1
|
Shen Y, Zhang H, Xue M, Zheng C, Chen Q. HSV-1 as a gene delivery platform for cancer gene therapy. Trends Pharmacol Sci 2025; 46:324-336. [PMID: 40069043 DOI: 10.1016/j.tips.2025.02.006] [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: 01/23/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 04/06/2025]
Abstract
Herpes simplex virus type 1 (HSV-1) is a DNA virus with strong replication capabilities, a large genomic payload (≥30 kb), and low toxicity, making it a prominent vector in cancer gene therapy. Clinically approved oncolytic HSV-1 (oHSV-1) variants, such as T-VEC and G47Δ, demonstrate safety and efficacy in localized tumors, but face challenges in treating metastatic disease. To address this issue, next-generation oHSV-1 designs focus on precision targeting and immune remodeling through the delivery of multiple exogenous genes. In this review, we provide an overview of the inherent characteristics of oHSV-1 as a gene delivery platform, focusing on its genetic modification strategies, safety challenges in clinical applications, and future directions to maximize its therapeutic potential.
Collapse
Affiliation(s)
- Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Hucheng Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology & Infection Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
2
|
Schock Vaiani J, Broekgaarden M, Coll JL, Sancey L, Busser B. In vivo vectorization and delivery systems for gene therapies and RNA-based therapeutics in oncology. NANOSCALE 2025; 17:5501-5525. [PMID: 39927415 DOI: 10.1039/d4nr05371k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Gene and RNA-based therapeutics represent a promising frontier in oncology, enabling targeted modulation of tumor-associated genes and proteins. This review explores the latest advances in payload vectorization and delivery systems developed for in vivo cancer treatments. We discuss viral and non-viral organic particles, including lipid based nanoparticles and polymeric structures, for the effective transport of plasmids, siRNA, and self-amplifying RNA therapeutics. Their physicochemical properties, strategies to overcome intracellular barriers, and innovations in cell-based carriers and engineered extracellular vesicles are highlighted. Moreover, we consider oncolytic viruses, novel viral capsid modifications, and approaches that refine tumor targeting and immunomodulation. Ongoing clinical trials and regulatory frameworks guide future directions and emphasize the need for safe, scalable production. The potential convergence of these systems with combination therapies paves the way toward personalized cancer medicine.
Collapse
Affiliation(s)
- Julie Schock Vaiani
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Mans Broekgaarden
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Jean-Luc Coll
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Lucie Sancey
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Benoit Busser
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
- Grenoble Alpes Univ. Hospital (CHUGA), 38043 Grenoble, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
3
|
Rahimian S, Mirkazemi K, Kamalinejad A, Doroudian M. Exosome-based advances in pancreatic cancer: The potential of mesenchymal stem cells. Crit Rev Oncol Hematol 2025; 207:104594. [PMID: 39732301 DOI: 10.1016/j.critrevonc.2024.104594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 12/08/2024] [Indexed: 12/30/2024] Open
Abstract
Pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), is one of the most challenging clinical conditions due to its late-stage diagnosis and poor survival rates. Mesenchymal stem cells (MSCs), used for targeted therapies, are being explored as a promising treatment because of their tumor-homing properties and potential contributions to the pancreatic cancer microenvironment. Understanding these interactions is crucial for developing effective treatments. In this study, we investigated how MSCs exhibit tropism towards tumors, influence the microenvironment through paracrine effects, and serve as potential drug delivery vehicles. We also examined their role in progression and therapeutic resistance in pancreatic cancer therapy. The cytotoxic effects of certain compounds on tumor cells, the use of genetically modified MSCs as drug carriers, and the potential of exosomal biomarkers like miRNAs and riRNAs for diagnosis and monitoring of pancreatic cancer were analyzed. Overall, MSC-based therapies, coupled with insights into tumor-stromal interactions, offer new avenues for improving outcomes in pancreatic cancer treatment. Additionally, the use of MSC-based therapies in clinical trials is discussed. While MSCs show promising potential for pancreatic cancer monitoring, diagnosis, and treatment, results so far have been limited.
Collapse
Affiliation(s)
- Sana Rahimian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Kimia Mirkazemi
- Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Armita Kamalinejad
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
4
|
Yang P, He S, Fan L, Ye L, Weng H. Risk factors for immunoresistance in advanced non-small cell lung cancer and the advantages of targeted therapy in improving prognosis. Am J Cancer Res 2025; 15:573-586. [PMID: 40084370 PMCID: PMC11897627 DOI: 10.62347/fgay1920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/17/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES The advent of immunotherapy has transformed the therapeutic landscape for advanced non-small cell lung cancer (NSCLC); nonetheless, the emergence of resistance to immunotherapy poses a considerable obstacle. Our research sought to identify factors contributing to immunotherapy resistance and to assess the effectiveness of subsequent treatments in patients with advanced NSCLC who have been exposed to immune checkpoint inhibitors (ICIs). METHODS This retrospective study analyzed data from 232 individuals with advanced NSCLC who were treated with ICIs during January 2020 to December 2023. Based on their response to ICIs, these patients were classified into two groups: immunoresistance group (IM group) and non-immunoresistance group (NIM group). Data collected included demographics, clinical parameters, cytokine profiles, tumor mutational burden (TMB), PD-L1 expression, overall survival (OS), progression-free survival (PFS), and adverse events. The association between risk factors and immunoresistance were assessed, and second-line treatment outcomes were evaluated. RESULTS Key risk factors for immunoresistance included lower TMB, higher levels of interleukin-10 (IL-10), and PD-L1 expression ≥ 50%. TMB was inversely correlated with immunoresistance (rho = -0.838, P < 0.001). In multivariate analysis, IL-10 remained a significant risk factor (OR = 33.654, P = 0.021), whereas TMB was protective (OR = 0.786, P < 0.001). Second-line targeted therapy significantly improved OS (8.72 ± 2.02 months) and PFS (5.37 ± 2.15 months) compared to chemotherapy (OS: 7.93 ± 2.13 months; PFS: 4.86 ± 1.68 months) (P < 0.05). The targeted therapy group experienced distinct side effects, notably increased hypertension and hand-foot syndrome, while chemotherapy group had higher rates of fatigue (P < 0.05). CONCLUSION Immunoresistance in advanced NSCLC is influenced by IL-10, TMB, and PD-L1 expression. Targeted therapies offer superior outcomes than chemotherapy, though side effect management remains crucial.
Collapse
Affiliation(s)
- Ping Yang
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| | - Shangxiang He
- Department of Medical Oncology, Shanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghai 200100, China
| | - Linyin Fan
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Ling Ye
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| | - Heng Weng
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| |
Collapse
|
5
|
Bigham A, Serrano-Ruiz M, Caporali M, Fasolino I, Peruzzini M, Ambrosio L, Raucci MG. Black phosphorus-based nanoplatforms for cancer therapy: chemistry, design, biological and therapeutic behaviors. Chem Soc Rev 2025; 54:827-897. [PMID: 39618201 DOI: 10.1039/d4cs00007b] [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: 01/21/2025]
Abstract
Cancer, a significant threat to human lives, has been the target of research for several decades. Although conventional therapies have drawbacks, such as side effects, low efficacy, and weak targeting, they have been applied extensively due to a lack of effective alternatives. The emergence of nanotechnology in medicine has opened up new possibilities and offered promising solutions for cancer therapy. In recent years, 2D nanomaterials have attracted enormous attention in nanomedicine due to their large surface-to-volume ratio, photo-responsivity, excellent electrical conductivity, etc. Among them, black phosphorus (BP) is a 2D nanomaterial consisting of multiple layers weakly bonded together through van der Waals forces. Its distinct structure makes BP suitable for biomedical applications, such as drug/gene carriers, PTT/PDT, and imaging agents. BP has demonstrated remarkable potential since its introduction in cancer therapy in 2015, particularly due to its selective anticancer activity even without the aid of near-infrared (NIR) or anticancer drugs. The present review makes efforts to cover and discuss studies published on the anticancer activity of BP. Based on the type of cancer, the subcategories are organized to shed light on the potential of BP nanosheets and BP quantum dots (BPQDs) against breast, brain, skin, prostate, and bone cancers, and a section is devoted to other cancer types. Since extensive attention has been paid to breast cancer cells and in vivo models, various subsections, including mono-, dual, and triple therapeutic approaches are established for this cancer type. Furthermore, the review outlines various synthesis approaches employed to produce BP nanomaterials, providing insights into key synthesis parameters. This review provides an up-to-date platform for the potential reader to understand what has been done about BP cancer therapy based on each disease, and the conclusions and outlook cover the directions in which this approach is going to proceed in the future.
Collapse
Affiliation(s)
- Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Viale John Fitzgerald Kennedy 54, Mostra d'Oltremare Padiglione 20, 80125 Naples, Italy.
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Manuel Serrano-Ruiz
- Institute for Chemistry of OrganoMetallic Compounds, National Research Council of Italy (ICCOM-CNR), Via Madonna del Piano 10, 5019 Sesto Fiorentino, Italy
| | - Maria Caporali
- Institute for Chemistry of OrganoMetallic Compounds, National Research Council of Italy (ICCOM-CNR), Via Madonna del Piano 10, 5019 Sesto Fiorentino, Italy
| | - Ines Fasolino
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Viale John Fitzgerald Kennedy 54, Mostra d'Oltremare Padiglione 20, 80125 Naples, Italy.
| | - Maurizio Peruzzini
- Institute for Chemistry of OrganoMetallic Compounds, National Research Council of Italy (ICCOM-CNR), Via Madonna del Piano 10, 5019 Sesto Fiorentino, Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Viale John Fitzgerald Kennedy 54, Mostra d'Oltremare Padiglione 20, 80125 Naples, Italy.
| | - Maria Grazia Raucci
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Viale John Fitzgerald Kennedy 54, Mostra d'Oltremare Padiglione 20, 80125 Naples, Italy.
| |
Collapse
|
6
|
Day IL, Tamboline M, Lipshutz GS, Xu S. Recent developments in translational imaging of in vivo gene therapy outcomes. Mol Ther 2024:S1525-0016(24)00849-9. [PMID: 39741403 DOI: 10.1016/j.ymthe.2024.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/18/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Gene therapy achieves therapeutic benefits by delivering genetic materials, packaged within a delivery vehicle, to target cells with defective genes. This approach has shown promise in treating various conditions, including cancer, metabolic disorders, and tissue-degenerative diseases. Over the past 5 years, molecular imaging has increasingly supported gene therapy development in both preclinical and clinical studies. High-quality images from positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and computed tomography (CT) enable quantitative and reliable monitoring of gene therapy. Most reported studies have applied imaging biomarkers to non-invasively evaluate the outcomes of gene therapy. This review aims to inform researchers in molecular imaging and gene therapy about the integration of these two disciplines. We highlight recent developments in using imaging biomarkers to monitor the outcome of in vivo gene therapy, where the therapeutic delivery vehicle is administered systemically. In addition, we discuss prospects for further incorporating imaging biomarkers to support the development and application of gene therapy.
