1
|
Aja PM, Agu PC, Ogbu C, Alum EU, Fasogbon IV, Musyoka AM, Ngwueche W, Egwu CO, Tusubira D, Ross K. RNA research for drug discovery: Recent advances and critical insight. Gene 2025; 947:149342. [PMID: 39983851 DOI: 10.1016/j.gene.2025.149342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
The field of RNA research has experienced significant changes and is now at the forefront of contemporary drug development. This narrative overview explores the scientific developments and historical turning points in RNA research, emphasising the field's critical significance in the development of novel therapeutics. Important discoveries like antisense oligonucleotides (ASOs), mRNA therapies, and RNA interference (RNAi) have created novel treatment options that can be targeted, such as the ground-breaking mRNA vaccinations against COVID-19. Advances in high-throughput sequencing, single-cell RNA sequencing, and epitranscriptomics have further unravelled the complexity of RNA biology, shedding light on the intricacies of gene regulation and cellular diversity. The integration of computational tools and bioinformatics has propelled the identification of RNA-based biomarkers and the development of RNA therapeutics. Despite significant progress, challenges such as RNA stability, delivery, and off-target effects persist, necessitating continuous innovation and ethical considerations. This review provides a critical insight into the current state and prospects of RNA research, emphasising its transformative potential in drug discovery. By examining the interplay between technological advancements and therapeutic applications, we underscore the promising horizon for RNA-based interventions in treating a myriad of diseases, marking a new era in precision medicine.
Collapse
Affiliation(s)
- Patrick Maduabuchi Aja
- Biochemistry Department, Biomedical Sciences Faculty, Kampala International University, P.O. Box Ishaka, Bushenyi, Uganda; Biochemistry Department, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria.
| | - Peter Chinedu Agu
- Biochemistry Department, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria; Department of Biochemistry, Faculty of Science, Evangel University, Nigeria
| | - Celestine Ogbu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Federal University of Health Sciences, Otukpo, Nigeria
| | - Esther Ugo Alum
- Publications and Extension Department, Kampala International University, P. O. Box 20000, Uganda; Biochemistry Department, Faculty of Science, Ebonyi State University, P.M.B. 053 Abakaliki, Ebonyi State, Nigeria
| | - Ilemobayo Victor Fasogbon
- Biochemistry Department, Biomedical Sciences Faculty, Kampala International University, P.O. Box Ishaka, Bushenyi, Uganda
| | - Angela Mumbua Musyoka
- Biochemistry Department, Biomedical Sciences Faculty, Kampala International University, P.O. Box Ishaka, Bushenyi, Uganda
| | - Wisdom Ngwueche
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chinedu Ogbonia Egwu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Ebonyi State, Nigeria
| | - Deusdedit Tusubira
- Department of Biochemistry, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom; Institute for Health Research, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
2
|
Song M, Park HY, Kim HJ, Kwon S. Enhanced in vitro transfection efficiency of mRNA-loaded polyplexes into natural killer cells through osmoregulation. Biomater Sci 2025; 13:2082-2091. [PMID: 40035713 DOI: 10.1039/d4bm01661k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
High expression of externally injected in vitro transcribed (IVT) mRNA in natural killer (NK) cells is a prerequisite for NK cell-mediated cell therapy. To enhance the transfection efficacy of IVT mRNA-loaded polyplexes, we exposed NK cells to a hypertonic condition during transfection, which facilitated endo/exocytosis to maintain the isotonic state of the cells. The transfection efficacy of IVT mRNA was significantly enhanced after 24 h, which was mainly due to the facilitated cellular uptake and endosomal escape of the polyplexes. Interestingly, osmotic alterations in NK cells significantly affect the expression levels of endosome-escape-related genes in ion channels. Treatment with a mild hypertonic condition exhibited negligible toxicity to NK cells, without disturbing the integrity of the cellular membranes or the innate cytotoxic abilities of NK cells against cancer cells. These results demonstrate that the hypertonic treatment of NK cells enhances the transfection efficacy of IVT mRNA to produce genetically engineered NK cells.
Collapse
Affiliation(s)
- Myeongkwan Song
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| | - Ha Yeon Park
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
- Biohybrid Systems Research Center (BSRC), Inha University, Incheon, 22212, Republic of Korea
| | - Soonjo Kwon
- Department of Biological Engineering, Inha University, 100 Inharo Michuhol-gu, Incheon, 22212, Republic of Korea.
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, 22212, Republic of Korea
| |
Collapse
|
3
|
Fatima M, An T, Hong KJ. Revolutionizing mRNA Vaccines Through Innovative Formulation and Delivery Strategies. Biomolecules 2025; 15:359. [PMID: 40149895 PMCID: PMC11940278 DOI: 10.3390/biom15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Modernization of existing methods for the delivery of mRNA is vital in advanced therapeutics. Traditionally, mRNA has faced obstacles of poor stability due to enzymatic degradation. This work examines cutting-edge formulation and emerging techniques for safer delivery of mRNA vaccines. Inspired by the success of lipid nanoparticles (LNP) in delivering mRNA vaccines for COVID-19, a variety of other formulations have been developed to deliver mRNA vaccines for diverse infections. The meritorious features of nanoparticle-based mRNA delivery strategies, including LNP, polymeric, dendrimers, polysaccharide-based, peptide-derived, carbon and metal-based, DNA nanostructures, hybrid, and extracellular vesicles, have been examined. The impact of these delivery platforms on mRNA vaccine delivery efficacy, protection from enzymatic degradation, cellular uptake, controlled release, and immunogenicity has been discussed in detail. Even with significant developments, there are certain limitations to overcome, including toxicity concerns, limited information about immune pathways, the need to maintain a cold chain, and the necessity of optimizing administration methods. Continuous innovation is essential for improving delivery systems for mRNA vaccines. Future research directions have been proposed to address the existing challenges in mRNA delivery and to expand their potential prophylactic and therapeutic application.
Collapse
Affiliation(s)
- Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea;
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
4
|
Fan J, Xiao Z, Dong Y, Ye F, Qiu Y, Zhang C, Yin X, Li Y, Wang T. Nanocarrier-Mediated RNA Delivery Platform as a Frontier Strategy for Hepatic Disease Treatment: Challenges and Opportunities. Adv Healthc Mater 2025; 14:e2402933. [PMID: 39723654 DOI: 10.1002/adhm.202402933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Hepatic diseases cause serious public health problems worldwide, and there is an urgent need to develop effective therapeutic agents. In recent years, significant progress is made in RNA therapy, and RNA molecules, such as mRNAs, siRNAs, miRNAs, and RNA aptamers, are shown to provide significant advantages in the treatment of hepatic diseases. However, the drawbacks of RNAs, such as their poor biological stability, easy degradation by nucleases in vivo, low bioavailability, and low concentrations in target tissues, significantly limit the clinical application of RNA-based drugs. Therefore, exploring and developing effective nanoscale delivery platforms for RNA therapeutics are of immense value. This review focuses on the different types of hepatic diseases and RNA therapeutics, summarizing various nanoscale delivery platforms and their strengths and weaknesses. Finally, the current status and future prospects of nanoscale delivery systems for RNA therapy are discussed.
Collapse
Affiliation(s)
- Jinhui Fan
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Zhicheng Xiao
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Yafen Dong
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201200, China
| | - Fei Ye
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Yan Qiu
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201200, China
| | - Chuan Zhang
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Xiaolan Yin
- Cancer center, Shanghai 411 hospital, China RongTong Medical Healthcare Group Co. Ltd./411 Hospital, Shanghai University, Shanghai, 200081, China
| | - Yi Li
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Tingfang Wang
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
5
|
Puttasiddaiah R, Basavegowda N, Lakshmanagowda NK, Raghavendra VB, Sagar N, Sridhar K, Dikkala PK, Bhaswant M, Baek KH, Sharma M. Emerging Nanoparticle-Based Diagnostics and Therapeutics for Cancer: Innovations and Challenges. Pharmaceutics 2025; 17:70. [PMID: 39861718 PMCID: PMC11768644 DOI: 10.3390/pharmaceutics17010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Malignant growth is expected to surpass other significant causes of death as one of the top reasons for dismalness and mortality worldwide. According to a World Health Organization (WHO) study, this illness causes approximately between 9 and 10 million instances of deaths annually. Chemotherapy, radiation, and surgery are the three main methods of treating cancer. These methods seek to completely eradicate all cancer cells while having the fewest possible unintended impacts on healthy cell types. Owing to the lack of target selectivity, the majority of medications have substantial side effects. On the other hand, nanomaterials have transformed the identification, diagnosis, and management of cancer. Nanostructures with biomimetic properties have been grown as of late, fully intent on observing and treating the sickness. These nanostructures are expected to be consumed by growth in areas with profound disease. Furthermore, because of their extraordinary physicochemical properties, which incorporate nanoscale aspects, a more prominent surface region, explicit geometrical features, and the ability to embody different substances within or on their outside surfaces, nanostructures are remarkable nano-vehicles for conveying restorative specialists to their designated regions. This review discusses recent developments in nanostructured materials such as graphene, dendrimers, cell-penetrating peptide nanoparticles, nanoliposomes, lipid nanoparticles, magnetic nanoparticles, and nano-omics in the diagnosis and management of cancer.
