1
|
Arregui-Almeida D, Coronel M, Analuisa K, Bastidas-Caldes C, Guerrero S, Torres M, Aluisa A, Debut A, Brämer-Escamilla W, Pilaquinga F. Banana fruit (Musa sp.) DNA-magnetite nanoparticles: Synthesis, characterization, and biocompatibility assays on normal and cancerous cells. PLoS One 2024; 19:e0311927. [PMID: 39401205 PMCID: PMC11472939 DOI: 10.1371/journal.pone.0311927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/26/2024] [Indexed: 10/17/2024] Open
Abstract
Magnet-mediated gene therapy has gained considerable interest from researchers as a novel alternative for treating genetic disorders, particularly through the use of superparamagnetic iron oxide nanoparticles (NPs)-such as magnetite NPs (Fe3O4NPs)-as non-viral genetic vectors. Despite their commercial availability for specific genetic transfection, such as in microglia cell lines, many potential uses remain unexplored. Still, ethical concerns surrounding the use of human DNA often impede genetic research. Hence, this study examined DNA-coated Fe3O4NPs (DNA-Fe₃O₄NPs) as potential transfection vectors for human foreskin fibroblasts (HFFs) and A549 (lung cancer) cell lines, using banana (Musa sp.) as a low-cost, and bioethically unproblematic DNA source. Following coprecipitation synthesis, DNA-Fe₃O₄NP characterization revealed a ζ-potential of 40.65 ± 4.10 mV, indicating good colloidal stability in aqueous media, as well as a superparamagnetic regime, evidenced by the absence of hysteresis in their magnetization curves. Successful DNA coating on the NPs was confirmed through infrared spectra and surface analysis results, while magnetite content was verified via characteristic X-ray diffraction peaks. Transmission electron microscopy (TEM) determined the average size of the DNA-Fe3O4NPs to be 14.69 ± 5.22 nm. TEM micrographs also showed no morphological changes in the DNA-Fe3O4NPs over a 30-day period. Confocal microscopy of HFF and A549 lung cancer cell lines incubated with fluoresceinamine-labeled DNA-Fe3O4NPs demonstrated their internalization into both the cytoplasm and nucleus. Neither uncoated Fe3O4NPs nor DNA-Fe3O4NPs showed cytotoxicity to A549 lung cancer cells at 1-50 μg/mL and 25-100 μg/mL, respectively, after 24 h. HFFs also maintained viability at 1-10 μg/mL for both NP types. In conclusion, DNA-Fe3O4NPs were successfully internalized into cells and exhibited no cytotoxicity in both healthy and cancerous cells across a range of concentrations. These NPs, capable of binding to various types of DNA and RNA, hold promise for applications in gene therapy.
Collapse
Affiliation(s)
- David Arregui-Almeida
- Escuela de Ciencias Químicas, Pontificia Universidad Católica del Ecuador, Quito, Pichincha, Ecuador
| | - Martín Coronel
- Escuela de Ciencias Químicas, Pontificia Universidad Católica del Ecuador, Quito, Pichincha, Ecuador
| | - Karina Analuisa
- Escuela de Ciencias Químicas, Pontificia Universidad Católica del Ecuador, Quito, Pichincha, Ecuador
| | | | - Santiago Guerrero
- Laboratorio de Ciencia de Datos Biomédicos, Universidad Internacional del Ecuador, Quito, Pichincha, Ecuador
| | - Marbel Torres
- Centro de Nanociencia y Nanotecnología CENCINAT, Universidad de las Fuerzas Armadas, ESPE, Sangolquí, Pichincha, Ecuador
| | - Andrea Aluisa
- Centro de Nanociencia y Nanotecnología CENCINAT, Universidad de las Fuerzas Armadas, ESPE, Sangolquí, Pichincha, Ecuador
| | - Alexis Debut
- Centro de Nanociencia y Nanotecnología CENCINAT, Universidad de las Fuerzas Armadas, ESPE, Sangolquí, Pichincha, Ecuador
| | - Werner Brämer-Escamilla
- Escuela de Ciencias Físicas y Nanotecnología, Universidad Yachay Tech, Urcuquí, Imbabura, Ecuador
| | - Fernanda Pilaquinga
- Escuela de Ciencias Químicas, Pontificia Universidad Católica del Ecuador, Quito, Pichincha, Ecuador
| |
Collapse
|
2
|
Mohd Abas MD, Mohd Asri MF, Yusafawi NAS, Rosman NAZ, Baharudin NAZ, Taher M, Susanti D, Khotib J. Advancements of gene therapy in cancer treatment: A comprehensive review. Pathol Res Pract 2024; 261:155509. [PMID: 39121791 DOI: 10.1016/j.prp.2024.155509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Cancer is the main contributor for mortality in the world. Conventional therapy that available as the treatment options are chemotherapy, radiotherapy and surgery. However, these treatments are hardly cell-specific most of the time. Nowadays, extensive research and investigations are made to develop cell-specific approaches prior to cancer treatment. Some of them are photodynamic therapy, hyperthermia, immunotherapy, stem cell transplantation and targeted therapy. This review article will be focusing on the development of gene therapy in cancer. The objective of gene therapy is to correct specific mutant genes causing the excessive proliferation of the cell that leads to cancer. There are lots of explorations in the approach to modify the gene. The delivery of this therapy plays a big role in its success. If the inserted gene does not find its way to the target, the therapy is considered a failure. Hence, vectors are needed and the common vectors used are viral, non viral or synthetic, polymer based and lipid based vectors. The advancement of gene therapy in cancer treatment will be focussing on the top three cancer cases in the world which are breast, lung and colon cancer. In breast cancer, the discussed therapy are CRISPR/Cas9, siRNA and gene silencing whereas in colon cancer miRNA and suicide gene therapy and in lung cancer, replacement of tumor suppressor gene, CRISPR/Cas9 and miRNA.