Collapse
Affiliation(s)
- Isabel L Day
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mikayla Tamboline
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
7
|
Pathak S, Singh V, Kumar G N, Jayandharan GR. AAV-mediated combination gene therapy of inducible Caspase 9 and miR-199a-5p is therapeutic in hepatocellular carcinoma. Cancer Gene Ther 2024; 31:1796-1803. [PMID: 39385010 DOI: 10.1038/s41417-024-00844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Advanced-stage hepatocellular carcinoma (HCC) remains an untreatable disease with an overall survival of less than one year. One of the critical molecular mediators contributing to increased resistance to therapy and relapse, is increased hypoxia-inducible factor 1α (HIF-1α) levels, leading to metastasis of tumor cells. Several microRNAs are known to be dysregulated and impact HIF-1α expression in HCC. An in silico analysis demonstrated that hsa-miR-199a-5p is downregulated at various stages of HCC and is known to repress HIF-1α expression. Based on this analysis, we developed a combinatorial suicide gene therapy by employing hepatotropic Adeno-associated virus-based vectors encoding an inducible caspase 9 (iCasp9) and miR-199a. The overexpression of miR-199a-5p alone significantly decreased ( ~ 2-fold vs. Mock treated cells, p < 0.05) HIF-1α mRNA levels, with a concomitant increase in cancer cell cytotoxicity in Huh7 cells in vitro and in xenograft models in vivo. To further enhance the efficacy of gene therapy, we evaluated the synergistic therapeutic effect of AAV8-miR-199a and AAV6-iCasp9 in a xenograft model of HCC. Our data revealed that mice receiving combination suicide gene therapy exhibited reduced expression of HIF-1α ( ~ 4-fold vs. Mock, p < 0.001), with a significant reduction in tumor growth when compared to mock-treated animals. These findings underscore the therapeutic potential of downregulating HIF-1α during suicide gene therapy for HCC.
Collapse
Affiliation(s)
- Subhajit Pathak
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Vijayata Singh
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Narendra Kumar G
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Giridhara R Jayandharan
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India.
| |
Collapse
|
8
|
Arjmand B, Alavi-Moghadam S, Khorsand G, Sarvari M, Arjmand R, Rezaei-Tavirani M, Rajaeinejad M, Mosaed R. Cell-Based Vaccines: Frontiers in Medical Technology for Cancer Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:480-499. [DOI: 10.1007/s40883-024-00338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025]
|
9
|
Artemyev V, Gubaeva A, Paremskaia AI, Dzhioeva AA, Deviatkin A, Feoktistova SG, Mityaeva O, Volchkov PY. Synthetic Promoters in Gene Therapy: Design Approaches, Features and Applications. Cells 2024; 13:1963. [PMID: 39682712 PMCID: PMC11640742 DOI: 10.3390/cells13231963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Gene therapy is a promising approach to the treatment of various inherited diseases, but its development is complicated by a number of limitations of the natural promoters used. The currently used strong ubiquitous natural promoters do not allow for the specificity of expression, while natural tissue-specific promoters have lowactivity. These limitations of natural promoters can be addressed by creating new synthetic promoters that achieve high levels of tissue-specific target gene expression. This review discusses recent advances in the development of synthetic promoters that provide a more precise regulation of gene expression. Approaches to the design of synthetic promoters are reviewed, including manual design and bioinformatic methods using machine learning. Examples of successful applications of synthetic promoters in the therapy of hereditary diseases and cancer are presented, as well as prospects for their clinical use.
Collapse
Affiliation(s)
- Valentin Artemyev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
| | - Anna Gubaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Anastasiia Iu. Paremskaia
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Amina A. Dzhioeva
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
| | - Andrei Deviatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Sofya G. Feoktistova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
| | - Olga Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia;
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
| | - Pavel Yu. Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia; (A.G.); (A.D.); (O.M.); (P.Y.V.)
- Faculty of Fundamental Medicine, Moscow State University, Lomonosovsky Pr., 27, 119991 Moscow, Russia
- Moscow Clinical Scientific Center N.A. A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
10
|
Wang X, Peng I, Peng CA. Eradication of Cancer Cells Using Doxifluridine and Mesenchymal Stem Cells Expressing Thymidine Phosphorylase. Bioengineering (Basel) 2024; 11:1194. [PMID: 39768012 PMCID: PMC11726915 DOI: 10.3390/bioengineering11121194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been developed over several decades as a targeted cancer treatment aimed at minimizing toxicity to healthy cells. This approach involves three key components: a non-toxic prodrug, a gene encoding an enzyme that converts the prodrug into an active chemotherapy drug, and a gene carrier to target cancer cells. In this study, the prodrug doxifluridine was enzymatically converted into the chemotherapy drug 5-fluorouracil via thymidine phosphorylase, using human mesenchymal stem cells (hMSCs) as delivery vehicles. The hMSCs were first transduced with thymidine phosphorylase-encoded lentiviral vectors produced by HEK293T cells, then co-cultured with A549 adenocarcinoma cells in the presence of doxifluridine. The results showed that after 3 days of prodrug treatment, cell viability in both A549 cancer cells and hMSCs dropped by about 50%, and by day 5, viability had decreased to 10%. In summary, exogenous thymidine phosphorylase expressed in hMSCs successfully converted the non-toxic prodrug doxifluridine into the chemotherapy agent 5-fluorouracil, effectively eliminating both cancer cells and hMSCs within a short period.
Collapse
Affiliation(s)
- Xutu Wang
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, USA
| | - Ian Peng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ching-An Peng
- Department of Chemical and Biological Engineering, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
11
|
Thapa R, Bhat AA, Gupta G, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Prasad GVS, Pramanik A, Ali H. CRBN-PROTACs in Cancer Therapy: From Mechanistic Insights to Clinical Applications. Chem Biol Drug Des 2024; 104:e70009. [PMID: 39496477 DOI: 10.1111/cbdd.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024]
Abstract
Cereblon (CRBN), a member of the E3 ubiquitin ligase complex, has gained significant attention as a therapeutic target in cancer. CRBN regulates the degradation of various proteins in cancer progression, including transcription factors and signaling molecules. PROTACs (proteolysis-targeting chimeras) are a novel approach that uses the cell's degradation system to remove disease-causing proteins selectively. CRBN-dependent PROTACs work by tagging harmful proteins for destruction through the ubiquitin-proteasome system. This strategy offers several advantages over traditional protein inhibition methods, including the potential to overcome drug resistance. Recent progress in developing CRBN-based PROTACs has shown promising preclinical results in both hematologic malignancies and solid tumors. Additionally, CRBN-based PROTACs have enhanced our understanding of CRBN's role in cancer, potentially serving as biomarkers for patient stratification and predicting therapeutic responses. In this review, we delineate the mechanisms of action for CRBN-dependent PROTACs (CRBN-PROTACs), summarize recent advances in preclinical and clinical applications, and provide our perspective on future development.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, UAE
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
12
|
Hoch CC, Hachani K, Han Y, Schmidl B, Wirth M, Multhoff G, Bashiri Dezfouli A, Wollenberg B. The future of interleukin gene therapy in head and neck cancers. Expert Opin Biol Ther 2024; 24:1057-1073. [PMID: 39291462 DOI: 10.1080/14712598.2024.2405568] [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: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Head and neck cancer (HNC), primarily head and neck squamous cell carcinomas, originates from the squamous epithelium in areas like the oral cavity, lip, larynx, and oropharynx. With high morbidity impacting critical functions, combined treatments like surgery, radiation, and chemotherapy often fall short in advanced stages, highlighting the need for innovative therapies. AREAS COVERED This review critically evaluates interleukin (IL) gene therapy for treating HNC. The discussion extends to key ILs in HNC, various gene therapy techniques and delivery methods. We particularly focus on the application of IL-2, IL-12, and IL-24 gene therapies, examining their mechanisms and outcomes in preclinical studies and clinical trials. The final sections address IL gene therapy challenges in HNC, exploring solutions and critically assessing future therapeutic directions. EXPERT OPINION Despite advancements in genomic and immunotherapy, significant challenges in HNC treatment persist, primarily due to the immunosuppressive nature of the tumor microenvironment and the adverse effects of current therapies. The therapeutic efficacy of IL gene therapy hinges on overcoming these hurdles through refined delivery methods that ensure targeted, tumor-specific gene expression. Future strategies should focus on refining gene delivery methods and combining IL gene therapy with other treatments to optimize efficacy and minimize toxicity.
Collapse
Affiliation(s)
- Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Yu Han
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Wirth
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Technical University of Munich (TranslaTUM), Munich, Germany
- Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
13
|
Fujimoto GR, Wooley DP, Byers KB, Yang OO, Behrman AJ, Winters TH, Hudson TW. Update on Managing the Risks of Exposure to Lentiviral and Retroviral Vectors. J Occup Environ Med 2024; 66:818-825. [PMID: 38913827 DOI: 10.1097/jom.0000000000003166] [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: 06/26/2024]
Abstract
OBJECTIVE This paper aims to review the risks associated with using lentiviral and retroviral vectors in research and clinical settings and to propose an update to an effective treatment plan. METHODS Risks of exposure were evaluated based on vector design, safety features, viral tropism, transgene, and means and modes of transmission. These risks were weighed against the potential risks and benefits of current HIV medications. RESULTS We recommend the following postexposure prophylactic treatment for significant lentiviral vector exposures: 1) dolutegravir 50 mg taken once a day for 7 days and 2) tenofovir disoproxil fumarate 300 mg taken once a day for 7 days (28 days of both medications for replication-competent vectors). CONCLUSIONS Because of the highly efficient delivery of transgenes by modern lentiviral and retroviral vectors, postexposure prophylaxis is indicated to prevent vector integration and oncogenic risks.