Collapse
Affiliation(s)
- Rachitha Puttasiddaiah
- Teresian College Research Centre, Teresian College, Siddarthanagar, Mysore 570011, India
| | - Nagaraj Basavegowda
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | | | | | - Niju Sagar
- Teresian College Research Centre, Teresian College, Siddarthanagar, Mysore 570011, India
| | - Kandi Sridhar
- Department of Food Technology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore 641021, India
| | - Praveen Kumar Dikkala
- Department of Food Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522502, India
| | - Maharshi Bhaswant
- New Industry Creation Hatchery Center, Tohoku University, Sendai 9808579, Japan
- Center for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai 600119, India
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Minaxi Sharma
- Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, Ningbo 315000, China
| |
Collapse
|
6
|
Paredes-Hernández U, Aguilar-Peña LV, Isaac-Olivé K, Ocampo-García B, Contreras I, Estrada JA, Izquierdo G, Morales-Avila E, Aranda-Lara L. Enhancing photodynamic and radionuclide therapy by small interfering RNA (siRNA)-RAD51 transfection via self-emulsifying delivery systems (SNEDDS). Cytotherapy 2025; 27:66-77. [PMID: 39186024 DOI: 10.1016/j.jcyt.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AIMS Gene-silencing by small interfering RNA (siRNA) is an attractive therapy to regulate cancer death, tumor recurrence or metastasis. Because siRNAs are easily degraded, it is necessary to develop transport and delivery systems to achieve efficient tumor targeting. Self-nanoemulsifying systems (SNEDDS) have been successfully used for pDNA transport and delivery, so they may be useful for siRNA. The aim of this work is to introduce siRNA-RAD51 into a SNEDDS prepared with Phospholipon-90G, Labrafil-M1944-CS and Cremophor-RH40 and evaluate its efficacy in preventing homologous recombination of DNA double-strand breaks caused by photodynamic therapy (PDT) and ionizing radiation (IR). METHODS The siRNA-RAD51 was loaded into SNEDDS using chitosan. Transfection capacity was estimated by comparison with Lipofectamine-2000. RESULTS SNEDDS(siRNA-RAD51) induced gene silencing effect on the therapies evaluated by cell viability and clonogenic assays using T47D breast cancer cells. CONCLUSIONS SNEDDS(siRNA-RAD51) shown to be an effective siRNA-delivery system to decrease cellular resistance in PDT or IR.
Collapse
Affiliation(s)
- Ulises Paredes-Hernández
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Leslie V Aguilar-Peña
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Keila Isaac-Olivé
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Blanca Ocampo-García
- Departamento de Materiales Radiactivos, Instituto Nacional de Investigaciones Nucleares, Ocoyoacac, Estado de México, Mexico
| | - Irazú Contreras
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - José A Estrada
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Germán Izquierdo
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - Enrique Morales-Avila
- Laboratorio de Toxicología y Farmacia, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| | - Liliana Aranda-Lara
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico.
| |
Collapse
|
7
|
Yuan Y, Sun W, Xie J, Zhang Z, Luo J, Han X, Xiong Y, Yang Y, Zhang Y. RNA nanotherapeutics for hepatocellular carcinoma treatment. Theranostics 2025; 15:965-992. [PMID: 39776807 PMCID: PMC11700867 DOI: 10.7150/thno.102964] [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: 08/29/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality worldwide, particularly due to the limited effectiveness of current therapeutic options for advanced-stage disease. The efficacy of traditional treatments is often compromised by the intricate liver microenvironment and the inherent heterogeneity. RNA-based therapeutics offer a promising alternative, utilizing the innovative approach of targeting aberrant molecular pathways and modulating the tumor microenvironment. The integration of nanotechnology in this field, through the development of advanced nanocarrier delivery systems, especially lipid nanoparticles (LNPs), polymer nanoparticles (PNPs), and bioinspired vectors, enhances the precision and efficacy of RNA therapies. This review highlights the significant progress in RNA nanotherapeutics for HCC treatment, covering micro RNA (miRNA), small interfering RNA (siRNA), message RNA (mRNA), and small activating RNA (saRNA) mediated gene silencing, therapeutic protein restoration, gene activation, cancer vaccines, and concurrent therapy. It further comprehensively discusses the prevailing challenges within this therapeutic landscape and provides a forward-looking perspective on the potential of RNA nanotherapeutics to transform HCC treatment.
Collapse
Affiliation(s)
- Yihang Yuan
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
- Department of General Surgery Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School Nanjing University, Nanjing 210008, China
| | - Weijie Sun
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Jiaqi Xie
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Ziheng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Jing Luo
- Department of Urology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiangfei Han
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yongfu Xiong
- Department of Hepatobiliary Surgery, Academician (Expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong 637600, China
| | - Yang Yang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Yang Zhang
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Kanarskaya MA, Novikova SV, Lomzov AA. Hybrid RNA/DNA Concatemers and Self-Limited Complexes: Structure and Prospects for Therapeutic Applications. Molecules 2024; 29:5896. [PMID: 39769985 PMCID: PMC11677838 DOI: 10.3390/molecules29245896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The development of new convenient tools for the design of multicomponent nucleic acid (NA) complexes is one of the challenges in biomedicine and NA nanotechnology. In this paper, we analyzed the formation of hybrid RNA/DNA concatemers and self-limited complexes by a pair of oligonucleotides using UV melting, circular dichroism spectroscopy, and a gel shift assay. Effects of the size of the linker between duplex-forming segments of the oligonucleotides on complexes' shape and number of subunits were compared and systematized for RNA/DNA, DNA/DNA, and RNA/RNA assemblies. The data on complex types summarized here as heat maps offer a convenient tool for the design of NA constructs. General rules found for RNA/DNA, DNA/DNA, and RNA/RNA complexes allow not only designing complexes with desired structures but also purposefully transforming their geometry. The A-form of the double helix of the studied RNA/DNA complexes was confirmed by circular dichroism analysis. Moreover, we show for the first time efficient degradation of RNA in hybrid self-limited complexes by RNase H and imidazole. The results open up new prospects for the design of supramolecular complexes as tools for nanotechnology, nanomachinery, and biomedical applications.
Collapse
Affiliation(s)
- Maria A. Kanarskaya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia; (M.A.K.); (S.V.N.)
- Department of Physics, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Sofia V. Novikova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia; (M.A.K.); (S.V.N.)
- Department of Physics, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexander A. Lomzov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia; (M.A.K.); (S.V.N.)
- Department of Physics, Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
9
|
Yip T, Qi X, Yan H, Chang Y. Therapeutic applications of RNA nanostructures. RSC Adv 2024; 14:28807-28821. [PMID: 39263430 PMCID: PMC11387945 DOI: 10.1039/d4ra03823a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
RNA-based therapeutics have gained wide public interest in recent years. RNA is a versatile molecule that exists in many forms including mRNA, siRNA, miRNA, ribozymes, and other non-coding RNAs and is primarily applied for gene therapy. RNA is also used as a modular building block to construct RNA nanostructures. The programmable nature of RNA nanostructures enables the generation of simple, modulable, and multi-functional RNA-based therapeutics. Although the therapeutic application of RNA may be limited due to its structural instability, advances in RNA nanotechnology have improved the stability of RNA nanostructures for greater application. Various strategies have been developed to enhance the stability of RNA nanostructures enabling their application in vivo. In this review, we examine the therapeutic applications of RNA nanostructures. Non-immunogenic RNA nanostructures can be rationally designed with functional RNA molecules to modulate gene expression for gene therapy. On the other hand, nucleic acids can be sensed by cellular receptors to elicit an innate immune response, for which certain DNA and RNA motifs can function as adjuvants. Taking advantage of this adjuvant potential, RNA nanostructures can be used for immunotherapy and be designed for cancer vaccines. Thus, we examine the therapeutic application of immunogenic RNA nanostructures for cancer immunotherapy. RNA nanostructures represent promising platforms to design new nanodrugs, gene therapeutics, immunotherapeutic adjuvants, and cancer vaccines. Ongoing research in the field of RNA nanotechnology will continue to empower the development of RNA nanostructure-based therapeutics with high efficacy and limited toxicity.
Collapse
Affiliation(s)
- Theresa Yip
- School of Life Sciences, Arizona State University Tempe AZ 85281 USA
- Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University Tempe AZ 85281 USA
| | - Xiaodong Qi
- Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University Tempe AZ 85281 USA
- School of Molecular Sciences, Arizona State University Tempe AZ 85281 USA
| | - Hao Yan
- Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University Tempe AZ 85281 USA
- School of Molecular Sciences, Arizona State University Tempe AZ 85281 USA
| | - Yung Chang
- School of Life Sciences, Arizona State University Tempe AZ 85281 USA
- Biodesign Center for Molecular Design and Biomimetics, Biodesign Institute, Arizona State University Tempe AZ 85281 USA
| |
Collapse
|
10
|
Ghosh S, Bhaskar R, Mishra R, Arockia Babu M, Abomughaid MM, Jha NK, Sinha JK. Neurological insights into brain-targeted cancer therapy and bioinspired microrobots. Drug Discov Today 2024; 29:104105. [PMID: 39029869 DOI: 10.1016/j.drudis.2024.104105] [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: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
Cancer, a multifaceted and pernicious disease, continuously challenges medicine, requiring innovative treatments. Brain cancers pose unique and daunting challenges due to the intricacies of the central nervous system and the blood-brain barrier. In this era of precision medicine, the convergence of neurology, oncology, and cutting-edge technology has given birth to a promising avenue - targeted cancer therapy. Furthermore, bioinspired microrobots have emerged as an ingenious approach to drug delivery, enabling precision and control in cancer treatment. This Keynote review explores the intricate web of neurological insights into brain-targeted cancer therapy and the paradigm-shifting world of bioinspired microrobots. It serves as a critical and comprehensive overview of these evolving fields, aiming to underscore their integration and potential for revolutionary cancer treatments.
Collapse
Affiliation(s)
- Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Richa Mishra
- Department of Computer Science and Engineering, Parul University, Vadodara, Gujrat 391760, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
11
|
Zhang L, Lou W, Wang J. Advances in nucleic acid therapeutics: structures, delivery systems, and future perspectives in cancer treatment. Clin Exp Med 2024; 24:200. [PMID: 39196428 PMCID: PMC11358240 DOI: 10.1007/s10238-024-01463-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Cancer has emerged as a significant threat to human health. Nucleic acid therapeutics regulate the gene expression process by introducing exogenous nucleic acid fragments, offering new possibilities for tumor remission and even cure. Their mechanism of action and high specificity demonstrate great potential in cancer treatment. However, nucleic acid drugs face challenges such as low stability and limited ability to cross physiological barriers in vivo. To address these issues, various nucleic acid delivery vectors have been developed to enhance the stability and facilitate precise targeted delivery of nucleic acid drugs within the body. In this review article, we primarily introduce the structures and principles of nucleic acid drugs commonly used in cancer therapy, as well as their cellular uptake and intracellular transportation processes. We focus on the various vectors commonly employed in nucleic acid drug delivery, highlighting their research progress and applications in recent years. Furthermore, we propose potential trends and prospects of nucleic acid drugs and their carriers in the future.