Collapse
Affiliation(s)
- Muhammad Dhiyauddin Mohd Abas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Muhammad Fareez Mohd Asri
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Anis Suffiah Yusafawi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Anis Zahra Rosman
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Arifah Zahidah Baharudin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Muhammad Taher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Deny Susanti
- Department of Chemistry, Faculty of Science, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Airlangga University, Surabaya 60115, Indonesia.
| |
Collapse
|
3
|
Malik P, Rani R, Solanki R, Patel VH, Mukherjee TK. Understanding the feasibility of chemotherapeutic and immunotherapeutic targets against non-small cell lung cancers: an update of resistant responses and recent combinatorial therapies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:850-895. [PMID: 37970206 PMCID: PMC10645466 DOI: 10.37349/etat.2023.00171] [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: 02/26/2023] [Accepted: 05/17/2023] [Indexed: 11/17/2023] Open
Abstract
Despite consistent progress in prompt diagnosis and curative therapies in the last decade, lung cancer (LC) continues to threaten mankind, accounting for nearly twice the casualties compared to prostate, breast, and other cancers. Statistics associate ~25% of 2021 cancer-related deaths with LC, more than 80% of which are explicitly caused by tobacco smoking. Prevailing as small and non-small cell pathologies, with respective occurring frequency of nearly 15% and 80-85%, non-small cell LCs (NSCLCs) are prominently distinguished into lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), subtypes. Since the first use of epidermal growth factor receptor (EGFR) inhibitor gefitinib for NSCLC treatment in 2002, immense progress has been made for targeted therapies with the next generation of drugs spanning across the chronological generations of small molecule inhibitors. The last two years have overseen the clinical approval of more than 10 therapeutic agents as first-line NSCLC medications. However, uncertain mutational aberrations as well as systemic resistant responses, and abysmal overall survival curtail the combating efficacies. Of late, immune checkpoint inhibitors (ICIs) against various molecules including programmed cell death-1 (PD-1) and its ligand (PD-L1) have been demonstrated as reliable LC treatment targets. Keeping these aspects in mind, this review article discusses the success of NSCLC chemo and immunotherapies with their characteristic effectiveness and future perspectives.
Collapse
Affiliation(s)
- Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Ruma Rani
- Indian Council of Agricultural Research (ICAR)-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Raghu Solanki
- School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | | | | |
Collapse
|
4
|
Girigoswami A, Girigoswami K. Potential Applications of Nanoparticles in Improving the Outcome of Lung Cancer Treatment. Genes (Basel) 2023; 14:1370. [PMID: 37510275 PMCID: PMC10379962 DOI: 10.3390/genes14071370] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Lung cancer is managed using conventional therapies, including chemotherapy, radiation therapy, or a combination of both. Each of these therapies has its own limitations, such as the indiscriminate killing of normal as well as cancer cells, the solubility of the chemotherapeutic drugs, rapid clearance of the drugs from circulation before reaching the tumor site, the resistance of cancer cells to radiation, and over-sensitization of normal cells to radiation. Other treatment modalities include gene therapy, immunological checkpoint inhibitors, drug repurposing, and in situ cryo-immune engineering (ICIE) strategy. Nanotechnology has come to the rescue to overcome many shortfalls of conventional therapies. Some of the nano-formulated chemotherapeutic drugs, as well as nanoparticles and nanostructures with surface modifications, have been used for effective cancer cell killing and radio sensitization, respectively. Nano-enabled drug delivery systems act as cargo to deliver the sensitizer molecules specifically to the tumor cells, thereby enabling the radiation therapy to be more effective. In this review, we have discussed the different conventional chemotherapies and radiation therapies used for inhibiting lung cancer. We have also discussed the improvement in chemotherapy and radiation sensitization using nanoparticles.
Collapse
Affiliation(s)
- Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Kelambakkam, Chennai 603103, India
| |
Collapse
|
5
|
Gholami L, Ivari JR, Nasab NK, Oskuee RK, Sathyapalan T, Sahebkar A. Recent Advances in Lung Cancer Therapy Based on Nanomaterials: A Review. Curr Med Chem 2023; 30:335-355. [PMID: 34375182 DOI: 10.2174/0929867328666210810160901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer is one of the commonest cancers with a significant mortality rate for both genders, particularly in men. Lung cancer is recognized as one of the leading causes of death worldwide, which threatens the lives of over 1.6 million people every day. Although cancer is the leading cause of death in industrialized countries, conventional anticancer medications are unlikely to increase patients' life expectancy and quality of life significantly. In recent years, there are significant advances in the development and applications of nanotechnology in cancer treatment. The superiority of nanostructured approaches is that they act more selectively than traditional agents. This progress led to the development of a novel field of cancer treatment known as nanomedicine. Various formulations based on nanocarriers, including lipids, polymers, liposomes, nanoparticles and dendrimers have opened new horizons in lung cancer therapy. The application and expansion of nano-agents lead to an exciting and challenging research era in pharmaceutical science, especially for the delivery of emerging anti-cancer agents. The objective of this review is to discuss the recent advances in three types of nanoparticle formulations for lung cancer treatments modalities, including liposomes, polymeric micelles, and dendrimers for efficient drug delivery. Afterward, we have summarized the promising clinical data on nanomaterials based therapeutic approaches in ongoing clinical studies.