Collapse
Affiliation(s)
- Gary R Fujimoto
- From the Occupational and Internal Medicine Consultant, Los Altos, California (G.R.F.); Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, Ohio (D.P.W.); Dana-Farber Cancer Institute, Boston, Massachusetts (K.B.B.); Division of Infectious Diseases, Department of Medicine, and Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California (O.O.Y.); Division of Occupational Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.B.); Occupational Health Programs at the Boston Children's Hospital and Dana Farber Cancer Institute, Research Occupational Health Program and the National Emerging Infectious Disease Laboratories, Boston University, and the Harvard T.H. Chan School of Public Health, Boston, Massachusetts (T.H.W.); and Department of Occupational and Environmental Health, University of California Irvine, Irvine, California (T.W.H.)
| | | | | | | | | | | | | |
Collapse
|
14
|
Da Silva RCDS, Simon NDA, Dos Santos AA, Olegário GDM, Da Silva JF, Sousa NO, Corbacho MAT, de Melo FF. Personalized medicine: Clinical oncology on molecular view of treatment. World J Clin Oncol 2024; 15:992-1001. [PMID: 39193152 PMCID: PMC11346063 DOI: 10.5306/wjco.v15.i8.992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 07/10/2024] [Indexed: 08/16/2024] Open
Abstract
Cancer, the second leading global cause of death, impacts both physically and emotionally. Conventional treatments such as surgeries, chemotherapy, and radiotherapy have adverse effects, driving the need for more precise approaches. Precision medicine enables more targeted treatments. Genetic mapping, alongside other molecular biology approaches, identifies specific genes, contributing to accurate prognoses. The review addresses, in clinical use, a molecular perspective on treatment. Biomarkers like alpha-fetoprotein, beta-human chorionic gonadotropin, 5-hydroxyindoleacetic acid, programmed death-1, and cytotoxic T lymphocyte-associated protein 4 are explored, providing valuable information. Bioinformatics, with an emphasis on artificial intelligence, revolutionizes the analysis of biological data, offering more accurate diagnoses. Techniques like liquid biopsy are emphasized for early detection. Precision medicine guides therapeutic strategies based on the molecular characteristics of the tumor, as evidenced in the molecular subtypes of breast cancer. Classifications allow personalized treatments, highlighting the role of trastuzumab and endocrine therapies. Despite the benefits, challenges persist, including high costs, tumor heterogeneity, and ethical issues. Overcoming obstacles requires collaboration, ensuring that advances in molecular biology translate into accessible benefits for all.
Collapse
Affiliation(s)
| | - Nathalia de Andrade Simon
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| | - André Alves Dos Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| | - Gabriel De Melo Olegário
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| | - Jayne Ferreira Da Silva
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| | - Naide Oliveira Sousa
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| | | | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória Da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
15
|
Pote MS, Singh D, M. A A, Suchita J, Gacche RN. Cancer metastases: Tailoring the targets. Heliyon 2024; 10:e35369. [PMID: 39170575 PMCID: PMC11336595 DOI: 10.1016/j.heliyon.2024.e35369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Metastasis is an intricate and formidable pathophysiological process encompassing the dissemination of cancer cells from the primary tumour body to distant organs. It stands as a profound and devastating phenomenon that constitutes the primary driver of cancer-related mortality. Despite great strides of advancements in cancer research and treatment, tailored anti-metastasis therapies are either lacking or have shown limited success, necessitating a deeper understanding of the intrinsic elements driving cancer invasiveness. This comprehensive review presents a contemporary elucidation of pivotal facets within the realm of cancer metastasis, commencing with the intricate processes of homing and invasion. The process of angiogenesis, which supports tumour growth and metastasis, is addressed, along with the pre-metastatic niche, wherein the primary tumour prepares for a favorable microenvironment at distant sites for subsequent metastatic colonization. The landscape of metastasis-related genetic and epigenetic mechanisms, involvement of metastasis genes and metastasis suppressor genes, and microRNAs (miRNA) are also discussed. Furthermore, immune modulators' impact on metastasis and their potential as therapeutic targets are addressed. The interplay between cancer cells and the immune system, including immune evasion mechanisms employed by metastatic cells, is discussed, highlighting the importance of targeting immune modulation in arresting metastatic progression. Finally, this review presents promising treatment opportunities derived from the insights gained into the mechanisms of metastasis. Identifying novel therapeutic targets and developing innovative strategies to disrupt the metastatic cascade holds excellent potential for improving patient outcomes and ultimately reducing cancer-related mortality.
Collapse
Affiliation(s)
| | | | | | | | - Rajesh N. Gacche
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
16
|
Alyafeai E, Qaed E, Al-Mashriqi HS, Almaamari A, Almansory AH, Futini FA, Sultan M, Tang Z. Molecular dynamics of DNA repair and carcinogen interaction: Implications for cancer initiation, progression, and therapeutic strategies. Mutat Res 2024; 829:111883. [PMID: 39265237 DOI: 10.1016/j.mrfmmm.2024.111883] [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: 05/01/2024] [Revised: 07/07/2024] [Accepted: 09/05/2024] [Indexed: 09/14/2024]
Abstract
The integrity of the genetic material in human cells is continuously challenged by environmental agents and endogenous stresses. Among these, environmental carcinogens are pivotal in initiating complex DNA lesions that can lead to malignant transformations if not properly repaired. This review synthesizes current knowledge on the molecular dynamics of DNA repair mechanisms and their interplay with various environmental carcinogens, providing a comprehensive overview of how these interactions contribute to cancer initiation and progression. We examine key DNA repair pathways including base excision repair, nucleotide excision repair, and double-strand break repair and their regulatory networks, highlighting how defects in these pathways can exacerbate carcinogen-induced damage. Further, we discuss how understanding these molecular interactions offers novel insights into potential therapeutic strategies. This includes leveraging synthetic lethality concepts and designing targeted therapies that exploit specific DNA repair vulnerabilities in cancer cells. By integrating recent advances in molecular biology, genetics, and oncology, this review aims to illuminate the complex landscape of DNA repair and carcinogen-induced carcinogenesis, setting the stage for future research and therapeutic innovations.
Collapse
Affiliation(s)
- Eman Alyafeai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Eskandar Qaed
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, Dalian 116044, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | | | - Ahmed Almaamari
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Anisa H Almansory
- Biological department, Faculty of Science, University of Sana'a, Yemen
| | - Fatima Al Futini
- Department of Food Science, Faculty of Food Science & Technology, University Putra Malaysia (UPM), Malaysia
| | - Marwa Sultan
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Zeyao Tang
- Collage of Pharmacy, Department of Pharmacology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
17
|
Lee HJ, Choi HJ, Jeong YJ, Na YH, Hong JT, Han JM, Hoe HS, Lim KH. Developing theragnostics for Alzheimer's disease: Insights from cancer treatment. Int J Biol Macromol 2024; 269:131925. [PMID: 38685540 DOI: 10.1016/j.ijbiomac.2024.131925] [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: 01/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The prevalence of Alzheimer's disease (AD) and its associated economic and societal burdens are on the rise, but there are no curative treatments for AD. Interestingly, this neurodegenerative disease shares several biological and pathophysiological features with cancer, including cell-cycle dysregulation, angiogenesis, mitochondrial dysfunction, protein misfolding, and DNA damage. However, the genetic factors contributing to the overlap in biological processes between cancer and AD have not been actively studied. In this review, we discuss the shared biological features of cancer and AD, the molecular targets of anticancer drugs, and therapeutic approaches. First, we outline the common biological features of cancer and AD. Second, we describe several anticancer drugs, their molecular targets, and their effects on AD pathology. Finally, we discuss how protein-protein interactions (PPIs), receptor inhibition, immunotherapy, and gene therapy can be exploited for the cure and management of both cancer and AD. Collectively, this review provides insights for the development of AD theragnostics based on cancer drugs and molecular targets.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Hee-Jeong Choi
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Yoo Joo Jeong
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yoon-Hee Na
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Ji Min Han
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| | - Hyang-Sook Hoe
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| |
Collapse
|
18
|
Khristiani Rahayu A, Fibriani A, Irasonia Tan M. Exploring the potential of black cumin derived nanovesicles for miRNA drug delivery. Eur J Pharm Biopharm 2024; 199:114275. [PMID: 38582178 DOI: 10.1016/j.ejpb.2024.114275] [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: 01/06/2024] [Revised: 03/14/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Liposomes is a non-viral vector drug delivery system. Nevertheless, the existing commercial liposomes are quite expensive and not always affordable, particularly in developing countries. To address this challenge, plant-derived nanoparticles offer a cost-effective alternative while maintaining similar drug delivery capabilities. Hence, this study aimed to explore the potential of nanovesicles derived from black cumin (Nigella sativa) as a miRNA delivery system. Gradient sucrose-centrifugation was utilized to separate the nanovesicles derived from black cumin. Subsequently, these isolated nanovesicles, originating from black cumin, underwent centrifugation at a speed of 11,000 rpm. The miRNAs were encapsulated within these nanovesicles through the ethanol injection method. Morphological examinations of the nanovesicles derived from black cumin and DOTAP, as the positive control, were conducted using TEM and SEM. Furthermore, the cytotoxicity of the nanovesicles derived from black cumin was evaluated through the MTT assay on the MCF-7 cell line. Lastly, the process of internalization for both the black cumin-derived nanovesicles and DOTAP was visualized using a confocal microscope. Results demonstrated the successful isolation of nanovesicles from black cumin using the sucrose gradient method. These particles exhibited a spherical shape with diameters ranging from 100 nm to 200 nm, featuring a negative surface charge. When MCF-7 cells were exposed to black cumin-derived nanovesicles at a concentration of 12 mg/mL, cell viability reached 89.8 %, showing no significant difference compared to the positive control (p > 0.05). Furthermore, the MCF-7 cell line effectively internalized the black cumin-derived nanovesicles after a 45-minute incubation period. Notably, the encapsulation of miRNA within these nanovesicles demonstrated an impressive entrapment efficiency of 76.4 %. Subsequent transfection of miRNA-loaded black cumin-derived nanovesicles resulted in a substantial inhibition of MCF-7 cell viability, reducing it to 67 % after 48 h of treatment. These findings underscore the potential of black cumin-derived nanovesicles as potential nanovectors for the encapsulation and delivery of miRNA within drug delivery systems, offering a cost-effective and accessible solution for advanced drug delivery technologies, particularly in developing country.
Collapse
Affiliation(s)
- Adelina Khristiani Rahayu
- Doctoral Program of Biology, School of Life Sciences and Technology, Institut Teknologi Bandung, Jl. Ganesha No. 10, Bandung, Jawa Barat, Indonesia
| | - Azzania Fibriani
- School of Life Sciences and Technology, Jl. Ganesha No. 10, Bandung, Institut Teknologi Bandung, Indonesia
| | - Marselina Irasonia Tan
- School of Life Sciences and Technology, Jl. Ganesha No. 10, Bandung, Institut Teknologi Bandung, Indonesia.
| |
Collapse
|
19
|
Reddy AJ, Herschl MH, Geng X, Kolli S, Lu AX, Kumar A, Hsu PD, Levine S, Ioannidis NM. Strategies for effectively modelling promoter-driven gene expression using transfer learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.24.529941. [PMID: 36909524 PMCID: PMC10002662 DOI: 10.1101/2023.02.24.529941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The ability to deliver genetic cargo to human cells is enabling rapid progress in molecular medicine, but designing this cargo for precise expression in specific cell types is a major challenge. Expression is driven by regulatory DNA sequences within short synthetic promoters, but relatively few of these promoters are cell-type-specific. The ability to design cell-type-specific promoters using model-based optimization would be impactful for research and therapeutic applications. However, models of expression from short synthetic promoters (promoter-driven expression) are lacking for most cell types due to insufficient training data in those cell types. Although there are many large datasets of both endogenous expression and promoter-driven expression in other cell types, which provide information that could be used for transfer learning, transfer strategies remain largely unexplored for predicting promoter-driven expression. Here, we propose a variety of pretraining tasks, transfer strategies, and model architectures for modelling promoter-driven expression. To thoroughly evaluate various methods, we propose two benchmarks that reflect data-constrained and large dataset settings. In the data-constrained setting, we find that pretraining followed by transfer learning is highly effective, improving performance by 24-27%. In the large dataset setting, transfer learning leads to more modest gains, improving performance by up to 2%. We also propose the best architecture to model promoter-driven expression when training from scratch. The methods we identify are broadly applicable for modelling promoter-driven expression in understudied cell types, and our findings will guide the choice of models that are best suited to designing promoters for gene delivery applications using model-based optimization. Our code and data are available at https://github.com/anikethjr/promoter_models.