Collapse
Affiliation(s)
- Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
12
|
Zhang Z, Li S, Wang H, Shan Y. The Effects of the Carrier and Ligand Spatial Conformation on RNA Nanodrug Cell Delivery. Anal Chem 2024; 96:13226-13233. [PMID: 39096242 DOI: 10.1021/acs.analchem.4c02270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Small interfering RNA (siRNA) highlights the immense therapeutic potential for cancer treatment. The major challenge in siRNA therapy is the effective RNA nanodrug delivery system, which is facilitated by the ligand and the carrier. In this study, we analyzed the binding specificity of linear RGD and circular RGD to αVβ3 integrins by mapping the morphology using super-resolution direct stochastic optical reconstruction microscopy. Meanwhile, the binding dynamics was investigated using single-molecule force spectroscopy. Then, the effects of the ligand and carrier on RNA nanodrug cell entry dynamic parameters were evaluated at the single particle level by the force tracing technique. Furthermore, the delivery efficiency of RNA nanodrugs was assessed using AFM-based nanoindentation at the single cell level. This report will provide valuable insights for rational design strategies aiming to achieve improved efficiency for nanodrug delivery systems.
Collapse
Affiliation(s)
- Zhuang Zhang
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Siying Li
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yuping Shan
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China
| |
Collapse
|
13
|
Gondaliya P, Sayyed AA, Yan IK, Driscoll J, Ziemer A, Patel T. Targeting PD-L1 in cholangiocarcinoma using nanovesicle-based immunotherapy. Mol Ther 2024; 32:2762-2777. [PMID: 38859589 PMCID: PMC11405167 DOI: 10.1016/j.ymthe.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/07/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
This study demonstrates the potential of using biological nanoparticles to deliver RNA therapeutics targeting programmed death-ligand 1 (PD-L1) as a treatment strategy for cholangiocarcinoma (CCA). RNA therapeutics offer prospects for intracellular immune modulation, but effective clinical translation requires appropriate delivery strategies. Milk-derived nanovesicles were decorated with epithelial cellular adhesion molecule (EpCAM) aptamers and used to deliver PD-L1 small interfering RNA (siRNA) or Cas9 ribonucleoproteins directly to CCA cells. In vitro, nanovesicle treatments reduced PD-L1 expression in CCA cells while increasing degranulation, cytokine release, and tumor cell cytotoxicity when tumor cells were co-cultured with T cells or natural killer cells. Similarly, immunomodulation was observed in multicellular spheroids that mimicked the tumor microenvironment. Combining targeted therapeutic vesicles loaded with siRNA to PD-L1 with gemcitabine effectively reduced tumor burden in an immunocompetent mouse CCA model compared with controls. This proof-of-concept study demonstrates the potential of engineered targeted nanovesicle platforms for delivering therapeutic RNA cargoes to tumors, as well as their use in generating effective targeted immunomodulatory therapies for difficult-to-treat cancers such as CCA.
Collapse
Affiliation(s)
- Piyush Gondaliya
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Adil Ali Sayyed
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Irene K Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Julia Driscoll
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Abbye Ziemer
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
14
|
Yan W, Cao Y, Yin Q, Li Y. Biomimetic Nucleic Acid Drug Delivery Systems for Relieving Tumor Immunosuppressive Microenvironment. Pharmaceutics 2024; 16:1028. [PMID: 39204373 PMCID: PMC11360391 DOI: 10.3390/pharmaceutics16081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Immunotherapy combats tumors by enhancing the body's immune surveillance and clearance of tumor cells. Various nucleic acid drugs can be used in immunotherapy, such as DNA expressing cytokines, mRNA tumor vaccines, small interfering RNAs (siRNA) knocking down immunosuppressive molecules, and oligonucleotides that can be used as immune adjuvants. Nucleic acid drugs, which are prone to nuclease degradation in the circulation and find it difficult to enter the target cells, typically necessitate developing appropriate vectors for effective in vivo delivery. Biomimetic drug delivery systems, derived from viruses, bacteria, and cells, can protect the cargos from degradation and clearance, and deliver them to the target cells to ensure safety. Moreover, they can activate the immune system through their endogenous activities and active components, thereby improving the efficacy of antitumor immunotherapeutic nucleic acid drugs. In this review, biomimetic nucleic acid delivery systems for relieving a tumor immunosuppressive microenvironment are introduced. Their immune activation mechanisms, including upregulating the proinflammatory cytokines, serving as tumor vaccines, inhibiting immune checkpoints, and modulating intratumoral immune cells, are elaborated. The advantages and disadvantages, as well as possible directions for their clinical translation, are summarized at last.
Collapse
Affiliation(s)
- Wenlu Yan
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Cao
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (W.Y.); (Y.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
15
|
Bouazzaoui A, Abdellatif AA. Vaccine delivery systems and administration routes: Advanced biotechnological techniques to improve the immunization efficacy. Vaccine X 2024; 19:100500. [PMID: 38873639 PMCID: PMC11170481 DOI: 10.1016/j.jvacx.2024.100500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/21/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
Collapse
Affiliation(s)
- Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
- Science and Technology Unit, Umm Al Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Ahmed A.H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, 51452 Qassim, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, 71524 Assiut, Egypt
| |
Collapse
|
16
|
Fazel F, Doost JS, Raj S, Boodhoo N, Karimi K, Sharif S. The mRNA vaccine platform for veterinary species. Vet Immunol Immunopathol 2024; 274:110803. [PMID: 39003921 DOI: 10.1016/j.vetimm.2024.110803] [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: 05/02/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Vaccination has proven to be an effective means of controlling pathogens in animals. Since the introduction of veterinary vaccines in the 19th century, several generations of vaccines have been introduced. These vaccines have had a positive impact on global animal health and production. Despite, the success of veterinary vaccines, there are still some pathogens for which there are no effective vaccines available, such as African swine fever. Further, animal health is under the constant threat of emerging and re-emerging pathogens, some of which are zoonotic and can pose a threat to human health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has highlighted the need for new vaccine platforms that are safe and efficacious, but also importantly, are adaptable and can be modified rapidly to match the circulating pathogens. mRNA vaccines have been shown to be an effective vaccine platform against various viral and bacterial pathogens. This review will cover some of the recent advances in the field of mRNA vaccines for veterinary species. Moreover, various mRNA vaccines and their delivery methods, as well as their reported efficacy, will be discussed. Current limitations and future prospects of this vaccine platform in veterinary medicine will also be discussed.
Collapse
Affiliation(s)
- Fatemeh Fazel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Sugandha Raj
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
17
|
Choi Y, Seok SH, Yoon HY, Ryu JH, Kwon IC. Advancing cancer immunotherapy through siRNA-based gene silencing for immune checkpoint blockade. Adv Drug Deliv Rev 2024; 209:115306. [PMID: 38626859 DOI: 10.1016/j.addr.2024.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024]
Abstract
Cancer immunotherapy represents a revolutionary strategy, leveraging the patient's immune system to inhibit tumor growth and alleviate the immunosuppressive effects of the tumor microenvironment (TME). The recent emergence of immune checkpoint blockade (ICB) therapies, particularly following the first approval of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors like ipilimumab, has led to significant growth in cancer immunotherapy. The extensive explorations on diverse immune checkpoint antibodies have broadened the therapeutic scope for various malignancies. However, the clinical response to these antibody-based ICB therapies remains limited, with less than 15% responsiveness and notable adverse effects in some patients. This review introduces the emerging strategies to overcome current limitations of antibody-based ICB therapies, mainly focusing on the development of small interfering ribonucleic acid (siRNA)-based ICB therapies and innovative delivery systems. We firstly highlight the diverse target immune checkpoint genes for siRNA-based ICB therapies, incorporating silencing of multiple genes to boost anti-tumor immune responses. Subsequently, we discuss improvements in siRNA delivery systems, enhanced by various nanocarriers, aimed at overcoming siRNA's clinical challenges such as vulnerability to enzymatic degradation, inadequate pharmacokinetics, and possible unintended target interactions. Additionally, the review presents various combination therapies that integrate chemotherapy, phototherapy, stimulatory checkpoints, ICB antibodies, and cancer vaccines. The important point is that when used in combination with siRNA-based ICB therapy, the synergistic effect of traditional therapies is strengthened, improving host immune surveillance and therapeutic outcomes. Conclusively, we discuss the insights into innovative and effective cancer immunotherapeutic strategies based on RNA interference (RNAi) technology utilizing siRNA and nanocarriers as a novel approach in ICB cancer immunotherapy.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
18
|
Liu J, Du C, Chen H, Huang W, Lei Y. Nano-Micron Combined Hydrogel Microspheres: Novel Answer for Minimal Invasive Biomedical Applications. Macromol Rapid Commun 2024; 45:e2300670. [PMID: 38400695 DOI: 10.1002/marc.202300670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/05/2024] [Indexed: 02/25/2024]
Abstract
Hydrogels, key in biomedical research for their hydrophilicity and versatility, have evolved with hydrogel microspheres (HMs) of micron-scale dimensions, enhancing their role in minimally invasive therapeutic delivery, tissue repair, and regeneration. The recent emergence of nanomaterials has ushered in a revolutionary transformation in the biomedical field, which demonstrates tremendous potential in targeted therapies, biological imaging, and disease diagnostics. Consequently, the integration of advanced nanotechnology promises to trigger a new revolution in the realm of hydrogels. HMs loaded with nanomaterials combine the advantages of both hydrogels and nanomaterials, which enables multifaceted functionalities such as efficient drug delivery, sustained release, targeted therapy, biological lubrication, biochemical detection, medical imaging, biosensing monitoring, and micro-robotics. Here, this review comprehensively expounds upon commonly used nanomaterials and their classifications. Then, it provides comprehensive insights into the raw materials and preparation methods of HMs. Besides, the common strategies employed to achieve nano-micron combinations are summarized, and the latest applications of these advanced nano-micron combined HMs in the biomedical field are elucidated. Finally, valuable insights into the future design and development of nano-micron combined HMs are provided.