Collapse
Affiliation(s)
- Leila Gholami
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Rouhani Ivari
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khandan Nasab
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Retroviral Replicating Vector Toca 511 ( Vocimagene Amiretrorepvec) for Prodrug Activator Gene Therapy of Lung Cancer. Cancers (Basel) 2022; 14:cancers14235820. [PMID: 36497300 PMCID: PMC9736610 DOI: 10.3390/cancers14235820] [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: 11/04/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Therapeutic efficacy of retroviral replicating vector (RRV)-mediated prodrug activator gene therapy has been demonstrated in a variety of tumor models, but clinical investigation of this approach has so far been restricted to glioma and gastrointestinal malignancies. In the present study, we evaluated replication kinetics, transduction efficiency, and therapeutic efficacy of RRV in experimental models of lung cancer. RRV delivering GFP as a reporter gene showed rapid viral replication in a panel of lung cancer cells in vitro, as well as robust intratumoral replication and high levels of tumor transduction in subcutaneous and orthotopic pleural dissemination models of lung cancer in vivo. Toca 511 (vocimagene amiretrorepvec), a clinical-stage RRV encoding optimized yeast cytosine deaminase (yCD) which converts the prodrug 5-fluorocytosine (5-FC) to the active drug 5-fluorouracil (5-FU), showed potent cytotoxicity in lung cancer cells upon exposure to 5-FC prodrug. In vivo, Toca 511 achieved significant tumor growth inhibition following 5-FC treatment in subcutaneous and orthotopic pleural dissemination models of lung cancer in both immunodeficient and immunocompetent hosts, resulting in significantly increased overall survival. This study demonstrates that RRV can serve as highly efficient vehicles for gene delivery to lung cancer, and indicates the translational potential of RRV-mediated prodrug activator gene therapy with Toca 511/5-FC as a novel therapeutic strategy for pulmonary malignancies.
Collapse
|
7
|
Chen S, Guan L, Zhao X, Yang J, Chen L, Guo M, Zhao J, Chen C, Zhou Y, Han Y, Xu L. Optimized thyroid transcription factor-1 core promoter-driven microRNA-7 expression effectively inhibits the growth of human non-small-cell lung cancer cells. J Zhejiang Univ Sci B 2022; 23:915-930. [PMID: 36379611 PMCID: PMC9676096 DOI: 10.1631/jzus.b2200116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/22/2022] [Indexed: 07/22/2023]
Abstract
Targeted gene therapy has become a promising approach for lung cancer treatment. In our previous work, we reported that the targeted expression of microRNA-7 (miR-7) operated by thyroid transcription factor-1 (TTF-1) promoter inhibited the growth of human lung cancer cells in vitro and in vivo; however, the intervention efficiency needed to be further improved. In this study, we identified the core promoter of TTF-1 (from -1299 bp to -871 bp) by 5' deletion assay and screened out the putative transcription factors nuclear factor-1 (NF-1) and activator protein-1 (AP-1). Further analysis revealed that the expression level of NF-1, but not AP-1, was positively connected with the activation of TTF-1 core promoter in human non-small-cell lung cancer (NSCLC) cells. Moreover, the silencing of NF-1 could reduce the expression level of miR-7 operated by TTF-1 core promoter. Of note, we optimized four distinct sequences to form additional NF-1-binding sites (TGGCA) in the sequence of TTF-1 core promoter (termed as optTTF-1 promoter), and verified the binding efficiency of NF-1 on the optTTF-1 promoter by electrophoretic mobility shift assay (EMSA). As expected, the optTTF-1 promoter could more effectively drive miR-7 expression and inhibit the growth of human NSCLC cells in vitro, accompanied by a reduced transduction of NADH dehydrogenase (ubiquinone) 1α subcomplex 4 (NDUFA4)/protein kinase B (Akt) pathway. Consistently, optTTF-1 promoter-driven miR-7 expression could also effectively abrogate the growth and metastasis of tumor cells in a murine xenograft model of human NSCLC. Finally, no significant changes were detected in the biological indicators or the histology of some important tissues and organs, including heart, liver, and spleen. On the whole, our study revealed that the optimized TTF-1 promoter could more effectively operate miR-7 to influence the growth of human NSCLC cells, providing a new basis for the development of microRNA-based targeting gene therapy against clinical lung cancer.
Collapse
Affiliation(s)
- Shipeng Chen
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Lian Guan
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Xu Zhao
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Jing Yang
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Longqing Chen
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ya Zhou
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China. ,
- Department of Medical Physics, Zunyi Medical University, Zunyi 563000, China. ,
| | - Yong Han
- Department of Physiology, Zunyi Medical University, Zunyi 563000, China. ,
| | - Lin Xu
- Special Key Laboratory of Gene Detection and Therapy & Base for Talents in Biotherapy of Guizhou Province, Zunyi 563000, China.
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
8
|
Sivarajah S, Emerick K, Kaufman HL. What Surgeons Need to Know About Gene Therapy for Cancer. Adv Surg 2022; 56:151-168. [PMID: 36096566 DOI: 10.1016/j.yasu.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The broad field of gene therapy offers numerous innovative approaches for cancer treatment. An understanding of the different modalities including gene replacement therapy, cancer vaccines, oncolytic viruses, cellular therapy, and gene editing is essential for managing patients with neoplastic disease. As in other areas of oncology, the surgeon plays a pivotal role in the diagnosis and treatment of the disease. This review focuses on what the clinical surgeon needs to know to optimize the benefit of gene therapy for patients with cancer.