Collapse
|
20
|
Maturana CJ. Engineered compact pan-neuronal promoter from Alphaherpesvirus LAP2 enhances target gene expression in the mouse brain and reduces tropism in the liver. Gene Ther 2024; 31:335-344. [PMID: 38012300 PMCID: PMC11090813 DOI: 10.1038/s41434-023-00430-0] [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: 07/09/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Small promoters capable of driving potent neuron-restricted gene expression are required to support successful brain circuitry and clinical gene therapy studies. However, converting large promoters into functional MiniPromoters, which can be used in vectors with limited capacity, remains challenging. In this study, we describe the generation of a novel version of alphaherpesvirus latency-associated promoter 2 (LAP2), which facilitates precise transgene expression exclusively in the neurons of the mouse brain while minimizing undesired targeting in peripheral tissues. Additionally, we aimed to create a compact neural promoter to facilitate packaging of larger transgenes. Our results revealed that MiniLAP2 (278 bp) drives potent transgene expression in all neurons in the mouse brain, with little to no expression in glial cells. In contrast to the native promoter, MiniLAP2 reduced tropism in the spinal cord and liver. No expression was detected in the kidney or skeletal muscle. In summary, we developed a minimal pan-neuronal promoter that drives specific and robust transgene expression in the mouse brain when delivered intravenously via AAV-PHP.eB vector. The use of this novel MiniPromoter may broaden the range of deliverable therapeutics and improve their safety and efficacy by minimizing the potential for off-target effects.
Collapse
Affiliation(s)
- Carola J Maturana
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
21
|
Chao G, Zukin S, Fortuna PRJ, Boettner B, Church GM. Progress and limitations in engineering cellular adhesion for research and therapeutics. Trends Cell Biol 2024; 34:277-287. [PMID: 37580241 DOI: 10.1016/j.tcb.2023.07.007] [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: 05/03/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/16/2023]
Abstract
Intercellular interactions form the cornerstone of multicellular biology. Despite advances in protein engineering, researchers artificially directing physical cell interactions still rely on endogenous cell adhesion molecules (CAMs) alongside off-target interactions and unintended signaling. Recently, methods for directing cellular interactions have been developed utilizing programmable domains such as coiled coils (CCs), nanobody-antigen, and single-stranded DNA (ssDNA). We first discuss desirable molecular- and systems-level properties in engineered CAMs, using the helixCAM platform as a benchmark. Next, we propose applications for engineered CAMs in immunology, developmental biology, tissue engineering, and neuroscience. Biologists in various fields can readily adapt current engineered CAMs to establish control over cell interactions, and their utilization in basic and translational research will incentivize further expansion in engineered CAM capabilities.
Collapse
Affiliation(s)
- George Chao
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
| | - Stefan Zukin
- Wyss Institute, Harvard Medical School, Boston, MA, USA
| | | | | | - George M Church
- Department of Genetics, Harvard Medical School, Boston, MA, USA; Wyss Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Guan X, Pei Y, Song J. DNA-Based Nonviral Gene Therapy─Challenging but Promising. Mol Pharm 2024; 21:427-453. [PMID: 38198640 DOI: 10.1021/acs.molpharmaceut.3c00907] [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] [Indexed: 01/12/2024]
Abstract
Over the past decades, significant progress has been made in utilizing nucleic acids, including DNA and RNA molecules, for therapeutic purposes. For DNA molecules, although various DNA delivery systems have been established, viral vector systems are the go-to choice for large-scale commercial applications. However, viral systems have certain disadvantages such as immune response, limited payload capacity, insertional mutagenesis and pre-existing immunity. In contrast, nonviral systems are less immunogenic, not size limited, safer, and easier for manufacturing compared with viral systems. What's more, nonviral DNA vectors have demonstrated their capacity to mediate specific protein expression in vivo for diverse therapeutic objectives containing a wide range of diseases such as cancer, rare diseases, neurodegenerative diseases, and infectious diseases, yielding promising therapeutic outcomes. However, exogenous plasmid DNA is prone to degrade and has poor immunogenicity in vivo. Thus, various strategies have been developed: (i) designing novel plasmids with special structures, (ii) optimizing plasmid sequences for higher expression, and (iii) developing more efficient nonviral DNA delivery systems. Based on these strategies, many interesting clinical results have been reported. This Review discusses the development of DNA-based nonviral gene therapy, including novel plasmids, nonviral delivery systems, clinical advances, and prospects. These developments hold great potential for enhancing the efficacy and safety of nonviral gene therapy and expanding its applications in the treatment of various diseases.
Collapse
Affiliation(s)
- Xiaocai Guan
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yufeng Pei
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jie Song
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
23
|
Tavakoli F, Ghavimi MA, Fakhrzadeh V, Abdolzadeh D, Afshari A, Eslami H. Evaluation of salivary transferrin in patients with oral squamous cell carcinoma. Clin Exp Dent Res 2024; 10:e809. [PMID: 37964689 PMCID: PMC10860556 DOI: 10.1002/cre2.809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
OBJECTIVES About 94% of oral cancers are squamous cell carcinomas (OSCCs). Its occurrence is age-related due to some factors. Salivary biomarkers have good susceptibility to OSCC's early diagnosis. Moreover, since the clinical diagnosis of advanced stages of OSCC is feasible, its prognosis is very poor. MATERIAL AND METHODS According to inclusion and exclusion criteria, 40 OSCC patients and 40 healthy people were selected, and 5 mL of saliva were prepared from each person. The quantity of saline transferrin was computed. After that, the data were analyzed. RESULTS Our study results demonstrated that the mean and standard deviation of the salivary transferrin in the control group were 1.234 mL and 0.374, respectively, and in the case group, it was equal to 2.512 mL for the mean and 0.463 for the standard deviation. There was a statistically substantial difference between the mean of the salivary transferrin variable in the two study groups. CONCLUSION In conclusion, the mean concentration of salivary transferrin in the case group was higher than in the control group.
Collapse
Affiliation(s)
- Fatemeh Tavakoli
- Department of Oral and Maxillofacial Medicine, School of DentistryShiraz University of Medical SciencesShirazIran
| | - Mohammad Ali Ghavimi
- Department, of Oral and Maxillofacial Surgery, School of DentistryTabriz University of Medical SciencesTabrizIran
| | - Vahid Fakhrzadeh
- Department of Prosthodontics, School of DentistryTabriz University of Medical SciencesTabrizIran
| | - Dorna Abdolzadeh
- School of DentistryTabriz University of Medical SciencesTabrizIran
| | - Aylar Afshari
- School of DentistryShiraz University of Medical SciencesShirazIran
| | - Hosein Eslami
- Department of Oral and Maxillofacial Medicine, School of DentistryTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
24
|
Batool A, Rashid W, Fatima K, Khan SU. Mechanisms of Cancer Resistance to Various Therapies. DRUG RESISTANCE IN CANCER: MECHANISMS AND STRATEGIES 2024:31-75. [DOI: 10.1007/978-981-97-1666-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Pathak S, Singh V, Kumar N, Jayandharan GR. Inducible caspase 9-mediated suicide gene therapy using AAV6 vectors in a murine model of breast cancer. Mol Ther Methods Clin Dev 2023; 31:101166. [PMID: 38149057 PMCID: PMC10750187 DOI: 10.1016/j.omtm.2023.101166] [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: 07/21/2023] [Accepted: 11/20/2023] [Indexed: 12/28/2023]
Abstract
Breast carcinoma has one of the highest incidence rates (11.7%), with significant clinical heterogeneity. Although conventional chemotherapy and surgical resection are the current standard of care, the resistance and recurrence, after these interventions, necessitate alternate therapeutic approaches. Cancer gene therapy for breast cancer with the suicide gene is an attractive option due to their directed delivery into the tumor. In this study, we have developed a novel treatment strategy against breast cancer with recombinant adeno-associated virus (AAV) serotype 6 vectors carrying a suicide gene, inducible Caspase 9 (iCasp9). Upon treatment with AAV6-iCasp9 vectors and the chemical inducer of dimerizer, AP20187, the viability of murine breast cancer cells (4T1) was significantly reduced to ∼40%-60% (mock control 100%). Following intratumoral delivery of AAV6-iCasp9 vectors in an orthotopic breast cancer mouse model, we observed a significant increase in iCasp9 transgene expression and a significant reduction in tumor growth rate. At the molecular level, immunohistochemical analysis demonstrated subsequent activation of the effector caspase 3 and cellular death. These data highlight the potential of AAV6-iCasp9-based suicide gene therapy for aggressive breast cancer in patients.
Collapse
Affiliation(s)
- Subhajit Pathak
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Vijayata Singh
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Narendra Kumar
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Giridhara R. Jayandharan
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
26
|
Hu D, Tian Y, Xu J, Xie D, Wang Y, Liu M, Wang Y, Yang L. Oncolytic viral therapy as promising immunotherapy against glioma. MEDCOMM – FUTURE MEDICINE 2023; 2. [DOI: 10.1002/mef2.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/22/2023] [Indexed: 03/19/2025]
Abstract
AbstractGlioma is a common primary central nervous system malignant tumor in clinical, traditional methods such as surgery and chemoradiotherapy are not effective in treatment. Therefore, more effective treatments need to be found. Oncolytic viruses (OVs) are a new type of immunotherapy that selectively infects and kills tumor cells instead of normal cells. OVs can mediate antitumor immune responses through a variety of mechanisms, and have the ability to activate antitumor immune responses, transform the tumor microenvironment from “cold” to “hot,” and enhance the efficacy of immune checkpoint inhibitors. Recently, a large number of preclinical and clinical studies have shown that OVs show great prospects in the treatment of gliomas. In this review, we summarize the current status of glioma therapies with a focus on OVs. First, this article introduces the current status of treatment of glioma and their respective shortcomings. Then, the important progress of OVs of in clinical trials of glioma is summarized. Finally, the urgent challenges of oncolytic virus treatment for glioma are sorted out, and related solutions are proposed. This review will help to further promote the use of OVs in the treatment of glioma.
