have seen a large and increasing interest in scientific developments that combine methods and materials from nanotechnology with questions and applications in bioelectronics. This follows with a number of broader trends: the rapid increase in functionality for materials at the nanoscale; a growing recognition of the importance of electric fields in diverse physiological processes; and continuous improvements in technologies that are naturally complementary with bioelectronics, such as optogenetics. Here, a progress report is provided on several of the most exciting recent developments in this field. The three critical functions of biointerface formation, biological modulation, and biological sensing using newly developed nanoscale materials are considered.
Collapse
Affiliation(s)
- Erik N Schaumann
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Bozhi Tian
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|