Collapse
Affiliation(s)
- Shanmugappiriya Sivarajah
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, 243 Charles Street Boston, MA 02114 USA
| | - Kevin Emerick
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, 243 Charles Street Boston, MA 02114 USA
| | - Howard L Kaufman
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, 55 Fruit Street, Yawkey 7E, Boston, MA 02114, USA.
| |
Collapse
|
9
|
Effects of inhalable gene transfection as a novel gene therapy for non-small cell lung cancer and malignant pleural mesothelioma. Sci Rep 2022; 12:8634. [PMID: 35606391 PMCID: PMC9126906 DOI: 10.1038/s41598-022-12624-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/13/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractGene therapy using vectors has attracted attention in recent years for the treatment of cancers caused by gene mutations. Besides, new treatments are imperative for lung cancer, including non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM), due to its high mortality. We developed a minimally invasive and orally inhalable tumor suppressor gene drug (SFD-p16 and SFD-p53) with non-viral vectors for lung cancer treatment by combining tumor suppressor genes with an inhalant powder that can deliver active ingredients directly to the lung. We used NSCLC (A549 and H1299) and MPM (H2052) cell lines in an air–liquid interface culture. Transfection of A549 and H2052 cells with SFD-p16 significantly increased p16 mRNA expression levels and decreased cell proliferation in both cell lines. Similar results were obtained with transfection of H1299 with the inhalable gene drug SFD-p53. In an in vivo experiment, a mouse model of lung cancer with orthotopically transplanted luciferase-expressing A549 cells was subjected to intratracheal insufflation of SFD-p16. Consequently, SFD-p16 effectively and directly affected lung cancer. This study suggests that inhalable gene drugs are effective treatments for NSCLC and MPM. We expect inhalable gene drugs to present a novel gene therapy agent for lung cancer that patients can self-administer.
Collapse
|
10
|
Singh V, Khan N, Jayandharan GR. Vector engineering, strategies and targets in cancer gene therapy. Cancer Gene Ther 2022; 29:402-417. [PMID: 33859378 DOI: 10.1038/s41417-021-00331-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/23/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023]
Abstract
Understanding the molecular basis of disease and the design of rationally designed molecular therapies has been the holy grail in the management of human cancers. Gene-based therapies are an important avenue for achieving a possible cure. Focused research in the last three decades has provided significant clues to optimize the potential of cancer gene therapy. The development of gene therapies with a high potential to kill the target cells at the lowest effective dose possible, the development of vectors with significant ability to target cancer-associated antigen, the application of adjunct therapies to target dysregulated microRNA, and embracing a hybrid strategy with a combination of gene therapy and low-dose chemotherapy in a disease-specific manner will be pivotal. This article outlines the advances and challenges in the field with emphasis on the biology and scope of vectors used for gene transfer, newer targets identified, and their outcome in preclinical and clinical studies.
Collapse
Affiliation(s)
- Vijayata Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India
| | - Nusrat Khan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India
| | - Giridhara R Jayandharan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, UP, India. .,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur, UP, India.
| |
Collapse
|
11
|
Bisserier M, Sun XQ, Fazal S, Turnbull IC, Bonnet S, Hadri L. Novel Insights into the Therapeutic Potential of Lung-Targeted Gene Transfer in the Most Common Respiratory Diseases. Cells 2022; 11:984. [PMID: 35326434 PMCID: PMC8947048 DOI: 10.3390/cells11060984] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Over the past decades, a better understanding of the genetic and molecular alterations underlying several respiratory diseases has encouraged the development of new therapeutic strategies. Gene therapy offers new therapeutic alternatives for inherited and acquired diseases by delivering exogenous genetic materials into cells or tissues to restore physiological protein expression and/or activity. In this review, we review (1) different types of viral and non-viral vectors as well as gene-editing techniques; and (2) the application of gene therapy for the treatment of respiratory diseases and disorders, including pulmonary arterial hypertension, idiopathic pulmonary fibrosis, cystic fibrosis, asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease, non-small-cell lung cancer, and COVID-19. Further, we also provide specific examples of lung-targeted therapies and discuss the major limitations of gene therapy.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Xiao-Qing Sun
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Shahood Fazal
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Irene C. Turnbull
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec, QC G1V4G5, Canada;
- Department of Medicine, Laval University, Québec, QC G1V4G5, Canada
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| |
Collapse
|
12
|
Gao F, Mu X, Wu H, Chen L, Liu J, Zhao Y. Calreticulin (CALR)-induced activation of NF-ĸB signaling pathway boosts lung cancer cell proliferation. Bioengineered 2022; 13:6856-6865. [PMID: 35264066 PMCID: PMC8974040 DOI: 10.1080/21655979.2022.2040874] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Calreticulin (CALR) is known to be aberrantly expressed in lung though the etiology underlying this phenomenon remains undetermined. The (Cancer Genome Atlas) databases were adopted to evaluate the expression status of CALR in pan-cancer, including Lung adenocarcinoma (LUAD) and Lung squamous cell carcinoma (LUSC) accompanied with Genotype-Tissue Expression project (GETx) database. Receiver operating characteristic (ROC) curves and Kaplan-Meier survival curve were plotted to assess its clinical significance in lung cancer. CCK8 and colony formation assays were conducted in addition to in vivo assays. The impact of CALR expression on NF-ĸB-mediated luciferase activity was detected by Luciferase assays. The regulatory relationship between CALR and NF-ĸB was further verified by NF-ĸB inhibitor treatment. LUAD and LUSC tissues reflected marked elevation in the mRNA levels of CALR. ROC analysis showed that CALR expression had a diagnostic value for LUAD or LUSC patients. High-CARL patients demonstrated inferior survival compared to that of Low-CALR patients. Functional assays revealed increased proliferative behaviors of A549 and H1299 cells associated with highly amplified while CALR gene inactivation could reduce the proliferation of both cells. CALR depletion decreased xenograft tumor growth. NF-ĸB transcriptional activity was found to be stimulated with CALR overexpression and reduced in CALR-deficient lung cancer cells, thereby clearly indicating CALR-dependent NF-ĸB activation. NF-ĸB specific inhibitors further validated enhanced NF-ĸB activity mediated by CALR overexpression. Conclusively, our results the role of CALR in lung cancer cells, indicating that highly expressed CALR proliferation at least by activation of NF-ĸB signaling pathway.