Collapse
Affiliation(s)
- Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
19
|
Abdul Manap AS, Wisham AA, Wong FW, Ahmad Najmi HR, Ng ZF, Diba RS. Mapping the function of MicroRNAs as a critical regulator of tumor-immune cell communication in breast cancer and potential treatment strategies. Front Cell Dev Biol 2024; 12:1390704. [PMID: 38726321 PMCID: PMC11079208 DOI: 10.3389/fcell.2024.1390704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Among women, breast cancer ranks as the most prevalent form of cancer, and the presence of metastases significantly reduces prognosis and diminishes overall survival rates. Gaining insights into the biological mechanisms governing the conversion of cancer cells, their subsequent spread to other areas of the body, and the immune system's monitoring of tumor growth will contribute to the advancement of more efficient and targeted therapies. MicroRNAs (miRNAs) play a critical role in the interaction between tumor cells and immune cells, facilitating tumor cells' evasion of the immune system and promoting cancer progression. Additionally, miRNAs also influence metastasis formation, including the establishment of metastatic sites and the transformation of tumor cells into migratory phenotypes. Specifically, dysregulated expression of these genes has been associated with abnormal expression of oncogenes and tumor suppressor genes, thereby facilitating tumor development. This study aims to provide a concise overview of the significance and function of miRNAs in breast cancer, focusing on their involvement as tumor suppressors in the antitumor immune response and as oncogenes in metastasis formation. Furthermore, miRNAs hold tremendous potential as targets for gene therapy due to their ability to modulate specific pathways that can either promote or suppress carcinogenesis. This perspective highlights the latest strategies developed for miRNA-based therapies.
Collapse
Affiliation(s)
- Aimi Syamima Abdul Manap
- Department of Biomedical Science, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | | | - Fei Wen Wong
- Faculty of Biosciences, MAHSA University, Kuala Langat, Selangor, Malaysia
| | | | - Zhi Fei Ng
- Faculty of Biosciences, MAHSA University, Kuala Langat, Selangor, Malaysia
| | | |
Collapse
|
20
|
Huang HYR, Badar S, Said M, Shah S, Bharadwaj HR, Ramamoorthy K, Alrawashdeh MM, Haroon F, Basit J, Saeed S, Aji N, Tse G, Roy P, Bardhan M. The advent of RNA-based therapeutics for metabolic syndrome and associated conditions: a comprehensive review of the literature. Mol Biol Rep 2024; 51:493. [PMID: 38580818 DOI: 10.1007/s11033-024-09457-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: 01/19/2024] [Accepted: 03/18/2024] [Indexed: 04/07/2024]
Abstract
Metabolic syndrome (MetS) is a prevalent and intricate health condition affecting a significant global population, characterized by a cluster of metabolic and hormonal disorders disrupting lipid and glucose metabolism pathways. Clinical manifestations encompass obesity, dyslipidemia, insulin resistance, and hypertension, contributing to heightened risks of diabetes and cardiovascular diseases. Existing medications often fall short in addressing the syndrome's multifaceted nature, leading to suboptimal treatment outcomes and potential long-term health risks. This scenario underscores the pressing need for innovative therapeutic approaches in MetS management. RNA-based treatments, employing small interfering RNAs (siRNAs), microRNAs (miRNAs), and antisense oligonucleotides (ASOs), emerge as promising strategies to target underlying biological abnormalities. However, a summary of research available on the role of RNA-based therapeutics in MetS and related co-morbidities is limited. Murine models and human studies have been separately interrogated to determine whether there have been recent advancements in RNA-based therapeutics to offer a comprehensive understanding of treatment available for MetS. In a narrative fashion, we searched for relevant articles pertaining to MetS co-morbidities such as cardiovascular disease, fatty liver disease, dementia, colorectal cancer, and endocrine abnormalities. We emphasize the urgency of exploring novel therapeutic avenues to address the intricate pathophysiology of MetS and underscore the potential of RNA-based treatments, coupled with advanced delivery systems, as a transformative approach for achieving more comprehensive and efficacious outcomes in MetS patients.
Collapse
Affiliation(s)
- Helen Ye Rim Huang
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sarah Badar
- Department of Biomedical Science, The University of the West Scotland, Paisley, Scotland
| | - Mohammad Said
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Siddiqah Shah
- Faculty of Medicine and Health Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Krishna Ramamoorthy
- Department of Biochemistry and Microbiology, Rutgers University-New Brunswick, Brunswick, NJ, USA
| | | | | | - Jawad Basit
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Sajeel Saeed
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Narjiss Aji
- Faculty of Medicine and Health, McGill University, Montreal, QC, Canada
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Priyanka Roy
- Directorate of Factories, Department of Labour, Government of West Bengal, Kolkata, India
| | - Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA.
| |
Collapse
|
21
|
Kim T, Han HS, Yang K, Kim YM, Nam K, Park KH, Choi SY, Park HW, Choi KY, Roh YH. Nanoengineered Polymeric RNA Nanoparticles for Controlled Biodistribution and Efficient Targeted Cancer Therapy. ACS NANO 2024; 18:7972-7988. [PMID: 38445578 DOI: 10.1021/acsnano.3c10732] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
RNA nanotechnology, including rolling circle transcription (RCT), has gained increasing interest as a fascinating siRNA delivery nanoplatform for biostable and tumor-targetable RNA-based therapies. However, due to the lack of fine-tuning technologies for RNA nanostructures, the relationship between physicochemical properties and siRNA efficacy of polymeric siRNA nanoparticles (PRNs) with different sizes has not yet been fully elucidated. Herein, we scrutinized the effects of size/surface chemistry-tuned PRNs on the biological and physiological interactions with tumors. PRNs with adjusted size and surface properties were prepared using sequential engineering processes: RCT, condensation, and nanolayer deposition of functional biopolymers. Through the RCT process, nanoparticles of three sizes with a diameter of 50-200 nm were fabricated and terminated with three types of biopolymers: poly-l-lysine (PLL), poly-l-glutamate (PLG), and hyaluronic acid (HA) for different surface properties. Among the PRNs, HA-layered nanoparticles with a diameter of ∼200 nm exhibited the most effective systemic delivery, resulting in superior anticancer effects in an orthotopic breast tumor model due to the CD44 receptor targeting and optimized nanosized structure. Depending on the type of PRNs, the in vivo siRNA delivery with protein expression inhibition differed by up to approximately 20-fold. These findings indicate that the types of layered biopolymers and the PRNs size mediate efficient polymeric siRNA delivery to the targeted tumors, resulting in high RNAi-induced therapeutic efficacy. This RNA-nanotechnology-based size/surface editing can overcome the limitations of siRNA therapeutics and represents a potent built-in module method to design RNA therapeutics tailored for targeted cancer therapy.
Collapse
Affiliation(s)
- Taehyung Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hwa Seung Han
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, 7 Jukjeon-gil, Gangneung-si, Gangwon 25457, Republic of Korea
| | - Kyungjik Yang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Young Min Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Keonwook Nam
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kyung Hoon Park
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Seung Young Choi
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, 7 Jukjeon-gil, Gangneung-si, Gangwon 25457, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ki Young Choi
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, 7 Jukjeon-gil, Gangneung-si, Gangwon 25457, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
22
|
Li X, Jin K, Cheng TC, Liao YC, Lee WJ, Bhullar AS, Chen LC, Rychahou P, Phelps MA, Ho YS, Guo P. RNA four-way junction (4WJ) for spontaneous cancer-targeting, effective tumor-regression, metastasis suppression, fast renal excretion and undetectable toxicity. Biomaterials 2024; 305:122432. [PMID: 38176263 PMCID: PMC10994150 DOI: 10.1016/j.biomaterials.2023.122432] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/14/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024]
Abstract
The field of RNA therapeutics has been emerging as the third milestone in pharmaceutical drug development. RNA nanoparticles have displayed motile and deformable properties to allow for high tumor accumulation with undetectable healthy organ accumulation. Therefore, RNA nanoparticles have the potential to serve as potent drug delivery vehicles with strong anti-cancer responses. Herein, we report the physicochemical basis for the rational design of a branched RNA four-way junction (4WJ) nanoparticle that results in advantageous high-thermostability and -drug payload for cancer therapy, including metastatic tumors in the lung. The 4WJ nanostructure displayed versatility through functionalization with an anti-cancer chemical drug, SN38, for the treatment of two different cancer models including colorectal cancer xenograft and orthotopic lung metastases of colon cancer. The resulting 4WJ RNA drug complex spontaneously targeted cancers effectively for cancer inhibition with and without ligands. The 4WJ displayed fast renal excretion, rapid body clearance, and little organ accumulation with undetectable toxicity and immunogenicity. The safety parameters were documented by organ histology, blood biochemistry, and pathological analysis. The highly efficient cancer inhibition, undetectable drug toxicity, and favorable Chemical, Manufacturing, and Control (CMC) production of RNA nanoparticles document a candidate with high potential for translation in cancer therapy.
Collapse
Affiliation(s)
- Xin Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Kai Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan
| | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110031, Taiwan
| | - Wen-Jui Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Abhjeet S Bhullar
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Li-Ching Chen
- Department of Biological Science & Technology, China Medical University, Taichung, 406040, Taiwan
| | - Piotr Rychahou
- Markey Cancer Center, Department of Surgery, University of Kentucky, Lexington, KY, 40536, USA
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Yuan Soon Ho
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, 406040, Taiwan.
| | - Peixuan Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA; Center for RNA Nanotechnology and Nanomedicine, The Ohio State University, Columbus, OH, 43210, USA; James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
23
|
Khairnar P, Kolipaka T, Pandey G, Phatale V, Shah S, Srinivasarao DA, Saraf S, Srivastava S. Nanosponge-mediated oligonucleotide delivery: A cutting-edge technology towards cancer management. J Drug Deliv Sci Technol 2024; 91:105226. [DOI: 10.1016/j.jddst.2023.105226] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
24
|
Mohanty P, Panda P, Acharya RK, Pande B, Bhaskar LVKS, Verma HK. Emerging perspectives on RNA virus-mediated infections: from pathogenesis to therapeutic interventions. World J Virol 2023; 12:242-255. [PMID: 38187500 PMCID: PMC10768389 DOI: 10.5501/wjv.v12.i5.242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/25/2023] Open
Abstract
RNA viruses continue to pose significant threats to global public health, necessitating a profound understanding of their pathogenic mechanisms and the development of effective therapeutic interventions. This manuscript provides a comprehensive overview of emerging perspectives on RNA virus-mediated infections, spanning from the intricate intricacies of viral pathogenesis to the forefront of innovative therapeutic strategies. A critical exploration of antiviral drugs sets the stage, highlighting the diverse classes of compounds that target various stages of the viral life cycle, underscoring the ongoing efforts to combat viral infections. Central to this discussion is the exploration of RNA-based therapeutics, with a spotlight on messenger RNA (mRNA)-based approaches that have revolutionized the landscape of antiviral interventions. Furthermore, the manuscript delves into the intricate world of delivery systems, exploring inno-vative technologies designed to enhance the efficiency and safety of mRNA vaccines. By analyzing the challenges and advancements in delivery mechanisms, this review offers a roadmap for future research and development in this critical area. Beyond conventional infectious diseases, the document explores the expanding applications of mRNA vaccines, including their promising roles in cancer immunotherapy and personalized medicine approaches. This manuscript serves as a valuable resource for researchers, clinicians, and policymakers alike, offering a nuanced perspective on RNA virus pathogenesis and the cutting-edge therapeutic interventions. By synthesizing the latest advancements and challenges, this review contributes significantly to the ongoing discourse in the field, driving the development of novel strategies to combat RNA virus-mediated infections effectively.