Collapse
Affiliation(s)
- Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yaomei Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- College of Bioengineering Sichuan University of Science & Engineering Zigong China
| | - Jie Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Daoyuan Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yuanda Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| |
Collapse
|
27
|
Wang Z, Sun P, Li Z, Xiao S. Clinical Advances and Future Directions of Oncolytic Virotherapy for Head and Neck Cancer. Cancers (Basel) 2023; 15:5291. [PMID: 37958464 PMCID: PMC10650136 DOI: 10.3390/cancers15215291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Oncolytic viruses (OVs), without harming normal tissues, selectively infect and replicate within tumor cells, to release immune molecules and tumor antigens, achieving immune-mediated destruction of tumors and making them one of the most promising immunotherapies for cancer. Many clinical studies have demonstrated that OVs can provide clinical benefits for patients with different types of tumors, at various stages, including metastatic and previously untreatable cases. When OVs are used in combination with chemotherapy, radiotherapy, immunotherapy, and other treatments, they can synergistically enhance the therapeutic effects. The concept of oncolytic virotherapy (OVT) was proposed in the early 20th century. With advancements in genetic engineering, genetically modified viruses can further enhance the efficacy of cancer immunotherapy. In recent years, global research on OV treatment of malignant tumors has increased dramatically. This article comprehensively reviews the findings from relevant research and clinical trials, providing an overview of the development of OVT and its application in the clinical treatment of head and neck cancer. The aim is to offer insights for future clinical and fundamental research on OVT.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Stomatology, Wenzhou Medical University Renji College, Wenzhou 325000, China
| | - Peng Sun
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China; (P.S.); (Z.L.)
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315000, China
| | - Zhiyong Li
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325000, China; (P.S.); (Z.L.)
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315000, China
| | - Shaowen Xiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
28
|
Choi H, Kang M, Lee KH, Kim YS. Elevated level of PLRG1 is critical for the proliferation and maintenance of genome stability of tumor cells. BMB Rep 2023; 56:612-617. [PMID: 37817442 PMCID: PMC10689083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
Pleiotropic regulator 1 (PLRG1), a highly conserved element in the spliceosome, can form a NineTeen Complex (NTC) with Prp19, SPF27, and CDC5L. This complex plays crucial roles in both pre-mRNA splicing and DNA repair processes. Here, we provide evidence that PLRG1 has a multifaceted impact on cancer cell proliferation. Comparing its expression levels in cancer and normal cells, we observed that PLRG1 was upregulated in various tumor tissues and cell lines. Knockdown of PLRG1 resulted in tumor-specific cell death. Depletion of PLRG1 had notable effects, including mitotic arrest, microtubule instability, endoplasmic reticulum (ER) stress, and accumulation of autophagy, ultimately culminating in apoptosis. Our results also demonstrated that PLRG1 downregulation contributed to DNA damage in cancer cells, which we confirmed through experimental validation as DNA repair impairment. Interestingly, when PLRG1 was decreased in normal cells, it induced G1 arrest as a self-protective mechanism, distinguishing it from effects observed in cancer cells. These results highlight multifaceted impacts of PLRG1 in cancer and underscore its potential as a novel anti-cancer strategy by selectively targeting cancer cells. [BMB Reports 2023; 56(11): 612-617].
Collapse
Affiliation(s)
- Hyunji Choi
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
| | | | - Kee-Ho Lee
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Yeon-Soo Kim
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
- R&D Center, artiCure Inc., Daejeon 34134, Korea
| |
Collapse
|
29
|
Wang W, Jiang S, Zhao Y, Zhu G. Echinacoside: A promising active natural products and pharmacological agents. Pharmacol Res 2023; 197:106951. [PMID: 37804927 DOI: 10.1016/j.phrs.2023.106951] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Echinacoside, a natural phenylethanoid glycoside, was discovered and isolated from the garden plant Echinacea angustifolia DC., belonging to the Compositae family, approximately sixty years ago. Extensive investigations have revealed that it possesses a wide array of pharmacologically beneficial activities for human health, particularly notable for its neuroprotective and anticancer activity. Several crucial concerns surfaced, encompassing the recognition of active metabolites that exhibited inadequate bioavailability in their prototype form, the establishment of precise molecular signal pathways or targets associated with the aforementioned effects of echinacoside, and the scarcity of dependable clinical trials. Hence, the question remains unanswered as to whether scientific research can effectively utilize this natural compound. To support future studies on this natural product, it is imperative to provide a systematic overview and insights into potential future prospects. The current review provides a comprehensive analysis of the existing knowledge on echinacoside, encompassing its wide distribution, structural diversity and metabolism, diverse therapeutic applications, and improvement of echinacoside bioavailability for its potential utilization.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujun Jiang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
30
|
Khan R, Oskouian B, Lee JY, Hodgin JB, Yang Y, Tassew G, Saba JD. AAV-SPL 2.0, a Modified Adeno-Associated Virus Gene Therapy Agent for the Treatment of Sphingosine Phosphate Lyase Insufficiency Syndrome. Int J Mol Sci 2023; 24:15560. [PMID: 37958544 PMCID: PMC10648410 DOI: 10.3390/ijms242115560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Sphingosine-1-phosphate lyase insufficiency syndrome (SPLIS) is an inborn error of metabolism caused by inactivating mutations in SGPL1, the gene encoding sphingosine-1-phosphate lyase (SPL), an essential enzyme needed to degrade sphingolipids. SPLIS features include glomerulosclerosis, adrenal insufficiency, neurological defects, ichthyosis, and immune deficiency. Currently, there is no cure for SPLIS, and severely affected patients often die in the first years of life. We reported that adeno-associated virus (AAV) 9-mediated SGPL1 gene therapy (AAV-SPL) given to newborn Sgpl1 knockout mice that model SPLIS and die in the first few weeks of life prolonged their survival to 4.5 months and prevented or delayed the onset of SPLIS phenotypes. In this study, we tested the efficacy of a modified AAV-SPL, which we call AAV-SPL 2.0, in which the original cytomegalovirus (CMV) promoter driving the transgene is replaced with the synthetic "CAG" promoter used in several clinically approved gene therapy agents. AAV-SPL 2.0 infection of human embryonic kidney (HEK) cells led to 30% higher SPL expression and enzyme activity compared to AAV-SPL. Newborn Sgpl1 knockout mice receiving AAV-SPL 2.0 survived ≥ 5 months and showed normal neurodevelopment, 85% of normal weight gain over the first four months, and delayed onset of proteinuria. Over time, treated mice developed nephrosis and glomerulosclerosis, which likely resulted in their demise. Our overall findings show that AAV-SPL 2.0 performs equal to or better than AAV-SPL. However, improved kidney targeting may be necessary to achieve maximally optimized gene therapy as a potentially lifesaving SPLIS treatment.
Collapse
Affiliation(s)
- Ranjha Khan
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA
| | - Babak Oskouian
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA
| | - Joanna Y Lee
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA
| | - Jeffrey B Hodgin
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yingbao Yang
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Gizachew Tassew
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA
| | - Julie D Saba
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
31
|
Supuramanian SS, Dsa S, Harihar S. Molecular interaction of metastasis suppressor genes and tumor microenvironment in breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:912-932. [PMID: 37970212 PMCID: PMC10645471 DOI: 10.37349/etat.2023.00173] [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: 02/27/2023] [Accepted: 08/03/2023] [Indexed: 11/17/2023] Open
Abstract
Breast cancer (BC) is a leading cause of cancer-related deaths in women worldwide where the process of metastasis is a major contributor to the mortality associated with this disease. Metastasis suppressor genes are a group of genes that play a crucial role in preventing or inhibiting the spread of cancer cells. They suppress the metastasis process by inhibiting colonization and by inducing dormancy. These genes function by regulating various cellular processes in the tumor microenvironment (TME), such as cell adhesion, invasion, migration, and angiogenesis. Dysregulation of metastasis suppressor genes can lead to the acquisition of an invasive and metastatic phenotype and lead to poor prognostic outcomes. The components of the TME generally play a necessary in the metastasis progression of tumor cells. This review has identified and elaborated on the role of a few metastatic suppressors associated with the TME that have been shown to inhibit metastasis in BC by different mechanisms, such as blocking certain cell signaling molecules involved in cancer cell migration, invasion, enhancing immune surveillance of cancer cells, and promoting the formation of a protective extracellular matrix (ECM). Understanding the interaction of metastatic suppressor genes and the components of TME has important implications for the development of novel therapeutic strategies to target the metastatic cascade. Targeting these genes or their downstream signaling pathways offers a promising approach to inhibiting the spread of cancer cells and improves patient outcomes.
Collapse
Affiliation(s)
| | - Sid Dsa
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| |
Collapse
|
32
|
Yuan K, Zhang Y, Yu Y, Xu Y, Xian S. Anchoring Filament Protein Ladinin-1 is an Immunosuppressive Microenvironment and Cold Tumor Correlated Prognosticator in Lung Adenocarcinoma. Biochem Genet 2023; 61:2173-2202. [PMID: 37005975 DOI: 10.1007/s10528-023-10370-4] [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: 02/13/2023] [Accepted: 03/19/2023] [Indexed: 04/04/2023]
Abstract
Anchoring filament protein ladinin-1 (LAD1) codes for an anchor filament protein in the basement membrane. Here, we have aimed to determine its potential role in LUAD. According to the comprehensive analyses conducted in this study, we studied the expression, prognostic significance, function, methylation, copy number variations, and the immune cell infiltration of LAD1 in LUAD. A higher level of LAD1 gene expression was observed in the LUAD tumor tissues compared to the normal lung tissues (p < 0.001). Furthermore, the multivariate analysis indicated that a higher LAD1 gene expression level was the independent prognostic factor. Additionally, the DNA methylation level of the LAD1 was inversely linked to its expression (p < 0.001). We noted that the patients affected due to LAD1 hypomethylation showed a very low overall survival rate compared to the patients with a higher LAD1 methylation score (p < 0.05). Moreover, the results of the immunity analysis indicated that the LAD1 expression might be inversely linked to the immune cell infiltration degree, expression of the infiltrated immune cells, and the PD-L1 levels. Lastly, we supplemented some verification to increase the rigor of the study. The results suggested that high expression of LAD1 may be related to cold tumors. Hence, this indirectly reflects that the immunotherapy effect of LUAD patients with high LAD1 expression might be worse. Based on the role played by the LAD1 in the tumor immune microenvironment, it can be considered a potential biomarker for predicting the immunotherapy response to LUAD.