Collapse
Affiliation(s)
- Fangfang Gao
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| | - Xiaoqian Mu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| | - Huijuan Wu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| | - Lijuan Chen
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| | - Jie Liu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| | - Yanqiu Zhao
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, China
| |
Collapse
|
13
|
Wang J, Mo J, Xie Y, Wang C. Ultrasound microbubbles-mediated miR-216b affects MALAT1-miRNA axis in non-small cell lung cancer cells. Tissue Cell 2021; 74:101703. [PMID: 34896788 DOI: 10.1016/j.tice.2021.101703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/19/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022]
Abstract
MiR-216b is ectopically expressed in various cancers. Ultrasound microbubbles (UTMBs) are an effective method for miRNA delivery. This article mainly explored the involvement of lncRNA in the effects of UTMBs-mediated miR-216b on non-small cell lung cancer (NSCLC) progression. Expressions and relationship of miR-216b and MALAT1 were examined using quantitative real-time polymerase chain reaction (qRT-PCR), Pearson, TargetScan, and dual-luciferase reporter assay. After the transfection with liposome- or UTMBs-mediated miR-216b mimic (M) or MALAT1 overexpression plasmid alone or together, levels of miR-216b and MALAT1, cell biological behaviors, as well as expressions of apoptosis- and epithelial mesenchymal transition (EMT)-related markers were examined using qRT-PCR, cell functional experiments, and western blot. Besides, we used qRT-PCR to quantify the expressions of multiple downstream miRNAs of MALAT1. MiR-216b expression was weakened yet MALAT1 expression was enhanced in NSCLC tissues, and miR-216b was negatively bound to MALAT1. TargetScan analysis manifested that miR-216b, targeted by MALAT1, was down-regulated in NSCLC cells. UTMBs-mediated miR-216b M further intensified miR-216b level yet weakened cell biological behaviors. The inhibitory effect of UTMBs-mediated miR-216b M on cell biological behaviors and MALAT1 expression was greatly better relative to that of miR-216b M. Moreover, miR-216b restrained the cell biological behaviors by repressing MALAT1 expression. We further manifested that miR-216b facilitated the expressions of apoptosis-related markers, but restrained those of EMT-related markers by repressing MALAT1 expression. Moreover, UTMBs-mediated miR-216b M enhanced the expressions of downstream multiple miRNAs of MALAT1, but this tendency was reversed by co-transfection of overexpressed MALAT1 and miR-216b M. Collectively, UTMBs-mediated miR-216b M restrained NSCLC cell growth by modulating the MALAT1-miRNA axis.
Collapse
Affiliation(s)
- Jian Wang
- Thoracic Surgery Department, Shenzhen People's Hospital, China
| | - Jianming Mo
- Pulmonary and Critical Care Medicine Department, Peking University Shenzhen Hospital, China
| | - Yuancai Xie
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, China.
| | - Chunguang Wang
- Thoracic Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
14
|
Artzy-Schnirman A, Arber Raviv S, Doppelt Flikshtain O, Shklover J, Korin N, Gross A, Mizrahi B, Schroeder A, Sznitman J. Advanced human-relevant in vitro pulmonary platforms for respiratory therapeutics. Adv Drug Deliv Rev 2021; 176:113901. [PMID: 34331989 PMCID: PMC7611797 DOI: 10.1016/j.addr.2021.113901] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 02/08/2023]
Abstract
Over the past years, advanced in vitro pulmonary platforms have witnessed exciting developments that are pushing beyond traditional preclinical cell culture methods. Here, we discuss ongoing efforts in bridging the gap between in vivo and in vitro interfaces and identify some of the bioengineering challenges that lie ahead in delivering new generations of human-relevant in vitro pulmonary platforms. Notably, in vitro strategies using foremost lung-on-chips and biocompatible "soft" membranes have focused on platforms that emphasize phenotypical endpoints recapitulating key physiological and cellular functions. We review some of the most recent in vitro studies underlining seminal therapeutic screens and translational applications and open our discussion to promising avenues of pulmonary therapeutic exploration focusing on liposomes. Undeniably, there still remains a recognized trade-off between the physiological and biological complexity of these in vitro lung models and their ability to deliver assays with throughput capabilities. The upcoming years are thus anticipated to see further developments in broadening the applicability of such in vitro systems and accelerating therapeutic exploration for drug discovery and translational medicine in treating respiratory disorders.