Collapse
Affiliation(s)
- Pratik Mohanty
- Department of Bioscience and Bioengineering, Indian Institute of Technology, Guwahati 781039, Assam, India
| | - Poojarani Panda
- Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Rakesh Kumar Acharya
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Bilaspur 495009, Chhattisgarh, India
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Science, Raipur 492001, chhattisgarh, India
| | - LVKS Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Bilaspur 495009, Chhattisgarh, India
| | - Henu Kumar Verma
- Lung Health and Immunity, Helmholtz Zentrum Munich, Munich 85764, Bayren, Germany
| |
Collapse
|
25
|
Xu Y, Zheng Y, Ding X, Wang C, Hua B, Hong S, Huang X, Lin J, Zhang P, Chen W. PEGylated pH-responsive peptide-mRNA nano self-assemblies enhance the pulmonary delivery efficiency and safety of aerosolized mRNA. Drug Deliv 2023; 30:2219870. [PMID: 37336779 DOI: 10.1080/10717544.2023.2219870] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 06/21/2023] Open
Abstract
Inhalable messenger RNA (mRNA) has demonstrated great potential in therapy and vaccine development to confront various lung diseases. However, few gene vectors could overcome the airway mucus and intracellular barriers for successful pulmonary mRNA delivery. Apart from the low pulmonary gene delivery efficiency, nonnegligible toxicity is another common problem that impedes the clinical application of many non-viral vectors. PEGylated cationic peptide-based mRNA delivery vector is a prospective approach to enhance the pulmonary delivery efficacy and safety of aerosolized mRNA by oral inhalation administration. In this study, different lengths of hydrophilic PEG chains were covalently linked to an amphiphilic, water-soluble pH-responsive peptide, and the peptide/mRNA nano self-assemblies were characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM). The in vitro mRNA binding and release, cellular uptake, transfection, and cytotoxicity were studied, and finally, a proper PEGylated peptide with enhanced pulmonary mRNA delivery efficiency and improved safety in mice was identified. These results showed that a proper N-terminus PEGylation strategy using 12-monomer linear monodisperse PEG could significantly improve the mRNA transfection efficiency and biocompatibility of the non-PEGylated cationic peptide carrier, while a longer PEG chain modification adversely decreased the cellular uptake and transfection on A549 and HepG2 cells, emphasizing the importance of a proper PEG chain length selection. Moreover, the optimized PEGylated peptide showed a significantly enhanced mRNA pulmonary delivery efficiency and ameliorated safety profiles over the non-PEGylated peptide and LipofectamineTM 2000 in mice. Our results reveal that the PEGylated peptide could be a promising mRNA delivery vector candidate for inhaled mRNA vaccines and therapeutic applications for the prevention and treatment of different respiratory diseases in the future.
Collapse
Affiliation(s)
- Yingying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China
| | - Yijing Zheng
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xuqiu Ding
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Chengyan Wang
- Institute of Laboratory Animal Center, Fujian Medical University, Fuzhou, China
| | - Bin Hua
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Shilian Hong
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaoman Huang
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jiali Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Peng Zhang
- Department of Pharmacy, The Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen, China
| | - Wei Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
26
|
Khalifah BA, Alghamdi SA, Alhasan AH. Unleashing the potential of catalytic RNAs to combat mis-spliced transcripts. Front Bioeng Biotechnol 2023; 11:1244377. [PMID: 38047291 PMCID: PMC10690607 DOI: 10.3389/fbioe.2023.1244377] [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: 06/22/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
Human transcriptome can undergo RNA mis-splicing due to spliceopathies contributing to the increasing number of genetic diseases including muscular dystrophy (MD), Alzheimer disease (AD), Huntington disease (HD), myelodysplastic syndromes (MDS). Intron retention (IR) is a major inducer of spliceopathies where two or more introns remain in the final mature mRNA and account for many intronic expansion diseases. Potential removal of such introns for therapeutic purposes can be feasible when utilizing bioinformatics, catalytic RNAs, and nano-drug delivery systems. Overcoming delivery challenges of catalytic RNAs was discussed in this review as a future perspective highlighting the significance of utilizing synthetic biology in addition to high throughput deep sequencing and computational approaches for the treatment of mis-spliced transcripts.
Collapse
Affiliation(s)
- Bashayer A. Khalifah
- Institute for Bioengineering, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
- Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Ali H. Alhasan
- Institute for Bioengineering, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
- College of Science and General Studies, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Cheng L, Yu J, Hao T, Wang W, Wei M, Li G. Advances in Polymeric Micelles: Responsive and Targeting Approaches for Cancer Immunotherapy in the Tumor Microenvironment. Pharmaceutics 2023; 15:2622. [PMID: 38004600 PMCID: PMC10675796 DOI: 10.3390/pharmaceutics15112622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
In recent years, to treat a diverse array of cancer forms, considerable advancements have been achieved in the field of cancer immunotherapies. However, these therapies encounter multiple challenges in clinical practice, such as high immune-mediated toxicity, insufficient accumulation in cancer tissues, and undesired off-target reactions. To tackle these limitations and enhance bioavailability, polymer micelles present potential solutions by enabling precise drug delivery to the target site, thus amplifying the effectiveness of immunotherapy. This review article offers an extensive survey of recent progress in cancer immunotherapy strategies utilizing micelles. These strategies include responsive and remodeling approaches to the tumor microenvironment (TME), modulation of immunosuppressive cells within the TME, enhancement of immune checkpoint inhibitors, utilization of cancer vaccine platforms, modulation of antigen presentation, manipulation of engineered T cells, and targeting other components of the TME. Subsequently, we delve into the present state and constraints linked to the clinical utilization of polymeric micelles. Collectively, polymer micelles demonstrate excellent prospects in tumor immunotherapy by effectively addressing the challenges associated with conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Lichun Cheng
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Jiankun Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Tangna Hao
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Wenshuo Wang
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Guiru Li
- Department of Pharmacy, The Second Hospital of Dalian Medical University, Dalian 116027, China; (L.C.); (T.H.); (W.W.)
| |
Collapse
|
28
|
Lotfi M, Morshedi Rad D, Mashhadi SS, Ashouri A, Mojarrad M, Mozaffari-Jovin S, Farrokhi S, Hashemi M, Lotfi M, Ebrahimi Warkiani M, Abbaszadegan MR. Recent Advances in CRISPR/Cas9 Delivery Approaches for Therapeutic Gene Editing of Stem Cells. Stem Cell Rev Rep 2023; 19:2576-2596. [PMID: 37723364 PMCID: PMC10661828 DOI: 10.1007/s12015-023-10585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 09/20/2023]
Abstract
Rapid advancement in genome editing technologies has provided new promises for treating neoplasia, cardiovascular, neurodegenerative, and monogenic disorders. Recently, the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system has emerged as a powerful gene editing tool offering advantages, including high editing efficiency and low cost over the conventional approaches. Human pluripotent stem cells (hPSCs), with their great proliferation and differentiation potential into different cell types, have been exploited in stem cell-based therapy. The potential of hPSCs and the capabilities of CRISPR/Cas9 genome editing has been paradigm-shifting in medical genetics for over two decades. Since hPSCs are categorized as hard-to-transfect cells, there is a critical demand to develop an appropriate and effective approach for CRISPR/Cas9 delivery into these cells. This review focuses on various strategies for CRISPR/Cas9 delivery in stem cells.
Collapse
Affiliation(s)
- Malihe Lotfi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dorsa Morshedi Rad
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Samaneh Sharif Mashhadi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Ashouri
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Mozaffari-Jovin
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Farrokhi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Lotfi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia.
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, Australia.
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Xie X, Yue T, Gu W, Cheng W, He L, Ren W, Li F, Piao JG. Recent Advances in Mesoporous Silica Nanoparticles Delivering siRNA for Cancer Treatment. Pharmaceutics 2023; 15:2483. [PMID: 37896243 PMCID: PMC10609930 DOI: 10.3390/pharmaceutics15102483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Silencing genes using small interfering (si) RNA is a promising strategy for treating cancer. However, the curative effect of siRNA is severely constrained by low serum stability and cell membrane permeability. Therefore, improving the delivery efficiency of siRNA for cancer treatment is a research hotspot. Recently, mesoporous silica nanoparticles (MSNs) have emerged as bright delivery vehicles for nucleic acid drugs. A comprehensive understanding of the design of MSN-based vectors is crucial for the application of siRNA in cancer therapy. We discuss several surface-functionalized MSNs' advancements as effective siRNA delivery vehicles in this paper. The advantages of using MSNs for siRNA loading regarding considerations of different shapes, various options for surface functionalization, and customizable pore sizes are highlighted. We discuss the recent investigations into strategies that efficiently improve cellular uptake, facilitate endosomal escape, and promote cargo dissociation from the MSNs for enhanced intracellular siRNA delivery. Also, particular attention was paid to the exciting progress made by combining RNAi with other therapies to improve cancer therapeutic outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fanzhu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (X.X.); (T.Y.); (W.G.); (W.C.); (L.H.); (W.R.)