Collapse
Affiliation(s)
- Kun Yuan
- Department of Respiratory and Critical Care Medicine, Chengdu First People's Hospital, Chengdu, 610095, China
| | - Yiping Zhang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Yilin Yu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Yuanji Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Shuang Xian
- China Drug Development and Medical Affairs Center, Eli Lilly and Company, Shanghai, 20040, China.
| |
Collapse
|
33
|
Wang X, Qiao Z, Aramini B, Lin D, Li X, Fan J. Potential biomarkers for immunotherapy in non-small-cell lung cancer. Cancer Metastasis Rev 2023; 42:661-675. [PMID: 37121931 DOI: 10.1007/s10555-022-10074-y] [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: 09/07/2022] [Accepted: 12/09/2022] [Indexed: 05/02/2023]
Abstract
For individuals with advanced or metastatic non-small cell lung cancer (NSCLC), the primary treatment is platinum-based doublet chemotherapy. Immune checkpoint inhibitors (ICIs), primarily PD-1/PD-L1 and CTLA-4, have been found to be effective in patients with NSCLC who have no EGFR/ALK mutations. Furthermore, ICIs are considered a standard therapy. The quantity of fresh immunogenic antigens discovered by cytotoxic T cells was measured by PD-L1 expression and tumor mutational burden (TMB), which were the first biomarkers assessed in clinical trials. However, immunotherapy did not have response efficacy markers similar to targeted therapy, highlighting the significance of newly developed biomarkers. This investigation aims to review the research on immunotherapy for NSCLC, focusing primarily on the impact of biomarkers on efficacy prediction to determine whether biomarkers may be utilized to evaluate the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Xing Wang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai, China
| | - Ziyun Qiao
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai, China
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine-DIMES of the Alma Mater Studiorum, G.B. Morgagni-L. Pierantoni Hospital, University of Bologna, Forlì, Italy
| | - Dong Lin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai, China
| | - Xiaolong Li
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai, China
| | - Jiang Fan
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai, China.
| |
Collapse
|
34
|
Terrazas-Armendáriz LD, Alvizo-Báez CA, Luna-Cruz IE, Hernández-González BA, Uscanga-Palomeque AC, Ruiz-Robles MA, Pérez Tijerina EG, Rodríguez-Padilla C, Tamez-Guerra R, Alcocer-González JM. Systemic Delivery of Magnetogene Nanoparticle Vector for Gene Expression in Hypoxic Tumors. Pharmaceutics 2023; 15:2232. [PMID: 37765201 PMCID: PMC10536535 DOI: 10.3390/pharmaceutics15092232] [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: 08/01/2023] [Revised: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer is a disease that causes millions of deaths per year worldwide because conventional treatments have disadvantages such as unspecific tumor selectivity and unwanted toxicity. Most human solid tumors present hypoxic microenvironments and this promotes multidrug resistance. In this study, we present "Magnetogene nanoparticle vector" which takes advantage of the hypoxic microenvironment of solid tumors to increase selective gene expression in tumor cells and reduce unwanted toxicity in healthy cells; this vector was guided by a magnet to the tumor tissue. Magnetic nanoparticles (MNPs), chitosan (CS), and the pHRE-Luc plasmid with a hypoxia-inducible promoter were used to synthesize the vector called "Magnetogene nanoparticles" by ionic gelation. The hypoxic functionality of Magnetogene vector nanoparticles was confirmed in the B16F10 cell line by measuring the expression of the luciferase reporter gene under hypoxic and normoxic conditions. Also, the efficiency of the Magnetogene vector was confirmed in vivo. Magnetogene was administered by intravenous injection (IV) in the tail vein and directed through an external magnetic field at the site of tumor growth in C57Bl/6 mice. A Magnetogene vector with a size of 50 to 70 nm was directed and retained at the tumor area and gene expression was higher at the tumor site than in the others tissues, confirming the selectivity of this vector towards hypoxic tumor areas. This nanosystem, that we called the "Magnetogene vector" for systemic delivery and specific gene expression in hypoxic tumors controlled by an external magnetic designed to target hypoxic regions of tumors, can be used for cancer-specific gene therapies.
Collapse
Affiliation(s)
- Luis Daniel Terrazas-Armendáriz
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Cynthia Aracely Alvizo-Báez
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Itza Eloisa Luna-Cruz
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Becky Annette Hernández-González
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Ashanti Concepción Uscanga-Palomeque
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Mitchel Abraham Ruiz-Robles
- Centro de Investigación en Ciencias Fisico Matematicas, Facultad de Ciencias Físico Matematicas, Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de los Garza 66451, NL, Mexico; (M.A.R.-R.); (E.G.P.T.)
| | - Eduardo Gerardo Pérez Tijerina
- Centro de Investigación en Ciencias Fisico Matematicas, Facultad de Ciencias Físico Matematicas, Universidad Autónoma de Nuevo León, Ciudad Universitaria, San Nicolás de los Garza 66451, NL, Mexico; (M.A.R.-R.); (E.G.P.T.)
| | - Cristina Rodríguez-Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Reyes Tamez-Guerra
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| | - Juan Manuel Alcocer-González
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66450, NL, Mexico; (L.D.T.-A.); (C.A.A.-B.); (I.E.L.-C.); (B.A.H.-G.); (A.C.U.-P.); (C.R.-P.); (R.T.-G.)
| |
Collapse
|
35
|
Montaño-Samaniego M, Sánchez-Cedillo J, Lucas-González A, Bravo-Estupiñan DM, Alarcón-Hernández E, Rivera-Gutiérrez S, Balderas-López JA, Ibáñez-Hernández M. Targeted Expression to Liver of an antimiR-33 Sponge as a Gene Therapy Strategy against Hypercholesterolemia: In Vitro Study. Curr Issues Mol Biol 2023; 45:7043-7057. [PMID: 37754229 PMCID: PMC10527677 DOI: 10.3390/cimb45090445] [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: 07/11/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 09/28/2023] Open
Abstract
Atherosclerosis is the leading cause of cardiovascular diseases in Mexico and worldwide. The membrane transporters ABCA1 and ABCG1 are involved in the reverse transport of cholesterol and stimulate the HDL synthesis in hepatocytes, therefore the deficiency of these transporters promotes the acceleration of atherosclerosis. MicroRNA-33 (miR-33) plays an important role in lipid metabolism and exerts a negative regulation on the transporters ABCA1 and ABCG1. It is known that by inhibiting the function of miR-33 with antisense RNA, HDL levels increase and atherogenic risk decreases. Therefore, in this work, a genetic construct, pPEPCK-antimiR-33-IRES2-EGFP, containing a specific antimiR-33 sponge with two binding sites for miR-33 governed under the PEPCK promoter was designed, constructed, and characterized, the identity of which was confirmed by enzymatic restriction, PCR, and sequencing. Hep G2 and Hek 293 FT cell lines, as well as a mouse hepatocyte primary cell culture were transfected with this plasmid construction showing expression specificity of the PEPCK promoter in hepatic cells. An analysis of the relative expression of miR-33 target messengers showed that the antimiR-33 sponge indirectly induces the expression of its target messengers (ABCA1 and ABCG1). This strategy could open new specific therapeutic options for hypercholesterolemia and atherosclerosis, by blocking the miR-33 specifically in hepatocytes.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (M.M.-S.); (J.S.-C.); (A.L.-G.); (D.M.B.-E.)
- Laboratorio de Técnicas Fototérmicas, Departamento de Ciencias Básicas, Unidad Politécnica Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Mexico City 07340, Mexico;
| | - Jorge Sánchez-Cedillo
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (M.M.-S.); (J.S.-C.); (A.L.-G.); (D.M.B.-E.)
| | - Amellalli Lucas-González
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (M.M.-S.); (J.S.-C.); (A.L.-G.); (D.M.B.-E.)
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (M.M.-S.); (J.S.-C.); (A.L.-G.); (D.M.B.-E.)
- Laboratorio de Quimiosensibilidad Tumoral, Facultad de Microbiología, Universidad de Costa Rica, San Jose 11501-2060, Costa Rica
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Sandra Rivera-Gutiérrez
- Laboratorio de Microbiología Molecular, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - José Abraham Balderas-López
- Laboratorio de Técnicas Fototérmicas, Departamento de Ciencias Básicas, Unidad Politécnica Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, Mexico City 07340, Mexico;
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (M.M.-S.); (J.S.-C.); (A.L.-G.); (D.M.B.-E.)
| |
Collapse
|
36
|
Irianto T, Gaipl US, Rückert M. Immune modulation during anti-cancer radio(immuno)therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:239-277. [PMID: 38225105 DOI: 10.1016/bs.ircmb.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Cancer can affect all human organs and tissues and ranks as a prominent cause of death as well as an obstruction to increasing life expectancy. A notable breakthrough in oncology has been the inclusion of the immune system in fighting cancer, potentially prolonging life and providing long-term benefits. The concept of "immunotherapy" has been discussed from the 19th and early 20th centuries by Wilhelm Busch, William B. Coley and Paul Ehrlich. This involves distinct approaches, including vaccines, non-specific cytokines and adoptive cell therapies. However, despite the advances made in recent years, questions on how to select the best therapeutic options or how to select the best combinations to improve clinical outcomes are still relevant for scientists and clinicians. More than half of cancer patients receive radiotherapy (RT) as part of their treatment. With the advances in RT and immunotherapy approaches, it is reasonable to consider how to enhance immunotherapy with radiation and vice versa, and to investigate whether combinations of these therapies would be beneficial. In this chapter, we will discuss how the immune system responds to cancer cells and different cancer therapies with a focus on combination of RT and immunotherapy (radioimmunotherapy, RIT).
Collapse
Affiliation(s)
- Teresa Irianto
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Michael Rückert
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
37
|
MacKenzie TMG, Cisneros R, Maynard RD, Snyder MP. Reverse-ChIP Techniques for Identifying Locus-Specific Proteomes: A Key Tool in Unlocking the Cancer Regulome. Cells 2023; 12:1860. [PMID: 37508524 PMCID: PMC10377898 DOI: 10.3390/cells12141860] [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: 05/29/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
A phenotypic hallmark of cancer is aberrant transcriptional regulation. Transcriptional regulation is controlled by a complicated array of molecular factors, including the presence of transcription factors, the deposition of histone post-translational modifications, and long-range DNA interactions. Determining the molecular identity and function of these various factors is necessary to understand specific aspects of cancer biology and reveal potential therapeutic targets. Regulation of the genome by specific factors is typically studied using chromatin immunoprecipitation followed by sequencing (ChIP-Seq) that identifies genome-wide binding interactions through the use of factor-specific antibodies. A long-standing goal in many laboratories has been the development of a 'reverse-ChIP' approach to identify unknown binding partners at loci of interest. A variety of strategies have been employed to enable the selective biochemical purification of sequence-defined chromatin regions, including single-copy loci, and the subsequent analytical detection of associated proteins. This review covers mass spectrometry techniques that enable quantitative proteomics before providing a survey of approaches toward the development of strategies for the purification of sequence-specific chromatin as a 'reverse-ChIP' technique. A fully realized reverse-ChIP technique holds great potential for identifying cancer-specific targets and the development of personalized therapeutic regimens.