Collapse
Affiliation(s)
- Arbel Artzy-Schnirman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Sivan Arber Raviv
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | | | - Jeny Shklover
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Adi Gross
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Boaz Mizrahi
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Avi Schroeder
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel.
| |
Collapse
|
15
|
Niu Z, Zhang W, Shi J, Li X, Wu H. Effect of silencing C-erbB-2 on esophageal carcinoma cell biological behaviors by inhibiting IGF-1 pathway activation. J Cardiothorac Surg 2021; 16:194. [PMID: 34233689 PMCID: PMC8265138 DOI: 10.1186/s13019-021-01540-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/13/2021] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE C-erbB-2 has been confirmed to be an oncogene that participates in cell growth, differentiation and division of tumors. We are wondered if its silenced expression can exert an anti-tumor effect. Therefore, this study is conducted to investigate the mechanism of C-erbB-2 silencing and IGF-1 pathway on esophageal carcinoma (EC) cell biological behaviors. METHODS The objects of study were 84 EC patients from Heping Hospital Affiliated to Changzhi Medical College, with the collection of EC tissue and adjacent normal tissue (> 5 cm away from cancer tissue). C-erbB-2 protein expression in EC tissues was detected by immunohistochemistry. Human EC cell line Eca-109 was purchased from Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. Based on different transfection protocols, EC cells with logarithmic growth phase of 3-5 passages were divided into blank control group, oe-C-erbB-2 NC group, siRNA C-erbB-2 NC group, oe-C-erbB-2 group, siRNA C-erbB-2 group, OSI-906 group, Rg5 group, Rg5 + siRNA C-erbB-2 NC group and Rg5 + siRNA C-erbB-2 group. Cell proliferation was detected by MTT assay; cell cycle distribution and apoptosis by flow cytometry; C-erbB-2, IGF-1, IGF-1R and Akt mRNA and protein expressions by qRT-PCR and western blot; and cell invasion and migration by Transwell assay and scratch test. Tumor growth was observed in male BALB/c nude mice (Shanghai Experimental Animal Center) based on Eca109 cell implantation, raising, and measurement. RESULTS C-erbB-2, IGF-1, IGF-1R and Akt expression were higher in EC tissues than those in adjacent tissues (all P < 0.05). Compared with blank control group, both si-C-erbB-2 and OSI-906 groups had decreased IGF-1, IGF-1R and Akt mRNA and protein expressions, decreased cell proliferation, migration and invasion, prolonged G0/G1 phase, shortened S phase, increased cell apoptosis, and inhibited tumor growth (all P < 0.05); while opposite trends were detected in C-erbB-2 vector and Rg5 groups (all P < 0.05), without statistical differences in siRNA C-erbB-2 + Rg5 group (all P > 0.05). CONCLUSION Silencing C-erbB-2 expression may inhibit EC cell proliferation, promote cell apoptosis and block cell cycle progression by inhibiting IGF-1 pathway activation. The beneficial effect of silencing C-erbB-2 expression can be reversed by promoting the activation of IGF-1 pathway. Findings in our study may provide potential reference for understanding the molecular mechanism of EC and supply possible axis for preventing the development of EC from the perspective of molecular biology.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Apoptosis/genetics
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Female
- Flow Cytometry
- Gene Expression Regulation, Neoplastic/physiology
- Gene Silencing/physiology
- Humans
- Immunohistochemistry
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplasm Transplantation
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/genetics
- Receptor, IGF Type 1
- Transfection
- Mice
Collapse
Affiliation(s)
- Zhigao Niu
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China.
| | - Wenping Zhang
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Jialun Shi
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Xiangdong Li
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Hanlei Wu
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| |
Collapse
|
16
|
Chaudhary S, Singh A, Kumar P, Kaushik M. Strategic targeting of non-small-cell lung cancer utilizing genetic material-based delivery platforms of nanotechnology. J Biochem Mol Toxicol 2021; 35:e22784. [PMID: 33826765 DOI: 10.1002/jbt.22784] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 02/04/2021] [Accepted: 03/24/2021] [Indexed: 02/06/2023]
Abstract
Several limitations of conventional cancer treatment such as non-specific targeting, solubility problems, and ineffective entry of chemotherapeutics into cancer cells can be overcome by using nanotechnology targeted drug delivery systems. Some combinations of biomolecules and nanoparticles have proven to be excellent therapeutics for Non-small cell lung cancer (NSCLC) in the last decades. Targeted gene delivery has shown in vivo as well as in vitro promising results with therapeutic efficacy. Gene therapy has shown enhanced transfection efficiency and better targeting potential on several NSCLC cell lines. Still, there are several challenges in nanoparticle-mediated gene therapy, which include stability of biomolecules and nanoparticles during delivery, managing their biodistribution, and reducing the possible cytotoxic effects of the nanoparticles, which need to be solved before clinical trials. Evaluation of therapeutic efficacy of biomolecules and nanoparticle combination in gene therapy must be established to expand the application of nano-gene therapy in cancer treatment.
Collapse
Affiliation(s)
- Swati Chaudhary
- Department of Applied Sciences, Maharaja Surajmal Institute of Technology, GGSIP University, New Delhi, India
| | - Amit Singh
- Department of Chemistry, University of Delhi, Delhi, India.,Nano-bioconjugate Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi, India
| | - Pankaj Kumar
- Department of Chemistry, University of Delhi, Delhi, India.,Nano-bioconjugate Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi, India
| | - Mahima Kaushik
- Nano-bioconjugate Chemistry Lab, Cluster Innovation Centre, University of Delhi, Delhi, India
| |
Collapse
|
17
|
Santos RVC, de Sena WLB, Dos Santos FA, da Silva Filho AF, da Rocha Pitta MG, da Rocha Pitta MG, de Melo Rego MB, Pereira MC. Potential Therapeutic Agents Against Par-4 Target for Cancer Treatment: Where Are We Going? Curr Drug Targets 2020; 20:635-654. [PMID: 30474528 DOI: 10.2174/1389450120666181126122440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023]
Abstract
One of the greatest challenges of cancer therapeutics nowadays is to find selective targets successfully. Prostate apoptosis response-4 (Par-4) is a selective tumor suppressor protein with an interesting therapeutic potential due to its specificity on inducing apoptosis in cancer cells. Par-4 activity and levels can be downregulated in several tumors and cancer cell types, indicating poor prognosis and treatment resistance. Efforts to increase Par-4 expression levels have been studied, including its use as a therapeutic protein by transfection with adenoviral vectors or plasmids. However, gene therapy is very complex and still presents many hurdles to be overcome. We decided to review molecules and drugs with the capacity to upregulate Par-4 and, thereby, be an alternative to reach this druggable target. In addition, Par-4 localization and function are reviewed in some cancers, clarifying how it can be used as a therapeutic target.