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (X.X.); (T.Y.); (W.G.); (W.C.); (L.H.); (W.R.)
| |
Collapse
|
30
|
Chehelgerdi M, Chehelgerdi M, Allela OQB, Pecho RDC, Jayasankar N, Rao DP, Thamaraikani T, Vasanthan M, Viktor P, Lakshmaiya N, Saadh MJ, Amajd A, Abo-Zaid MA, Castillo-Acobo RY, Ismail AH, Amin AH, Akhavan-Sigari R. Progressing nanotechnology to improve targeted cancer treatment: overcoming hurdles in its clinical implementation. Mol Cancer 2023; 22:169. [PMID: 37814270 PMCID: PMC10561438 DOI: 10.1186/s12943-023-01865-0] [Citation(s) in RCA: 249] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023] Open
Abstract
The use of nanotechnology has the potential to revolutionize the detection and treatment of cancer. Developments in protein engineering and materials science have led to the emergence of new nanoscale targeting techniques, which offer renewed hope for cancer patients. While several nanocarriers for medicinal purposes have been approved for human trials, only a few have been authorized for clinical use in targeting cancer cells. In this review, we analyze some of the authorized formulations and discuss the challenges of translating findings from the lab to the clinic. This study highlights the various nanocarriers and compounds that can be used for selective tumor targeting and the inherent difficulties in cancer therapy. Nanotechnology provides a promising platform for improving cancer detection and treatment in the future, but further research is needed to overcome the current limitations in clinical translation.
Collapse
Affiliation(s)
- Mohammad Chehelgerdi
- Novin Genome (NG) Institute, Research and Development Center for Biotechnology, Shahrekord, Chaharmahal and Bakhtiari, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Chaharmahal and Bakhtiari, Iran.
| | - Matin Chehelgerdi
- Novin Genome (NG) Institute, Research and Development Center for Biotechnology, Shahrekord, Chaharmahal and Bakhtiari, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Chaharmahal and Bakhtiari, Iran
| | | | | | - Narayanan Jayasankar
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Devendra Pratap Rao
- Department of Chemistry, Coordination Chemistry Laboratory, Dayanand Anglo-Vedic (PG) College, Kanpur-208001, U.P, India
| | - Tamilanban Thamaraikani
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Manimaran Vasanthan
- Department of Pharmaceutics, SRM Institute of Science and Technology, SRM College Of Pharmacy, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Patrik Viktor
- Keleti Károly Faculty of Business and Management, Óbuda University, Tavaszmező U. 15-17, 1084, Budapest, Hungary
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Ayesha Amajd
- Faculty of Organization and Management, Silesian University of Technology, 44-100, Gliwice, Poland
- Department of Mechanical Engineering, CEMMPRE, University of Coimbra, Polo II, 3030-788, Coimbra, Portugal
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, 82817, Jazan, Saudi Arabia
| | | | - Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, 82817, Jazan, Saudi Arabia
| | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warsaw, Poland
| |
Collapse
|
31
|
Liu J, Jiang J, Deng C, Huang X, Huang S, Liu Z, Yang J, Mo J, Chen HJ, Wang J, Xie X. Nanochannel Electro-Injection as a Versatile Platform for Efficient RNA/DNA Programming on Dendritic Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303088. [PMID: 37381646 DOI: 10.1002/smll.202303088] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/03/2023] [Indexed: 06/30/2023]
Abstract
The utilization of dendritic cell (DC) vaccines is a promising approach in cancer immunotherapy, and the modification of DCs for the expression of tumor-associated antigens is critical for successful cancer immunotherapy. A safe and efficient method for delivering DNA/RNA into DCs without inducing maturation is beneficial to achieve successful DC transformation for cell vaccine applications, yet remains challenging. This work presents a nanochannel electro-injection (NEI) system for the safe and efficient delivery of a variety of nucleic acid molecules into DCs. The device is based on track-etched nanochannel membrane as key components, where the nano-sized channels localize the electric field on the cell membrane, enabling lower voltage (<30 V) for cell electroporation. The pulse conditions of NEI are examined so that the transfection efficiency (>70%) and biosafety (viability >85%) on delivering fluorescent dyes, plasmid DNA, messenger RNA, and circular RNA (circRNA) into DC2.4 are optimized. Primary mouse bone marrow DC can also be transfected with circRNA with 68.3% efficiency, but without remarkably affecting cellular viability or inducing DC maturation. These results suggest that NEI can be a safe and efficient transfection platform for in vitro transformation of DCs and possesses a promising potential for developing DC vaccines against cancer.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Juan Jiang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Caiguanxi Deng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Shuang Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Zhengjie Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Jiang Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Jingshan Mo
- School of Electronic and Information Engineering, Guangdong Ocean University, Zhanjiang, 524088, P. R. China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Ji Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Xi Xie
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
32
|
Lv T, Meng Y, Liu Y, Han Y, Xin H, Peng X, Huang J. RNA nanotechnology: A new chapter in targeted therapy. Colloids Surf B Biointerfaces 2023; 230:113533. [PMID: 37713955 DOI: 10.1016/j.colsurfb.2023.113533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/14/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
Nanoparticles have been widely studied in the fields of biotechnology, pharmacy, optics and medicine and have broad application prospects. Numerous studies have shown significant interest in utilizing nanoparticles for chemically coating or coupling drugs, aiming to address the challenges of drug delivery, including degradability and uncertainty. Furthermore, the utilization of lipid nanoparticles loaded with novel coronavirus antigen mRNA to control the COVID-19 pandemic has led to a notable surge in research on nanoparticle vaccines. Hence, nanoparticles have emerged as a crucial delivery system for disease prevention and treatment, bearing immense significance. Current research highlights that nanoparticles offer superior efficacy and potential compared to conventional drug treatment and prevention methods. Notably, for drug delivery applications, it is imperative to utilize biodegradable nanoparticles. This paper reviews the structures and characteristics of various biodegradable nanoparticles and their applications in biomedicine in order to inspire more researchers to further explore the functions of nanoparticles. RNA plays a pivotal role in regulating the occurrence and progression of diseases, but its inherent susceptibility to degradation poses a challenge. In light of this, we conducted a comprehensive review of the research advancements concerning RNA-containing biodegradable nanoparticles in the realm of disease prevention and treatment, focusing on cancer, inflammatory diseases, and viral infections.
Collapse
Affiliation(s)
- Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yingying Meng
- Department of Gastroenterology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yifan Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Department of Oncology, Jingzhou Hospital Affifiliated to Yangtze University, Jingzhou, Hubei, China
| | - Yukun Han
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affifiliated Hospital, Yangtze University, Jingzhou, Hubei, China
| | - Hongwu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China; Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China.
| | - Jinbai Huang
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affifiliated Hospital, Yangtze University, Jingzhou, Hubei, China.
| |
Collapse
|
33
|
Fu J, Dong H, Wu J, Jin Y. Emerging Progress of RNA-Based Antitumor Therapeutics. Int J Biol Sci 2023; 19:3159-3183. [PMID: 37416764 PMCID: PMC10321292 DOI: 10.7150/ijbs.83732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
RNA-based therapeutics (e.g., mRNAs, siRNAs, microRNAs, ASOs, and saRNAs) have considerable potential for tumor treatment. The development and optimization of RNA modifications and delivery systems enable the stable and efficient delivery of RNA cargos in vivo to elicit an antitumor response. Targeted RNA-based therapeutics with multiple specificities and high efficacies are now available. In this review, we discuss progress in RNA-based antitumor therapeutics, including mRNAs, siRNAs, miRNAs, ASOs, saRNAs, RNA aptamers, and CRISPR-based gene editing. We focus on the immunogenicity, stability, translation efficiency, and delivery of RNA drugs, and summarize their optimization and the development of delivery systems. In addition, we describe the mechanisms by which RNA-based therapeutics induce antitumor responses. Furthermore, we review the merits and limitations of RNA cargos and their therapeutic potential for cancers.
Collapse
Affiliation(s)
- Jiayan Fu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection, Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Haiyang Dong
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection, Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, Zhejiang, China
| | - Yongfeng Jin
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, 310058, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection, Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 310006, Hangzhou, China
| |
Collapse
|
34
|
Luo H, Zhang H, Mao J, Cao H, Tao Y, Zhao G, Zhang Z, Zhang N, Liu Z, Zhang J, Luo P, Xia Y, Cheng Y, Xie Z, Cheng Q, Liu G. Exosome-based nanoimmunotherapy targeting TAMs, a promising strategy for glioma. Cell Death Dis 2023; 14:235. [PMID: 37012233 PMCID: PMC10070666 DOI: 10.1038/s41419-023-05753-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
Exosomes, the cell-derived small extracellular vehicles, play a vital role in intracellular communication by reciprocally transporting DNA, RNA, bioactive protein, chains of glucose, and metabolites. With great potential to be developed as targeted drug carriers, cancer vaccines and noninvasive biomarkers for diagnosis, treatment response evaluation, prognosis prediction, exosomes show extensive advantages of relatively high drug loading capacity, adjustable therapeutic agents release, enhanced permeation and retention effect, striking biodegradability, excellent biocompatibility, low toxicity, etc. With the rapid progression of basic exosome research, exosome-based therapeutics are gaining increasing attention in recent years. Glioma, the standard primary central nervous system (CNS) tumor, is still up against significant challenges as current traditional therapies of surgery resection combined with radiotherapy and chemotherapy and numerous efforts into new drugs showed little clinical curative effect. The emerging immunotherapy strategy presents convincing results in many tumors and is driving researchers to exert its potential in glioma. As the crucial component of the glioma microenvironment, tumor-associated macrophages (TAMs) significantly contribute to the immunosuppressive microenvironment and strongly influence glioma progression via various signaling molecules, simultaneously providing new insight into therapeutic strategies. Exosomes would substantially assist the TAMs-centered treatment as drug delivery vehicles and liquid biopsy biomarkers. Here we review the current potential exosome-mediated immunotherapeutics targeting TAMs in glioma and conclude the recent investigation on the fundamental mechanisms of diversiform molecular signaling events by TAMs that promote glioma progression.