Collapse
Affiliation(s)
| | - Rocío Cisneros
- Sarafan ChEM-H/IMA Postbaccalaureate Fellow in Target Discovery, Stanford University, Stanford, CA 94305, USA
| | - Rajan D Maynard
- Genetics Department, Stanford University, Stanford, CA 94305, USA
| | - Michael P Snyder
- Genetics Department, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
38
|
Hu Y, Zhang H, Guo Z, Zhou J, Zhang W, Gong M, Wu J. CKM and TERT dual promoters drive CRISPR-dCas9 to specifically inhibit the malignant behavior of osteosarcoma cells. Cell Mol Biol Lett 2023; 28:52. [PMID: 37415116 DOI: 10.1186/s11658-023-00464-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Improvements in treatment and chemotherapy have increased the survival rate of osteosarcoma, but overall efficacy remains low, highlighting the need for new gene therapy methods. Clustered regularly interspaced short palindromic repeats-deactivated Cas9 (CRISPR-dCas9) technology offers a promising strategy, but targeting osteosarcoma cells precisely is a challenge. We designed a system to achieve specific expression of CRISPR-dCas9-KRAB in osteosarcoma cells by using the creatine kinase muscle (CKM) promoter to drive dCas9-KRAB and the telomerase reverse transcriptase (TERT) promoter to drive single guide (sg)RNA expression. We inhibited the MDM2 proto-oncogene using this system in vitro, which efficiently inhibited the malignant behavior of osteosarcoma cells and induced apoptosis without affecting normal cells. In vivo experiments demonstrated that this system effectively inhibited the growth of subcutaneously transplanted tumors in nude mice. These findings provide a new method for precise identification and intervention of osteosarcoma with significant implications for the development of gene therapy methods for other cancers. Future research should focus on optimizing this system for clinical translation.
Collapse
Affiliation(s)
- Yawei Hu
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Hao Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Zengfeng Guo
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Jianhua Zhou
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Wang Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Ming Gong
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Jiawen Wu
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China.
| |
Collapse
|
39
|
Heo J, Liang JD, Kim CW, Woo HY, Shih IL, Su TH, Lin ZZ, Yoo SY, Chang S, Urata Y, Chen PJ. Safety and dose escalation of the targeted oncolytic adenovirus OBP-301 for refractory advanced liver cancer: Phase I clinical trial. Mol Ther 2023; 31:2077-2088. [PMID: 37060176 PMCID: PMC10362399 DOI: 10.1016/j.ymthe.2023.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/29/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023] Open
Abstract
OBP-301 is an oncolytic adenovirus modified to replicate within cancer cells and lyse them. This open-label, non-comparative, phase I dose-escalation trial aimed to assess its safety and optimal dosage in 20 patients with advanced hepatocellular carcinoma. Good tolerance was shown with a maximum tolerated dose of 6 × 1012 viral particles. The most common treatment-emergent adverse events were influenza-like illness, pyrexia, fatigue, decreased platelet count, abdominal distension, and anemia. Cohorts 4 and 5 had approximately 50% higher levels of CD8+ T cells in the peripheral blood after injection. The best target response occurred in 14 patients, 4 of whom had progressive disease. Multiple intratumoral injections of OBP-301 were well tolerated in patients with advanced hepatocellular carcinoma. The stable disease rate for the injected tumors was greater than the overall response rate, even with no obvious tumor response. OBP-301 might have a greater impact on local response as histological examination revealed that the presence of OBP-301 was consistent with the necrotic area at the injection site. Increased infiltration of CD8+ T cells and <1% PD-L1 expression were observed in tumors after injection. Improved antitumor efficacy might be achieved in future studies via viral injection with volume adjustment and in combination with other immuno-therapeutics.
Collapse
Affiliation(s)
- Jeong Heo
- Department of Internal Medicine, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| | - Ja-Der Liang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chang Won Kim
- Department of Radiology, College of Medicine, Pusan National University, Busan, Republic of Korea
| | - Hyun Young Woo
- Department of Internal Medicine, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - I-Lun Shih
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Tung-Hung Su
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Zhong-Zhe Lin
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan, Republic of Korea
| | | | | | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
40
|
Lundstrom K. Viral vectors engineered for gene therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:1-41. [PMID: 37541721 DOI: 10.1016/bs.ircmb.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
Gene therapy has seen major progress in recent years. Viral vectors have made a significant contribution through efficient engineering for improved delivery and safety. A large variety of indications such as cancer, cardiovascular, metabolic, hematological, neurological, muscular, ophthalmological, infectious diseases, and immunodeficiency have been targeted. Viral vectors based on adenoviruses, adeno-associated viruses, herpes simplex viruses, retroviruses including lentiviruses, alphaviruses, flaviviruses, measles viruses, rhabdoviruses, Newcastle disease virus, poxviruses, picornaviruses, reoviruses, and polyomaviruses have been used. Proof-of-concept has been demonstrated for different indications in animal models. Therapeutic efficacy has also been achieved in clinical trials. Several viral vector-based drugs have been approved for the treatment of cancer, and hematological, metabolic, and neurological diseases. Moreover, viral vector-based vaccines have been approved against COVID-19 and Ebola virus disease.
Collapse
|
41
|
He S, Hou Y, Hou L, Chen N, Yang X, Wang H, Han P, Fan Y, Zhao J, Zhang J, Geng J. Targeted RASSF1A expression inhibits proliferation of HER2‑positive breast cancer cells in vitro. Exp Ther Med 2023; 25:245. [PMID: 37153885 PMCID: PMC10160914 DOI: 10.3892/etm.2023.11944] [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: 11/16/2022] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Human epidermal growth factor receptor 2-positive (HER2+) breast cancer, which accounts for 15-20% of all breast cancer, is associated with tumor recurrence and poor prognosis. RAS association domain family protein 1 subtype A (RASSF1A) is a tumor suppressor that is silenced in a variety of human cancers. The present study aimed to investigate the role of RASSF1A in HER2+ breast cancer and the therapeutic potential of RASSF1A-based targeted gene therapy for this malignancy. RASSF1A expression in human HER2+ breast cancer tissues and cell lines was evaluated by reverse transcription PCR and western blot analysis. The associations between tumorous RASSF1A level and tumor grade, TNM stage, tumor size, lymph node metastasis and five-year survival were examined. HER2+ and HER2-negative (HER2-) breast cancer cells were transfected with a lentiviral vector (LV-5HH-RASSF1A) that could express RASSF1A under the control of five copies of the hypoxia-responsive element (5HRE) and one copy of the HER2 promoter (HER2p). Cell proliferation was evaluated by the MTT and colony formation assays. It was found that tumorous RASSF1A level was negatively associated with tumor grade (P=0.014), TNM stage (P=0.0056), tumor size (P=0.014) and lymph node metastasis (P=0.029) and positively associated with five-year survival (P=0.038) in HER2+ breast cancer patients. Lentiviral transfection of HER2+ breast cancer cells resulted in increased RASSF1A expression and decreased cell proliferation, especially under hypoxic conditions. However, lentiviral transfection of HER2-breast cancer cells did not affect RASSF1A expression. In conclusion, these findings verified the clinical significance of RASSF1A as a tumor suppressor in HER2+ breast cancer and supported LV-5HH-RASSF1A as a potential targeted gene therapy for this malignancy.
Collapse
Affiliation(s)
- Sai He
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Yanni Hou
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Leina Hou
- Department of Anesthesiology, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Nan Chen
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaomin Yang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Huxia Wang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Pihua Han
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Yongguo Fan
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Zhao
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jingyuan Zhang
- Department of Breast Cancer, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Jie Geng
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
- Correspondence to: Dr Jie Geng, Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xiwu Road, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
42
|
Dutt Y, Pandey RP, Dutt M, Gupta A, Vibhuti A, Vidic J, Raj VS, Chang CM, Priyadarshini A. Therapeutic applications of nanobiotechnology. J Nanobiotechnology 2023; 21:148. [PMID: 37149615 PMCID: PMC10163736 DOI: 10.1186/s12951-023-01909-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023] Open
Abstract
Nanobiotechnology, as a novel and more specialized branch of science, has provided a number of nanostructures such as nanoparticles, by utilizing the methods, techniques, and protocols of other branches of science. Due to the unique features and physiobiological characteristics, these nanostructures or nanocarriers have provided vast methods and therapeutic techniques, against microbial infections and cancers and for tissue regeneration, tissue engineering, and immunotherapies, and for gene therapies, through drug delivery systems. However, reduced carrying capacity, abrupt and non-targeted delivery, and solubility of therapeutic agents, can affect the therapeutic applications of these biotechnological products. In this article, we explored and discussed the prominent nanobiotechnological methods and products such as nanocarriers, highlighted the features and challenges associated with these products, and attempted to conclude if available nanostructures offer any scope of improvement or enhancement. We aimed to identify and emphasize the nanobiotechnological methods and products, with greater prospect and capacity for therapeutic improvements and enhancements. We found that novel nanocarriers and nanostructures, such as nanocomposites, micelles, hydrogels, microneedles, and artificial cells, can address the associated challenges and inherited drawbacks, with help of conjugations, sustained and stimuli-responsive release, ligand binding, and targeted delivery. We recommend that nanobiotechnology, despite having few challenges and drawbacks, offers immense opportunities that can be harnessed in delivering quality therapeutics with precision and prediction. We also recommend that, by exploring the branched domains more rigorously, bottlenecks and obstacles can also be addressed and resolved in return.
Collapse
Affiliation(s)
- Yogesh Dutt
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India
| | - Ramendra Pati Pandey
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India.
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India.
| | - Mamta Dutt
- Mamta Dental Clinic, Opposite Sector 29, Main Badkhal Road, Faridabad, Haryana, 121002, India
| | - Archana Gupta
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India
| | - Arpana Vibhuti
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India
| | - Jasmina Vidic
- Université Paris-Saclay, Micalis Institute, INRAE, AgroParisTech, 78350, Jouy-en-Josas, France
| | - V Samuel Raj
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India
| | - Chung-Ming Chang
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, No.259, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33302, Taiwan (ROC).
| | - Anjali Priyadarshini
- Department of Microbiology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India.