Collapse
Affiliation(s)
- Renata Virgínia Cavalcanti Santos
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Wanessa Layssa Batista de Sena
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Flaviana Alves Dos Santos
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Antônio Felix da Silva Filho
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | | | - Maira Galdino da Rocha Pitta
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Moacyr Barreto de Melo Rego
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| | - Michelly Cristiny Pereira
- Laboratory of Immunomodulation and New Therapeutical Approaches, Research Centre for Therapeutic Innovation Suely Galdino (NUPIT-SG), Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
18
|
DeMaio A, Sterman D. Bronchoscopic intratumoural therapies for non-small cell lung cancer. Eur Respir Rev 2020; 29:200028. [PMID: 32554757 PMCID: PMC9488902 DOI: 10.1183/16000617.0028-2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/07/2020] [Indexed: 12/20/2022] Open
Abstract
The past decade has brought remarkable improvements in the treatment of non-small cell lung cancer (NSCLC) with novel therapies, such as immune checkpoint inhibitors, although response rates remain suboptimal. Direct intratumoural injection of therapeutic agents via bronchoscopic approaches poses the unique ability to directly target the tumour microenvironment and offers several theoretical advantages over systemic delivery including decreased toxicity. Increases in understanding of the tumour microenvironment and cancer immunology have identified many potential options for intratumoural therapy, especially combination immunotherapies. Herein, we review advances in the development of novel bronchoscopic treatments for NSCLC over the past decade with a focus on the potential of intratumoural immunotherapy alone or in combination with systemic treatments.
Collapse
Affiliation(s)
- Andrew DeMaio
- NYU PORT (Pulmonary Oncology Research Team), Division of Pulmonary, Critical Care, and Sleep Medicine, NYU Langone Health/NYU Grossman School of Medicine, New York, NY, United States
| | - Daniel Sterman
- NYU PORT (Pulmonary Oncology Research Team), Division of Pulmonary, Critical Care, and Sleep Medicine, NYU Langone Health/NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Holt GE, Daftarian P. Non-small-cell lung cancer homing peptide-labeled dendrimers selectively transfect lung cancer cells. Immunotherapy 2019; 10:1349-1360. [PMID: 30474481 DOI: 10.2217/imt-2018-0078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM Lung cancer gene therapies require reagents to selectively transfect lung tumors after systemic administration. MATERIALS & METHODS We created a reagent called NSCLC-NP by attaching a peptide with binding affinity for lung cancer to polyamidoamine dendrimers. The positively charged dendrimers electrostatically bind negatively charged nucleic acids, inhibit endogenous nucleases and transfect cells targeted by the attached peptide. RESULTS In vitro, NSCLC-NP complexed to DNA plasmids bound and transfected three human lung cancer cell lines producing protein expression of the plasmid's gene. In vivo, systemically administered NSCLC-NP selectively transfected lung cancer cells growing in RAG1KO mice. CONCLUSION The capability of NSCLC-NP to selectively transfect lung cancer allows its future use as a vehicle to implement human lung cancer gene therapy strategies.
Collapse
Affiliation(s)
- Gregory E Holt
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, University of Miami, Miami, FL, USA.,Department of Medicine, Division of Pulmonology, Miami VA Medical Center, Miami, FL, USA
| | - Pirouz Daftarian
- Department of Ophthalmology, University of Miami, FL, USA.,JSR Micro Life Sciences, Sunnyvale, CA 94089, USA
| |
Collapse
|
20
|
Lee AY, Cho MH, Kim S. Recent advances in aerosol gene delivery systems using non-viral vectors for lung cancer therapy. Expert Opin Drug Deliv 2019; 16:757-772. [PMID: 31282221 DOI: 10.1080/17425247.2019.1641083] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Lung cancer commonly occurs at a high incidence worldwide. Application of aerosol gene delivery systems using various kinds of vectors can improve the patient's quality of life by prolonging the survival rate. AREAS COVERED This review provides a recent update on aerosol gene delivery strategies using various kinds of vectors and gene-modification technologies. Peptide-mediated gene therapy achieves specific targeting of cells and highly improves efficacy. Promoter-operating expression and the CRISPR/Cas9 system are novel gene therapy strategies for effective lung cancer treatment. Furthermore, hybrid systems with a combination of vectors or drugs have been recently applied as new trends in gene therapy. EXPERT OPINION Although aerosol gene delivery has many advantages, physiological barriers in the lungs pose formidable challenges. Targeted gene delivery and gene-editing technology are promising strategies for lung cancer therapy. These strategies may allow the development of safety and high efficiency for clinical application. Recently, hybrid gene therapy combining novel and specific vectors has been developed as an advanced strategy. Although gene therapy for lung cancer is being actively researched, aerosol gene therapy strategies are currently lacking, and further studies on aerosol gene therapy are needed to treat lung cancer.