Collapse
Affiliation(s)
- Hong Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jinning Mao
- Health management center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, Changsha, China
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhiwen Zhang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuguo Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Liu J, Li G, Guo H, Ni C, Gao Y, Cao X, Xia J, Shi X, Guo R. Dual-Responsive Core-Shell Tecto Dendrimers Enable Efficient Gene Editing of Cancer Cells to Boost Immune Checkpoint Blockade Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12809-12821. [PMID: 36853989 DOI: 10.1021/acsami.2c22584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immune checkpoint blockade (ICB) therapy has become a promising strategy in treating multiple tumor types, but the therapeutic efficacy is still unsatisfactory due to the temporary and inefficient blocking and the poor immune responsiveness. Herein, we report the development of dual reactive oxygen species (ROS)- and pH-responsive core-shell tecto dendrimers loaded with gold nanoparticles (for short, Au CSTDs) to deliver a plasmid-clustered regularly interspersed short palindromic repeats (CRISPR)/Cas9 system for the permanent disruption of the programmed death ligand 1 (PD-L1) gene in cancer cells to boost cancer immunotherapy. In our work, Au CSTDs were constructed using lactobionic acid (LA)-modified generation 5 poly(amidoamine) dendrimers entrapped with gold nanoparticles as cores and phenylboronic acid (PBA)-conjugated generation 3 dendrimers as shells via the formation of responsive phenylborate ester bonds between PBA and LA. The plasmid-CRISPR/Cas9 system can be efficiently compacted and specifically taken up by cancer cells overexpressing sialic acids due to the PBA-mediated targeting and be responsively released in cancer cells by the responsive dissociation of the Au CSTDs, leading to the successful endosomal escape and the efficient knockout of the PD-L1 gene. Further in vivo delivery in a mouse melanoma model reveals that the developed Au CSTDs/plasmid-CRISPR/Cas9 complexes can be specifically accumulated at the tumor site for enhanced computed tomography (CT) imaging of tumors, owing to the X-ray attenuation effect of Au, and disrupt the PD-L1 expression in tumor cells, thus promoting the ICB-based antitumor immunity. The designed dual-responsive Au CSTDs may be developed as a versatile tool for genetic engineering of other cell types to achieve different therapeutic effects for expanded space of biomedical applications.
Collapse
Affiliation(s)
- Junjie Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Gaoming Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, China
| | - Cheng Ni
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Jindong Xia
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
36
|
He J, Wu W. Comprehensive landscape and future perspectives of long noncoding RNAs (lncRNAs) in colorectal cancer (CRC): Based on a bibliometric analysis. Noncoding RNA Res 2023; 8:33-52. [PMID: 36311994 PMCID: PMC9582894 DOI: 10.1016/j.ncrna.2022.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
This review aimed to use bibliometric analysis to sort out, analyze and summarize the knowledge foundation and hot topics in the field of long noncoding RNAs (lncRNAs) in colorectal cancer (CRC), and point out future trends to inspire related research and innovation. We used CiteSpace to analyze publication outputs, countries, institutions, authors, journals, references, and keywords. Knowledge foundations, hotspots, and future trends were then depicted. The overall research showed the trend of biomedical-oriented multidisciplinary. Much evidence indicates that lncRNA plays the role of oncogene or tumor suppressor in the occurrence and development of CRC. Besides, many lncRNAs have multiple mechanisms. lncRNAs and metastasis of CRC, lncRNAs and drug resistance of CRC, and the clinical application of lncRNAs in CRC are current research hotspots. Through insight into the development trend of lncRNAs in CRC, this study will help researchers extract hidden valuable information for further research.
Collapse
Affiliation(s)
- Jia He
- Faculty Affairs and Human Resources Management Department, Southwest Medical University, Luzhou, China
| | - Wenhan Wu
- Department of General Surgery (Gastrointestinal Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
37
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
38
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
39
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
40
|
Wang T, Zhao H, Jing S, Fan Y, Sheng G, Ding Q, Liu C, Wu H, Liu Y. Magnetofection of miR-21 promoted by electromagnetic field and iron oxide nanoparticles via the p38 MAPK pathway contributes to osteogenesis and angiogenesis for intervertebral fusion. J Nanobiotechnology 2023; 21:27. [PMID: 36694219 PMCID: PMC9875474 DOI: 10.1186/s12951-023-01789-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Magnetofection-mediated gene delivery shows great therapeutic potential through the regulation of the direction and degree of differentiation. Lumbar degenerative disc disease (DDD) is a serious global orthopaedic problem. However, even though intervertebral fusion is the gold standard for the treatment of DDD, its therapeutic effect is unsatisfactory. Here, we described a novel magnetofection system for delivering therapeutic miRNAs to promote osteogenesis and angiogenesis in patients with lumbar DDD. RESULTS Co-stimulation with electromagnetic field (EMF) and iron oxide nanoparticles (IONPs) enhanced magnetofection efficiency significantly. Moreover, in vitro, magnetofection of miR-21 into bone marrow mesenchymal stem cells (BMSCs) and human umbilical endothelial cells (HUVECs) influenced their cellular behaviour and promoted osteogenesis and angiogenesis. Then, gene-edited seed cells were planted onto polycaprolactone (PCL) and hydroxyapatite (HA) scaffolds (PCL/HA scaffolds) and evolved into the ideal tissue-engineered bone to promote intervertebral fusion. Finally, our results showed that EMF and polyethyleneimine (PEI)@IONPs were enhancing transfection efficiency by activating the p38 MAPK pathway. CONCLUSION Our findings illustrate that a magnetofection system for delivering miR-21 into BMSCs and HUVECs promoted osteogenesis and angiogenesis in vitro and in vivo and that magnetofection transfection efficiency improved significantly under the co-stimulation of EMF and IONPs. Moreover, it relied on the activation of p38 MAPK pathway. This magnetofection system could be a promising therapeutic approach for various orthopaedic diseases.
Collapse
Affiliation(s)
- Tianqi Wang
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hongqi Zhao
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shaoze Jing
- grid.470966.aThird Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032 China
| | - Yang Fan
- grid.412793.a0000 0004 1799 5032Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Gaohong Sheng
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qing Ding
- grid.412793.a0000 0004 1799 5032Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Chaoxu Liu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hua Wu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yang Liu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
41
|
Chen X, Wei C, Huang L, Syrigos K, Li Y, Li P. Non-coding RNAs regulate mitochondrial dynamics in the development of gastric cancer. Front Mol Biosci 2023; 10:1107651. [PMID: 36714260 PMCID: PMC9877238 DOI: 10.3389/fmolb.2023.1107651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Gastric cancer (GC) is a malignant cancer that reduces life expectancy worldwide. Although treatment strategies have improved, patients with GC still have poor prognoses. Hence, it is necessary to understand the molecular mechanisms of GC and to find new therapeutic targets. Mitochondrial dynamics and mitochondrial dysfunction are associated with cancer cell growth and progression. Numerous studies have reported that non-coding RNAs (ncRNAs) can participate in the occurrence and development of GC by regulating mitochondrial dynamics. Elucidating the crosstalk between ncRNAs and mitochondria would be helpful in preventing and treating GC. Herein, we review and summarize the functions of oncogenes and tumor suppressors in suppressing ncRNAs and regulating mitochondrial dynamics in GC tumor growth, proliferation, invasion and metastasis. This review provides new insights into the pathogenesis of and intervention for GC.
Collapse
Affiliation(s)
- Xiatian Chen
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Chuang Wei
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liting Huang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Yuzhen Li
- Basic Medical Department, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
42
|
Wang J, Sha CM, Dokholyan NV. Combining Experimental Restraints and RNA 3D Structure Prediction in RNA Nanotechnology. Methods Mol Biol 2023; 2709:51-64. [PMID: 37572272 PMCID: PMC10680996 DOI: 10.1007/978-1-0716-3417-2_3] [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] [Indexed: 08/14/2023]
Abstract
Precise RNA tertiary structure prediction can aid in the design of RNA nanoparticles. However, most existing RNA tertiary structure prediction methods are limited to small RNAs with relatively simple secondary structures. Large RNA molecules usually have complex secondary structures, including multibranched loops and pseudoknots, allowing for highly flexible RNA geometries and multiple stable states. Various experiments and bioinformatics analyses can often provide information about the distance between atoms (or residues) in RNA, which can be used to guide the prediction of RNA tertiary structure. In this chapter, we will introduce a platform, iFoldNMR, that can incorporate non-exchangeable imino protons resonance data from NMR as restraints for RNA 3D structure prediction. We also introduce an algorithm, DVASS, which optimizes distance restraints for better RNA 3D structure prediction.
Collapse
Affiliation(s)
- Jian Wang
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Congzhou M Sha
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Engineering Science and Mechanics, Penn State University, State College, PA, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Engineering Science and Mechanics, Penn State University, State College, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Chemistry, Penn State University, State College, PA, USA.
- Department of Biomedical Engineering, Penn State University, State College, PA, USA.
| |
Collapse
|
43
|
Ojha A, Jaiswal S, Bharti P, Mishra SK. Nanoparticles and Nanomaterials-Based Recent Approaches in Upgraded Targeting and Management of Cancer: A Review. Cancers (Basel) 2022; 15:cancers15010162. [PMID: 36612158 PMCID: PMC9817889 DOI: 10.3390/cancers15010162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
Along with the extensive improvement in tumor biology research and different therapeutic developments, cancer remains a dominant and deadly disease. Tumor heterogeneity, systemic toxicities, and drug resistance are major hurdles in cancer therapy. Chemotherapy, radiotherapy, phototherapy, and surgical therapy are some prominent areas of cancer treatment. During chemotherapy for cancer, chemotherapeutic agents are distributed all over the body and also damage normal cells. With advancements in nanotechnology, nanoparticles utilized in all major areas of cancer therapy offer the probability to advance drug solubility, and stability, extend drug half-lives in plasma, reduce off-target effects, and quintessence drugs at a target site. The present review compiles the use of different types of nanoparticles in frequently and recently applied therapeutics of cancer therapy. A recent area of cancer treatment includes cancer stem cell therapy, DNA/RNA-based immunomodulation therapy, alteration of the microenvironment, and cell membrane-mediated biomimetic approach. Biocompatibility and bioaccumulation of nanoparticles is the major impediment in nano-based therapy. More research is required to develop the next generation of nanotherapeutics with the incorporation of new molecular entities, such as kinase inhibitors, siRNA, mRNA, and gene editing. We assume that nanotherapeutics will dramatically improve patient survival, move the model of cancer treatment, and develop certainty in the foreseeable future.