- Department of Biotechnology, SRM University, 39, Rajiv Gandhi Education City, Post Office P.S. Rai, Sonepat, Haryana, 131029, India.
| |
Collapse
|
43
|
Wang Y, Zhou SK, Wang Y, Lu ZD, Zhang Y, Xu CF, Wang J. Engineering tumor-specific gene nanomedicine to recruit and activate T cells for enhanced immunotherapy. Nat Commun 2023; 14:1993. [PMID: 37031188 PMCID: PMC10082825 DOI: 10.1038/s41467-023-37656-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/27/2023] [Indexed: 04/10/2023] Open
Abstract
PD-1/PD-L1 blockade therapy that eliminates T-cell inhibition signals is successful, but poor benefits are often observed. Increasing T-cell infiltration and quantity of PD-1/PD-L1 inhibitors in tumor can improve efficacy but remains challenging. Here, we devise tumor-specific gene nanomedicines to mobilize tumor cells to secrete CXCL9 (T-cell chemokine) and anti-PD-L1 scFv (αPD-L1, PD-L1 blocking agent) for enhanced immunotherapy. The tyrosinase promoter-driven NPTyr-C9AP can specifically co-express CXCL9 and αPD-L1 in melanoma cells, thereby forming a CXCL9 gradient for T-cell recruitment and high intratumoral αPD-L1 concentration for enhancing T-cell activation. As a result, NPTyr-C9AP shows strong antimelanoma effects. Moreover, specific co-expression of CXCL9 and αPD-L1 in various tumor cells is achieved by replacing the tyrosinase promoter of NPTyr-C9AP with a survivin promoter, which increases T-cell infiltration and activation and therapeutic efficacy in multiple tumors in female mice. This study provides a strategy to maximize the immunotherapeutic outcome regardless of the heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Yue Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
| | - Shi-Kun Zhou
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
| | - Yan Wang
- School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Zi-Dong Lu
- School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Yue Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P.R. China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P.R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P.R. China.
| |
Collapse
|
44
|
Huang Z, Guo H, Lin L, Li S, Yang Y, Han Y, Huang W, Yang J. Application of oncolytic virus in tumor therapy. J Med Virol 2023; 95:e28729. [PMID: 37185868 DOI: 10.1002/jmv.28729] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023]
Abstract
Oncolytic viruses (OVs) can selectively kill tumor cells without affecting normal cells, as well as activate the innate and adaptive immune systems in patients. Thus, they have been considered as a promising measure for safe and effective cancer treatment. Recently, a few genetically engineered OVs have been developed to further improve the effect of tumor elimination by expressing specific immune regulatory factors and thus enhance the body's antitumor immunity. In addition, the combined therapies of OVs and other immunotherapies have been applied in clinical. Although there are many studies on this hot topic, a comprehensive review is missing on illustrating the mechanisms of tumor clearance by OVs and how to modify engineered OVs to further enhance their antitumor effects. In this study, we provided a review on the mechanisms of immune regulatory factors in OVs. In addition, we reviewed the combined therapies of OVs with other therapies including radiotherapy and CAR-T or TCR-T cell therapy. The review is useful in further generalize the usage of OV in cancer treatment.
Collapse
Affiliation(s)
- Zhijian Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hongen Guo
- Department of Dermatology, Dermatology Hospital of Fuzhou, Fujian, Fuzhou, China
| | - Lin Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Shixiong Li
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yong Yang
- Department of Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuanyuan Han
- Center of Tree Shrew Germplasm Resources, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Weiwei Huang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd, Beijing, China
- Academician Workstation, Changsha Medical University, Changsha, China
| |
Collapse
|
45
|
Lundstrom K. Application of DNA Replicons in Gene Therapy and Vaccine Development. Pharmaceutics 2023; 15:pharmaceutics15030947. [PMID: 36986808 PMCID: PMC10054396 DOI: 10.3390/pharmaceutics15030947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
DNA-based gene therapy and vaccine development has received plenty of attention lately. DNA replicons based on self-replicating RNA viruses such as alphaviruses and flaviviruses have been of particular interest due to the amplification of RNA transcripts leading to enhanced transgene expression in transfected host cells. Moreover, significantly reduced doses of DNA replicons compared to conventional DNA plasmids can elicit equivalent immune responses. DNA replicons have been evaluated in preclinical animal models for cancer immunotherapy and for vaccines against infectious diseases and various cancers. Strong immune responses and tumor regression have been obtained in rodent tumor models. Immunization with DNA replicons has provided robust immune responses and protection against challenges with pathogens and tumor cells. DNA replicon-based COVID-19 vaccines have shown positive results in preclinical animal models.
Collapse
|
46
|
Viral Vectors in Gene Therapy: Where Do We Stand in 2023? Viruses 2023; 15:v15030698. [PMID: 36992407 PMCID: PMC10059137 DOI: 10.3390/v15030698] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Viral vectors have been used for a broad spectrum of gene therapy for both acute and chronic diseases. In the context of cancer gene therapy, viral vectors expressing anti-tumor, toxic, suicide and immunostimulatory genes, such as cytokines and chemokines, have been applied. Oncolytic viruses, which specifically replicate in and kill tumor cells, have provided tumor eradication, and even cure of cancers in animal models. In a broader meaning, vaccine development against infectious diseases and various cancers has been considered as a type of gene therapy. Especially in the case of COVID-19 vaccines, adenovirus-based vaccines such as ChAdOx1 nCoV-19 and Ad26.COV2.S have demonstrated excellent safety and vaccine efficacy in clinical trials, leading to Emergency Use Authorization in many countries. Viral vectors have shown great promise in the treatment of chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, hemophilia, β-thalassemia, and sickle cell disease (SCD). Proof-of-concept has been established in preclinical studies in various animal models. Clinical gene therapy trials have confirmed good safety, tolerability, and therapeutic efficacy. Viral-based drugs have been approved for cancer, hematological, metabolic, neurological, and ophthalmological diseases as well as for vaccines. For example, the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, the oncolytic HSV T-VEC for melanoma, lentivirus-based treatment of ADA-SCID disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease have been approved for human use.
Collapse
|
47
|
Lores S, Gámez-Chiachio M, Cascallar M, Ramos-Nebot C, Hurtado P, Alijas S, López López R, Piñeiro R, Moreno-Bueno G, de la Fuente M. Effectiveness of a novel gene nanotherapy based on putrescine for cancer treatment. Biomater Sci 2023. [PMID: 36790445 DOI: 10.1039/d2bm01456d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Gene therapy has long been proposed for cancer treatment. However, the use of therapeutic nucleic acids presents several limitations such as enzymatic degradation, rapid clearance, and poor cellular uptake and efficiency. In this work we propose the use of putrescine, a precursor for higher polyamine biosynthesis for the preparation of cationic nanosystems for cancer gene therapy. We have formulated and characterized putrescine-sphingomyelin nanosystems (PSN) and studied their endocytic pathway and intracellular trafficking in cancer cells. After loading a plasmid DNA (pDNA) encoding the apoptotic Fas Ligand (FasL), we proved their therapeutic activity by measuring the cell death rate after treatment of MDA-MB-231 cells. We have also used xenografted zebrafish embryos as a first in vivo approach to demonstrate the efficacy of the proposed PSN-pDNA formulation in a more complex model. Finally, intratumoral and intraperitoneal administration to mice-bearing MDA-MB-231 xenografts resulted in a significant decrease in tumour cell growth, highlighting the potential of the developed gene therapy nanoformulation for the treatment of triple negative breast cancer.
Collapse
Affiliation(s)
- Saínza Lores
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain
| | - Manuel Gámez-Chiachio
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - María Cascallar
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - Carmen Ramos-Nebot
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain
| | - Pablo Hurtado
- Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Rafael López López
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Roberto Piñeiro
- Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,Roche-CHUS Join Unit. Translational Medical Oncology Group (ONCOMET), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain.
| | - Gema Moreno-Bueno
- Translational Cancer Research Laboratory, Department of Biochemistry, Autonomous University of Madrid, School of Medicine, "Alberto Sols" Biomedical Research Institute CSIC-UAM, IdiPaz, Arturo Duperier 4, 28029, Madrid, Spain. .,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,MD Anderson International Foundation, Gómez Hemans s/n, 28033 Madrid, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, Santiago de Compostela, 15706, A Coruña, Spain. .,Universidade de Santiago de Compostela (USC), Praza do Obradoiro, s/n, Santiago de Compostela, 15782, A Coruña, Spain.,Biomedical Cancer Research Network (CIBERONC), 28029 Madrid, Spain.,DIVERSA Technologies SL, Edificio Emprendia, Universidade de Santiago de Compostela, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
48
|
The Dilemma of HSV-1 Oncolytic Virus Delivery: The Method Choice and Hurdles. Int J Mol Sci 2023; 24:ijms24043681. [PMID: 36835091 PMCID: PMC9962028 DOI: 10.3390/ijms24043681] [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: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as effective gene therapy and immunotherapy drugs. As an important gene delivery platform, the integration of exogenous genes into OVs has become a novel path for the advancement of OV therapy, while the herpes simplex virus type 1 (HSV-1) is the most commonly used. However, the current mode of administration of HSV-1 oncolytic virus is mainly based on the tumor in situ injection, which limits the application of such OV drugs to a certain extent. Intravenous administration offers a solution to the systemic distribution of OV drugs but is ambiguous in terms of efficacy and safety. The main reason is the synergistic role of innate and adaptive immunity of the immune system in the response against the HSV-1 oncolytic virus, which is rapidly cleared by the body's immune system before it reaches the tumor, a process that is accompanied by side effects. This article reviews different administration methods of HSV-1 oncolytic virus in the process of tumor treatment, especially the research progress in intravenous administration. It also discusses immune constraints and solutions of intravenous administration with the intent to provide new insights into HSV-1 delivery for OV therapy.
Collapse
|
49
|
Аpplication of massive parallel reporter analysis in biotechnology and medicine. КЛИНИЧЕСКАЯ ПРАКТИКА 2023. [DOI: 10.17816/clinpract115063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The development and functioning of an organism relies on tissue-specific gene programs. Genome regulatory elements play a key role in the regulation of such programs, and disruptions in their function can lead to the development of various pathologies, including cancers, malformations and autoimmune diseases. The emergence of high-throughput genomic studies has led to massively parallel reporter analysis (MPRA) methods, which allow the functional verification and identification of regulatory elements on a genome-wide scale. Initially MPRA was used as a tool to investigate fundamental aspects of epigenetics, but the approach also has great potential for clinical and practical biotechnology. Currently, MPRA is used for validation of clinically significant mutations, identification of tissue-specific regulatory elements, search for the most promising loci for transgene integration, and is an indispensable tool for creating highly efficient expression systems, the range of application of which extends from approaches for protein development and design of next-generation therapeutic antibody superproducers to gene therapy. In this review, the main principles and areas of practical application of high-throughput reporter assays will be discussed.
Collapse
|
50
|
Lundstrom K. Gene Therapy Cargoes Based on Viral Vector Delivery. Curr Gene Ther 2023; 23:111-134. [PMID: 36154608 DOI: 10.2174/1566523222666220921112753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.
Collapse
|