Collapse
Affiliation(s)
- Ah Young Lee
- a Center for Molecular Recognition Research, Materials and Life Science Research Division , Korea Institute of Science and Technology (KIST) , Seoul , Korea
| | - Myung-Haing Cho
- b Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine , Seoul National University , Seoul , Republic of Korea
| | - Sanghwa Kim
- c Cancer Biology Laboratory , Institut Pasteur Korea , Seongnam-si , Korea
| |
Collapse
|
21
|
Lin XR, Zhou XL, Feng Q, Pan XY, Song SL, Fang H, Lei J, Yang JL. CIK cell-based delivery of recombinant adenovirus KGHV500 carrying the anti-p21Ras scFv gene enhances the anti-tumor effect and safety in lung cancer. J Cancer Res Clin Oncol 2019; 145:1123-1132. [PMID: 30796510 DOI: 10.1007/s00432-019-02857-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Adenovirus (Ads) is one of the most popular vectors used in gene therapy for the treatment of cancer. However, systemic therapy is limited by circulating antiviral antibodies and poor viral delivery in vivo. In this study, we used cytokine-induced killer (CIK) cells as delivery vehicles of Ads KGHV500 carrying the anti-p21Ras scFv gene to treat Ras gene-related lung cancer and investigate the anti-tumor effect in vitro and in vivo. METHODS The human lung cancer cell line A549 was employed to investigate the anti-tumor activity of recombinant Ads KGHV500 harboring the anti-p21Ras scFv gene using MTT, wound healing, transwell invasion, and apoptosis assays in vitro. Next, CIK cells were used as delivery vehicles to deliver KGHV500 carrying the anti-p21Ras scFv gene to treat A549-transplanted tumors in nude mice, and viral replication, p21Ras scFv expression, and the therapeutic efficacy were assessed. RESULTS In vitro studies showed that KGHV500 had potent anti-tumor activity. In addition, in vivo, this combination therapy significantly inhibited the growth of lung cancer xenografts compared with mice treated with KGHV500 alone. KGHV500 and anti-p21Ras scFv were observed in tumor tissue, but were nearly undetectable in normal tissues. CONCLUSIONS The co-delivery of anti-p21Ras scFv by CIK cells and KGHV500 could increase the anti-tumor effect and safety, and possess considerable advantages for the treatment of Ras-related cancer.
Collapse
Affiliation(s)
- Xin-Rui Lin
- Graduate School, Kunming Medical University, Chenggong District, Kunming, People's Republic of China.,Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Xin-Liang Zhou
- Department of Pathology, Medical Faculty, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Qiang Feng
- Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Xin-Yan Pan
- Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Shu-Ling Song
- Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Hong Fang
- Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Jin Lei
- Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China
| | - Ju-Lun Yang
- Graduate School, Kunming Medical University, Chenggong District, Kunming, People's Republic of China. .,Department of Pathology, 920th Hospital of the Joint Logistics Support Force of PLA, 212 Daguan Road, Kunming, 650032, Yunnan, People's Republic of China. .,Department of Pathology, Medical Faculty, Kunming University of Science and Technology, Kunming, People's Republic of China.
| |
Collapse
|
22
|
Qi QM, Xue YC, Lv J, Sun D, Du JX, Cai SQ, Li YH, Gu TC, Wang MB. Ginkgolic acids induce HepG2 cell death via a combination of apoptosis, autophagy and the mitochondrial pathway. Oncol Lett 2018; 15:6400-6408. [PMID: 29725398 PMCID: PMC5920365 DOI: 10.3892/ol.2018.8177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
Ginkgolic acids may induce malignant cell death via the B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax)/Bcl-2 apoptosis pathway. Concurrently, apoptosis, autophagy and mitochondrial dysfunction may also be involved in bringing about this endpoint. The anticancer effect of Ginkgolic acids (GAs) was investigated using the HepG2 cell line. The median lethal dose of the GAs of the HepG2 was measured via an MTT assay, the dose-response curves were evaluated and changes in cell morphology were monitored by microscopy. Autophagy in HepG2 cells was down regulated using 3-methyladenine (3-MA) or Beclin-1-specific small interfering RNA (siRNA) and the expression of apoptosis associated proteins caspase-3, Bax/Bcl-2, and the autophagy-associated protein 5 and microtubule-associated protein 1A/1B-light chain 3 in the GA-treated HepG2 cells were all measured by western blot analysis. The level of apoptosis in the GA-treated cells was also assessed using terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling (TUNEL) assay, and the mitochondrial membrane potential (Δψm) was detected by immunofluorescence. The results of the MTT and TUNEL assays indicated that the proliferation of HepG2 cells treated with GAs was significantly reduced compared with the control group, and the rate of the inhibition was dose-dependent. Western blot analysis indicated that treatment with the Gas induced apoptosis and autophagy in the HepG2 cells. The Δψm of the GA-treated HepG2 cells was decreased compared with the control, as monitored by immunofluorescence. However, upon the administration of 3-MA or Beclin-1-specific siRNAs (inhibitors of the autophagy), the expression levels of the apoptosis- and autophagy-associated proteins were decreased. In conclusion, the results of the present study indicated that GAs are potent anticancer agents that function through a combination of the apoptosis, autophagy and mitochondrial pathways.
Collapse
Affiliation(s)
- Qian-Ming Qi
- Department of Medical Laboratory, The 359th Hospital of The People's Liberation Army, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yin-Cun Xue
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Jian Lv
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Di Sun
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Jian-Xin Du
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Sheng-Qiang Cai
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Yun-He Li
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Tian-Cun Gu
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Mu-Bing Wang
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China,Correspondence to: Dr Mu-Bing Wang, Department of General Surgery, Jingjiang People's Hospital, 28 Zhongzhou Road, Taizhou, Jiangsu 214500, P.R. China, E-mail:
| |
Collapse
|