Collapse
Affiliation(s)
- Anupama Ojha
- Department of Allied Health Science, Mahayogi Gorakhnath University, Gorakhpur 273007, India
| | - Sonali Jaiswal
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
| | - Priyanka Bharti
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
| | - Sarad Kumar Mishra
- Department of Biotechnology, DDU Gorakhpur University, Gorakhpur 273009, India
- Correspondence:
| |
Collapse
|
44
|
Katopodi T, Petanidis S, Tsavlis D, Anestakis D, Charalampidis C, Chatziprodromidou I, Eskitzis P, Zarogoulidis P, Kosmidis C, Matthaios D, Porpodis K. Engineered multifunctional nanocarriers for controlled drug delivery in tumor immunotherapy. Front Oncol 2022; 12:1042125. [PMID: 36338748 PMCID: PMC9634039 DOI: 10.3389/fonc.2022.1042125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
The appearance of chemoresistance in cancer is a major issue. The main barriers to conventional tumor chemotherapy are undesirable toxic effects and multidrug resistance. Cancer nanotherapeutics were developed to get around the drawbacks of conventional chemotherapy. Through clinical evaluation of thoughtfully developed nano delivery systems, cancer nanotherapeutics have recently offered unmatched potential to comprehend and combat drug resistance and toxicity. In different design approaches, including passive targeting, active targeting, nanomedicine, and multimodal nanomedicine combination therapy, were successful in treating cancer in this situation. Even though cancer nanotherapy has achieved considerable technological development, tumor biology complexity and heterogeneity and a lack of full knowledge of nano-bio interactions remain important hurdles to future clinical translation and commercialization. The recent developments and advancements in cancer nanotherapeutics utilizing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are covered in this article. Additionally, an evaluation of different nanotherapeutics-based approaches to cancer treatment, such as tumor microenvironment targeted techniques, sophisticated delivery methods for the precise targeting of cancer stem cells, as well as an update on clinical studies are discussed. Lastly, the potential for cancer nanotherapeutics to overcome tumor relapse and the therapeutic effects and targeted efficacies of modern nanosystems are analyzed.
Collapse
Affiliation(s)
- Theodora Katopodi
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Savvas Petanidis
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Drosos Tsavlis
- Department of Medicine, Laboratory of Experimental Physiology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Doxakis Anestakis
- Department of Histology, Medical School, University of Cyprus, Nicosia, Cyprus
| | | | | | | | - Paul Zarogoulidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christoforos Kosmidis
- Third Department of Surgery, “AHEPA“ University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Konstantinos Porpodis
- Pulmonary Department-Oncology Unit, “G. Papanikolaou” General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
45
|
Wu S, Liu C, Bai S, Lu Z, Liu G. Broadening the Horizons of RNA Delivery Strategies in Cancer Therapy. Bioengineering (Basel) 2022; 9:576. [PMID: 36290544 PMCID: PMC9598637 DOI: 10.3390/bioengineering9100576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
RNA-based therapy is a promising and innovative strategy for cancer treatment. However, poor stability, immunogenicity, low cellular uptake rate, and difficulty in endosomal escape are considered the major obstacles in the cancer therapy process, severely limiting the development of clinical translation and application. For efficient and safe transport of RNA into cancer cells, it usually needs to be packaged in appropriate carriers so that it can be taken up by the target cells and then be released to the specific location to perform its function. In this review, we will focus on up-to-date insights of the RNA-based delivery carrier and comprehensively describe its application in cancer therapy. We briefly discuss delivery obstacles in RNA-mediated cancer therapy and summarize the advantages and disadvantages of different carriers (cationic polymers, inorganic nanoparticles, lipids, etc.). In addition, we further summarize and discuss the current RNA therapeutic strategies approved for clinical use. A comprehensive overview of various carriers and emerging delivery strategies for RNA delivery, as well as the current status of clinical applications and practice of RNA medicines are classified and integrated to inspire fresh ideas and breakthroughs.
Collapse
Affiliation(s)
- Shuaiying Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shuang Bai
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhixiang Lu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
46
|
Abosalha AK, Boyajian J, Ahmad W, Islam P, Ghebretatios M, Schaly S, Thareja R, Arora K, Prakash S. Clinical pharmacology of siRNA therapeutics: current status and future prospects. Expert Rev Clin Pharmacol 2022; 15:1327-1341. [PMID: 36251525 DOI: 10.1080/17512433.2022.2136166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Small interfering RNA (siRNA) has emerged as a powerful tool for post-transcriptional downregulation of multiple genes for various therapies. Naked siRNA molecules are surrounded by several barriers that tackle their optimum delivery to target tissues such as limited cellular uptake, short circulation time, degradation by endonucleases, glomerular filtration, and capturing by the reticuloendothelial system (RES). AREAS COVERED This review provides insights into studies that investigate various siRNA-based therapies, focusing on the mechanism, delivery strategies, bioavailability, pharmacokinetic, and pharmacodynamics of naked and modified siRNA molecules. The clinical pharmacology of currently approved siRNA products is also discussed. EXPERT OPINION Few siRNA-based products have been approved recently by the Food and Drug Administration (FDA) and other regulatory agencies after approximately twenty years following its discovery due to the associated limitations. The absorption, distribution, metabolism, and excretion of siRNA therapeutics are highly restricted by several obstacles, resulting in rapid clearance of siRNA-based therapeutic products from systemic circulation before reaching the cytosol of targeted cells. The siRNA therapeutics however are very promising in many diseases, including gene therapy and SARS-COV-2 viral infection. The design of suitable delivery vehicles and developing strategies toward better pharmacokinetic parameters may solve the challenges of siRNA therapies.
Collapse
Affiliation(s)
- Ahmed Khaled Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada.,Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Jacqueline Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Waqar Ahmad
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Merry Ghebretatios
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| |
Collapse
|
47
|
Chen Z, Krishnamachary B, Mironchik Y, Ray Banerjee S, Pomper MG, Bhujwalla ZM. PSMA-specific degradable dextran for multiplexed immunotargeted siRNA therapeutics against prostate cancer. NANOSCALE 2022; 14:14014-14022. [PMID: 36093754 PMCID: PMC9844541 DOI: 10.1039/d2nr02200a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Small interfering RNA (siRNA) is ideal for gene silencing through a sequence-specific RNA interference process. The redundancy and complexity of molecular pathways in cancer create a need for multiplexed targeting that can be achieved with multiplexed siRNA delivery. Here, we delivered multiplexed siRNA with a PSMA-targeted biocompatible dextran nanocarrier to downregulate CD46 and PD-L1 in PSMA expressing prostate cancer cells. The selected gene targets, PD-L1 and CD46, play important roles in the escape of cancer cells from immune surveillance. PSMA, abundantly expressed by prostate cancer cells, allowed the prostate cancer-specific delivery of the nanocarrier. The nanocarrier was modified with acid cleavable acetal bonds for a rapid release of siRNA. Cell imaging and flow cytometry studies confirmed the PSMA-specific delivery of CD46 and PD-L1 siRNA to high PSMA expressing PC-3 PIP cells. Immunoblot, qRT-PCR and flow cytometry methods confirmed the downregulation of CD46 and PD-L1 following treatment with multiplexed siRNA.
Collapse
Affiliation(s)
- Zhihang Chen
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Balaji Krishnamachary
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Yelena Mironchik
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Sangeeta Ray Banerjee
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Zaver M Bhujwalla
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
48
|
De Mey W, Esprit A, Thielemans K, Breckpot K, Franceschini L. RNA in Cancer Immunotherapy: Unlocking the Potential of the Immune System. Clin Cancer Res 2022; 28:3929-3939. [PMID: 35583609 PMCID: PMC9475240 DOI: 10.1158/1078-0432.ccr-21-3304] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/24/2022] [Accepted: 05/03/2022] [Indexed: 01/07/2023]
Abstract
Recent advances in the manufacturing, modification, purification, and cellular delivery of ribonucleic acid (RNA) have enabled the development of RNA-based therapeutics for a broad array of applications. The approval of two SARS-CoV-2-targeting mRNA-based vaccines has highlighted the advances of this technology. Offering rapid and straightforward manufacturing, clinical safety, and versatility, this paves the way for RNA therapeutics to expand into cancer immunotherapy. Together with ongoing trials on RNA cancer vaccination and cellular therapy, RNA therapeutics could be introduced into clinical practice, possibly stewarding future personalized approaches. In the present review, we discuss recent advances in RNA-based immuno-oncology together with an update on ongoing clinical applications and their current challenges.
Collapse
Affiliation(s)
- Wout De Mey
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Arthur Esprit
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.,Corresponding Author: Karine Breckpot, Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium. Phone: 32-2-477-45-66; E-mail:
| | - Lorenzo Franceschini
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
49
|
Bansal A, Kaushik S, Kukreti S. Non-canonical DNA structures: Diversity and disease association. Front Genet 2022; 13:959258. [PMID: 36134025 PMCID: PMC9483843 DOI: 10.3389/fgene.2022.959258] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
A complete understanding of DNA double-helical structure discovered by James Watson and Francis Crick in 1953, unveil the importance and significance of DNA. For the last seven decades, this has been a leading light in the course of the development of modern biology and biomedical science. Apart from the predominant B-form, experimental shreds of evidence have revealed the existence of a sequence-dependent structural diversity, unusual non-canonical structures like hairpin, cruciform, Z-DNA, multistranded structures such as DNA triplex, G-quadruplex, i-motif forms, etc. The diversity in the DNA structure depends on various factors such as base sequence, ions, superhelical stress, and ligands. In response to these various factors, the polymorphism of DNA regulates various genes via different processes like replication, transcription, translation, and recombination. However, altered levels of gene expression are associated with many human genetic diseases including neurological disorders and cancer. These non-B-DNA structures are expected to play a key role in determining genetic stability, DNA damage and repair etc. The present review is a modest attempt to summarize the available literature, illustrating the occurrence of non-canonical structures at the molecular level in response to the environment and interaction with ligands and proteins. This would provide an insight to understand the biological functions of these unusual DNA structures and their recognition as potential therapeutic targets for diverse genetic diseases.
Collapse
Affiliation(s)
- Aparna Bansal
- Nucleic Acid Research Lab, Department of Chemistry, University of Delhi, Delhi, India
- Department of Chemistry, Hansraj College, University of Delhi, Delhi, India
| | - Shikha Kaushik
- Nucleic Acid Research Lab, Department of Chemistry, University of Delhi, Delhi, India
- Department of Chemistry, Rajdhani College, University of Delhi, New Delhi, India
| | - Shrikant Kukreti
- Nucleic Acid Research Lab, Department of Chemistry, University of Delhi, Delhi, India
- *Correspondence: Shrikant Kukreti,
| |
Collapse
|
50
|
Wu C, Zhang Y, Li F, Bei S, Pan M, Feng L. Precise engineering of cholesterol-loaded chitosan micelles as a promising nanocarrier system for co-delivery drug-siRNA for the treatment of gastric cancer therapy. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
|